1
|
Jardon KM, Umanets A, Gijbels A, Trouwborst I, Hul GB, Siebelink E, Vliex LM, Bastings JJ, Argamasilla R, Chenal E, Venema K, Afman LA, Goossens GH, Blaak EE. Distinct gut microbiota and metabolome features of tissue-specific insulin resistance in overweight and obesity. Gut Microbes 2025; 17:2501185. [PMID: 40336254 PMCID: PMC12064058 DOI: 10.1080/19490976.2025.2501185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 02/24/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Insulin resistance (IR) is an early marker of cardiometabolic deterioration which may develop heterogeneously in key metabolic organs, including the liver (LIR) and skeletal muscle (MIR). This tissue-specific IR is characterized by distinct metabolic signatures, but the role of the gut microbiota in its etiology remains unclear. Here, we profiled the gut microbiota, its metabolites and the plasma metabolome in individuals with either a LIR or MIR phenotype (n = 233). We observed distinct microbial community structures LIR and MIR, and higher short-chain fatty acid (SCFA) producing bacteria, fecal SCFAs and branched-chain fatty acids and a higher postprandial plasma glucagon-like-peptide-1 response in LIR. In addition, we found variations in metabolome profiles and phenotype-specific associations between microbial taxa and functional metabolite groups. Overall, our study highlights association between gut microbiota and its metabolites composition with IR heterogeneity that can be targeted in precision-based strategies to improve cardiometabolic health. Clinicaltrials.gov registration: NCT03708419.
Collapse
Affiliation(s)
- Kelly M. Jardon
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Alexander Umanets
- Centre for Healthy Eating & Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
- Chair Group Youth Food and Health, Faculty of Science and Engineering, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Anouk Gijbels
- TiFN, Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Inez Trouwborst
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Gabby B. Hul
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Els Siebelink
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Lars M.M. Vliex
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jacco J.A.J. Bastings
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | | | - Koen Venema
- Centre for Healthy Eating & Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Lydia A. Afman
- TiFN, Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ellen E. Blaak
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
2
|
Wanders L, Gijbels A, Hul GB, Feskens EJM, Afman LA, Blaak EE, Hopman MTE, Goossens GH, Thijssen DHJ. Impact of a 12-week personalized dietary intervention on vascular function and cardiovascular risk factors. Diabetes Obes Metab 2025; 27:2601-2612. [PMID: 40013435 PMCID: PMC11965023 DOI: 10.1111/dom.16261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/28/2025]
Abstract
AIMS Individuals with liver insulin-resistant (LIR) or muscle insulin-resistant (MIR) phenotypes may respond differently to dietary interventions. Given the interaction between insulin resistance and cardiovascular risk, this sub-analysis of the PERSON study examined whether a personalized diet according to MIR or LIR phenotypes improves vascular function and cardiovascular disease risk factors. MATERIALS AND METHODS We randomized 119 participants to a 12-week low-fat, high-protein, high-fibre diet (LFHP; may be optimal for LIR) or Mediterranean diet (high in monounsaturated fat, HMUFA; may be optimal for MIR). Randomization linked the insulin-resistant (IR) phenotype to the proposed optimal diet, leading to PhenoDiet A (MIR-HMUFA and LIR-LFHP) and PhenoDiet B (MIR-LFHP and LIR-HMUFA). Before and after the intervention, vascular function (carotid artery reactivity) and cardiovascular risk factors (blood pressure, total cholesterol, HDL-cholesterol and Framingham risk score) were examined. A 7-point oral glucose tolerance test was performed to determine insulin resistance (Matsuda index and HOMA-IR) and disposition index. RESULTS Following drop-out (n = 18), 101 participants finished the intervention (54 women, 61 ± 7 years, 27.6 [26.4;30.0] kg/m2), with n = 80 available for the primary outcome of vascular function. Overall, the dietary interventions significantly decreased blood pressure, total cholesterol, HDL-cholesterol and the Framingham risk score (all p < 0.05), while vascular function was not affected (p = 0.485). Insulin resistance (p ≤ 0.001), but not disposition index (p = 0.362), was significantly improved after intervention. The Matsuda index (p = 0.078) tended to increase more and total cholesterol (p = 0.052) tended to decrease more in PhenoDiet group B than A, but other changes in outcome parameters were not significantly different between PhenoDiet groups. The LFHP diet resulted in more pronounced improvements in cholesterol, diastolic blood pressure (DBP) and insulin resistance compared with the HMUFA diet (all p < 0.05). CONCLUSION A 12-week diet improves metabolic and cardiovascular outcomes, but not vascular function in insulin-resistant adults with overweight or obesity. Whilst the LFHP diet resulted in greater improvements in cardiometabolic risk markers than the HMUFA diet, we found no significant differences between the PhenoDiet groups.
Collapse
Affiliation(s)
- Lisa Wanders
- Radboud Institute for Health Sciences, Department of PhysiologyRadboud university medical centerNijmegenThe Netherlands
- TiFNWageningenThe Netherlands
| | - Anouk Gijbels
- TiFNWageningenThe Netherlands
- Division of Human Nutrition and HealthWageningen UniversityWageningenThe Netherlands
| | - Gabby B. Hul
- Department of Human BiologyInstitute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Edith J. M. Feskens
- Division of Human Nutrition and HealthWageningen UniversityWageningenThe Netherlands
| | - Lydia A. Afman
- Division of Human Nutrition and HealthWageningen UniversityWageningenThe Netherlands
| | - Ellen E. Blaak
- Department of Human BiologyInstitute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Maria T. E. Hopman
- Radboud Institute for Health Sciences, Department of PhysiologyRadboud university medical centerNijmegenThe Netherlands
| | - Gijs H. Goossens
- Department of Human BiologyInstitute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Dick H. J. Thijssen
- Radboud Institute for Health Sciences, Department of PhysiologyRadboud university medical centerNijmegenThe Netherlands
- Research Institute for Sport and Exercise SciencesLiverpool John Moores UniversityLiverpoolUK
| |
Collapse
|
3
|
Doulamis IP, Pan B, Tzani A, Plutzky J, Wong GW, Kilic A, Wolf RM. Transcriptomic signatures of subcutaneous adipose tissue in patients with diabetes and coronary artery disease: a pilot study. Front Cardiovasc Med 2025; 12:1524605. [PMID: 39974596 PMCID: PMC11836038 DOI: 10.3389/fcvm.2025.1524605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/09/2025] [Indexed: 02/21/2025] Open
Abstract
The exact role of subcutaneous adipose tissue in the interplay between type 2 diabetes (T2D) and coronary artery disease (CAD) is yet to be determined. A prospective cohort study of adult patients with and without T2D undergoing CABG was performed. Subcutaneous adipose tissue was collected during the procedure and RNA seq analysis was performed. A total of 741 differentially expressed genes (DEGs) were identified (332 up- and 409 down-regulated in the T2D group). Our results demonstrated that pathways related to apoptosis and immune response were significantly dysregulated in the adipose tissue of T2D subjects. The main molecular pathways involved were CXCR, NOTCH, STAT, NFKB1 and FGFR pathways, which have a well-documented role in diabetes and CAD. SPI1 and MTF1 were two novel upstream transcription factors identified which have been suggested to be involved in the inflammatory cascade and insulin regulation in diabetes. Three miRNAs were differentially expressed between the two groups (miR-27a, miR-335 and miR-146). These preliminary results provide fertile ground for further research of potential targets for patients with diabetes and coronary artery disease.
Collapse
Affiliation(s)
- Ilias P. Doulamis
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Bernard Pan
- Department of Pediatrics, Division of Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Aspasia Tzani
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jorge Plutzky
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - G. William Wong
- Department of Physiology, Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ahmet Kilic
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Risa M. Wolf
- Department of Pediatrics, Division of Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Li G, Meex RCR, Goossens GH. The role of tissue oxygenation in obesity-related cardiometabolic complications. Rev Endocr Metab Disord 2025; 26:19-30. [PMID: 39298040 PMCID: PMC11790814 DOI: 10.1007/s11154-024-09910-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 09/21/2024]
Abstract
Obesity is a complex, multifactorial, chronic disease that acts as a gateway to a range of other diseases. Evidence from recent studies suggests that changes in oxygen availability in the microenvironment of metabolic organs may exert an important role in the development of obesity-related cardiometabolic complications. In this review, we will first discuss results from observational and controlled laboratory studies that examined the relationship between reduced oxygen availability and obesity-related metabolic derangements. Next, the effects of alterations in oxygen partial pressure (pO2) in the adipose tissue, skeletal muscle and the liver microenvironment on physiological processes in these key metabolic organs will be addressed, and how this might relate to cardiometabolic complications. Since many obesity-related chronic diseases, including type 2 diabetes mellitus, cardiovascular diseases, chronic kidney disease, chronic obstructive pulmonary disease and obstructive sleep apnea, are characterized by changes in pO2 in the tissue microenvironment, a better understanding of the metabolic impact of altered tissue oxygenation can provide valuable insights into the complex interplay between environmental and biological factors involved in the pathophysiology of metabolic impairments. This may ultimately contribute to the development of novel strategies to prevent and treat obesity-related cardiometabolic diseases.
Collapse
Affiliation(s)
- Geng Li
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands
| | - Ruth C R Meex
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands
| | - Gijs H Goossens
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands.
| |
Collapse
|
5
|
Zadgaonkar U. The Interplay Between Adipokines and Body Composition in Obesity and Metabolic Diseases. Cureus 2025; 17:e78050. [PMID: 40013194 PMCID: PMC11863173 DOI: 10.7759/cureus.78050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
The worldwide health system faces challenges from obesity and related metabolic disorders because they exhibit both rising rates of occurrence and intricate pathophysiological mechanisms. This work examines how adipokines interact with body composition during obesity to control important metabolic functions. Bioactive molecules produced by adipose tissue function as adipokines which regulate essential biological pathways that control inflammation response and insulin sensitivity alongside energy balance management and immune system operation. The disruption of adipokine secretion and function leads directly to metabolic disorders which include insulin resistance and persistent inflammation characteristic of obesity-related conditions. This article investigates the therapeutic possibilities of adipokine pathway manipulation through new pharmacological approaches and lifestyle changes alongside personalized medicine developments. Researchers analyze adipokines as important biomarkers for patient disease classification and their application in creating individualized treatment plans. The review highlights existing research deficiencies and obstacles that stand in the way of applying adipokine discoveries to clinical settings. This article integrates existing research to show how adipokine regulation helps prevent obesity-related metabolic issues and suggests directions for future studies to enhance treatment results.
Collapse
Affiliation(s)
- Umesh Zadgaonkar
- Department of Nutrition and Dietetics, Global University, Itanagar, IND
| |
Collapse
|
6
|
Vieira DFM, Torres CVDS, Secaf ADF, Palma MDM, Gouvea GDL, Jorge E, Reis RB, Muglia V. Comparison of Morphological and Functional MRI Assessments of Periprostatic Fat for Predicting Prostate Cancer Aggressiveness. Int Braz J Urol 2025; 51:e20240318. [PMID: 39556849 PMCID: PMC11869916 DOI: 10.1590/s1677-5538.ibju.2024.0318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/04/2024] [Indexed: 11/20/2024] Open
Abstract
PURPOSE The objective of this study was to evaluate whether morphological (linear measurements) and functional (ADC value) assessments of periprostatic fat can predict the aggressiveness of prostate cancer (PCa) over a 5-year follow-up period. MATERIAL AND METHODS This retrospective study included patients with histologically proven PCa who underwent 3.0T MRI between July 2016 and June 2018. Clinical and demographic data collected included PSA, PSA density (dPSA), ISUP grade, clinical and pathological staging, and treatment details. MRI-derived parameters were assessed by an experienced radiologist, who measured subcutaneous and periprostatic fat thickness, and calculated ADC values from ROI plots in periprostatic fat. Clinical and MRI parameters were analyzed for associations with biochemical recurrence, systemic metastasis, and PCa-related mortality. RESULTS After applying exclusion criteria, 109 patients were included. Using the Cox model, dPSA (p<0.01), systemic disease at diagnosis (p<0.01), and mean ADC (p<0.02) were independent predictors of overall survival (OS). For progression-free survival (PFS), only dPSA (p<0.01) and systemic disease at diagnosis (p<0.01) were significant predictors. In the Poisson Model for systemic recurrence risk, dPSA had a relative risk (RR) of 1.04 (95%CI 1.0-1.07, p=0.03), systemic disease at diagnosis had an RR of 63.3 (95%CI 3.7-86.4, p<0.01), and average ADC had an RR of 3.42 (95%CI 1.52-7.69, p<0.01). CONCLUSIONS The ADC value of periprostatic fat may serve as an additional tool for PCa risk stratification, correlating with poorer outcomes such as systemic recurrence and overall survival. If validated by external, prospective, multicenter studies, these findings could impact future therapeutic decisions.
Collapse
Affiliation(s)
- David Freire Maia Vieira
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoDepartamento de ImagemRibeirão PretoSPBrasilDepartamento de Imagem, Oncologia e Hematologia - Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo - USP, Ribeirão Preto, SP, Brasil
| | - Cecília Vidal de Souza Torres
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoDepartamento de ImagemRibeirão PretoSPBrasilDepartamento de Imagem, Oncologia e Hematologia - Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo - USP, Ribeirão Preto, SP, Brasil
| | - André de Freitas Secaf
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoDepartamento de ImagemRibeirão PretoSPBrasilDepartamento de Imagem, Oncologia e Hematologia - Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo - USP, Ribeirão Preto, SP, Brasil
| | - Matheus de Moraes Palma
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoDepartamento de ImagemRibeirão PretoSPBrasilDepartamento de Imagem, Oncologia e Hematologia - Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo - USP, Ribeirão Preto, SP, Brasil
| | - Gabriel de Lion Gouvea
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoDepartamento de ImagemRibeirão PretoSPBrasilDepartamento de Imagem, Oncologia e Hematologia - Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo - USP, Ribeirão Preto, SP, Brasil
| | - Elias Jorge
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoDepartamento de ImagemRibeirão PretoSPBrasilDepartamento de Imagem, Oncologia e Hematologia - Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo - USP, Ribeirão Preto, SP, Brasil
| | - Rodolfo Borges Reis
- Universidade de São PauloFaculdade de Medicina de Ribeirao PretoDepartamento de Cirurgia e AnatomiaRibeirão PretoSPBrasilDepartamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirao Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Valdair Muglia
- Universidade de São PauloClínica Médica Faculdade de Medicina de Ribeirão PretoRibeirão PretoSPBrasilClínica Médica Faculdade de Medicina de Ribeirão Preto - Universidade de São Paulo - USP, Ribeirão Preto, SP, Brasil
| |
Collapse
|
7
|
Ziadlou R, Pandian GN, Hafner J, Akdis CA, Stingl G, Maverakis E, Brüggen M. Subcutaneous adipose tissue: Implications in dermatological diseases and beyond. Allergy 2024; 79:3310-3325. [PMID: 39206504 PMCID: PMC11657049 DOI: 10.1111/all.16295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Subcutaneous adipose tissue (SAT) is the deepest component of the three-layered cutaneous integument. While mesenteric adipose tissue-based immune processes have gained recognition in the context of the metabolic syndrome, SAT has been traditionally considered primarily for energy storage, with less attention to its immune functions. SAT harbors a reservoir of immune and stromal cells that significantly impact metabolic and immunologic processes not only in the skin, but even on a systemic level. These processes include wound healing, cutaneous and systemic infections, immunometabolic, and autoimmune diseases, inflammatory skin diseases, as well as neoplastic conditions. A better understanding of SAT immune functions in different processes, could open avenues for novel therapeutic interventions. Targeting SAT may not only address SAT-specific diseases but also offer potential treatments for cutaneous or even systemic conditions. This review aims to provide a comprehensive overview on SAT's structure and functions, highlight recent advancements in understanding its role in both homeostatic and pathological conditions within and beyond the skin, and discuss the main questions for future research in the field.
Collapse
Affiliation(s)
- Reihane Ziadlou
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
- Christine Kühne Center for Allergy Research and Education CK‐CAREDavosSwitzerland
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichZurichSwitzerland
| | - Ganesh N. Pandian
- Institute for Integrated Cell‐Material Science (WPI‐iCeMS)Kyoto UniversityKyotoJapan
| | - Jürg Hafner
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
| | - Cezmi A. Akdis
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
- Christine Kühne Center for Allergy Research and Education CK‐CAREDavosSwitzerland
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichZurichSwitzerland
| | - Georg Stingl
- Department of DermatologyMedical University of ViennaViennaAustria
| | | | - Marie‐Charlotte Brüggen
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
- Christine Kühne Center for Allergy Research and Education CK‐CAREDavosSwitzerland
| |
Collapse
|
8
|
Bandres-Meriz J, Sanz-Cuadrado M, Hurtado de Mendoza I, Majali-Martinez A, Honeder S, Cindrova-Davies T, Birner-Gruenberger R, Dalgaard L, Desoye G. MCM proteins are up-regulated in placentas of women with reduced insulin sensitivity. Biosci Rep 2024; 44:BSR20240430. [PMID: 39268985 PMCID: PMC11461181 DOI: 10.1042/bsr20240430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
In the first trimester of pregnancy the human placenta grows rapidly, making it sensitive to changes in the intrauterine environment. To test whether exposure to an environment in utero often associated with obesity modifies placental proteome and function, we performed untargeted proteomics (LC-MS/MS) in placentas from 19 women (gestational age 35-48 days, i.e. 5+0-6+6 weeks). Maternal clinical traits (body mass index, leptin, glucose, C-peptide and insulin sensitivity) and gestational age were recorded. DNA replication and cell cycle pathways were enriched in the proteome of placentas of women with low maternal insulin sensitivity. Driving these pathways were the minichromosome maintenance (MCM) proteins MCM2, MCM3, MCM4, MCM5, MCM6 and MCM7 (MCM-complex). These proteins are part of the pre-replicative complex and participate in DNA damage repair. Indeed, MCM6 and γH2AX (DNA-damage marker) protein levels correlated in first trimester placental tissue (r = 0.514, P<0.01). MCM6 and γH2AX co-localized to nuclei of villous cytotrophoblast cells, the proliferative cell type of the placenta, suggesting increased DNA damage in this cell type. To mimic key features of the intrauterine obesogenic environment, a first trimester trophoblast cell line, i.e., ACH-3P, was exposed to high insulin (10 nM) or low oxygen tension (2.5% O2). There was a significant correlation between MCM6 and γH2AX protein levels, but these were independent of insulin or oxygen exposure. These findings show that chronic exposure in utero to reduced maternal insulin sensitivity during early pregnancy induces changes in the early first trimester placental proteome. Pathways related to DNA replication, cell cycle and DNA damage repair appear especially sensitive to such an in utero environment.
Collapse
Affiliation(s)
- Julia Bandres-Meriz
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | | | | | - Alejandro Majali-Martinez
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
- Departamento de Medicina, Facultad de Ciencias Biomédicas y de la Salud. Universidad Europea de Madrid, Madrid, Spain
| | - Sophie Elisabeth Honeder
- Institute of Pathology, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, Vienna, Austria
| | - Tereza Cindrova-Davies
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, U.K
| | - Ruth Birner-Gruenberger
- Institute of Pathology, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, Vienna, Austria
| | | | - Gernot Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| |
Collapse
|
9
|
Mashayekhi M, Sheng Q, Bailin SS, Massier L, Zhong J, Shi M, Wanjalla CN, Wang TJ, Ikizler TA, Niswender KD, Gabriel CL, Palacios J, Turgeon-Jones R, Reynolds CF, Luther JM, Brown NJ, Das S, Dahlman I, Mosley JD, Koethe JR, Rydén M, Bachmann KN, Shah RV. The subcutaneous adipose transcriptome identifies a molecular signature of insulin resistance shared with visceral adipose. Obesity (Silver Spring) 2024; 32:1526-1540. [PMID: 38967296 PMCID: PMC11269023 DOI: 10.1002/oby.24064] [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: 02/09/2024] [Revised: 04/05/2024] [Accepted: 04/23/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE The objective of this study was to identify the transcriptional landscape of insulin resistance (IR) in subcutaneous adipose tissue (SAT) in humans across the spectrum of obesity. METHODS We used SAT RNA sequencing in 220 individuals with metabolic phenotyping. RESULTS We identified a 35-gene signature with high predictive accuracy for homeostatic model of IR that was expressed across a variety of non-immune cell populations. We observed primarily "protective" IR associations for adipocyte transcripts and "deleterious" associations for macrophage transcripts, as well as a high concordance between SAT and visceral adipose tissue (VAT). Multiple SAT genes exhibited dynamic expression 5 years after weight loss surgery and with insulin stimulation. Using available expression quantitative trait loci in SAT and/or VAT, we demonstrated similar genetic effect sizes of SAT and VAT on type 2 diabetes and BMI. CONCLUSIONS SAT is conventionally viewed as a metabolic buffer for lipid deposition during positive energy balance, whereas VAT is viewed as a dominant contributor to and prime mediator of IR and cardiometabolic disease risk. Our results implicate a dynamic transcriptional architecture of IR that resides in both immune and non-immune populations in SAT and is shared with VAT, nuancing the current VAT-centric concept of IR in humans.
Collapse
Affiliation(s)
- Mona Mashayekhi
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
| | - Quanhu Sheng
- Vanderbilt University Medical Center, Department of Biostatistics, Nashville, Tennessee, USA
| | - Samuel S. Bailin
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, USA
| | - Lucas Massier
- Karolinska Institutet, Department of Medicine, Huddinge (H7), Stockholm, Sweden
| | - Jiawei Zhong
- Karolinska Institutet, Department of Medicine, Huddinge (H7), Stockholm, Sweden
| | - Mingjian Shi
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Celestine N. Wanjalla
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, USA
| | - Thomas J. Wang
- UT Southwestern Medical Center, Department of Internal Medicine, Dallas, Texas, USA
| | - T. Alp Ikizler
- Vanderbilt University Medical Center, Department of Medicine, Division of Nephrology and Hypertension, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Kevin D. Niswender
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Curtis L. Gabriel
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Nashville, Tennessee, USA
| | - Julia Palacios
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
| | - Rachel Turgeon-Jones
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
| | - Cassandra F. Reynolds
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Department of Medicine, Cardiology Division, Nashville, Tennessee, USA
| | - James M. Luther
- Vanderbilt University Medical Center, Department of Medicine, Division of Nephrology and Hypertension, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Saumya Das
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Ingrid Dahlman
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan D. Mosley
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John R. Koethe
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Mikael Rydén
- Karolinska Institutet, Department of Medicine, Huddinge (H7), Stockholm, Sweden
| | - Katherine N. Bachmann
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Ravi V. Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Department of Medicine, Cardiology Division, Nashville, Tennessee, USA
| |
Collapse
|
10
|
Smeehuijzen L, Gijbels A, Nugteren-Boogaard JP, Vrieling F, Boutagouga Boudjadja M, Trouwborst I, Jardon KM, Hul GB, Feskens EJM, Blaak EE, Goossens GH, Afman LA, Stienstra R. Immunometabolic Signatures of Circulating Monocytes in Humans With Obesity and Insulin Resistance. Diabetes 2024; 73:1112-1121. [PMID: 38656918 DOI: 10.2337/db23-0970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/14/2024] [Indexed: 04/26/2024]
Abstract
Obesity is associated with chronic inflammation and metabolic complications, including insulin resistance (IR). Immune cells drive inflammation through the rewiring of intracellular metabolism. However, the impact of obesity-related IR on the metabolism and functionality of circulating immune cells, like monocytes, remains poorly understood. To increase insight into the interindividual variation of immunometabolic signatures among individuals and their role in the development of IR, we assessed systemic and tissue-specific IR and circulating immune markers, and we characterized metabolic signatures and cytokine secretion of circulating monocytes from 194 individuals with a BMI ≥25 kg/m2. Monocyte metabolic signatures were defined using extracellular acidification rates (ECARs) to estimate glycolysis and oxygen consumption rates (OCRs) for oxidative metabolism. Although monocyte metabolic signatures and function based on cytokine secretion varied greatly among study participants, they were strongly associated with each other. The ECAR-to-OCR ratio, representing the balance between glycolysis and oxidative metabolism, was negatively associated with fasting insulin levels, systemic IR, and liver-specific IR. These results indicate that monocytes from individuals with IR were relatively more dependent on oxidative metabolism, whereas monocytes from more insulin-sensitive individuals were more dependent on glycolysis. Additionally, circulating CXCL11 was negatively associated with the degree of systemic IR and positively with the ECAR-to-OCR ratio in monocytes, suggesting that individuals with high IR and a monocyte metabolic dependence on oxidative metabolism also have lower levels of circulating CXCL11. Our findings suggest that monocyte metabolism is related to obesity-associated IR progression and deepen insights into the interplay between innate immune cell metabolism and IR development in humans. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Lisa Smeehuijzen
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Anouk Gijbels
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
- Top Institute Food and Nutrition, Wageningen, the Netherlands
| | | | - Frank Vrieling
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | | | - Inez Trouwborst
- Top Institute Food and Nutrition, Wageningen, the Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Kelly M Jardon
- Top Institute Food and Nutrition, Wageningen, the Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Gabby B Hul
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Lydia A Afman
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Rinke Stienstra
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
- Department of Internal Medicine (463), Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
11
|
Oussaada SM, Kilicarslan M, de Weijer BA, Gilijamse PW, Şekercan A, Virtue S, Janssen IMC, van de Laar A, Demirkiran A, van Wagensveld BA, Houdijk APJ, Jongejan A, Moerland PD, Verheij J, Geijtenbeek TB, Bloks VW, de Goffau MC, Romijn JA, Nieuwdorp M, Vidal-Puig A, Ter Horst KW, Serlie MJ. Tissue-specific inflammation and insulin sensitivity in subjects with obesity. Diabetes Res Clin Pract 2024; 211:111663. [PMID: 38616042 DOI: 10.1016/j.diabres.2024.111663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Abstract
Obesity is associated with low-grade inflammation and insulin resistance (IR). The contribution of adipose tissue (AT) and hepatic inflammation to IR remains unclear. We conducted a study across three cohorts to investigate this relationship. The first cohort consists of six women with normal weight and twenty with obesity. In women with obesity, we found an upregulation of inflammatory markers in subcutaneous and visceral adipose tissue, isolated AT macrophages, and the liver, but no linear correlation with tissue-specific insulin sensitivity. In the second cohort, we studied 24 women with obesity in the upper vs lower insulin sensitivity quartile. We demonstrated that several omental and mesenteric AT inflammatory genes and T cell-related pathways are upregulated in IR, independent of BMI. The third cohort consists of 23 women and 18 men with obesity, studied before and one year after bariatric surgery. Weight loss following surgery was associated with downregulation of multiple immune pathways in subcutaneous AT and skeletal muscle, alongside notable metabolic improvements. Our results show that obesity is characterised by systemic and tissue-specific inflammation. Subjects with obesity and IR show a more pronounced inflammation phenotype, independent of BMI. Bariatric surgery-induced weight loss is associated with reduced inflammation and improved metabolic health.
Collapse
Affiliation(s)
- S M Oussaada
- Amsterdam UMC Location University of Amsterdam, Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - M Kilicarslan
- Amsterdam UMC Location University of Amsterdam, Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - B A de Weijer
- Amsterdam UMC Location University of Amsterdam, Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - P W Gilijamse
- Amsterdam UMC Location University of Amsterdam, Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - A Şekercan
- Amsterdam UMC Location University of Amsterdam, Department of Public Health, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Department of Surgery, Meibergdreef 9, Amsterdam, the Netherlands
| | - S Virtue
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - I M C Janssen
- Nederlandse Obesitas Kliniek, Departement of Science, Huis ter Heide, the Netherlands
| | - A van de Laar
- Spaarne Gasthuis, Department of Surgery, Haarlem, the Netherlands
| | - A Demirkiran
- Red Cross Hospital, Department of Gastrointestinal Surgery, Beverwijk, the Netherlands
| | - B A van Wagensveld
- NMC Royal Hospital, Department of Surgery, Abu Dhabi, United Arab Emirates
| | - A P J Houdijk
- Northwest Clinics, Department of Surgery, Alkmaar, the Netherlands
| | - A Jongejan
- Amsterdam UMC Location University of Amsterdam, Epidemiology and Data Science, Amsterdam, the Netherlands; Amsterdam Public Health, Methodology, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, the Netherlands
| | - P D Moerland
- Amsterdam UMC Location University of Amsterdam, Epidemiology and Data Science, Amsterdam, the Netherlands; Amsterdam Public Health, Methodology, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, the Netherlands
| | - J Verheij
- Amsterdam UMC Location University of Amsterdam, Department of Pathology, Amsterdam, the Netherlands
| | - T B Geijtenbeek
- Amsterdam UMC Location University of Amsterdam, Laboratory for Experimental Immunology, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Cancer Immunology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Immunology, Amsterdam, the Netherlands
| | - V W Bloks
- University Medical Center Groningen, Department of Paediatrics, University of Groningen, Groningen, the Netherlands
| | - M C de Goffau
- Amsterdam UMC Location University of Amsterdam, Department of Experimental Vascular Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Wellcome Trust Sanger Institute, Hinxton, UK; Amsterdam UMC, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 9, Amsterdam, the Netherlands
| | - J A Romijn
- Amsterdam UMC Location University of Amsterdam, Department of Internal Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - M Nieuwdorp
- Amsterdam UMC Location University of Amsterdam, Department of Vascular Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - A Vidal-Puig
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - K W Ter Horst
- Amsterdam UMC Location University of Amsterdam, Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - M J Serlie
- Amsterdam UMC Location University of Amsterdam, Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands; Section of Endocrinology, Yale School of Medicine, New Haven, USA.
| |
Collapse
|
12
|
Trouwborst I, Jardon KM, Gijbels A, Hul G, Feskens EJM, Afman LA, Linge J, Goossens GH, Blaak EE. Body composition and body fat distribution in tissue-specific insulin resistance and in response to a 12-week isocaloric dietary macronutrient intervention. Nutr Metab (Lond) 2024; 21:20. [PMID: 38594756 PMCID: PMC11003022 DOI: 10.1186/s12986-024-00795-y] [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: 12/05/2023] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Body composition and body fat distribution are important predictors of cardiometabolic diseases. The etiology of cardiometabolic diseases is heterogenous, and partly driven by inter-individual differences in tissue-specific insulin sensitivity. OBJECTIVES To investigate (1) the associations between body composition and whole-body, liver and muscle insulin sensitivity, and (2) changes in body composition and insulin sensitivity and their relationship after a 12-week isocaloric diet high in mono-unsaturated fatty acids (HMUFA) or a low-fat, high-protein, high-fiber (LFHP) diet. METHODS This subcohort analysis of the PERSON study includes 93 individuals (53% women, BMI 25-40 kg/m2, 40-75 years) who participated in this randomized intervention study. At baseline and after 12 weeks of following the LFHP, or HMUFA diet, we performed a 7-point oral glucose tolerance test to assess whole-body, liver, and muscle insulin sensitivity, and whole-body magnetic resonance imaging to determine body composition and body fat distribution. Both diets are within the guidelines of healthy nutrition. RESULTS At baseline, liver fat content was associated with worse liver insulin sensitivity (β [95%CI]; 0.12 [0.01; 0.22]). Only in women, thigh muscle fat content was inversely related to muscle insulin sensitivity (-0.27 [-0.48; -0.05]). Visceral adipose tissue (VAT) was inversely associated with whole-body, liver, and muscle insulin sensitivity. Both diets decreased VAT, abdominal subcutaneous adipose tissue (aSAT), and liver fat, but not whole-body and tissue-specific insulin sensitivity with no differences between diets. Waist circumference, however, decreased more following the LFHP diet as compared to the HMUFA diet (-3.0 vs. -0.5 cm, respectively). After the LFHP but not HMUFA diet, improvements in body composition were positively associated with improvements in whole-body and liver insulin sensitivity. CONCLUSIONS Liver and muscle insulin sensitivity are distinctly associated with liver and muscle fat accumulation. Although both LFHP and HMUFA diets improved in body fat, VAT, aSAT, and liver fat, only LFHP-induced improvements in body composition are associated with improved insulin sensitivity. TRIAL REGISTRATION NCT03708419 (clinicaltrials.gov).
Collapse
Affiliation(s)
- Inez Trouwborst
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center +, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- TI Food and Nutrition (TiFN), Wageningen, The Netherlands
| | - Kelly M Jardon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center +, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- TI Food and Nutrition (TiFN), Wageningen, The Netherlands
| | - Anouk Gijbels
- TI Food and Nutrition (TiFN), Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Gabby Hul
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center +, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Lydia A Afman
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Jennifer Linge
- AMRA Medical AB, Linköping, Sweden
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center +, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center +, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
- TI Food and Nutrition (TiFN), Wageningen, The Netherlands.
| |
Collapse
|
13
|
Gijbels A, Erdős B, Trouwborst I, Jardon KM, Adriaens ME, Goossens GH, Blaak EE, Feskens EJM, Afman LA. Hepatic insulin resistance and muscle insulin resistance are characterized by distinct postprandial plasma metabolite profiles: a cross-sectional study. Cardiovasc Diabetol 2024; 23:97. [PMID: 38493102 PMCID: PMC10944619 DOI: 10.1186/s12933-024-02188-0] [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] [Received: 09/19/2023] [Accepted: 03/02/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Tissue-specific insulin resistance (IR) predominantly in muscle (muscle IR) or liver (liver IR) has previously been linked to distinct fasting metabolite profiles, but postprandial metabolite profiles have not been investigated in tissue-specific IR yet. Given the importance of postprandial metabolic impairments in the pathophysiology of cardiometabolic diseases, we compared postprandial plasma metabolite profiles in response to a high-fat mixed meal between individuals with predominant muscle IR or liver IR. METHODS This cross-sectional study included data from 214 women and men with BMI 25-40 kg/m2, aged 40-75 years, and with predominant muscle IR or liver IR. Tissue-specific IR was assessed using the muscle insulin sensitivity index (MISI) and hepatic insulin resistance index (HIRI), which were calculated from the glucose and insulin responses during a 7-point oral glucose tolerance test. Plasma samples were collected before (T = 0) and after (T = 30, 60, 120, 240 min) consumption of a high-fat mixed meal and 247 metabolite measures, including lipoproteins, cholesterol, triacylglycerol (TAG), ketone bodies, and amino acids, were quantified using nuclear magnetic resonance spectroscopy. Differences in postprandial plasma metabolite iAUCs between muscle and liver IR were tested using ANCOVA with adjustment for age, sex, center, BMI, and waist-to-hip ratio. P-values were adjusted for a false discovery rate (FDR) of 0.05 using the Benjamini-Hochberg method. RESULTS Sixty-eight postprandial metabolite iAUCs were significantly different between liver and muscle IR. Liver IR was characterized by greater plasma iAUCs of large VLDL (p = 0.004), very large VLDL (p = 0.002), and medium-sized LDL particles (p = 0.026), and by greater iAUCs of TAG in small VLDL (p = 0.025), large VLDL (p = 0.003), very large VLDL (p = 0.002), all LDL subclasses (all p < 0.05), and small HDL particles (p = 0.011), compared to muscle IR. In liver IR, the postprandial plasma fatty acid (FA) profile consisted of a higher percentage of saturated FA (p = 0.013), and a lower percentage of polyunsaturated FA (p = 0.008), compared to muscle IR. CONCLUSION People with muscle IR or liver IR have distinct postprandial plasma metabolite profiles, with more unfavorable postprandial metabolite responses in those with liver IR compared to muscle IR.
Collapse
Affiliation(s)
- Anouk Gijbels
- Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands.
| | - Balázs Erdős
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Paul-Henri Spaaklaan 1, 6229 EN, Maastricht, The Netherlands
| | - Inez Trouwborst
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Kelly M Jardon
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Michiel E Adriaens
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Paul-Henri Spaaklaan 1, 6229 EN, Maastricht, The Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Ellen E Blaak
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Lydia A Afman
- Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| |
Collapse
|
14
|
Hornero-Ramirez H, Aubin A, Michalski MC, Vinoy S, Caussy C, Nazare JA. Multifunctional dietary interventions, low-grade inflammation and cardiometabolic profile: a scoping review. Front Immunol 2024; 15:1304686. [PMID: 38476230 PMCID: PMC10927766 DOI: 10.3389/fimmu.2024.1304686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/07/2024] [Indexed: 03/14/2024] Open
Abstract
Background Growing evidence highlights the significant impact of diet to modify low-grade inflammation closely linked to cardiometabolic profile. Multifunctionnal diets, combining several compounds have been shown to beneficially impact metabolic parameters. Objective This study synthesizes the knowledge on the impact of RCTs combining dietary multifunctional compounds on low-grade inflammation in humans. We investigate whether the effects of dietary multifunctional interventions on inflammatory markers were parallel to alterations of cardiometabolic parameters. Methodology We considered both the integrated dietary interventions (ID, i.e. global diets such as Mediterranean, Nordic…) and the dietary interventions based on selected bioactive mix (BM) compounds, in healthy individuals and those at cardiometabolic risk. Out of 221 screened publications, we selected 27 studies: 11 for BM (polyphenols and/or omega-3 fatty acids and/or antioxidants and/or dietary fiber) and 16 for ID (Mediterranean, paleo, Nordic, Dietary Approaches to Stop Hypertension (DASH) diet…). Results ID studies reflected significant improvements in inflammatory markers (CRP, IL-6, IL-10, IL-1b), concomitantly with beneficial changes in metabolic parameters. In BM studies, pronounced effects on low-grade inflammatory markers were observed, while improvements in metabolic parameters were not consistent. Both types of studies suggested a favorable impact on oxidative stress, a factor closely linked to the inflammatory profile. Conclusion Our findings showed that multifunctional RCT diets have differential role in managing low-grade inflammation and cardiometabolic health, with a large heterogeneity in explored inflammatory markers. Further research is imperative to elucidate the link between low-grade inflammation and other cardiometabolic risk factors, such as intestinal inflammation or postprandial inflammatory dynamics, aiming to attain a comprehensive understanding of the mechanisms involved in these processes. These future investigations not only have the potential to deepen our insights into the connections among these elements but also pave the way for significant advancements in the prevention and management of conditions related to the cardiovascular and metabolic systems.
Collapse
Affiliation(s)
- Hugo Hornero-Ramirez
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, CarMeN lab, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Adrien Aubin
- Département Endocrinologie, Diabète et Nutrition, Hôpital Lyon Sud, Pierre-Bénite, Hospices Civils de Lyon, Pierre Bénite, France
| | - Marie-Caroline Michalski
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, CarMeN lab, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Sophie Vinoy
- Nutrition Research, Paris Saclay Tech Center, Mondelez International R&D, 91400, Saclay, France
| | - Cyrielle Caussy
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, CarMeN lab, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
- Département Endocrinologie, Diabète et Nutrition, Hôpital Lyon Sud, Pierre-Bénite, Hospices Civils de Lyon, Pierre Bénite, France
| | - Julie-Anne Nazare
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, CarMeN lab, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| |
Collapse
|
15
|
Kajita K, Ishii I, Mori I, Asano M, Fuwa M, Morita H. Sphingosine 1-Phosphate Regulates Obesity and Glucose Homeostasis. Int J Mol Sci 2024; 25:932. [PMID: 38256005 PMCID: PMC10816022 DOI: 10.3390/ijms25020932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
One of the major global health and welfare issues is the treatment of obesity and associated metabolic disorders, such as type 2 diabetes mellitus and nonalcoholic fatty liver disease. Obesity, caused by the excessive accumulation of triglycerides in adipose tissues, induces adipocyte dysfunction, followed by inflammation, in adipose tissues and lipotoxicity in nonadipose tissues. Several studies have shown that obesity and glucose homeostasis are influenced by sphingolipid mediators, including ceramide and sphingosine 1-phosphate (S1P). Cellular accumulation of ceramide impairs pancreatic β-cell survival, confers insulin resistance in the liver and the skeletal muscle, and deteriorates adipose tissue inflammation via unknown molecular mechanisms. The roles of S1P are more complicated, because there are five cell-surface S1P receptors (S1PRs: S1P1-5) which have altered functions, different cellular expression patterns, and inapparent intracellular targets. Recent findings, including those by our group, support the notable concept that the pharmacological activation of S1P1 or S1P3 improves obesity and associated metabolic disorders, whereas that of S1P2 has the opposite effect. In addition, the regulation of S1P production by sphingosine kinase (SphK) is an essential factor affecting glucose homeostasis. This review summarizes the current knowledge on SphK/S1P/S1PR signaling in and against obesity, insulin resistance, and associated disorders.
Collapse
Affiliation(s)
- Kazuo Kajita
- Department of Health and Nutrition, Faculty of Home Economics, Gifu Women’s University, 80 Taromaru, Gifu 501-2592, Japan
| | - Isao Ishii
- Department of Health Chemistry, Showa Pharmaceutical University, 3-3165 Higashitamagawagakuen, Machida 194-8543, Japan
| | - Ichiro Mori
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| | - Motochika Asano
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| | - Masayuki Fuwa
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| | - Hiroyuki Morita
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| |
Collapse
|
16
|
Steinbach E, Belda E, Alili R, Adriouch S, Dauriat CJG, Donatelli G, Dumont JL, Pacini F, Tuszynski T, Pelloux V, Jacques F, Creusot L, Coles E, Taillandier P, Vazquez Gomez M, Masi D, Mateo V, André S, Kordahi M, Rouault C, Zucker JD, Sokol H, Genser L, Chassaing B, Le Roy T, Clément K. Comparative analysis of the duodenojejunal microbiome with the oral and fecal microbiomes reveals its stronger association with obesity and nutrition. Gut Microbes 2024; 16:2405547. [PMID: 39679619 DOI: 10.1080/19490976.2024.2405547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 12/17/2024] Open
Abstract
The intestinal microbiota is increasingly recognized as a crucial player in the development and maintenance of various chronic conditions, including obesity and associated metabolic diseases. While most research focuses on the fecal microbiota due to its easier accessibility, the small intestine, as a major site for nutrient sensing and absorption, warrants further investigation to determine its microbiota composition and functions. Here, we conducted a clinical research project in 30 age- and sex-matched participants with (n = 15) and without (n = 15) obesity. Duodenojejunal fluid was obtained by aspiration during endoscopy. Phenotyping included clinical variables related to metabolic status, lifestyle, and psychosocial factors using validated questionnaires. We performed metagenomic analyses of the oral, duodenojejunal, and fecal microbiome, alongside metabolomic data from duodenojejunal fluid and feces, integrating these data with clinical and lifestyle information. Our results highlight significant associations between duodenojejunal microbiota composition and usual dietary intake, as well as clinical phenotypes, with larger effect sizes than the associations between these variables and fecal microbiota. Notably, we found that the duodenojejunal microbiota of patients with obesity exhibited higher diversity and showed distinct differences in the abundance of several duodenojejunal microbiota species compared with individuals without obesity. Our findings support the relevance of studying the role of the small intestinal microbiota in the pathogenesis of nutrition-related diseases.
Collapse
Affiliation(s)
- Emilie Steinbach
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Eugeni Belda
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, Sorbonne Université, Institut de Recherche pour le Développement (IRD), Paris, France
| | - Rohia Alili
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Solia Adriouch
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Charlène J G Dauriat
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM, CNRS UMR8104, Université de Paris, Paris, France
| | - Gianfranco Donatelli
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Endoscopy Department, Peupliers Hospital, Ramsay-Santé, Paris, France
| | - Jean-Loup Dumont
- Endoscopy Department, Peupliers Hospital, Ramsay-Santé, Paris, France
| | - Filippo Pacini
- Endoscopy Department, Peupliers Hospital, Ramsay-Santé, Paris, France
| | - Thierry Tuszynski
- Endoscopy Department, Peupliers Hospital, Ramsay-Santé, Paris, France
| | - Véronique Pelloux
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Flavien Jacques
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Laura Creusot
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
| | - Emavieve Coles
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Paul Taillandier
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Marta Vazquez Gomez
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Davide Masi
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - Véronique Mateo
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Sébastien André
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Melissa Kordahi
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM, CNRS UMR8104, Université de Paris, Paris, France
| | - Christine Rouault
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Jean-Daniel Zucker
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, Sorbonne Université, Institut de Recherche pour le Développement (IRD), Paris, France
| | - Harry Sokol
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
- INRAE, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France
| | - Laurent Genser
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Sorbonne Université, Department of Hepato-Biliary and Pancreatic Surgery, Assistance Publique-Hôpitaux de Paris, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM, CNRS UMR8104, Université de Paris, Paris, France
| | - Tiphaine Le Roy
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
| | - Karine Clément
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches, NutriOmics Research Unit, Sorbonne Université, Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
17
|
Shannon CE, Ní Chathail MB, Mullin SM, Meehan A, McGillicuddy FC, Roche HM. Precision nutrition for targeting pathophysiology of cardiometabolic phenotypes. Rev Endocr Metab Disord 2023; 24:921-936. [PMID: 37402955 PMCID: PMC10492734 DOI: 10.1007/s11154-023-09821-5] [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: 06/22/2023] [Indexed: 07/06/2023]
Abstract
Obesity is a heterogenous disease accompanied by a broad spectrum of cardiometabolic risk profiles. Traditional paradigms for dietary weight management do not address biological heterogeneity between individuals and have catastrophically failed to combat the global pandemic of obesity-related diseases. Nutritional strategies that extend beyond basic weight management to instead target patient-specific pathophysiology are warranted. In this narrative review, we provide an overview of the tissue-level pathophysiological processes that drive patient heterogeneity to shape distinct cardiometabolic phenotypes in obesity. Specifically, we discuss how divergent physiology and postprandial phenotypes can reveal key metabolic defects within adipose, liver, or skeletal muscle, as well as the integrative involvement of the gut microbiome and the innate immune system. Finally, we highlight potential precision nutritional approaches to target these pathways and discuss recent translational evidence concerning the efficacy of such tailored dietary interventions for different obesity phenotypes, to optimise cardiometabolic benefits.
Collapse
Affiliation(s)
- Christopher E Shannon
- Nutrigenomics Research Group, UCD Conway Institute, and Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Republic of Ireland
- School of Medicine, University College Dublin, Dublin, Republic of Ireland
- Division of Diabetes, Department of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Méabh B Ní Chathail
- Nutrigenomics Research Group, UCD Conway Institute, and Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Republic of Ireland
| | - Sinéad M Mullin
- Nutrigenomics Research Group, UCD Conway Institute, and Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Republic of Ireland
| | - Andrew Meehan
- School of Medicine, University College Dublin, Dublin, Republic of Ireland
| | | | - Helen M Roche
- Nutrigenomics Research Group, UCD Conway Institute, and Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Republic of Ireland.
- Institute for Global Food Security, Queen's University Belfast, Belfast, Northern Ireland.
| |
Collapse
|
18
|
Blaak EE, Goossens GH. Metabolic phenotyping in people living with obesity: Implications for dietary prevention. Rev Endocr Metab Disord 2023; 24:825-838. [PMID: 37581871 PMCID: PMC10492670 DOI: 10.1007/s11154-023-09830-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
Given the increasing number of people living with obesity and related chronic metabolic disease, precision nutrition approaches are required to increase the effectiveness of prevention strategies. This review addresses these approaches in different metabolic phenotypes (metabotypes) in obesity. Although obesity is typically associated with an increased cardiometabolic disease risk, some people with obesity are relatively protected against the detrimental effects of excess adiposity on cardiometabolic health, also referred to as 'metabolically healthy obesity' (MHO). Underlying mechanisms, the extent to which MHO is a transient state as well as lifestyle strategies to counteract the transition from MHO to metabolically unhealthy obesity (MUO) are discussed. Based on the limited resources that are available for dietary lifestyle interventions, it may be reasonable to prioritize interventions for people with MUO, since targeting high-risk patients for specific nutritional, lifestyle or weight-loss strategies may enhance the cost-effectiveness of these interventions. Additionally, the concept of tissue insulin resistant (IR) metabotypes is discussed, representing distinct etiologies towards type 2 diabetes (T2D) as well as cardiovascular disease (CVD). Recent evidence indicates that these tissue IR metabotypes, already present in individuals with obesity with a normal glucose homeostasis, respond differentially to diet. Modulation of dietary macronutrient composition according to these metabotypes may considerably improve cardiometabolic health benefits. Thus, nutritional or lifestyle intervention may improve cardiometabolic health, even with only minor or no weight loss, which stresses the importance of focusing on a healthy lifestyle and not on weight loss only. Targeting different metabotypes towards T2D and cardiometabolic diseases may lead to more effective lifestyle prevention and treatment strategies. Age and sex-related differences in tissue metabotypes and related microbial composition and functionality (fermentation), as important drivers and/or mediators of dietary intervention response, have to be taken into account. For the implementation of these approaches, more prospective trials are required to provide the knowledge base for precision nutrition in the prevention of chronic metabolic diseases.
Collapse
Affiliation(s)
- Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
19
|
Cossins BC, van den Munckhof I, Rutten JHW, van der Graaf M, Stienstra R, Joosten LAB, Netea MG, Li Y, Riksen NP. Sex-specific Association Between Adipose Tissue Inflammation and Vascular and Metabolic Complications of Obesity. J Clin Endocrinol Metab 2023; 108:2537-2549. [PMID: 37014796 PMCID: PMC10505527 DOI: 10.1210/clinem/dgad193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/12/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
CONTEXT Adipose tissue (AT) inflammation predisposes to insulin resistance and metabolic syndrome in obesity. OBJECTIVE To investigate the association between adipocyte size, AT inflammation, systemic inflammation, and metabolic and atherosclerotic complications of obesity in a sex-specific manner. DESIGN Cross-sectional cohort study. SETTING University hospital in the Netherlands. PARTICIPANTS A total of 302 adult subjects with a body mass index (BMI) ≥ 27 kg/m2. MAIN OUTCOME MEASURES We obtained subcutaneous abdominal fat biopsies and systematically assessed, in a sex-specific manner, associations of several parameters of AT inflammation (including adipocyte size, macrophage content, crown-like structures, and gene expression) to biomarkers of systemic inflammation, leukocyte number and function, and to the presence of metabolic syndrome, insulin resistance, and carotid atherosclerotic plaques, assessed with ultrasound. RESULTS Adipocyte size was associated with metabolic syndrome and AT macrophage content with insulin resistance. In contrast, none of the AT parameters was associated with carotid atherosclerosis, although mRNA expression of the anti-inflammatory IL-37 was associated with a lower intima-media thickness. We revealed profound sex-specific differences, with an association between BMI and adipocyte size, and between adipocyte size and metabolic syndrome in men only. Also, only men showed an association between adipocyte size, AT expression of leptin and MCP-1, and AT macrophage numbers, and between AT inflammation (crown-like structure number) and several circulating inflammatory proteins, including high specificity C-reactive protein, and IL-6. CONCLUSIONS Inflammation in abdominal subcutaneous adipose tissue is more related to the metabolic than the atherosclerotic complications of obesity, and there are profound sex-specific differences in the association between BMI, adipocyte size, AT inflammation, and systemic inflammation, which are much stronger in men than women.
Collapse
Affiliation(s)
- Benjamin C Cossins
- Department of Internal Medicine & Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Inge van den Munckhof
- Department of Internal Medicine & Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine & Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Marinette van der Graaf
- Department of Medical Imaging, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine & Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, 6708 WE Wageningen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine & Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine & Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53127 Bonn, Germany
| | - Yang Li
- Department of Internal Medicine & Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM) & TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Niels P Riksen
- Department of Internal Medicine & Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| |
Collapse
|
20
|
Wanders L, Gijbels A, Bakker EA, Trouwborst I, Jardon KM, Manusama KCM, Hul GB, Feskens EJM, Afman LA, Blaak EE, Hopman MTE, Goossens GH, Thijssen DHJ. Physical activity and sedentary behavior show distinct associations with tissue-specific insulin sensitivity in adults with overweight. Acta Physiol (Oxf) 2023; 237:e13945. [PMID: 36745002 DOI: 10.1111/apha.13945] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/21/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023]
Abstract
AIM The aim of this study is to investigate associations between the physical activity (PA) spectrum (sedentary behavior to exercise) and tissue-specific insulin resistance (IR). METHODS We included 219 participants for analysis (median [IQR]: 61 [55; 67] years, BMI 29.6 [26.9; 32.0] kg/m2 ; 60% female) with predominant muscle or liver IR, as determined using a 7-point oral glucose tolerance test (OGTT). PA and sedentary behavior were measured objectively (ActivPAL) across 7 days. Context-specific PA was assessed with the Baecke questionnaire. Multiple linear regression models (adjustments include age, sex, BMI, site, season, retirement, and dietary intake) were used to determine associations between the PA spectrum and hepatic insulin resistance index (HIRI), muscle insulin sensitivity index (MISI) and whole-body IR (HOMA-IR, Matsuda index). RESULTS In fully adjusted models, objectively measured total PA (standardized regression coefficient β = 0.17, p = 0.020), light-intensity PA (β = 0.15, p = 0.045) and moderate-to-vigorous intensity PA (β = 0.13, p = 0.048) were independently associated with Matsuda index, but not HOMA-IR (p > 0.05). A higher questionnaire-derived sport index and leisure index were associated with significantly lower whole-body IR (Matsuda, HOMA-IR) in men but not in women. Results varied across tissues: more time spent sedentary (β = -0.24, p = 0.045) and a higher leisure index (β = 0.14, p = 0.034) were respectively negatively and positively associated with MISI, but not HIRI. A higher sport index was associated with lower HIRI (β = -0.30, p = 0.007, in men only). CONCLUSION While we confirm a beneficial association between PA and whole-body IR, our findings indicate that associations between the PA spectrum and IR seem distinct depending on the primary site of insulin resistance (muscle or liver).
Collapse
Affiliation(s)
- Lisa Wanders
- Radboud Institute for Health Sciences, Department of Physiology, Radboud university medical center, Nijmegen, the Netherlands
- TiFN, Wageningen, the Netherlands
| | - Anouk Gijbels
- TiFN, Wageningen, the Netherlands
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Esmée A Bakker
- Radboud Institute for Health Sciences, Department of Physiology, Radboud university medical center, Nijmegen, the Netherlands
| | - Inez Trouwborst
- TiFN, Wageningen, the Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Kelly M Jardon
- TiFN, Wageningen, the Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Koen C M Manusama
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Gabby B Hul
- TiFN, Wageningen, the Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Lydia A Afman
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Ellen E Blaak
- TiFN, Wageningen, the Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Maria T E Hopman
- Radboud Institute for Health Sciences, Department of Physiology, Radboud university medical center, Nijmegen, the Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Dick H J Thijssen
- Radboud Institute for Health Sciences, Department of Physiology, Radboud university medical center, Nijmegen, the Netherlands
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
21
|
Trouwborst I, Gijbels A, Jardon KM, Siebelink E, Hul GB, Wanders L, Erdos B, Péter S, Singh-Povel CM, de Vogel-van den Bosch J, Adriaens ME, Arts ICW, Thijssen DHJ, Feskens EJM, Goossens GH, Afman LA, Blaak EE. Cardiometabolic health improvements upon dietary intervention are driven by tissue-specific insulin resistance phenotype: A precision nutrition trial. Cell Metab 2023; 35:71-83.e5. [PMID: 36599304 DOI: 10.1016/j.cmet.2022.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/12/2022] [Accepted: 11/13/2022] [Indexed: 01/05/2023]
Abstract
Precision nutrition based on metabolic phenotype may increase the effectiveness of interventions. In this proof-of-concept study, we investigated the effect of modulating dietary macronutrient composition according to muscle insulin-resistant (MIR) or liver insulin-resistant (LIR) phenotypes on cardiometabolic health. Women and men with MIR or LIR (n = 242, body mass index [BMI] 25-40 kg/m2, 40-75 years) were randomized to phenotype diet (PhenoDiet) group A or B and followed a 12-week high-monounsaturated fatty acid (HMUFA) diet or low-fat, high-protein, and high-fiber diet (LFHP) (PhenoDiet group A, MIR/HMUFA and LIR/LFHP; PhenoDiet group B, MIR/LFHP and LIR/HMUFA). PhenoDiet group B showed no significant improvements in the primary outcome disposition index, but greater improvements in insulin sensitivity, glucose homeostasis, serum triacylglycerol, and C-reactive protein compared with PhenoDiet group A were observed. We demonstrate that modulating macronutrient composition within the dietary guidelines based on tissue-specific insulin resistance (IR) phenotype enhances cardiometabolic health improvements. Clinicaltrials.gov registration: NCT03708419, CCMO registration NL63768.068.17.
Collapse
Affiliation(s)
- Inez Trouwborst
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands
| | - Anouk Gijbels
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Kelly M Jardon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands
| | - Els Siebelink
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Gabby B Hul
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands
| | - Lisa Wanders
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Balázs Erdos
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | | | | | | | - Michiel E Adriaens
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | - Ilja C W Arts
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | - Dick H J Thijssen
- Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, the Netherlands; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Lydia A Afman
- TI Food and Nutrition (TIFN), Wageningen, the Netherlands; Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; TI Food and Nutrition (TIFN), Wageningen, the Netherlands.
| |
Collapse
|
22
|
Oghabian A, van der Kolk BW, Marttinen P, Valsesia A, Langin D, Saris WH, Astrup A, Blaak EE, Pietiläinen KH. Baseline gene expression in subcutaneous adipose tissue predicts diet-induced weight loss in individuals with obesity. PeerJ 2023; 11:e15100. [PMID: 36992941 PMCID: PMC10042157 DOI: 10.7717/peerj.15100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/28/2023] [Indexed: 03/31/2023] Open
Abstract
Background Weight loss effectively reduces cardiometabolic health risks among people with overweight and obesity, but inter-individual variability in weight loss maintenance is large. Here we studied whether baseline gene expression in subcutaneous adipose tissue predicts diet-induced weight loss success. Methods Within the 8-month multicenter dietary intervention study DiOGenes, we classified a low weight-losers (low-WL) group and a high-WL group based on median weight loss percentage (9.9%) from 281 individuals. Using RNA sequencing, we identified the significantly differentially expressed genes between high-WL and low-WL at baseline and their enriched pathways. We used this information together with support vector machines with linear kernel to build classifier models that predict the weight loss classes. Results Prediction models based on a selection of genes that are associated with the discovered pathways 'lipid metabolism' (max AUC = 0.74, 95% CI [0.62-0.86]) and 'response to virus' (max AUC = 0.72, 95% CI [0.61-0.83]) predicted the weight-loss classes high-WL/low-WL significantly better than models based on randomly selected genes (P < 0.01). The performance of the models based on 'response to virus' genes is highly dependent on those genes that are also associated with lipid metabolism. Incorporation of baseline clinical factors into these models did not noticeably enhance the model performance in most of the runs. This study demonstrates that baseline adipose tissue gene expression data, together with supervised machine learning, facilitates the characterization of the determinants of successful weight loss.
Collapse
Affiliation(s)
- Ali Oghabian
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Birgitta W. van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pekka Marttinen
- Helsinki Institute for Information Technology HIIT, Department of Computer Science, Aalto University, Espoo, Finland
| | | | - Dominique Langin
- Department of Biochemistry, Toulouse University Hospitals, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - W. H. Saris
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Arne Astrup
- Healthy Weight Center, Novo Nordisk Fonden, Copenhagen, Denmark
| | - Ellen E. Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Healthy Weight Hub, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Aragón-Vela J, Alcalá-Bejarano Carrillo J, Moreno-Racero A, Plaza-Diaz J. The Role of Molecular and Hormonal Factors in Obesity and the Effects of Physical Activity in Children. Int J Mol Sci 2022; 23:15413. [PMID: 36499740 PMCID: PMC9737554 DOI: 10.3390/ijms232315413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/27/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Obesity and overweight are defined as abnormal fat accumulations. Adipose tissue consists of more than merely adipocytes; each adipocyte is closely coupled with the extracellular matrix. Adipose tissue stores excess energy through expansion. Obesity is caused by the abnormal expansion of adipose tissue as a result of adipocyte hypertrophy and hyperplasia. The process of obesity is controlled by several molecules, such as integrins, kindlins, or matrix metalloproteinases. In children with obesity, metabolomics studies have provided insight into the existence of unique metabolic profiles. As a result of low-grade inflammation in the system, abnormalities were observed in several metabolites associated with lipid, carbohydrate, and amino acid pathways. In addition, obesity and related hormones, such as leptin, play an instrumental role in regulating food intake and contributing to childhood obesity. The World Health Organization states that physical activity benefits the heart, the body, and the mind. Several noncommunicable diseases, such as cardiovascular disease, cancer, and diabetes, can be prevented and managed through physical activity. In this work, we reviewed pediatric studies that examined the molecular and hormonal control of obesity and the influence of physical activity on children with obesity or overweight. The purpose of this review was to examine some orchestrators involved in this disease and how they are related to pediatric populations. A larger number of randomized clinical trials with larger sample sizes and long-term studies could lead to the discovery of new key molecules as well as the detection of significant factors in the coming years. In order to improve the health of the pediatric population, omics analyses and machine learning techniques can be combined in order to improve treatment decisions.
Collapse
Affiliation(s)
- Jerónimo Aragón-Vela
- Department of Health Sciences, Area of Physiology, Building B3, Campus s/n “Las Lagunillas”, University of Jaén, 23071 Jaén, Spain
| | - Jesús Alcalá-Bejarano Carrillo
- Department of Health, University of the Valley of Mexico, Robles 600, Tecnologico I, San Luis Potosí 78220, Mexico
- Research and Advances in Molecular and Cellular Immunology, Center of Biomedical Research, University of Granada, Avda, del Conocimiento s/n, 18016 Armilla, Spain
| | - Aurora Moreno-Racero
- Research and Advances in Molecular and Cellular Immunology, Center of Biomedical Research, University of Granada, Avda, del Conocimiento s/n, 18016 Armilla, Spain
| | - Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Instituto de Investigación Biosanitaria IBS, Granada, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| |
Collapse
|
24
|
Exploring Anti-Type 2 Diabetes Mellitus Mechanism of Gegen Qinlian Decoction by Network Pharmacology and Experimental Validation. DISEASE MARKERS 2022; 2022:1927688. [PMID: 36284987 PMCID: PMC9588339 DOI: 10.1155/2022/1927688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022]
Abstract
Purpose. Gegen Qinlian Decoction (GGQL) has been employed to treat type 2 diabetes mellitus (T2DM) in the clinical practice of traditional Chinese medicine. However, the underlying mechanism of GGQL in the treatment of T2DM remains unknown. This study was aimed at exploring the pharmacological mechanisms of GGQL against T2DM via network pharmacology analysis combined with experimental validation. Methods. The effective components of GGQL were screened, and the target was predicted by using traditional Chinese medicine systems pharmacology database and analysis platform (TCMSP). The candidate targets of GGQL were predicted by network pharmacological analysis, and crucial targets were chosen by the protein-protein interaction (PPI) network. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses were performed to predict the core targets and pathways of GGQL against T2DM. Then, T2DM mice were induced by a high-fat diet combined with streptozotocin. The model and GGQL groups were given normal saline and GGQL aqueous solution (10 and 20 g/kg/d) intragastric administration, respectively, for 8 weeks. The mice in the GGQLT groups were administered with GGQLT at 10 and 20 g/kg/d, respectively. The pathological changes in liver tissues were observed by hematoxylin-eosin staining. The protein expression of TNF-α and NF-κB was verified by western blotting. Results. A total of 204 common targets of GGQL for the treatment of T2DM were obtained from 140 active ingredients and 212 potential targets of T2DM. GO and KEGG enrichment analysis involved 119 signaling pathways, mainly in inflammatory TNF signaling pathways. Animal experiments showed that GGQL significantly reduced the serum levels of body mass, fasting blood glucose, fasting insulin, HOMA-IR, TNF-α, and IL-17. The liver pathological section showed that GGQL could improve the vacuolar degeneration and lipid deposition in the liver of T2DM mice. Mechanistically, GGQL downregulated the mRNA expression of TNF-α and NF-κB. Conclusions. This study demonstrated that GGQL may exert antidiabetic effects against T2DM by suppressing TNF-α signaling pathway activation, thus providing a basis for its potential use in clinical practice and further study in treating T2DM.
Collapse
|
25
|
Sabaratnam R, Skov V, Paulsen SK, Juhl S, Kruse R, Hansen T, Halkier C, Kristensen JM, Vind BF, Richelsen B, Knudsen S, Dahlgaard J, Beck-Nielsen H, Kruse TA, Højlund K. A Signature of Exaggerated Adipose Tissue Dysfunction in Type 2 Diabetes Is Linked to Low Plasma Adiponectin and Increased Transcriptional Activation of Proteasomal Degradation in Muscle. Cells 2022; 11:cells11132005. [PMID: 35805088 PMCID: PMC9265693 DOI: 10.3390/cells11132005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/12/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
Insulin resistance in skeletal muscle in type 2 diabetes (T2D) is characterized by more pronounced metabolic and molecular defects than in obesity per se. There is increasing evidence that adipose tissue dysfunction contributes to obesity-induced insulin resistance in skeletal muscle. Here, we used an unbiased approach to examine if adipose tissue dysfunction is exaggerated in T2D and linked to diabetes-related mechanisms of insulin resistance in skeletal muscle. Transcriptional profiling and biological pathways analysis were performed in subcutaneous adipose tissue (SAT) and skeletal muscle biopsies from 17 patients with T2D and 19 glucose-tolerant, age and weight-matched obese controls. Findings were validated by qRT-PCR and western blotting of selected genes and proteins. Patients with T2D were more insulin resistant and had lower plasma adiponectin than obese controls. Transcriptional profiling showed downregulation of genes involved in mitochondrial oxidative phosphorylation and the tricarboxylic-acid cycle and increased expression of extracellular matrix (ECM) genes in SAT in T2D, whereas genes involved in proteasomal degradation were upregulated in the skeletal muscle in T2D. qRT-PCR confirmed most of these findings and showed lower expression of adiponectin in SAT and higher expression of myostatin in muscle in T2D. Interestingly, muscle expression of proteasomal genes correlated positively with SAT expression of ECM genes but inversely with the expression of ADIPOQ in SAT and plasma adiponectin. Protein content of proteasomal subunits and major ubiquitin ligases were unaltered in the skeletal muscle of patients with T2D. A transcriptional signature of exaggerated adipose tissue dysfunction in T2D, compared with obesity alone, is linked to low plasma adiponectin and increased transcriptional activation of proteasomal degradation in skeletal muscle.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, DK-4000 Roskilde, Denmark;
| | - Søren K. Paulsen
- Department of Pathology, Viborg Regional Hospital, DK-8800 Viborg, Denmark;
| | - Stine Juhl
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Rikke Kruse
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Thea Hansen
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Cecilie Halkier
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Jonas M. Kristensen
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Molecular Physiology Section, Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Birgitte F. Vind
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
| | - Bjørn Richelsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, DK-8200 Aarhus N, Denmark;
| | - Steen Knudsen
- Allarity Therapeutics Europe, DK-2970 Hørsholm, Denmark;
| | - Jesper Dahlgaard
- Program for Mind and Body in Mental Health, Research Centre for Health and Welfare Technology, VIA University College, DK-8200 Aarhus, Denmark;
- Department of Clinical Medicine, Aarhus University, DK-8200 Aarhus, Denmark
| | - Henning Beck-Nielsen
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
| | - Torben A. Kruse
- Department of Clinical Genetics, Odense University Hospital, DK-5000 Odense C, Denmark;
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
- Correspondence: ; Tel.: +45-2532-0648
| |
Collapse
|
26
|
Vliex LMM, Le GN, Fassarella M, Reijnders D, Goossens GH, Zoetendal EG, Penders J, Blaak EE. Fecal carriage of vanB antibiotic resistance gene affects adipose tissue function under vancomycin use. Gut Microbes 2022; 14:2083905. [PMID: 35695620 PMCID: PMC9196849 DOI: 10.1080/19490976.2022.2083905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Detrimental consequences of antibiotic treatment may include long-lasting disruption of the gut microbiota. Previous studies found no negative effects of antibiotics on metabolic health, although individualized responses were observed. Here, we aimed to investigate the subject-specific response to vancomycin use in tissue-specific insulin sensitivity by stratifying individuals based on the presence of antibiotic resistance genes (ARGs) or opportunistic pathogens (OPs) in the baseline fecal microbiota. Quantitative Polymerase Chain Reaction (qPCR) was used to detect ARGs and OPs in DNA isolated from fecal samples of 56 males with overweight/obesity (Body Mass Index: 25-35 kg/m2) and impaired glucose metabolism (fasting plasma glucose ≥5.6 mmol/L and/or 2-hour glucose 7.8-11.1 mmol/L). A two-step hyperinsulinemic-euglycemic clamp was performed to determine tissue-specific insulin sensitivity. Abdominal subcutaneous adipose tissue (AT) gene expression was assessed using Affymetrix microarray. Gut microbial composition was determined using the Human Intestinal Tract Chip (HITChip) microarray. At baseline, the vancomycin resistance gene vanB was present in 60% of our population. In individuals that were vanB-negative at baseline, AT insulin sensitivity (insulin-mediated suppression of plasma free fatty acids) improved during vancomycin use, while it decreased among vanB-positive individuals (% change post versus baseline: 14.1 ± 5.6 vs. -6.7 ± 7.5% (p = .042)). The vancomycin-induced increase in AT insulin sensitivity was accompanied by downregulation of inflammatory pathways and enrichment of extracellular matrix remodeling pathways in AT. In the vanB-positive group, well-known vanB-carrying bacteria, Enterococcus and Streptococcus, expanded in the gut microbiome. In conclusion, microbiome composition and adipose tissue biology were differentially affected by vancomycin treatment based on fecal vanB carriage.
Collapse
Affiliation(s)
- Lars M. M. Vliex
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Giang N. Le
- Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Marina Fassarella
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Dorien Reijnders
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Erwin G. Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ellen E. Blaak
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands,CONTACT Ellen E. Blaak Department of Human Biology, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
27
|
Detection of SARS-CoV-2 in subcutaneous fat but not visceral fat, and the disruption of fat lymphocyte homeostasis in both fat tissues in the macaque. Commun Biol 2022; 5:542. [PMID: 35661814 PMCID: PMC9166782 DOI: 10.1038/s42003-022-03503-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 05/19/2022] [Indexed: 12/13/2022] Open
Abstract
The well documented association between obesity and the severity of SARS-CoV-2 infection raises the question of whether adipose tissue (AT) is impacted during this infection. Using a model of SARS-CoV-2 infection in cynomolgus macaques, we detected the virus within subcutaneous AT (SCAT) but not in visceral AT (VAT) or epicardial AT on day 7 post-infection. We sought to determine the mechanisms responsible for this selective detection and observed higher levels of angiotensin-converting-enzyme-2 mRNA expression in SCAT than in VAT. Lastly, we evaluated the immunological consequences of SARS-CoV-2 infection on AT: both SCAT and VAT T cells showed a drastic reduction in CD69 expression, a standard marker of resident memory T cell in tissue, that is also involved in the migratory and metabolic properties of T cells. Our results demonstrate that in a model of mild infection, SCAT is selectively infected by SARS-CoV-2 although changes in the immune properties of AT are observed in both SCAT and VAT. Subcutaneous fat tissue expresses higher angiotensin-converting-enzyme 2 mRNA than visceral fat tissue and is selectively infected by SARS-Cov-2, while both fat tissues show drastic reduction in CD69 expression in T cells.
Collapse
|
28
|
Ghanbari F, Yazdanpanah N, Yazdanpanah M, Richards JB, Manousaki D. Connecting Genomics and Proteomics to Identify Protein Biomarkers for Adult and Youth-Onset Type 2 Diabetes: A Two-Sample Mendelian Randomization Study. Diabetes 2022; 71:1324-1337. [PMID: 35234851 DOI: 10.2337/db21-1046] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022]
Abstract
Type 2 diabetes shows an increasing prevalence in both adults and children. Identification of biomarkers for both youth and adult-onset type 2 diabetes is crucial for development of screening tools or drug targets. In this study, using two-sample Mendelian randomization (MR), we identified 22 circulating proteins causally linked to adult type 2 diabetes and 11 proteins with suggestive evidence for association with youth-onset type 2 diabetes. Among these, colocalization analysis further supported a role in type 2 diabetes for C-type mannose receptor 2 (MR odds ratio [OR] 0.85 [95% CI 0.79-0.92] per genetically predicted SD increase in protein level), MANS domain containing 4 (MR OR 0.90 [95% CI 0.88-0.92]), sodium/potassium-transporting ATPase subunit β2 (MR OR 1.10 [95% CI 1.06-1.15]), endoplasmic reticulum oxidoreductase 1β (MR OR 1.09 [95% CI 1.05-1.14]), spermatogenesis-associated protein 20 (MR OR 1.12 [95% CI 1.06-1.18]), haptoglobin (MR OR 0.96 [95% CI 0.94-0.98]), and α1-3-N-acetylgalactosaminyltransferase and α1-3-galactosyltransferase (MR OR 1.04 [95% CI 1.03-1.05]). Our findings support a causal role in type 2 diabetes for a set of circulating proteins, which represent promising type 2 diabetes drug targets.
Collapse
Affiliation(s)
- Faegheh Ghanbari
- Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Nahid Yazdanpanah
- Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Mojgan Yazdanpanah
- Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada
| | - J Brent Richards
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Department of Epidemiology and Biostatistics, McGill University, Montreal, Quebec, Canada
- Department of Twin Research, King's College London, London, U.K
| | - Despoina Manousaki
- Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada
- Departments of Pediatrics, Biochemistry and Molecular Medicine, University of Montreal, Montreal, Canada
| |
Collapse
|
29
|
de Hoogh IM, Pasman WJ, Boorsma A, van Ommen B, Wopereis S. Effects of a 13-Week Personalized Lifestyle Intervention Based on the Diabetes Subtype for People with Newly Diagnosed Type 2 Diabetes. Biomedicines 2022; 10:biomedicines10030643. [PMID: 35327447 PMCID: PMC8945461 DOI: 10.3390/biomedicines10030643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/15/2022] [Accepted: 03/07/2022] [Indexed: 12/16/2022] Open
Abstract
A type 2 diabetes mellitus (T2DM) subtyping method that determines the T2DM phenotype based on an extended oral glucose tolerance test is proposed. It assigns participants to one of seven subtypes according to their β-cell function and the presence of hepatic and/or muscle insulin resistance. The effectiveness of this subtyping approach and subsequent personalized lifestyle treatment in ameliorating T2DM was assessed in a primary care setting. Sixty participants, newly diagnosed with (pre)diabetes type 2 and not taking diabetes medication, completed the intervention. Retrospectively collected data of 60 people with T2DM from usual care were used as controls. Bodyweight (p < 0.01) and HbA1c (p < 0.01) were significantly reduced after 13 weeks in the intervention group, but not in the usual care group. The intervention group achieved 75.0% diabetes remission after 13 weeks (fasting glucose ≤ 6.9 mmol/L and HbA1c < 6.5% (48 mmol/mol)); for the usual care group, this was 22.0%. Lasting (two years) remission was especially achieved in subgroups with isolated hepatic insulin resistance. Our study shows that a personalized diagnosis and lifestyle intervention for T2DM in a primary care setting may be more effective in improving T2DM-related parameters than usual care, with long-term effects seen especially in subgroups with hepatic insulin resistance.
Collapse
|
30
|
Huang X, Maguire OA, Walker JM, Jiang CS, Carroll TS, Luo JD, Tonorezos E, Friedman DN, Cohen P. Therapeutic radiation exposure of the abdomen during childhood induces chronic adipose tissue dysfunction. JCI Insight 2021; 6:153586. [PMID: 34554929 PMCID: PMC8663557 DOI: 10.1172/jci.insight.153586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/22/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUNDChildhood cancer survivors who received abdominal radiotherapy (RT) or total body irradiation (TBI) are at increased risk for cardiometabolic disease, but the underlying mechanisms are unknown. We hypothesize that RT-induced adipose tissue dysfunction contributes to the development of cardiometabolic disease in the expanding population of childhood cancer survivors.METHODSWe performed clinical metabolic profiling of adult childhood cancer survivors previously exposed to TBI, abdominal RT, or chemotherapy alone, alongside a group of healthy controls. Study participants underwent abdominal s.c. adipose biopsies to obtain tissue for bulk RNA sequencing. Transcriptional signatures were analyzed using pathway and network analyses and cellular deconvolution.RESULTSIrradiated adipose tissue is characterized by a gene expression signature indicative of a complex macrophage expansion. This signature includes activation of the TREM2-TYROBP network, a pathway described in diseases of chronic tissue injury. Radiation exposure of adipose is further associated with dysregulated adipokine secretion, specifically a decrease in insulin-sensitizing adiponectin and an increase in insulin resistance-promoting plasminogen activator inhibitor-1. Accordingly, survivors exhibiting these changes have early signs of clinical metabolic derangement, such as increased fasting glucose and hemoglobin A1c.CONCLUSIONChildhood cancer survivors exposed to abdominal RT or TBI during treatment exhibit signs of chronic s.c. adipose tissue dysfunction, manifested as dysregulated adipokine secretion that may negatively impact their systemic metabolic health.FUNDINGThis study was supported by Rockefeller University Hospital; National Institute of General Medical Sciences (T32GM007739); National Center for Advancing Translational Sciences (UL1 TR001866); National Cancer Institute (P30CA008748); American Cancer Society (133831-CSDG-19-117-01-CPHPS); American Diabetes Association (1-17-ACE-17); and an anonymous donor (MSKCC).
Collapse
Affiliation(s)
- Xiaojing Huang
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA.,Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York, USA
| | - Olivia A Maguire
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York, USA.,Weill Cornell/Rockefeller/Sloan Kettering Tri-institutional MD-PhD Program, New York, New York, USA
| | | | | | - Thomas S Carroll
- Bioinformatics Resource Center, The Rockefeller University, New York, New York, USA
| | - Ji-Dung Luo
- Bioinformatics Resource Center, The Rockefeller University, New York, New York, USA
| | - Emily Tonorezos
- Office of Cancer Survivorship, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland, USA
| | | | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York, USA
| |
Collapse
|
31
|
Gijbels A, Trouwborst I, Jardon KM, Hul GB, Siebelink E, Bowser SM, Yildiz D, Wanders L, Erdos B, Thijssen DHJ, Feskens EJM, Goossens GH, Afman LA, Blaak EE. The PERSonalized Glucose Optimization Through Nutritional Intervention (PERSON) Study: Rationale, Design and Preliminary Screening Results. Front Nutr 2021; 8:694568. [PMID: 34277687 PMCID: PMC8278004 DOI: 10.3389/fnut.2021.694568] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
Background: It is well-established that the etiology of type 2 diabetes differs between individuals. Insulin resistance (IR) may develop in different tissues, but the severity of IR may differ in key metabolic organs such as the liver and skeletal muscle. Recent evidence suggests that these distinct tissue-specific IR phenotypes may also respond differentially to dietary macronutrient composition with respect to improvements in glucose metabolism. Objective: The main objective of the PERSON study is to investigate the effects of an optimal vs. suboptimal dietary macronutrient intervention according to tissue-specific IR phenotype on glucose metabolism and other health outcomes. Methods: In total, 240 overweight/obese (BMI 25 – 40 kg/m2) men and women (age 40 – 75 years) with either skeletal muscle insulin resistance (MIR) or liver insulin resistance (LIR) will participate in a two-center, randomized, double-blind, parallel, 12-week dietary intervention study. At screening, participants undergo a 7-point oral glucose tolerance test (OGTT) to determine the hepatic insulin resistance index (HIRI) and muscle insulin sensitivity index (MISI), classifying each participant as either “No MIR/LIR,” “MIR,” “LIR,” or “combined MIR/LIR.” Individuals with MIR or LIR are randomized to follow one of two isocaloric diets varying in macronutrient content and quality, that is hypothesized to be either an optimal or suboptimal diet, depending on their tissue-specific IR phenotype (MIR/LIR). Extensive measurements in a controlled laboratory setting as well as phenotyping in daily life are performed before and after the intervention. The primary study outcome is the difference in change in disposition index, which is the product of insulin sensitivity and first-phase insulin secretion, between participants who received their hypothesized optimal or suboptimal diet. Discussion: The PERSON study is one of the first randomized clinical trials in the field of precision nutrition to test effects of a more personalized dietary intervention based on IR phenotype. The results of the PERSON study will contribute knowledge on the effectiveness of targeted nutritional strategies to the emerging field of precision nutrition, and improve our understanding of the complex pathophysiology of whole body and tissue-specific IR. Clinical Trial Registration:https://clinicaltrials.gov/ct2/show/NCT03708419, clinicaltrials.gov as NCT03708419.
Collapse
Affiliation(s)
- Anouk Gijbels
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands.,Top Institute Food and Nutrition, Wageningen, Netherlands
| | - Inez Trouwborst
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Kelly M Jardon
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Gabby B Hul
- Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Els Siebelink
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands
| | - Suzanne M Bowser
- Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Dilemin Yildiz
- Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Lisa Wanders
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Balázs Erdos
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - Dick H J Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Lydia A Afman
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands
| | - Ellen E Blaak
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
32
|
Kalafati M, Lenz M, Ertaylan G, Arts ICW, Evelo CT, van Greevenbroek MMJ, Blaak EE, Adriaens M, Kutmon M. Assessing the Contribution of Relative Macrophage Frequencies to Subcutaneous Adipose Tissue. Front Nutr 2021; 8:675935. [PMID: 34136521 PMCID: PMC8200404 DOI: 10.3389/fnut.2021.675935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/16/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Macrophages play an important role in regulating adipose tissue function, while their frequencies in adipose tissue vary between individuals. Adipose tissue infiltration by high frequencies of macrophages has been linked to changes in adipokine levels and low-grade inflammation, frequently associated with the progression of obesity. The objective of this project was to assess the contribution of relative macrophage frequencies to the overall subcutaneous adipose tissue gene expression using publicly available datasets. Methods: Seven publicly available microarray gene expression datasets from human subcutaneous adipose tissue biopsies (n = 519) were used together with TissueDecoder to determine the adipose tissue cell-type composition of each sample. We divided the subjects in four groups based on their relative macrophage frequencies. Differential gene expression analysis between the high and low relative macrophage frequencies groups was performed, adjusting for sex and study. Finally, biological processes were identified using pathway enrichment and network analysis. Results: We observed lower frequencies of adipocytes and higher frequencies of adipose stem cells in individuals characterized by high macrophage frequencies. We additionally studied whether, within subcutaneous adipose tissue, interindividual differences in the relative frequencies of macrophages were reflected in transcriptional differences in metabolic and inflammatory pathways. Adipose tissue of individuals with high macrophage frequencies had a higher expression of genes involved in complement activation, chemotaxis, focal adhesion, and oxidative stress. Similarly, we observed a lower expression of genes involved in lipid metabolism, fatty acid synthesis, and oxidation and mitochondrial respiration. Conclusion: We present an approach that combines publicly available subcutaneous adipose tissue gene expression datasets with a deconvolution algorithm to calculate subcutaneous adipose tissue cell-type composition. The results showed the expected increased inflammation gene expression profile accompanied by decreased gene expression in pathways related to lipid metabolism and mitochondrial respiration in subcutaneous adipose tissue in individuals characterized by high macrophage frequencies. This approach demonstrates the hidden strength of reusing publicly available data to gain cell-type-specific insights into adipose tissue function.
Collapse
Affiliation(s)
- Marianthi Kalafati
- Deparment of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Michael Lenz
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Institute of Organismic and Molecular Evolution, Johannes Gutenberg University of Mainz, Mainz, Germany.,Preventive Cardiology and Preventive Medicine-Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gökhan Ertaylan
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Unit Health, Flemish Institute for Technological Research, Antwerp, Belgium
| | - Ilja C W Arts
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Department of Epidemiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Chris T Evelo
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Department of Bioinformatics-BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Ellen E Blaak
- Deparment of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Michiel Adriaens
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - Martina Kutmon
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Department of Bioinformatics-BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
33
|
van der Kolk BW, Muniandy M, Kaminska D, Alvarez M, Ko A, Miao Z, Valsesia A, Langin D, Vaittinen M, Pääkkönen M, Jokinen R, Kaye S, Heinonen S, Virtanen KA, Andersson DP, Männistö V, Saris WH, Astrup A, Rydén M, Blaak EE, Pajukanta P, Pihlajamäki J, Pietiläinen KH. Differential Mitochondrial Gene Expression in Adipose Tissue Following Weight Loss Induced by Diet or Bariatric Surgery. J Clin Endocrinol Metab 2021; 106:1312-1324. [PMID: 33560372 PMCID: PMC8063261 DOI: 10.1210/clinem/dgab072] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Mitochondria are essential for cellular energy homeostasis, yet their role in subcutaneous adipose tissue (SAT) during different types of weight-loss interventions remains unknown. OBJECTIVE To investigate how SAT mitochondria change following diet-induced and bariatric surgery-induced weight-loss interventions in 4 independent weight-loss studies. METHODS The DiOGenes study is a European multicenter dietary intervention with an 8-week low caloric diet (LCD; 800 kcal/d; n = 261) and 6-month weight-maintenance (n = 121) period. The Kuopio Obesity Surgery study (KOBS) is a Roux-en-Y gastric bypass (RYGB) surgery study (n = 172) with a 1-year follow-up. We associated weight-loss percentage with global and 2210 mitochondria-related RNA transcripts in linear regression analysis adjusted for age and sex. We repeated these analyses in 2 studies. The Finnish CRYO study has a 6-week LCD (800-1000 kcal/d; n = 19) and a 10.5-month follow-up. The Swedish DEOSH study is a RYGB surgery study with a 2-year (n = 49) and 5-year (n = 37) follow-up. RESULTS Diet-induced weight loss led to a significant transcriptional downregulation of oxidative phosphorylation (DiOGenes; ingenuity pathway analysis [IPA] z-scores: -8.7 following LCD, -4.4 following weight maintenance; CRYO: IPA z-score: -5.6, all P < 0.001), while upregulation followed surgery-induced weight loss (KOBS: IPA z-score: 1.8, P < 0.001; in DEOSH: IPA z-scores: 4.0 following 2 years, 0.0 following 5 years). We confirmed an upregulated oxidative phosphorylation at the proteomics level following surgery (IPA z-score: 3.2, P < 0.001). CONCLUSIONS Differentially regulated SAT mitochondria-related gene expressions suggest qualitative alterations between weight-loss interventions, providing insights into the potential molecular mechanistic targets for weight-loss success.
Collapse
Affiliation(s)
- Birgitta W van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Dorota Kaminska
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Arthur Ko
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Zong Miao
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Armand Valsesia
- Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Dominique Langin
- Institut National de la Santé et de la Recherche Médicale (Inserm), Université Paul Sabatier, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
- Department of Biochemistry, Toulouse University Hospitals, France
| | - Maija Vaittinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Mirva Pääkkönen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Finland
| | - Riikka Jokinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Sanna Kaye
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Kirsi A Virtanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
- Turku PET Center, Turku University Hospital, Turku, Finland
| | - Daniel P Andersson
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Wim H Saris
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, MD Maastricht, The Netherlands
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, MD Maastricht, The Netherlands
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
- Obesity Center, Abdominal center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
34
|
Molecular pathways behind acquired obesity: Adipose tissue and skeletal muscle multiomics in monozygotic twin pairs discordant for BMI. CELL REPORTS MEDICINE 2021; 2:100226. [PMID: 33948567 PMCID: PMC8080113 DOI: 10.1016/j.xcrm.2021.100226] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/31/2020] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
Tissue-specific mechanisms prompting obesity-related development complications in humans remain unclear. We apply multiomics analyses of subcutaneous adipose tissue and skeletal muscle to examine the effects of acquired obesity among 49 BMI-discordant monozygotic twin pairs. Overall, adipose tissue appears to be more affected by excess body weight than skeletal muscle. In heavier co-twins, we observe a transcriptional pattern of downregulated mitochondrial pathways in both tissues and upregulated inflammatory pathways in adipose tissue. In adipose tissue, heavier co-twins exhibit lower creatine levels; in skeletal muscle, glycolysis- and redox stress-related protein and metabolite levels remain higher. Furthermore, metabolomics analyses in both tissues reveal that several proinflammatory lipids are higher and six of the same lipid derivatives are lower in acquired obesity. Finally, in adipose tissue, but not in skeletal muscle, mitochondrial downregulation and upregulated inflammation are associated with a fatty liver, insulin resistance, and dyslipidemia, suggesting that adipose tissue dominates in acquired obesity. Multiomics analyses of adipose tissue and skeletal muscle in BMI-discordant twins Excess body weight downregulates mitochondrial pathways in both tissues Excess body weight upregulates proinflammatory pathways in both tissues Adipose tissue alterations are associated with metabolic health in acquired obesity
Collapse
|
35
|
Brennan L, de Roos B. Nutrigenomics: lessons learned and future perspectives. Am J Clin Nutr 2021; 113:503-516. [PMID: 33515029 DOI: 10.1093/ajcn/nqaa366] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
The omics technologies of metabolomics, transcriptomics, proteomics, and metagenomics are playing an increasingly important role in nutrition science. With the emergence of the concept of precision nutrition and the need to understand individual responses to dietary interventions, it is an opportune time to examine the impact of these tools to date in human nutrition studies. Advances in our mechanistic understanding of dietary interventions were realized through incorporation of metabolomics, proteomics, and, more recently, metagenomics. A common observation across the studies was the low intra-individual variability of the omics measurements and the high inter-individual variation. Harnessing this data for use in the development of precision nutrition will be important. Metabolomics in particular has played a key role in the development of biomarkers of food intake in an effort to enhance the accuracy of dietary assessments. Further work is needed to realize the full potential of such biomarkers and to demonstrate integration with current strategies, with the goal of overcoming the well-established limitations of self-reported approaches. Although many of the nutrigenomic studies performed to date were labelled as proof-of-concept or pilot studies, there is ample evidence to support the use of these technologies in nutrition science. Incorporating omic technologies from the start of study designs will ensure that studies are sufficiently powered for such data. Furthermore, multi-disciplinary collaborations are likely to become even more important to aid analyses and interpretation of the data.
Collapse
Affiliation(s)
- Lorraine Brennan
- Institute of Food and Health and Conway Institute, University College Dublin (UCD) School of Agriculture and Food Science, UCD, Belfield, Dublin, Ireland
| | - Baukje de Roos
- The Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| |
Collapse
|
36
|
Bassaganya-Riera J, Berry EM, Blaak EE, Burlingame B, le Coutre J, van Eden W, El-Sohemy A, German JB, Knorr D, Lacroix C, Muscaritoli M, Nieman DC, Rychlik M, Scholey A, Serafini M. Goals in Nutrition Science 2020-2025. Front Nutr 2021; 7:606378. [PMID: 33665201 PMCID: PMC7923694 DOI: 10.3389/fnut.2020.606378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Five years ago, with the editorial board of Frontiers in Nutrition, we took a leap of faith to outline the Goals for Nutrition Science - the way we see it (1). Now, in 2020, we can put ourselves to the test and take a look back. Without a doubt we got it right with several of the key directions. To name a few, Sustainable Development Goals (SDGs) for Food and Nutrition are part of the global public agenda, and the SDGs contribute to the structuring of international science and research. Nutritional Science has become a critical element in strengthening work on the SDGs, and the development of appropriate methodologies is built on the groundwork of acquiring and analyzing big datasets. Investigation of the Human Microbiome is providing novel insight on the interrelationship between nutrition, the immune system and disease. Finally, with an advanced definition of the gut-brain-axis we are getting a glimpse into the potential for Nutrition and Brain Health. Various milestones have been achieved, and any look into the future will have to consider the lessons learned from Covid-19 and the sobering awareness about the frailty of our food systems in ensuring global food security. With a view into the coming 5 years from 2020 to 2025, the editorial board has taken a slightly different approach as compared to the previous Goals article. A mind map has been created to outline the key topics in nutrition science. Not surprisingly, when looking ahead, the majority of scientific investigation required will be in the areas of health and sustainability. Johannes le Coutre, Field Chief Editor, Frontiers in Nutrition.
Collapse
Affiliation(s)
- Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory (NIMML) Institute, Blacksburg, VA, United States
| | - Elliot M Berry
- Braun School of Public Health, Hebrew University - Hadassah Medical School, Jerusalem, Israel
| | - Ellen E Blaak
- Department of Human Biology, Maastricht University, Maastricht, Netherlands
| | | | - Johannes le Coutre
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Willem van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Ahmed El-Sohemy
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - J Bruce German
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | - Dietrich Knorr
- Institute of Food Technology and Chemistry, Technische Universität Berlin, Berlin, Germany
| | - Christophe Lacroix
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - David C Nieman
- Human Performance Laboratory, Department of Biology, Appalachian State University, Kannapolis, NC, United States
| | - Michael Rychlik
- Technical University of Munich, Analytical Food Chemistry, Freising, Germany
| | - Andrew Scholey
- Centre for Human Psychopharmacology, Swinburne University, Melbourne, VIC, Australia
| | - Mauro Serafini
- Functional Food and Metabolic Stress Prevention Laboratory, Faculty of Biosciences and Technologies for Agriculture, Food and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
37
|
Kawai T, Autieri MV, Scalia R. Adipose tissue inflammation and metabolic dysfunction in obesity. Am J Physiol Cell Physiol 2020; 320:C375-C391. [PMID: 33356944 DOI: 10.1152/ajpcell.00379.2020] [Citation(s) in RCA: 886] [Impact Index Per Article: 177.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Several lines of preclinical and clinical research have confirmed that chronic low-grade inflammation of adipose tissue is mechanistically linked to metabolic disease and organ tissue complications in the overweight and obese organism. Despite this widely confirmed paradigm, numerous open questions and knowledge gaps remain to be investigated. This is mainly due to the intricately intertwined cross-talk of various pro- and anti-inflammatory signaling cascades involved in the immune response of expanding adipose depots, particularly the visceral adipose tissue. Adipose tissue inflammation is initiated and sustained over time by dysfunctional adipocytes that secrete inflammatory adipokines and by infiltration of bone marrow-derived immune cells that signal via production of cytokines and chemokines. Despite its low-grade nature, adipose tissue inflammation negatively impacts remote organ function, a phenomenon that is considered causative of the complications of obesity. The aim of this review is to broadly present an overview of adipose tissue inflammation by highlighting the most recent reports in the scientific literature and summarizing our overall understanding of the field. We also discuss key endogenous anti-inflammatory mediators and analyze their mechanistic role(s) in the pathogenesis and treatment of adipose tissue inflammation. In doing so, we hope to stimulate studies to uncover novel physiological, cellular, and molecular targets for the treatment of obesity.
Collapse
Affiliation(s)
- Tatsuo Kawai
- The Cardiovascular Research Center and The Limole Center for Integrated Lymphatic Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Michael V Autieri
- The Cardiovascular Research Center and The Limole Center for Integrated Lymphatic Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Rosario Scalia
- The Cardiovascular Research Center and The Limole Center for Integrated Lymphatic Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
38
|
Oosterman JE, Wopereis S, Kalsbeek A. The Circadian Clock, Shift Work, and Tissue-Specific Insulin Resistance. Endocrinology 2020; 161:5916887. [PMID: 33142318 DOI: 10.1210/endocr/bqaa180] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/25/2020] [Indexed: 01/02/2023]
Abstract
Obesity and type 2 diabetes (T2D) have become a global health concern. The prevalence of obesity and T2D is significantly higher in shift workers compared to people working regular hours. An accepted hypothesis is that the increased risk for metabolic health problems arises from aberrantly timed eating behavior, that is, eating out of synchrony with the biological clock. The biological clock is part of the internal circadian timing system, which controls not only the sleep/wake and feeding/fasting cycle, but also many metabolic processes in the body, including the timing of our eating behavior, and processes involved in glucose homeostasis. Rodent studies have shown that eating out of phase with the endogenous clock results in desynchronization between rhythms of the central and peripheral clock systems and between rhythms of different tissue clocks (eg, liver and muscle clock). Glucose homeostasis is a complex process that involves multiple organs. In the healthiest situation, functional rhythms of these organs are synchronized. We hypothesize that desynchronization between different metabolically active organs contributes to alterations in glucose homeostasis. Here we summarize the most recent information on desynchronization between organs due to shift work and shifted food intake patterns and introduce the concept of phenotypic flexibility, a validated test to assess the contribution of each organ to insulin resistance (IR) in humans. We propose this test as a way to provide further insight into the possible desynchronization between tissue clocks. Because different types of IR benefit from different therapeutic approaches, we also describe different chronotherapeutic strategies to promote synchrony within and between metabolically active organs.
Collapse
Affiliation(s)
- Johanneke E Oosterman
- Department of Microbiology and Systems Biology, Netherlands Organization for Applied Scientific Research (TNO), HE Zeist, the Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, AZ Amsterdam, the Netherlands
| | - Suzan Wopereis
- Department of Microbiology and Systems Biology, Netherlands Organization for Applied Scientific Research (TNO), HE Zeist, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, AZ Amsterdam, the Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), BA Amsterdam, the Netherlands
| |
Collapse
|
39
|
AlZaim I, Hammoud SH, Al-Koussa H, Ghazi A, Eid AH, El-Yazbi AF. Adipose Tissue Immunomodulation: A Novel Therapeutic Approach in Cardiovascular and Metabolic Diseases. Front Cardiovasc Med 2020; 7:602088. [PMID: 33282920 PMCID: PMC7705180 DOI: 10.3389/fcvm.2020.602088] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is a critical regulator of systemic metabolism and bodily homeostasis as it secretes a myriad of adipokines, including inflammatory and anti-inflammatory cytokines. As the main storage pool of lipids, subcutaneous and visceral adipose tissues undergo marked hypertrophy and hyperplasia in response to nutritional excess leading to hypoxia, adipokine dysregulation, and subsequent low-grade inflammation that is characterized by increased infiltration and activation of innate and adaptive immune cells. The specific localization, physiology, susceptibility to inflammation and the heterogeneity of the inflammatory cell population of each adipose depot are unique and thus dictate the possible complications of adipose tissue chronic inflammation. Several lines of evidence link visceral and particularly perivascular, pericardial, and perirenal adipose tissue inflammation to the development of metabolic syndrome, insulin resistance, type 2 diabetes and cardiovascular diseases. In addition to the implication of the immune system in the regulation of adipose tissue function, adipose tissue immune components are pivotal in detrimental or otherwise favorable adipose tissue remodeling and thermogenesis. Adipose tissue resident and infiltrating immune cells undergo metabolic and morphological adaptation based on the systemic energy status and thus a better comprehension of the metabolic regulation of immune cells in adipose tissues is pivotal to address complications of chronic adipose tissue inflammation. In this review, we discuss the role of adipose innate and adaptive immune cells across various physiological and pathophysiological states that pertain to the development or progression of cardiovascular diseases associated with metabolic disorders. Understanding such mechanisms allows for the exploitation of the adipose tissue-immune system crosstalk, exploring how the adipose immune system might be targeted as a strategy to treat cardiovascular derangements associated with metabolic dysfunctions.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Safaa H. Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | - Houssam Al-Koussa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Alaa Ghazi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
40
|
Xiao Y, Liu D, Cline MA, Gilbert ER. Chronic stress, epigenetics, and adipose tissue metabolism in the obese state. Nutr Metab (Lond) 2020; 17:88. [PMID: 33088334 PMCID: PMC7574417 DOI: 10.1186/s12986-020-00513-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
In obesity, endocrine and metabolic perturbations, including those induced by chronic activation of the hypothalamus-pituitary-adrenal axis, are associated with the accumulation of adipose tissue and inflammation. Such changes are attributable to a combination of genetic and epigenetic factors that are influenced by the environment and exacerbated by chronic activation of the hypothalamus-pituitary-adrenal axis. Stress exposure at different life stages can alter adipose tissue metabolism directly through epigenetic modification or indirectly through the manipulation of hypothalamic appetite regulation, and thereby contribute to endocrine changes that further disrupt whole-body energy balance. This review synthesizes current knowledge, with an emphasis on human clinical trials, to describe metabolic changes in adipose tissue and associated endocrine, genetic and epigenetic changes in the obese state. In particular, we discuss epigenetic changes induced by stress exposure and their contribution to appetite and adipocyte dysfunction, which collectively promote the pathogenesis of obesity. Such knowledge is critical for providing future directions of metabolism research and targets for treating metabolic disorders.
Collapse
Affiliation(s)
- Yang Xiao
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Dongmin Liu
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Mark A Cline
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA USA.,School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Elizabeth R Gilbert
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA USA.,School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| |
Collapse
|
41
|
Lynch GM, Murphy CH, Castro EDM, Roche HM. Inflammation and metabolism: the role of adiposity in sarcopenic obesity. Proc Nutr Soc 2020; 79:1-13. [PMID: 32669148 DOI: 10.1017/s0029665120007119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sarcopenic obesity is characterised by the double burden of diminished skeletal muscle mass and the presence of excess adiposity. From a mechanistic perspective, both obesity and sarcopenia are associated with sub-acute, chronic pro-inflammatory states that impede metabolic processes, disrupting adipose and skeletal functionality, which may potentiate disease. Recent evidence suggests that there is an important cross-talk between metabolism and inflammation, which has shifted focus upon metabolic-inflammation as a key emerging biological interaction. Dietary intake, physical activity and nutritional status are important environmental factors that may modulate metabolic-inflammation. This paradigm will be discussed within the context of sarcopenic obesity risk. There is a paucity of data in relation to the nature and the extent to which nutritional status affects metabolic-inflammation in sarcopenic obesity. Research suggests that there may be scope for the modulation of sarcopenic obesity with alterations in diet. The potential impact of increasing protein consumption and reconfiguration of dietary fat composition in human dietary interventions are evaluated. This review will explore emerging data with respect to if and how different dietary components may modulate metabolic-inflammation, particularly with respect to adiposity, within the context of sarcopenic obesity.
Collapse
Affiliation(s)
- G M Lynch
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - C H Murphy
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - E de Marco Castro
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - H M Roche
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
- Institute for Global Food Security, Queen's University Belfast, Belfast, UK
| |
Collapse
|
42
|
Valsesia A, Chakrabarti A, Hager J, Langin D, Saris WHM, Astrup A, Blaak EE, Viguerie N, Masoodi M. Integrative phenotyping of glycemic responders upon clinical weight loss using multi-omics. Sci Rep 2020; 10:9236. [PMID: 32514005 PMCID: PMC7280519 DOI: 10.1038/s41598-020-65936-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
Weight loss aims to improve glycemic control in obese but strong variability is observed. Using a multi-omics approach, we investigated differences between 174 responders and 201 non-responders, that had lost >8% body weight following a low-caloric diet (LCD, 800 kcal/d for 8 weeks). The two groups were comparable at baseline for body composition, glycemic control, adipose tissue transcriptomics and plasma ketone bodies. But they differed significantly in their response to LCD, including improvements in visceral fat, overall insulin resistance (IR) and tissue-specific IR. Transcriptomics analyses found down-regulation in key lipogenic genes (e.g. SCD, ELOVL5) in responders relative to non-responders; metabolomics showed increase in ketone bodies; while proteomics revealed differences in lipoproteins. Findings were consistent between genders; with women displaying smaller improvements owing to a better baseline metabolic condition. Integrative analyses identified a plasma omics model that was able to predict non-responders with strong performance (on a testing dataset, the Receiving Operating Curve Area Under the Curve (ROC AUC) was 75% with 95% Confidence Intervals (CI) [67%, 83%]). This model was based on baseline parameters without the need for intrusive measurements and outperformed clinical models (p = 0.00075, with a +14% difference on the ROC AUCs). Our approach document differences between responders and non-responders, with strong contributions from liver and adipose tissues. Differences may be due to de novo lipogenesis, keto-metabolism and lipoprotein metabolism. These findings are useful for clinical practice to better characterize non-responders both prior and during weight loss.
Collapse
Affiliation(s)
| | | | - Jörg Hager
- Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Dominique Langin
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,University of Toulouse, Paul Sabatier University, Toulouse, France.,Toulouse University Hospitals, Laboratory of Clinical Biochemistry, Toulouse, France
| | - Wim H M Saris
- Department of Human Biology, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+(MUMC+), Maastricht, The Netherlands
| | - Arne Astrup
- University of Copenhagen, Department of Nutrition, Exercise and Sports, Faculty of Science, Copenhagen, Denmark
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+(MUMC+), Maastricht, The Netherlands
| | - Nathalie Viguerie
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Mojgan Masoodi
- Nestlé Institute of Health Sciences, Lausanne, Switzerland. .,Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland.
| |
Collapse
|
43
|
Abstract
Lay Summary
A person’s diet may impact inflammatory bowel disease (IBD) risk. IBD is an inflammatory condition. We explore how certain foods may trigger, or indeed attenuate, inflammation in some IBD patients, but not others. Greater knowledge is needed underpinning personalized nutrition within effective medical management.
Collapse
Affiliation(s)
- Colm B Collins
- UCD School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Helen M Roche
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland,Institute for Global Food Security, Queen’s University Belfast, Northern Ireland, UK,Address correspondence to: Helen M. Roche, PhD, UCD Conway Institute and School of Public Health, Physiotherapy and Sports Science, University College Dublin, Belfield, Dublin 4, Ireland ()
| |
Collapse
|
44
|
Dalle S, Koppo K. Is inflammatory signaling involved in disease-related muscle wasting? Evidence from osteoarthritis, chronic obstructive pulmonary disease and type II diabetes. Exp Gerontol 2020; 137:110964. [PMID: 32407865 DOI: 10.1016/j.exger.2020.110964] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/15/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022]
Abstract
Muscle loss is an important feature that occurs in multiple pathologies including osteoarthritis (OA), chronic obstructive pulmonary disease (COPD) and type II diabetes (T2D). Despite differences in pathogenesis and disease-related complications, there are reasons to believe that some fundamental underlying mechanisms are inherent to the muscle wasting process, irrespective of the pathology. Recent evidence shows that inflammation, either local or systemic, contributes to the modulation of muscle mass and/or muscle strength, via an altered molecular profile in muscle tissue. However, it remains ambiguous to which extent and via which mechanisms inflammatory signaling affects muscle mass in disease. Therefore, the objective of the present review is to discuss the role of inflammation on skeletal muscle anabolism, catabolism and functionality in three pathologies that are characterized by an eventual loss in muscle mass (and muscle strength), i.e. OA, COPD and T2D. In OA and COPD, most rodent models confirmed that systemic (COPD) or muscle (OA) inflammation directly induces muscle loss or muscle dysfunctionality. However, in a patient population, the association between inflammation and muscular maladaptations are more ambiguous. For example, in T2D patients, systemic inflammation is associated with muscle loss whereas in OA patients this link has not consistently been established. T2D rodent models revealed that increased levels of advanced glycation end-products (AGEs) and a decreased mTORC1 activation play a key role in muscle atrophy, but it remains to be elucidated whether AGEs and mTORC1 are interconnected and contribute to muscle loss in T2D patients. Generally, if any, associations between inflammation and muscle are mainly based on observational and cross-sectional data. There is definitely a need for longitudinal evidence through well-powered randomized control trials that take into account confounders such as age, disease-phenotypes, comorbidities, physical (in) activity etc. This will allow to improve our understanding of the complex interaction between inflammatory signaling and muscle mass loss and hence contribute to the development of therapeutic strategies to combat muscle wasting in these diseases.
Collapse
Affiliation(s)
- Sebastiaan Dalle
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium
| | - Katrien Koppo
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium.
| |
Collapse
|
45
|
Current metabolic perspective on malnutrition in obesity: towards more subgroup-based nutritional approaches? Proc Nutr Soc 2020; 79:331-337. [PMID: 32122428 PMCID: PMC7663313 DOI: 10.1017/s0029665120000117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lifestyle intervention may be effective in reducing type 2 diabetes mellitus incidence and cardiometabolic risk. A more personalised nutritional approach based on an individual or subgroup-based metabolic profile may optimise intervention outcome. Whole body insulin resistance (IR) reflects defective insulin action in tissues such as muscle, liver, adipose tissue, gut and brain, which may precede the development of cardiometabolic diseases. IR may develop in different organs but the severity may vary between organs. Individuals with more pronounced hepatic IR have a distinct plasma metabolome and lipidome profile as compared with individuals with more pronounced muscle IR. Additionally, genes related to extracellular modelling were upregulated in abdominal subcutaneous adipose tissue in individuals with more pronounced hepatic IR, whilst genes related to inflammation as well as systemic low-grade inflammation were upregulated in individuals with primarily muscle IR. There are indications that these distinct IR phenotypes may also respond differentially to dietary macronutrient composition. Besides metabolic phenotype, microbial phenotype may be of importance in personalising the response to diet. In particular fibres or fibre mixtures, leading to a high distal acetate and SCFA production may have more pronounced effects on metabolic health. Notably, individuals with prediabetes may have a reduced response to diet-induced microbiota modulation with respect to host insulin sensitivity and metabolic health outcomes. Overall, we need more research to relate metabolic subphenotypes to intervention outcomes to define more optimal diets for individuals with or predisposed to chronic metabolic diseases.
Collapse
|