1
|
Lecoutre S, Rebière C, Maqdasy S, Lambert M, Dussaud S, Abatan JB, Dugail I, Gautier EL, Clément K, Marcelin G. Enhancing adipose tissue plasticity: progenitor cell roles in metabolic health. Nat Rev Endocrinol 2025; 21:272-288. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/07/2025]
Abstract
Adipose tissue demonstrates considerable plasticity and heterogeneity, enabling metabolic, cellular and structural adaptations to environmental signals. This adaptability is key for maintaining metabolic homeostasis. Impaired adipose tissue plasticity can lead to abnormal adipose tissue responses to metabolic cues, which contributes to the development of cardiometabolic diseases. In chronic obesity, white adipose tissue undergoes pathological remodelling marked by adipocyte hypertrophy, chronic inflammation and fibrosis, which are linked to local and systemic insulin resistance. Research data suggest that the capacity for healthy or unhealthy white adipose tissue remodelling might depend on the intrinsic diversity of adipose progenitor cells (APCs), which sense and respond to metabolic cues. This Review highlights studies on APCs as key determinants of adipose tissue plasticity, discussing differences between subcutaneous and visceral adipose tissue depots during development, growth and obesity. Modulating APC functions could improve strategies for treating adipose tissue dysfunction and metabolic diseases in obesity.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| | - Clémentine Rebière
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine, Karolinska Institutet Hospital, Stockholm, Sweden
| | - Mélanie Lambert
- Institut National de la Santé et de la Recherche Médicale, Bobigny, France
- Labex Inflamex, Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Bobigny, France
| | - Sébastien Dussaud
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Emmanuel L Gautier
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
- Department of Nutrition, Pitie-Salpêtriere Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| |
Collapse
|
2
|
Malin SK, Remchak ME, Heiston EM, Fabris C, Shah AM. Pancreatic β-cell Function is Higher in Morning Versus Intermediate Chronotypes With Obesity. Obes Sci Pract 2025; 11:e70064. [PMID: 40018087 PMCID: PMC11864105 DOI: 10.1002/osp4.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 03/01/2025] Open
Abstract
Objectives People with later chronotypes are at greater T2D risk, yet it is unknown if β-cell function differs among chronotypes. Thus we, assessed β-cell function in morning (MORN) and intermediate (INT) chronotypes with obesity. Methods Adults (n = 41, 9M, 55 ± 1.7 y, 36.8 ± 1.0 kg/m2) were grouped as MORN or INT per the Morningness-Eveningness Questionnaire. Glucose, insulin, C-peptide, GIP, and GLP-1(active) were collected every 30 min during a 120 min 75g-OGTT. Insulin secretion rates (ISR) were calculated (regularized deconvolution) to assess early (total area under the curve; tAUC0-30min) and total-phase (tAUC0-120min) glucose-stimulated insulin secretion (GSIS:ISR/Glucose). Skeletal muscle (glucose infusion rate/steady-state insulin) insulin sensitivity and hepatic (HOMA-IR) as well as adipose (Adipose-IR) insulin resistance were assessed during a 120 min euglycemic hyperinsulinemic clamp (40mU/m2/min, 90 mg/dL). β-cell function (disposition index (DI): GSIS adjusted insulin sensitivity) was determined. Body composition (DXA) and fitness (VO2max) were also measured. Results Age, body composition and VO2max were similar between groups, but INT had reduced muscle insulin sensitivity and higher hepatic and adipose IR (p < 0.05). INT had higher C-peptide tAUC0-30min (p = 0.04) and lower hepatic DI (tAUC0-30min p = 0.05 and tAUC0-120min p = 0.07, respectively). Early phase hepatic DI correlated with GLP-1 tAUC0-30min (r = 0.35, p < 0.02) and tAUC0-120min (r = -0.40, p = 0.04). Conclusions β-cell function was higher in MORN versus INT chronotypes. Further work is warranted to discern how chronotype impacts insulin secretion. Trial Registration NCT03355469.
Collapse
Affiliation(s)
- Steven K. Malin
- Department of Kinesiology & HealthRutgers UniversityNew BrunswickNew JerseyUSA
- Division of Endocrinology, Metabolism & NutritionRutgers UniversityNew BrunswickNew JerseyUSA
- New Jersey Institute for FoodNutrition and HealthRutgers UniversityNew BrunswickNew JerseyUSA
- Institute of Translational Medicine and ScienceRutgers UniversityNew BrunswickNew JerseyUSA
| | | | - Emily M. Heiston
- Department of Kinesiology & HealthRutgers UniversityNew BrunswickNew JerseyUSA
| | - Chiara Fabris
- Center for Diabetes TechnologySchool of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Ankit M. Shah
- Division of Endocrinology, Metabolism & NutritionRutgers UniversityNew BrunswickNew JerseyUSA
| |
Collapse
|
3
|
Jahan I, Shuvo AUH, Alimullah M, Rahman ASMN, Siddiqua S, Rafia S, Khan F, Ahmed KS, Hossain H, Akramuddaula K, Alam MA, Subhan N. Purple potato extract modulates fat metabolizing genes expression, prevents oxidative stress, hepatic steatosis, and attenuates high-fat diet-induced obesity in male rats. PLoS One 2025; 20:e0318162. [PMID: 40168333 PMCID: PMC11960900 DOI: 10.1371/journal.pone.0318162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 01/03/2025] [Indexed: 04/03/2025] Open
Abstract
OBJECTIVE In this investigation, the significance of purple potato (Solanum tuberosum L.) extract treatment was assessed against oxidative stress and fat metabolizing transcription factors in the liver of high-fat (HF) diet-fed rats. METHODS Wistar (male) rats were arranged into several groups and provided with a control and HF diet along with the purple potato extract. Body weights, oral glucose tolerance test (OGTT), insulin, plasma lipids, and oxidative stress-related indicators were analyzed in plasma and tissue samples. Additionally, real-time PCR was performed to evaluate the gene expression for oxidative stress and fat metabolism in the liver. Histological staining was also performed on pancreatic and hepatic tissues. RESULTS Purple potato extract lowered body weights and improved glucose utilization in the OGTT test in HF diet-fed rats. Purple potato extract also suppressed HF-diet-induced oxidative stress in plasma and hepatic tissues. Purple potato extract also restored the Nrf-2 expression in the liver, followed by the improved expression of HO-1, HO-2, and other antioxidant genes in HF diet-fed rats. In addition, genes involved in lipid metabolism were also positively modulated due to purple potato extract treatment. Furthermore, histological examination revealed the reduction of lipid accumulation and amelioration of inflammation due to the consumption of purple potato extract. CONCLUSION This investigation revealed that antioxidant-rich purple potato extract can modulate the antioxidant and fat metabolizing genes expression, ameliorated oxidative stress and glucose intolerance as well as lowered blood lipids in male rats.
Collapse
Affiliation(s)
- Ishrat Jahan
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Asif Ul Haque Shuvo
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Mirza Alimullah
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | | | | | - Shatil Rafia
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Ferdous Khan
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Khondoker Shahin Ahmed
- Chemical Research Division, BCSIR Laboratories, Dhaka, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Hemayet Hossain
- Chemical Research Division, BCSIR Laboratories, Dhaka, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | | | - Md Ashraful Alam
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | - Nusrat Subhan
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| |
Collapse
|
4
|
Andrade N, Rodrigues I, Carmo F, Campanher G, Bracchi I, Lopes J, Patrício E, Guimarães JT, Barreto-Peixoto JA, Costa ASG, Santo LE, Machado M, Soares TF, Machado S, Oliveira MBPP, Alves RC, Martel F, Silva C. Sustainable Utilization of Coffee Pulp, a By-Product of Coffee Production: Effects on Metabolic Syndrome in Fructose-Fed Rats. Antioxidants (Basel) 2025; 14:266. [PMID: 40227203 PMCID: PMC11939298 DOI: 10.3390/antiox14030266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 04/15/2025] Open
Abstract
Metabolic syndrome (MetS) is a cluster of metabolic abnormalities that include insulin resistance, impaired glucose tolerance, dyslipidemia, hypertension, and abdominal obesity. Coffee production generates large quantities of waste products, which pose a serious threat to the environment. However, coffee by-products, such as coffee pulp (CP), possess an undeniable wealth of bioactive components. Based on this, we investigated whether a 10-week dietary intervention with 250 mg/kg/d of CP could prevent or ameliorate MetS in high-fructose-fed rats. Consumption of CP by rats fed a high-fructose diet reduced body weight gain, lowered systolic blood pressure (SBP), fasting plasma glucose and insulin levels, and improved insulin resistance compared to rats fed a high-fructose diet alone. At the hepatic level, CP attenuated the increase in lipid storage, reduced lipid peroxidation, and improved glutathione levels when combined with a high-fructose diet. CP also affected the expression of key genes related to glucose and lipid metabolism in hepatic and adipose tissues, in rats fed a fructose-rich diet. This study demonstrates that CP ameliorates several consequences of high-fructose-induced MetS in the rat (weight gain, hypertension, glucose intolerance, insulin resistance, changes in liver, and adipose tissue function). Hence, our data provide evidence that CP consumption in the context of a high-fructose diet can be used to improve MetS management.
Collapse
Affiliation(s)
- Nelson Andrade
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-465 Porto, Portugal; (I.R.); (F.C.); (G.C.); (I.B.); (J.T.G.)
| | - Ilda Rodrigues
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-465 Porto, Portugal; (I.R.); (F.C.); (G.C.); (I.B.); (J.T.G.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-465 Porto, Portugal
| | - Francisca Carmo
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-465 Porto, Portugal; (I.R.); (F.C.); (G.C.); (I.B.); (J.T.G.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-465 Porto, Portugal
| | - Gabriela Campanher
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-465 Porto, Portugal; (I.R.); (F.C.); (G.C.); (I.B.); (J.T.G.)
- School of Medical Sciences, University of Örebro, Campus USÖ, S-701 82 Örebro, Sweden
| | - Isabella Bracchi
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-465 Porto, Portugal; (I.R.); (F.C.); (G.C.); (I.B.); (J.T.G.)
| | - Joanne Lopes
- Department of Pathology, São João Hospital Centre, 4200-319 Porto, Portugal; (J.L.); (E.P.)
| | - Emília Patrício
- Department of Pathology, São João Hospital Centre, 4200-319 Porto, Portugal; (J.L.); (E.P.)
| | - João T. Guimarães
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-465 Porto, Portugal; (I.R.); (F.C.); (G.C.); (I.B.); (J.T.G.)
- Department of Clinical Pathology, São João Hospital Centre, 4200-319 Porto, Portugal
| | - Juliana A. Barreto-Peixoto
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
| | - Anabela S. G. Costa
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
| | - Liliana Espírito Santo
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
| | - Marlene Machado
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
| | - Thiago F. Soares
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
| | - Susana Machado
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
| | - Maria Beatriz P. P. Oliveira
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
| | - Rita C. Alves
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-465 Porto, Portugal; (I.R.); (F.C.); (G.C.); (I.B.); (J.T.G.)
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-465 Porto, Portugal
| | - Cláudia Silva
- Laboratório Associado para a Química Verde—Tecnologias e Processos Limpos (REQUIMTE/LAQV), Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (N.A.); (J.A.B.-P.); (A.S.G.C.); (L.E.S.); (M.M.); (T.F.S.); (S.M.); (M.B.P.P.O.); (R.C.A.); (C.S.)
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-465 Porto, Portugal; (I.R.); (F.C.); (G.C.); (I.B.); (J.T.G.)
| |
Collapse
|
5
|
Sandforth L, Kullmann S, Sandforth A, Fritsche A, Jumpertz-von Schwartzenberg R, Stefan N, Birkenfeld AL. Prediabetes remission to reduce the global burden of type 2 diabetes. Trends Endocrinol Metab 2025:S1043-2760(25)00004-9. [PMID: 39955249 DOI: 10.1016/j.tem.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/12/2024] [Accepted: 01/15/2025] [Indexed: 02/17/2025]
Abstract
Prediabetes is a highly prevalent and increasingly common condition affecting a significant proportion of the global population. The heterogeneous nature of prediabetes presents a challenge in identifying individuals who particularly benefit from lifestyle or other therapeutic interventions aiming at preventing type 2 diabetes (T2D) and associated comorbidities. The phenotypic characteristics of individuals at risk for diabetes are associated with both specific risk profiles for progression and a differential potential to facilitate prediabetes remission and reduce the risk of future T2D. This review examines the current definition and global prevalence of prediabetes and evaluates the potential of prediabetes remission to reduce the alarming increase in the global burden of T2D.
Collapse
Affiliation(s)
- Leontine Sandforth
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Arvid Sandforth
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Reiner Jumpertz-von Schwartzenberg
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany; M3 Research Center, Malignom, Metabolome, Microbiome, 72076 Tübingen, Germany; Cluster of Excellence EXC 2124 'Controlling Microbes to Fight Infections' (CMFI), University of Tübingen, Tübingen, Germany
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany; Department of Diabetes, Life Sciences, and Medicine, Cardiovascular Medicine and Life Sciences, King's College London, London, UK.
| |
Collapse
|
6
|
Klein H, Zelicha H, Yaskolka Meir A, Rinott E, Tsaban G, Kaplan A, Chassidim Y, Gepner Y, Blüher M, Ceglarek U, Isermann B, Stumvoll M, Shelef I, Qi L, Li J, Hu FB, Stampfer MJ, Shai I. Visceral adipose tissue area and proportion provide distinct reflections of cardiometabolic outcomes in weight loss; pooled analysis of MRI-assessed CENTRAL and DIRECT PLUS dietary randomized controlled trials. BMC Med 2025; 23:57. [PMID: 39901232 PMCID: PMC11792534 DOI: 10.1186/s12916-025-03891-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 01/22/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Visceral adipose tissue (VAT) is well established as a pathogenic fat depot, whereas superficial subcutaneous adipose tissue (SAT) is associated with either an improved or neutral cardiovascular state. However, it is unclear to what extent VAT area (VATcm2) and its proportion of total abdominal adipose tissue (VAT%) are distinguished in predicting cardiometabolic status and clinical outcomes during weight loss. METHODS We integrated magnetic resonance imaging (MRI) measurements of VAT, deep-SAT, and superficial-SAT from two 18-month lifestyle weight loss clinical trials, CENTRAL and DIRECT PLUS (n = 572). RESULTS At baseline, the mean VATcm2 was 144.8cm2 and VAT% = 28.2%; over 18 months, participants lost 28cm2 VATcm2 (- 22.5%), and 1.3 VAT% units. Baseline VATcm2 and VAT% were similarly associated with metabolic syndrome, hypertension, and diabetes status, while VAT% better classified hypertriglyceridemia. Conversely, higher VATcm2 was associated with elevated high-sensitivity C-reactive protein (hsCRP), while VAT% was not. After 18 months of lifestyle intervention, both VATcm2 and VAT% loss were significantly associated with decreased triglycerides, HbA1c, ferritin, and liver enzymes, and increased HDL-c levels beyond weight loss (FDR < 0.05). Only VATcm2 loss was correlated with decreased HOMA-IR, chemerin, and leptin levels. CONCLUSIONS MRI follow-up of 572 participants over 18 months of weight loss intervention suggests that although increased VATcm2 and VAT% exhibit similar clinical manifestations, it might be preferable to examine VAT% when exploring lipid status, while VATcm2 may better reflect inflammatory and glycemic states. TRIAL REGISTRATION CENTRAL (Clinical-trials-identifier: NCT01530724); DIRECT PLUS (Clinical-trials-identifier: NCT03020186).
Collapse
Affiliation(s)
- Hadar Klein
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Hila Zelicha
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Anat Yaskolka Meir
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Ehud Rinott
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Gal Tsaban
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Alon Kaplan
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Yoash Chassidim
- Department of Engineering, Sapir Academic College, Shaar Hanegev, Israel
| | - Yftach Gepner
- Department of Health Promotion, School of Public Health, Faculty of Medicine and Sylvan Adams Sports Institute, Tel-Aviv University, Tel-Aviv, Israel
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | | | - Ilan Shelef
- Soroka University Medical Center, Beer-Sheva, Israel
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jun Li
- Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Meir J Stampfer
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Iris Shai
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
- Department of Medicine, University of Leipzig, Leipzig, Germany.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
7
|
Yildiz R, Ganbold K, Sparman NZR, Rajbhandari P. Immune Regulatory Crosstalk in Adipose Tissue Thermogenesis. Compr Physiol 2025; 15:e70001. [PMID: 39921241 DOI: 10.1002/cph4.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Brown adipose tissue (BAT) and thermogenic beige fat within white adipose tissue (WAT), collectively known as adaptive thermogenic fat, dissipate energy as heat, offering promising therapeutic potential to combat obesity and metabolic disorders. The specific biological functions of these fat depots are determined by their unique interaction with the microenvironments, composed of immune cells, endothelial cells, pericytes, and nerve fibers. Immune cells residing in these depots play a key role in regulating energy expenditure and systemic energy homeostasis. The dynamic microenvironment of thermogenic fat depots is essential for maintaining tissue health and function. Immune cells infiltrate both BAT and beige WAT, contributing to their homeostasis and activation through intricate cellular communications. Emerging evidence underscores the importance of various immune cell populations in regulating thermogenic adipose tissue, though many remain undercharacterized. This review provides a comprehensive overview of the immune cells that regulate adaptive thermogenesis and their complex interactions within the adipose niche, highlighting their potential to influence metabolic health and contribute to therapeutic interventions for obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Ramazan Yildiz
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Khatanzul Ganbold
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Njeri Z R Sparman
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
8
|
Michelsen TM, Skytte HN, Gunnes N, Holven KB, Christensen JJ, Roland MCP. Metabolic profiles in early pregnancy associated with metabolic pregnancy complications in women with obesity. J Reprod Immunol 2024; 166:104397. [PMID: 39577057 DOI: 10.1016/j.jri.2024.104397] [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: 06/25/2024] [Revised: 11/07/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
INTRODUCTION Higher maternal body mass index (BMI) is associated with metabolic disturbances and pregnancy complications. We aimed to examine whether metabolic profiles in early pregnancy were associated with metabolic pregnancy complications in women with obesity (BMI ≥ 30 kg/m2). MATERIAL AND METHODS Nested cohort study from a prospective longitudinal cohort (n = 1031) of women who were healthy prior to pregnancy and gave birth at Oslo University Hospital from 2002-2008. The sample comprised 81 women with obesity. Metabolic pregnancy complications included gestational diabetes mellitus, gestational hypertension and preeclampsia. In plasma samples from gestational weeks 14-16, 91 metabolites were analyzed by nuclear magnetic resonance spectroscopy. We performed a principal component analysis to reduce the metabolic dimensions. Logistic regression models were fitted to estimate crude and adjusted odds ratios (ORs) of metabolic pregnancy complications. RESULTS Twenty-four out of 81 women developed metabolic pregnancy complications (gestational hypertension, preeclampsia, and/or gestational diabetes). Two of five principal components (80 % explained variance) were significantly associated with metabolic pregnancy complications. The ratio of monounsaturated to total fatty acids increased the risk of metabolic pregnancy complications (OR 2.09, 95 % confidence interval [CI] 1.25-3.75), while the ratio of polyunsaturated to monounsaturated fatty acids decreased the risk (OR 0.54, 95 % CI 0.30-0.89). The ratio of omega-3 to total fatty acids (OR 0.59, 95 % CI 0.34-0.98) and the ratio of docosahexaenoic acid to total fatty acids (OR 0.57, 95 % CI 0.31-0.97) also decreased the risk of metabolic pregnancy complications. CONCLUSION Metabolic profile in early pregnancy was associated with risk of metabolic pregnancy complications in women with obesity. We observed the strongest associations between fatty acid composition and metabolic pregnancy complications.
Collapse
Affiliation(s)
- Trond Melbye Michelsen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Hege Nyhus Skytte
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Norwegian Research Centre for Women's Health, Oslo University Hospital, Oslo, Norway
| | - Nina Gunnes
- Norwegian Research Centre for Women's Health, Oslo University Hospital, Oslo, Norway
| | - Kirsten Bjørklund Holven
- Department of Nutrition, University of Oslo, Oslo, Norway; Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Oslo, Norway
| | | | - Marie Cecilie Paasche Roland
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, Norway; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
9
|
Weaver KD, Simon L, Molina PE, Souza-Smith F. The Role of Lymph-Adipose Crosstalk in Alcohol-Induced Perilymphatic Adipose Tissue Dysfunction. Int J Mol Sci 2024; 25:10811. [PMID: 39409139 PMCID: PMC11482484 DOI: 10.3390/ijms251910811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
Chronic alcohol use leads to metabolic dysfunction in adipose tissue. The underlying mechanisms and the contribution of alcohol-induced adipose tissue dysfunction to systemic metabolic dysregulation are not well understood. In our previous studies, we found that chronic alcohol feeding induces mesenteric lymphatic leakage, perilymphatic adipose tissue (PLAT) inflammation, and local insulin resistance in rats. The goal of this study was to further explore the link between alcohol-induced lymphatic leakage and PLAT immunometabolic dysregulation, locally and systemically, using in vivo and ex vivo approaches. Male rats received a Lieber-DeCarli liquid diet, of which 36% of the calories were from alcohol, for 10 weeks. Time-matched control animals were pair-fed. Adipokine levels were measured in PLAT, subcutaneous fat, plasma, and mesenteric lymph samples. Glucose tolerance was assessed after 10 weeks. Further, we used a novel ex vivo lymph-stimulated naïve PLAT explant approach to modeling lymph leakage to assess changes in adipokine secretion and expression of proinflammatory markers after stimulation with lymph from alcohol- or pair-fed animals. Our data show that chronic alcohol-fed rats presented PLAT-specific decreases in adiponectin and leptin levels, alterations in the expression of genes involved in lipid metabolic pathways, and associated impaired whole-body glucose homeostasis. Further, we found that direct naïve PLAT stimulation with lymph contents from alcohol-fed animals increased IL-6 expression in demonstrating the ability of lymph contents to differentially impact naïve adipose tissue. Overall, chronic alcohol feeding leads to depot-specific alterations in metabolic profile, impaired systemic glucose tolerance, and lymph-induced adipose tissue inflammation. The specific lymph components leading to PLAT immunometabolic dysregulation remain to be determined.
Collapse
Affiliation(s)
| | | | | | - Flavia Souza-Smith
- Department of Physiology, Louisiana State University Health Sciences Center-New Orleans, New Orleans, LA 70112, USA; (K.D.W.); (L.S.); (P.E.M.)
| |
Collapse
|
10
|
Odimegwu CL, Uwaezuoke SN, Chikani UN, Mbanefo NR, Adiele KD, Nwolisa CE, Eneh CI, Ndiokwelu CO, Okpala SC, Ogbuka FN, Odo KE, Ohuche IO, Obiora-Izuka CE. Targeting the Epigenetic Marks in Type 2 Diabetes Mellitus: Will Epigenetic Therapy Be a Valuable Adjunct to Pharmacotherapy? Diabetes Metab Syndr Obes 2024; 17:3557-3576. [PMID: 39323929 PMCID: PMC11423826 DOI: 10.2147/dmso.s479077] [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] [Received: 05/20/2024] [Accepted: 08/03/2024] [Indexed: 09/27/2024] Open
Abstract
Although genetic, environmental, and lifestyle factors largely contribute to type 2 diabetes mellitus (T2DM) risk, the role of epigenetics in its pathogenesis is now well established. The epigenetic mechanisms in T2DM mainly consist of DNA methylation, histone modifications and regulation by noncoding RNAs (ncRNAs). For instance, DNA methylation at CpG islands in the promoter regions of specific genes encoding insulin signaling and glucose metabolism suppresses these genes. Modulating the enzyme mediators of these epigenetic marks aims to restore standard gene expression patterns and improve glycemic control. In targeting these epigenetic marks, using epigenetic drugs such as DNA methyltransferase (DNAMT), histone deacetylase (HDAC) and histone acetyltransferase (HAT) inhibitors has led to variable success in humans and experimental murine models. Specifically, the United States' Food and Drug Administration (US FDA) has approved DNAMT inhibitors like 5-azacytidine and 5-aza-2'-deoxycytidine for use in diabetic retinopathy: a T2DM microvascular complication. These DNAMT inhibitors block the genes for methylation of mitochondrial superoxide dismutase 2 (SOD2) and matrix metallopeptidase 9 (MMP-9): the epigenetic marks in diabetic retinopathy. Traditional pharmacotherapy with metformin also have epigenetic effects in T2DM and positively alter disease outcomes when combined with epigenetic drugs like DNAMT and HDAC inhibitors, raising the prospect of using epigenetic therapy as a valuable adjunct to pharmacotherapy. However, introducing small interfering RNAs (siRNAs) in cells to silence specific target genes remains in the exploratory phase. Future research should focus on regulating gene expression in T2DM using long noncoding RNA (lncRNA) molecules, another type of ncRNA. This review discusses the epigenetics of T2DM and that of its macro- and microvascular complications, and the potential benefits of combining epigenetic therapy with pharmacotherapy for optimal results.
Collapse
Affiliation(s)
- Chioma Laura Odimegwu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Samuel Nkachukwu Uwaezuoke
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ugo N Chikani
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ngozi Rita Mbanefo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ken Daberechi Adiele
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | - Chizoma Ihuarula Eneh
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Chibuzo Obiora Ndiokwelu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Somkenechi C Okpala
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Francis N Ogbuka
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Kenneth E Odo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | | |
Collapse
|
11
|
Ahmed M. A clinician's perspective on the new organ mesentery and non-vascular mesenteropathies. Front Physiol 2024; 15:1336908. [PMID: 39296517 PMCID: PMC11408482 DOI: 10.3389/fphys.2024.1336908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 07/17/2024] [Indexed: 09/21/2024] Open
Abstract
Mesentery was discovered as a new organ in 2017. It is a continuous membranous tissue from the duodenojejunal flexure to the anorectal junction. It has distinct anatomy, physiology, and disease states. Primary mesenteropathies include vascular and non-vascular diseases. Some of them are common, and some of them are rarely seen in clinical practice. Secondary mesenteropathies occur when infection or malignancy in another organ spreads to the mesentery. Each entity has specific diagnostic and treatment protocols. Increased awareness of different mesenteropathies and an understanding of their various presentations at different stages of life can help in early diagnosis and improved clinical outcomes.
Collapse
|
12
|
Liu J, Li L, Xu D, Li Y, Chen T, Liu Y, Bao Y, Wang Y, Yang L, Li P, Xu L. Rab18 maintains homeostasis of subcutaneous adipose tissue to prevent obesity-induced metabolic disorders. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1170-1182. [PMID: 38523235 DOI: 10.1007/s11427-023-2367-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/15/2023] [Indexed: 03/26/2024]
Abstract
Metabolically healthy obesity refers to obese individuals who do not develop metabolic disorders. These people store fat in subcutaneous adipose tissue (SAT) rather than in visceral adipose tissue (VAT). However, the molecules participating in this specific scenario remain elusive. Rab18, a lipid droplet (LD)-associated protein, mediates the contact between the endoplasmic reticulum (ER) and LDs to facilitate LD growth and maturation. In the present study, we show that the protein level of Rab18 is specifically upregulated in the SAT of obese people and mice. Rab18 adipocyte-specific knockout (Rab18 AKO) mice had a decreased volume ratio of SAT to VAT compared with wildtype mice. When subjected to high-fat diet (HFD), Rab18 AKO mice had increased ER stress and inflammation, reduced adiponectin, and decreased triacylglycerol (TAG) accumulation in SAT. In contrast, TAG accumulation in VAT, brown adipose tissue (BAT) or liver of Rab18 AKO mice had a moderate increase without ER stress stimulation. Rab18 AKO mice developed insulin resistance and systematic inflammation. Rab18 AKO mice maintained body temperature in response to acute and chronic cold induction with a thermogenic SAT, similar to the counterpart mice. Furthermore, Rab18-deficient 3T3-L1 adipocytes were more prone to palmitate-induced ER stress, indicating the involvement of Rab18 in alleviating lipid toxicity. Rab18 AKO mice provide a good animal model to investigate metabolic disorders such as impaired SAT. In conclusion, our studies reveal that Rab18 is a key and specific regulator that maintains the proper functions of SAT by alleviating lipid-induced ER stress.
Collapse
Affiliation(s)
- Jiaming Liu
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Shanghai Qi Zhi Institute, Shanghai, 200232, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
| | - Liangkui Li
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Dijin Xu
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuqi Li
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Tao Chen
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, 341000, China
| | - Yeyang Liu
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, 200025, China
| | - Yan Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, China
| | - Longyan Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, China
| | - Peng Li
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Shanghai Qi Zhi Institute, Shanghai, 200232, China.
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Li Xu
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Shanghai Qi Zhi Institute, Shanghai, 200232, China.
| |
Collapse
|
13
|
Hagberg CE, Spalding KL. White adipocyte dysfunction and obesity-associated pathologies in humans. Nat Rev Mol Cell Biol 2024; 25:270-289. [PMID: 38086922 DOI: 10.1038/s41580-023-00680-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 02/10/2024]
Abstract
The prevalence of obesity and associated chronic diseases continues to increase worldwide, negatively impacting on societies and economies. Whereas the association between excess body weight and increased risk for developing a multitude of diseases is well established, the initiating mechanisms by which weight gain impairs our metabolic health remain surprisingly contested. In order to better address the myriad of disease states associated with obesity, it is essential to understand adipose tissue dysfunction and develop strategies for reinforcing adipocyte health. In this Review we outline the diverse physiological functions and pathological roles of human white adipocytes, examining our current knowledge of why white adipocytes are vital for systemic metabolic control, yet poorly adapted to our current obesogenic environment.
Collapse
Affiliation(s)
- Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kirsty L Spalding
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
14
|
Al lami Z, Kurtca M, Atique MU, Opekun AR, Siam MS, Jalal PK, Najafi B, Devaraj S, Mindikoglu AL. Dawn-to-dusk dry fasting decreases circulating inflammatory cytokines in subjects with increased body mass index. Metabol Open 2024; 21:100274. [PMID: 38455231 PMCID: PMC10918425 DOI: 10.1016/j.metop.2024.100274] [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: 11/11/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND The circadian rhythm involves numerous metabolic processes, including sleep/awakening, body temperature regulation, hormone secretion, hepatic function, cellular plasticity, and cytokine release (inflammation), that appear to have a dynamic relationship with all the processes above. Studies have linked various cytokines to the chronic state of low-grade inflammation and oxidative stress in obesity. Dawn-to-dusk dry fasting (DDDF) could alleviate the adverse effects of obesity by decreasing inflammation. This study examined the effects of DDDF on circulating inflammatory cytokines in subjects with increased body mass index (BMI). METHODS The current observational prospective study included adult subjects with a BMI equal to or greater than 25 kg/m2 who practiced the annual religious 30-day DDDF. Individuals with significant underlying medical conditions were excluded to limit confounding factors. All subjects were evaluated within two weeks before 30-day DDDF, within the fourth week of 30-day DDDF, and within two weeks after 30-day DDDF. Multiple cytokines and clinical health indicators were measured at each evaluation. RESULTS Thirteen subjects (10 men and three women) with a mean age of 32.9 years (SD = 9.7 years) and a mean BMI of 32 kg/m2 (SD = 4.6 kg/m2) were included. An overall associated decrease in the levels of multiple cytokines with DDDF was observed. A significant decrease in the mean interleukin 1 beta level was observed within the fourth week of 30-day DDDF (P = 0.045), which persisted even after the fasting period (P = 0.024). There was also a significant decrease in the mean levels of interleukin 15 (IL-15) (P = 0.014), interleukin 1 receptor antagonist (P = 0.041), macrophage-derived chemokine (MDC) (P = 0.013), and monokine induced by interferon gamma/chemokine (C-X-C motif) ligand 9 (P = 0.027) within the fourth week of 30-day DDDF and in the mean levels of fibroblast growth factor 2 (P = 0.010), interleukin 12 p40 subunit (P = 0.038), interleukin 22 (P = 0.025) and tumor necrosis factor alpha (P = 0.046) within two weeks after 30-DDDF. In terms of anthropometric parameters, there was a decrease in mean body weight (P = 0.032), BMI (P = 0.028), and hip circumference (P = 0.007) within the fourth week of 30-day DDDF and a decrease in mean weight (P = 0.026), BMI (P = 0.033) and hip circumference (P = 0.016) within two weeks after 30-day DDDF compared with the levels measured within two weeks before 30-day DDDF. Although there was no significant correlation between changes in weight and changes in circulating inflammatory cytokines, there was a significant positive correlation between changes in waist circumference and changes in specific inflammatory cytokines (e.g., IL-15, MDC, platelet-derived growth factor, soluble CD40L, vascular endothelial growth factor A) within the fourth week of 30-day DDDF and/or two weeks after 30-day DDDF. A significant decrease in mean average resting heart rate within the fourth week of 30-day DDDF was also observed (P = 0.023), and changes between average resting heart rate and changes in interleukin-8 levels within the fourth week of 30-day DDDF compared with baseline levels were positively correlated (r = 0.57, P = 0.042). CONCLUSION DDDF appears to be a unique and potent treatment to reduce low-grade chronic inflammation caused by obesity and visceral adiposity. Further studies with more extended follow-up periods are warranted to investigate the long-term anti-inflammatory benefits of DDDF in individuals with increased BMI.
Collapse
Affiliation(s)
- Zahraa Al lami
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - Miray Kurtca
- Clinical Chemistry and Point of Care Technology, Texas Children's Hospital and Health Centers, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Moin Uddin Atique
- Interdisciplinary Consortium on Advanced Motion Performance (iCAMP), Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Antone R. Opekun
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Division of Gastroenterology, Nutrition and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - Mohamad S. Siam
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - Prasun K. Jalal
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
- Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, USA
| | - Bijan Najafi
- Interdisciplinary Consortium on Advanced Motion Performance (iCAMP), Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Sridevi Devaraj
- Clinical Chemistry and Point of Care Technology, Texas Children's Hospital and Health Centers, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Ayse L. Mindikoglu
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
- Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
15
|
Sim JH, Kim KW, Ko Y, Moon YJ, Kwon HM, Jun IG, Kim SH, Kim KS, Song JG, Hwang GS. Association between visceral obesity and tumor recurrence in hepatocellular carcinoma recipients undergoing liver transplantation. Int J Obes (Lond) 2023; 47:1214-1223. [PMID: 37640894 DOI: 10.1038/s41366-023-01367-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 07/28/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Excessive visceral obesity in recipients of living donor liver transplantation (LDLT) is associated with mortality, and a recent study reported the correlation between visceral adiposity of male LDLT recipients and hepatocellular carcinoma (HCC) recurrence. However, there is no study on the relationship between the donor's visceral adiposity and surgical outcomes in LDLT recipients. We investigated the association of the visceral-to-subcutaneous fat area ratio (VSR) in donors and recipients with HCC recurrence and mortality in LDLT. METHODS We analyzed 1386 sets of donors and recipients who underwent LDLT between January 2008 and January 2018. The maximal chi-square method was used to determine the optimal cutoff values for VSR for predicting overall HCC recurrence and mortality. Cox regression analyses were performed to evaluate the association of donor VSR and recipient VSR with overall HCC recurrence and mortality in recipients. RESULTS The cutoff values of VSR was determined as 0.73 in males and 0.31 in females. High donor VSR was significantly associated with overall HCC recurrence (adjusted hazard ratio [HR]: 1.43, 95% confidence interval [CI]: 1.06-1.93, p = 0.019) and mortality (HR: 1.35, 95% CI: 1.03-1.76, p = 0.030). High recipient VSR was significantly associated with overall HCC recurrence (HR: 1.40, 95% CI: 1.04-1.88, p = 0.027) and mortality (HR: 1.50, 95% CI: 1.14-1.96, p = 0.003). CONCLUSIONS Both recipient VSR and donor VSR were significant risk factors for HCC recurrence and mortality in LDLT recipients. Preoperative donor VSR and recipient VSR may be strong predictors of the surgical outcomes of LDLT recipients with HCC.
Collapse
Affiliation(s)
- Ji-Hoon Sim
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung-Won Kim
- Department of Radiology, Asan Image Metrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - YouSun Ko
- Department of Radiology, Asan Image Metrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Young-Jin Moon
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hye-Mee Kwon
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In-Gu Jun
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Hoon Kim
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyoung-Sun Kim
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jun-Gol Song
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Gyu-Sam Hwang
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
16
|
Soileau LG, Nguyen A, Senthil A, Boullion JA, Talbot NC, Ahmadzadeh S, Shekoohi S, Kaye AD, Varrassi G. Bromocriptine and Colesevelam Hydrochloride: Novel Therapies for Type II Diabetes Mellitus. Cureus 2023; 15:e50138. [PMID: 38192911 PMCID: PMC10771968 DOI: 10.7759/cureus.50138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/07/2023] [Indexed: 01/10/2024] Open
Abstract
The increasing prevalence of type II diabetes mellitus (T2DM) is a worldwide healthcare concern. Over the years, our understanding of T2DM has grown considerably in uncovering the pathophysiology of the disease and, in turn, understanding how improved treatment methods can be used to slow disease progression. Some long-term complications that are responsible for most T2DM mortalities include cardiovascular disease, neurological decline, and renal failure. In treating T2DM, it is important that not only glycemic control be obtained but also control of associated complications. Bromocriptine and colesevelam hydrochloride have both been approved by the Food and Drug Administration (FDA) to treat T2DM but are not readily used in practice. These medications are known to treat glycemic dysregulation via unconventional mechanisms, which might contribute to their potential to provide protection against common diabetic complications such as cardiovascular disease. In order to ensure that these overlooked medications become more readily used, it is vital that more research be performed to further elucidate their efficacy in a clinical setting. Future studies should continue to provide clinicians a better understanding of the role these medications have on the treatment of T2DM such as their ability to be used in combination with other commonly used T2DM medications or as monotherapies.
Collapse
Affiliation(s)
- Lenise G Soileau
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Angela Nguyen
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Aarthi Senthil
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Jolie A Boullion
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Norris C Talbot
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | | |
Collapse
|
17
|
Christiansen MR, Kilpeläinen TO, McCaffery JM. Abdominal Obesity Genetic Variants Predict Waist Circumference Regain After Weight Loss. Diabetes 2023; 72:1424-1432. [PMID: 37494631 DOI: 10.2337/db23-0131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
Although many individuals are able to achieve weight loss, maintaining this loss over time is challenging. We aimed to study whether genetic predisposition to general or abdominal obesity predicts weight regain after weight loss. We examined the associations between genetic risk scores for higher BMI and higher waist-to-hip ratio adjusted for BMI (WHRadjBMI) with changes in weight and waist circumference up to 3 years after a 1-year weight loss program in participants (n = 822 women, n = 593 men) from the Look AHEAD (Action for Health in Diabetes) study who had lost ≥3% of their initial weight. Genetic predisposition to higher BMI or WHRadjBMI was not associated with weight regain after weight loss. However, the WHRadjBMI genetic score did predict an increase in waist circumference independent of weight change. To conclude, a genetic predisposition to higher WHRadjBMI predicts an increase in abdominal obesity after weight loss, whereas genetic predisposition to higher BMI is not predictive of weight regain. These results suggest that genetic effects on abdominal obesity may be more pronounced than those on general obesity during weight regain. ARTICLE HIGHLIGHTS Nearly all individuals who intentionally lose weight experience weight regain. Individuals with a higher genetic risk for abdominal adiposity experience increased regain in waist circumference after weight loss. Genetic predisposition to higher BMI does not predict weight regain after weight loss.
Collapse
Affiliation(s)
- Malene Revsbech Christiansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Jeanne M McCaffery
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT
| |
Collapse
|
18
|
Abstract
Gluconeogenesis is a critical biosynthetic process that helps maintain whole-body glucose homeostasis and becomes altered in certain medical diseases. We review gluconeogenic flux in various medical diseases, including common metabolic disorders, hormonal imbalances, specific inborn genetic errors, and cancer. We discuss how the altered gluconeogenic activity contributes to disease pathogenesis using data from experiments using isotopic tracer and spectroscopy methodologies. These in vitro, animal, and human studies provide insights into the changes in circulating levels of available gluconeogenesis substrates and the efficiency of converting those substrates to glucose by gluconeogenic organs. We highlight ongoing knowledge gaps, discuss emerging research areas, and suggest future investigations. A better understanding of altered gluconeogenesis flux may ultimately identify novel and targeted treatment strategies for such diseases.
Collapse
Affiliation(s)
- Ankit Shah
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA; ,
| | - Fredric E Wondisford
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA; ,
| |
Collapse
|
19
|
Zheng L, Sun A, Han S, Qi R, Wang R, Gong X, Xue M. Association between visceral obesity and 10-year risk of first atherosclerotic cardiovascular diseases events among American adults: National Health and Nutrition Examination Survey. Front Cardiovasc Med 2023; 10:1249401. [PMID: 37674809 PMCID: PMC10479018 DOI: 10.3389/fcvm.2023.1249401] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023] Open
Abstract
Background In the United States, the relationship between visceral obesity and the risk of developing atherosclerosis cardiovascular disease (ASCVD) for the first time in 10 years is unclear. Methods Data for this cross-sectional study came from the National Health and Nutrition Examination Survey (NHANES) from 2011 to 2020. We collected variable information related to 10-year ASCVD risk and visceral obesity reliable indicators [Visceral obesity index (VAI) and Lipid accumulation product (LAP)]. And we used multiple logistic regression to analyze the correlation of visceral obesity indicators (VAI and LAP) with 10-year ASCVD risk. In addition, we assessed the linear relationship between VAI or LAP and 10-year ASCVD risk by smoothing curve fitting. Finally, we conducted subgroup analysis and sensitivity analysis after excluding participants with extreme VAI and LAP values to ensure that we obtained accurate and reliable results. Results Our study included a total of 1,547 participants (mean age: 56.5 ± 10.1, 60% of males). The results of the multiple logistic regression showed that compared with participants with the lowest VAI in the 1st Quartile (≤0.79), the adjusted OR values for VAI and elevated 10-year ASCVD risk in Q3 (1.30-2.14), and Q4 (≥2.15) were 2.58 (95% CI: 1.24-5.36, P = 0.011), 15.14 (95% CI: 6.93-33.05, P < 0.001), respectively. Compared with participants with the lowest LAP in the 1st Quartile (≤28.29), the adjusted OR values for VAI and elevated 10-year ASCVD risk in Q3 (46.52-77.00), and Q4 (≥77.01) were 4.63 (95% CI: 2.18-9.82, P < 0.001), 16.94 (95% CI: 6.74-42.57, P < 0.001), respectively. Stratified analysis showed that the association between VAI or LAP and the first ASCVD event was more pronounced in males. Conclusion Higher VAI or LAP scores are significantly associated with elevated 10-year ASCVD risk in adults aged 40 to 79 in the USA, which suggested that monitoring visceral obesity is crucial to reduce the risk of a first ASCVD event.
Collapse
Affiliation(s)
- Liying Zheng
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aochuan Sun
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Senfu Han
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rongming Qi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Rumeng Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Gong
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mei Xue
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
20
|
Moraes DD, Mousovich-Neto F, Cury SS, Oliveira J, Souza JDS, Freire PP, Dal-Pai-Silva M, Mori MADS, Fernandez GJ, Carvalho RF. The Transcriptomic Landscape of Age-Induced Changes in Human Visceral Fat and the Predicted Omentum-Liver Connectome in Males. Biomedicines 2023; 11:biomedicines11051446. [PMID: 37239116 DOI: 10.3390/biomedicines11051446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/15/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Aging causes alterations in body composition. Specifically, visceral fat mass increases with age and is associated with age-related diseases. The pathogenic potential of visceral fat accumulation has been associated with its anatomical location and metabolic activity. Visceral fat may control systemic metabolism by secreting molecules that act in distal tissues, mainly the liver, through the portal vein. Currently, little is known about age-related changes in visceral fat in humans. Aiming to identify molecular and cellular changes occurring with aging in the visceral fat of humans, we analyzed publicly available transcriptomic data of 355 omentum samples from the Genotype-Tissue Expression portal (GTEx) of 20-79-year-old males and females. We identified the functional enrichment of genes associated with aging, inferred age-related changes in visceral fat cellularity by deconvolution analysis, profiled the senescence-associated secretory phenotype of visceral adipose tissue, and predicted the connectivity of the age-induced visceral fat secretome with the liver. We demonstrate that age induces alterations in visceral fat cellularity, synchronous to changes in metabolic pathways and a shift toward a pro-inflammatory secretory phenotype. Furthermore, our approach identified candidates such as ADIPOQ-ADIPOR1/ADIPOR2, FCN2-LPR1, and TF-TFR2 to mediate visceral fat-liver crosstalk in the context of aging. These findings cast light on how alterations in visceral fat with aging contribute to liver dysfunction and age-related disease etiology.
Collapse
Affiliation(s)
- Diogo de Moraes
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, UNESP, Botucatu 18618-689, SP, Brazil
- Department of Biochemistry and Tissue Biology, University of Campinas, Monteiro Lobato St., 255, Campinas 13083-862, SP, Brazil
| | - Felippe Mousovich-Neto
- Department of Biochemistry and Tissue Biology, University of Campinas, Monteiro Lobato St., 255, Campinas 13083-862, SP, Brazil
| | - Sarah Santiloni Cury
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, UNESP, Botucatu 18618-689, SP, Brazil
| | - Jakeline Oliveira
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, UNESP, Botucatu 18618-689, SP, Brazil
| | - Jeferson Dos Santos Souza
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, UNESP, Botucatu 18618-689, SP, Brazil
| | - Paula Paccielli Freire
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, UNESP, Botucatu 18618-689, SP, Brazil
| | - Maeli Dal-Pai-Silva
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, UNESP, Botucatu 18618-689, SP, Brazil
| | - Marcelo Alves da Silva Mori
- Department of Biochemistry and Tissue Biology, University of Campinas, Monteiro Lobato St., 255, Campinas 13083-862, SP, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas 13083-862, SP, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas 13083-862, SP, Brazil
| | - Geysson Javier Fernandez
- Grupo Biologia y Control de Enfermedades Infeciosas (BCEI), Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia (UdeA), Medellín 050010, Colombia
| | - Robson Francisco Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, UNESP, Botucatu 18618-689, SP, Brazil
| |
Collapse
|
21
|
Sim JH, Kim KW, Ko Y, Kwon HM, Moon YJ, Jun IG, Kim SH, Kim S, Song JG, Hwang GS. Association of sex-specific donor skeletal muscle index with surgical outcomes in living donor liver transplantation recipients. Liver Int 2023; 43:684-694. [PMID: 36377561 DOI: 10.1111/liv.15478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/28/2022] [Accepted: 11/13/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND A recent study reported a correlation between the muscle mass of male donors and graft failure in living donor liver transplantation (LDLT) recipients. We investigated the association of sex-specific donor skeletal muscle index (SMI) with mortality and graft failure in LDLT recipients. METHODS We retrospectively analysed 2750 sets of donors and recipients between January 2008 and January 2018. The recipient outcomes were analysed by dividing the data according to donor sex. Cox regression analyses were performed to evaluate the association between donor SMI by sex and 1-year mortality and graft failure in recipients. RESULTS In the male donor group, robust donor (increased SMI) was significantly associated with higher risks for mortality (hazard ratio [HR]: 1.03, 95% confidence interval [CI]: 1.00-1.06, p = .023) and graft failure (HR: 1.04, 95% CI: 1.01-1.06, p = .007) at 1 year. In the female donor group, the robust donor was significantly associated with lower risks for mortality (HR: 0.92, 95% CI: 0.87-0.97, p = .003) and graft failure (HR: 0.95, 95% CI: 0.90-1.00, p = .032) at 1 year. CONCLUSIONS Donor SMI was associated with surgical outcomes in recipients. Robust male and female donors were a significant negative and protective factor for grafts respectively.
Collapse
Affiliation(s)
- Ji-Hoon Sim
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung-Won Kim
- Department of Radiology, Asan Image Metrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - YouSun Ko
- Department of Radiology, Asan Image Metrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hye-Mee Kwon
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Jin Moon
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In-Gu Jun
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Hoon Kim
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seonok Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jun-Gol Song
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gyu-Sam Hwang
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
Wang CJ, Noble PB, Elliot JG, James AL, Wang KCW. From Beneath the Skin to the Airway Wall: Understanding the Pathological Role of Adipose Tissue in Comorbid Asthma-Obesity. Compr Physiol 2023; 13:4321-4353. [PMID: 36715283 DOI: 10.1002/cphy.c220011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article provides a contemporary report on the role of adipose tissue in respiratory dysfunction. Adipose tissue is distributed throughout the body, accumulating beneath the skin (subcutaneous), around organs (visceral), and importantly in the context of respiratory disease, has recently been shown to accumulate within the airway wall: "airway-associated adipose tissue." Excessive adipose tissue deposition compromises respiratory function and increases the severity of diseases such as asthma. The mechanisms of respiratory impairment are inflammatory, structural, and mechanical in nature, vary depending on the anatomical site of deposition and adipose tissue subtype, and likely contribute to different phenotypes of comorbid asthma-obesity. An understanding of adipose tissue-driven pathophysiology provides an opportunity for diagnostic advancement and patient-specific treatment. As an exemplar, the potential impact of airway-associated adipose tissue is highlighted, and how this may change the management of a patient with asthma who is also obese. © 2023 American Physiological Society. Compr Physiol 13:4321-4353, 2023.
Collapse
Affiliation(s)
- Carolyn J Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - John G Elliot
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
23
|
Börgeson E, Boucher J, Hagberg CE. Of mice and men: Pinpointing species differences in adipose tissue biology. Front Cell Dev Biol 2022; 10:1003118. [PMID: 36187476 PMCID: PMC9521710 DOI: 10.3389/fcell.2022.1003118] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The prevalence of obesity and metabolic diseases continues to rise, which has led to an increased interest in studying adipose tissue to elucidate underlying disease mechanisms. The use of genetic mouse models has been critical for understanding the role of specific genes for adipose tissue function and the tissue’s impact on other organs. However, mouse adipose tissue displays key differences to human fat, which has led, in some cases, to the emergence of some confounding concepts in the adipose field. Such differences include the depot-specific characteristics of visceral and subcutaneous fat, and divergences in thermogenic fat phenotype between the species. Adipose tissue characteristics may therefore not always be directly compared between species, which is important to consider when setting up new studies or interpreting results. This mini review outlines our current knowledge about the cell biological differences between human and mouse adipocytes and fat depots, highlighting some examples where inadequate knowledge of species-specific differences can lead to confounding results, and presenting plausible anatomic explanations that may underlie the differences. The article thus provides critical insights and guidance for researchers working primarily with only human or mouse fat tissue, and may contribute to new ideas or concepts in the important and evolving field of adipose biology.
Collapse
Affiliation(s)
- Emma Börgeson
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Region Vaestra Goetaland, Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jeremie Boucher
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Metabolic Disease, Evotec International GmbH, Göttingen, Germany
| | - Carolina E. Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- *Correspondence: Carolina E. Hagberg,
| |
Collapse
|
24
|
AYDIN MM, DAĞISTAN E, COŞGUN Z. Metabolik sendromda visseral ve subkutan yağ miktari ve hepatosteatozun bilgisayarli tomografi ile kantitatif değerlendirilmesi. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1037220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Purpose: We aimed to evaluate visceral and subcutaneous fat tissue and its association with hepatosteatosis on computed tomography (CT) scans to determine cut-off criteria for metabolic syndrome, measure abdominal obesity directly based on the visceral fat tissue area (VFTA) rather than indirectly based on waist circumference and obtain supportive findings by density measurements in addition to the VFTA measurements.Materials and Methods: The Hounsfield unit (HU) values, visceral, subcutaneous fat areas and HU values of 108 patients diagnosed with metabolic syndrome (MS) were determined according to the National Cholesterol Education Program Adult Treatment Panel III 2001 Criteria by retrospectively analyzing their abdominal CT images taken for various reasons. The relationships of the obtained values with each other and to MS were evaluated.Results: The strongest predictor of MS was VFTA, and 156.47 cm² was the most significant value with 74.1% sensitivity and 58.6% specificity. An HU value of -102.99 for visceral fat tissue density (VFTD) was found as the second most significant finding with 75% sensitivity and 57.6% specificity. The VFTA values of the patients with hepatosteatosis were higher, and increased VFTA values were associated with lower VFTD values.Conclusion: The most important supportive finding was the demonstration of the possibility of measuring abdominal obesity, which has an important place among criteria, directly by measuring VFTA, rather than indirectly based on waist circumference.
Collapse
Affiliation(s)
- Mehmet Maruf AYDIN
- SAĞLIK BİLİMLERİ ÜNİVERSİTESİ, SAMSUN SAĞLIK UYGULAMA VE ARAŞTIRMA MERKEZİ
| | | | | |
Collapse
|
25
|
Efthymiou V, Patti ME. It Is Not Just Fat: Dissecting the Heterogeneity of Adipose Tissue Function. Curr Diab Rep 2022; 22:177-187. [PMID: 35267142 DOI: 10.1007/s11892-022-01455-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/11/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW The purpose of the current review is to summarize findings from the most recent and impactful studies which investigated human and mouse adipose tissue transcriptomes at a single-cell level. We provide perspective about the potential importance of data derived from these single-cell technologies in improving our understanding of the adipose organ and metabolic disease and likely future directions of this approach. RECENT FINDINGS The majority of single-cell or single-nuclei studies of the adipose organ so far have focused on investigating the stromal-vascular fraction (SVF) of mouse subcutaneous and intraabdominal white and interscapular brown fat depots. Few studies have also evaluated the impact of additional factors as drivers of adipose phenotypes, such as high-fat diet-induced obesity, adolescence, aging, and cold exposure. Recent studies have also investigated human cell lines and human fat biopsies across a range of body mass index (BMI) and in response to insulin resistance or T2D. These studies have identified numerous previously unexplored subpopulations of adipocyte progenitors, immune cells, and mature adipocytes in both mice and men. Single-cell and single-nuclei technologies have brought an explosion of data that have advanced our understanding of the adipose organ in health and disease. However, we are still at the dawn of achieving a complete and comprehensive map of the mouse and human adipose organ. Multi-modal single-cell approaches to identify both anatomic localization of specific cellular populations and epigenetic mechanisms responsible for observed transcriptomic patterns are underway and will likely provide an even deeper understanding of the adipose organ in response to health and disease.
Collapse
Affiliation(s)
- Vissarion Efthymiou
- Department of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Mary-Elizabeth Patti
- Department of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, MA, 02215, USA.
- Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
26
|
Kirschner KM, Scholz H. WT1 in Adipose Tissue: From Development to Adult Physiology. Front Cell Dev Biol 2022; 10:854120. [PMID: 35372335 PMCID: PMC8965737 DOI: 10.3389/fcell.2022.854120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
Much of the fascination of the Wilms tumor protein (WT1) emanates from its unique roles in development and disease. Ubiquitous Wt1 deletion in adult mice causes multiple organ failure including a reduction of body fat. WT1 is expressed in fat cell progenitors in visceral white adipose tissue (WAT) but detected neither in energy storing subcutaneous WAT nor in heat producing brown adipose tissue (BAT). Our recent findings indicate that WT1 represses thermogenic genes and maintains the white adipose identity of visceral fat. Wt1 heterozygosity in mice is associated with molecular and morphological signs of browning including elevated levels of uncoupling protein 1 (UCP1) in epididymal WAT. Compared to their wild-type littermates, Wt1 heterozygous mice exhibit significantly improved whole-body glucose tolerance and alleviated hepatic steatosis under high-fat diet. Partial protection of heterozygous Wt1 knockout mice against metabolic dysfunction is presumably related to browning of their epididymal WAT. In the light of recent advancements, this article reviews the role of WT1 in the development of visceral WAT and its supposed function as a regulator of white adipose identity.
Collapse
|
27
|
Li Y, Li J, Yu H, Liu Y, Song H, Tian X, Liu D, Yan C, Han Y. HOXA5-miR-574-5p axis promotes adipogenesis and alleviates insulin resistance. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:200-210. [PMID: 34976438 PMCID: PMC8693313 DOI: 10.1016/j.omtn.2021.08.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 08/31/2021] [Indexed: 11/15/2022]
Abstract
Differentiation of preadipocytes into functional adipocytes could be a major target for repressing obesity-induced insulin resistance (IR). However, the molecular mechanisms involved in adipogenesis and the development of IR are unclear. We report, for the first time, that miR-574-5p, a novel miRNA, promotes adipogenesis to suppress IR. An increase in the level of miR-574-5p significantly induced the differentiation of preadipocytes into mature adipocytes. Conversely, reduction of miR-574-5p levels blocked the differentiation of preadipocytes in vitro. In a dual-luciferase reporter assay, it was shown that homeobox A5 (HOXA5) promoted the transcription of miR-574-5p to induce the differentiation of preadipocytes. Hdac9, a direct downstream target of miR-574-5p, was involved in the regulation of adipocyte differentiation. The overexpression of miR-574-5p also promoted adipogenesis in subcutaneous fat to alleviate IR in high-fat-diet-fed mice. Additionally, miR-574-5p expression was significantly higher in the subcutaneous adipose tissue of obese patients without type 2 diabetes than in those with type 2 diabetes. There was an increase in HOXA5 expression and a decrease in histone deacetylase 9 (HDAC9) expression in the subcutaneous fat of obese patients without type 2 diabetes. These results suggest that miR-574-5p may be a potential therapeutic target for combating obesity-related IR.
Collapse
Affiliation(s)
- Yuying Li
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang 110016, China
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jiayin Li
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang 110016, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110016, China
| | - Haibo Yu
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Yanxia Liu
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Haixu Song
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Xiaoxiang Tian
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Dan Liu
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Chenghui Yan
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Yaling Han
- Department of Cardiology and Cardiovascular Research Institute of PLA, General Hospital of Northern Theater Command, Shenyang 110016, China
| |
Collapse
|
28
|
Sakers A, De Siqueira MK, Seale P, Villanueva CJ. Adipose-tissue plasticity in health and disease. Cell 2022; 185:419-446. [PMID: 35120662 PMCID: PMC11152570 DOI: 10.1016/j.cell.2021.12.016] [Citation(s) in RCA: 439] [Impact Index Per Article: 146.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
Adipose tissue, colloquially known as "fat," is an extraordinarily flexible and heterogeneous organ. While historically viewed as a passive site for energy storage, we now appreciate that adipose tissue regulates many aspects of whole-body physiology, including food intake, maintenance of energy levels, insulin sensitivity, body temperature, and immune responses. A crucial property of adipose tissue is its high degree of plasticity. Physiologic stimuli induce dramatic alterations in adipose-tissue metabolism, structure, and phenotype to meet the needs of the organism. Limitations to this plasticity cause diminished or aberrant responses to physiologic cues and drive the progression of cardiometabolic disease along with other pathological consequences of obesity.
Collapse
Affiliation(s)
- Alexander Sakers
- Institute for Diabetes, Obesity & Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Mirian Krystel De Siqueira
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA.
| | - Claudio J Villanueva
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA.
| |
Collapse
|
29
|
Jiang K, Luan H, Pu X, Wang M, Yin J, Gong R. Association Between Visceral Adiposity Index and Insulin Resistance: A Cross-Sectional Study Based on US Adults. Front Endocrinol (Lausanne) 2022; 13:921067. [PMID: 35937809 PMCID: PMC9353944 DOI: 10.3389/fendo.2022.921067] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/22/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Visceral obesity index (VAI) is an empirical mathematical model used to evaluate the distribution and function of fat. Some studies have shown that VAI may be associated with the development of insulin resistance. In view of the differences in insulin resistance among different ethnic groups, this study attempts to analyze the special relationship between VAI and insulin resistance in American adults. METHODS We conducted a cross-sectional study through NHANES database. A total of 27309 patients over the age of 18 from the United States took part in the survey. It was divided into two groups: the IR-positive group and the IR-negative group. The association of VAI with IR was evaluated by logistic regression analyses mainly, including univariate analysis, multivariate regression analysis, curve fitting analysis and subgroup analysis. RESULTS The results showed that in the full-adjusted model, there is a strong positive association between VAI level and insulin resistance (OR: 1.28 (1.2~1.37), P<0.001) and there is a threshold effect. CONCLUSIONS This study suggests that higher VAI levels are associated with insulin resistance. VAI index may be used as a predictor of insulin resistance.
Collapse
Affiliation(s)
- Kai Jiang
- Department of Cardiovascular Medicine, The Bozhou Hospital Affiliated to Anhui Medical University, Bozhou, China
| | - Hong Luan
- Department of Cardiovascular Medicine, People’s Hospital of Ningxia Hui Autonomous Region, YinChuan, China
| | - Xiaolu Pu
- Medical College of Qinghai University, Xining, China
| | | | - Jiahui Yin
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Rongpeng Gong, ; ; Jiahui Yin,
| | - Rongpeng Gong
- Medical College of Qinghai University, Xining, China
- *Correspondence: Rongpeng Gong, ; ; Jiahui Yin,
| |
Collapse
|
30
|
Gagnon E, Pelletier W, Gobeil É, Bourgault J, Manikpurage HD, Maltais-Payette I, Abner E, Taba N, Esko T, Mitchell PL, Ghodsian N, Després JP, Vohl MC, Tchernof A, Thériault S, Arsenault BJ. Mendelian randomization prioritizes abdominal adiposity as an independent causal factor for liver fat accumulation and cardiometabolic diseases. COMMUNICATIONS MEDICINE 2022; 2:130. [PMID: 36249462 PMCID: PMC9561122 DOI: 10.1038/s43856-022-00196-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Background Observational studies have linked adiposity and especially abdominal adiposity to liver fat accumulation and non-alcoholic fatty liver disease. These traits are also associated with type 2 diabetes and coronary artery disease but the causal factor(s) underlying these associations remain unexplored. Methods We used a multivariable Mendelian randomization study design to determine whether body mass index and waist circumference were causally associated with non-alcoholic fatty liver disease using publicly available genome-wide association study summary statistics of the UK Biobank (n = 461,460) and of non-alcoholic fatty liver disease (8434 cases and 770,180 control). A multivariable Mendelian randomization study design was also used to determine the respective causal contributions of waist circumference and liver fat (n = 32,858) to type 2 diabetes and coronary artery disease. Results Using multivariable Mendelian randomization we show that waist circumference increase non-alcoholic fatty liver disease risk even when accounting for body mass index (odd ratio per 1-standard deviation increase = 2.35 95% CI = 1.31-4.22, p = 4.2e-03), but body mass index does not increase non-alcoholic fatty liver disease risk when accounting for waist circumference (0.86 95% CI = 0.54-1.38, p = 5.4e-01). In multivariable Mendelian randomization analyses accounting for liver fat, waist circumference remains strongly associated with both type 2 diabetes (3.27 95% CI = 2.89-3.69, p = 3.8e-80) and coronary artery disease (1.66 95% CI = 1.54-1.8, p = 3.4e-37). Conclusions These results identify waist circumference as a strong, independent, and causal contributor to non-alcoholic fatty liver disease, type 2 diabetes and coronary artery disease, thereby highlighting the importance of assessing body fat distribution for the prediction and prevention of cardiometabolic diseases.
Collapse
Affiliation(s)
- Eloi Gagnon
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada
| | - William Pelletier
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada
| | - Émilie Gobeil
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada
| | - Jérôme Bourgault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada
| | - Hasanga D Manikpurage
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada
| | - Ina Maltais-Payette
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada.,School of Nutrition, Université Laval, Québec, QC Canada
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Riia 23b, Tartu, 51010 Estonia
| | - Nele Taba
- Estonian Genome Center, Institute of Genomics, University of Tartu, Riia 23b, Tartu, 51010 Estonia.,Institute of Molecular and Cell Biology, University of Tartu, Riia 23, Tartu, 51,010 Estonia
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Riia 23b, Tartu, 51010 Estonia
| | - Patricia L Mitchell
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada
| | - Nooshin Ghodsian
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada
| | - Jean-Pierre Després
- VITAM - Centre de recherche en santé durable, Université Laval, Québec, QC Canada
| | - Marie-Claude Vohl
- School of Nutrition, Université Laval, Québec, QC Canada.,Centre NUTRISS, Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, QC Canada
| | - André Tchernof
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada.,School of Nutrition, Université Laval, Québec, QC Canada
| | - Sébastien Thériault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC Canada
| |
Collapse
|
31
|
The Association between Anthropometry Indices and Serum Concentrations of Gamma-Glutamyl Transferase, Alkaline Phosphatase, Alanine Aminotransferase, and Aspartate Aminotransferase. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2365399. [PMID: 34853788 PMCID: PMC8629636 DOI: 10.1155/2021/2365399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/09/2021] [Indexed: 02/08/2023]
Abstract
Background Nowadays, metabolic syndrome (MetS) has become a great public health crisis that threatens too many lives worldwide. Many previous studies have been investigated the association between MetS and anthropometric indices. This study is aimed at investigating the association between anthropometric indices with gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), alanine aminotransferase (ALT), and aspartate aminotransferase (AST). We were using data from a large population-based cohort of seemingly healthy women and men. Methods A total of 7216 participants were included in this study. The serum levels of GGT, ALP, ALT, and AST with bioimpedance measures were obtained at the time of enrollment. Multiple regression analysis was performed to assess the relationship between anthropometric indices and serum levels of the aforementioned laboratory tests. Results Serum levels of GGT, ALP, ALT, and AST significantly correlated with body mass index (BMI). Only ALP had a significant association with visceral fat area (VFA). AST, ALT, and ALP levels had a positive correlation with 50 kHz whole-body phase. Conclusion From the findings of this study, body mass index consistently appeared a good predictor of elevated hepatic enzymes and triglycerides. Thus, it can be helpful in clinical settings to identify patients at risk of nonalcoholic fatty liver disease, which is closely related to metabolic syndrome.
Collapse
|
32
|
Dey P, Chaudhuri SR, Efferth T, Pal S. The intestinal 3M (microbiota, metabolism, metabolome) zeitgeist - from fundamentals to future challenges. Free Radic Biol Med 2021; 176:265-285. [PMID: 34610364 DOI: 10.1016/j.freeradbiomed.2021.09.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
The role of the intestine in human health and disease has historically been neglected and was mostly attributed to digestive and absorptive functions. In the past two decades, however, discoveries related to human nutrition and intestinal host-microbe reciprocal interaction have established the essential role of intestinal health in the pathogenesis of chronic diseases and the overall wellbeing. That transfer of gut microbiota could be a means of disease phenotype transfer has revolutionized our understanding of chronic disease pathogenesis. This narrative review highlights the major concepts related to intestinal microbiota, metabolism, and metabolome (3M) that have facilitated our fundamental understanding of the association between the intestine, and human health and disease. In line with increased interest of microbiota-dependent modulation of human health by dietary phytochemicals, we have also discussed the emerging concepts beyond the phytochemical bioactivities which emphasizes the integral role of microbial metabolites of parent phytochemicals at extraintestinal tissues. Finally, this review concludes with challenges and future prospects in defining the 3M interactions and has emphasized the fact that, it takes 'guts' to stay healthy.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Sirshendu Pal
- Mukherjee Hospital, Mitra's Clinic and Nursing Home, Siliguri, West Bengal, India
| |
Collapse
|
33
|
Are sex disparities in COVID-19 a predictable outcome of failing men's health provision? Nat Rev Urol 2021; 19:47-63. [PMID: 34795426 PMCID: PMC8600906 DOI: 10.1038/s41585-021-00535-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 coronavirus, has taken a catastrophic toll on society, health-care systems and the economy. Notably, COVID-19 has been shown to be associated with a higher mortality rate in men than in women. This disparity is likely to be a consequence of a failure to invest in men’s health, as it has also been established that men have a lower life expectancy and poorer outcomes from non-communicable diseases than women. A variety of biological, social and economic factors have contributed to the sex disparities in mortality from COVID-19. A streamlined men’s health programme — with the urologist as the gatekeeper of men’s health — is needed to help prevent future tragedies of this nature. COVID-19 has been shown to be associated with a higher mortality rate in men than in women. In this Perspectives article, the authors posit that this disparity is due to a failure to invest in men’s health and discuss the biological, social and economic factors that have contributed to the sex disparities in mortality from COVID-19, as well as considering how a streamlined men’s health programme with the urologist in a central role could address these issues.
Collapse
|
34
|
Abstract
Obesity is a chronic and progressive process affecting whole-body energy balance and is associated with comorbidities development. In addition to increased fat mass, obesity induces white adipose tissue (WAT) inflammation and fibrosis, leading to local and systemic metabolic dysfunctions, such as insulin resistance (IR). Accordingly, limiting inflammation or fibrosis deposition may improve IR and glucose homeostasis. Although no targeted therapy yet exists to slow or reverse adipose tissue fibrosis, a number of findings have clarified the underlying cellular and molecular mechanisms. In this review, we highlight adipose tissue remodeling events shown to be associated with fibrosis deposition, with a focus on adipose progenitors involved in obesity-induced healthy as well as unhealthy WAT expansion. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Geneviève Marcelin
- INSERM, Nutrition and Obesities: Systemic Approach (NutriOmics) Research Unit, UMRS U1269, Sorbonne Université, Paris, France; ,
| | | | - Karine Clément
- INSERM, Nutrition and Obesities: Systemic Approach (NutriOmics) Research Unit, UMRS U1269, Sorbonne Université, Paris, France; , .,Nutrition Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
35
|
Huang C, Rosencrans RF, Bugescu R, Vieira CP, Hu P, Adu-Agyeiwaah Y, Gamble KL, Longhini ALF, Fuller PM, Leinninger GM, Grant MB. Depleting hypothalamic somatostatinergic neurons recapitulates diabetic phenotypes in mouse brain, bone marrow, adipose and retina. Diabetologia 2021; 64:2575-2588. [PMID: 34430981 PMCID: PMC9004546 DOI: 10.1007/s00125-021-05549-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/11/2021] [Indexed: 12/01/2022]
Abstract
AIMS/HYPOTHESIS Hypothalamic inflammation and sympathetic nervous system hyperactivity are hallmark features of the metabolic syndrome and type 2 diabetes. Hypothalamic inflammation may aggravate metabolic and immunological pathologies due to extensive sympathetic activation of peripheral tissues. Loss of somatostatinergic (SST) neurons may contribute to enhanced hypothalamic inflammation. METHODS The present data show that leptin receptor-deficient (db/db) mice exhibit reduced hypothalamic SST neurons, particularly in the periventricular nucleus. We model this finding, using adeno-associated virus delivery of diphtheria toxin subunit A (DTA) driven by an SST-cre system to deplete these neurons in Sstcre/gfp mice (SST-DTA). RESULTS SST-DTA mice exhibit enhanced hypothalamic c-Fos expression and brain inflammation as demonstrated by microglial and astrocytic activation. Bone marrow from SST-DTA mice undergoes skewed haematopoiesis, generating excess granulocyte-monocyte progenitors and increased proinflammatory (C-C chemokine receptor type 2; CCR2hi) monocytes. SST-DTA mice exhibited a 'diabetic retinopathy-like' phenotype: reduced visual function by optokinetic response (0.4 vs 0.25 cycles/degree; SST-DTA vs control mice); delayed electroretinogram oscillatory potentials; and increased percentages of retinal monocytes. Finally, mesenteric visceral adipose tissue from SST-DTA mice was resistant to catecholamine-induced lipolysis, displaying 50% reduction in isoprenaline (isoproterenol)-induced lipolysis compared with control littermates. Importantly, hyperglycaemia was not observed in SST-DTA mice. CONCLUSIONS/INTERPRETATION The isolated reduction in hypothalamic SST neurons was able to recapitulate several hallmark features of type 2 diabetes in disease-relevant tissues.
Collapse
Affiliation(s)
- Chao Huang
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert F Rosencrans
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Raluca Bugescu
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Cristiano P Vieira
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ping Hu
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yvonne Adu-Agyeiwaah
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Karen L Gamble
- Department of Psychiatry and Neurobehavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ana Leda F Longhini
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Patrick M Fuller
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Gina M Leinninger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Maria B Grant
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
36
|
Bake S, Pinson MR, Pandey S, Chambers JP, Mota R, Fairchild AE, Miranda RC, Sohrabji F. Prenatal alcohol-induced sex differences in immune, metabolic and neurobehavioral outcomes in adult rats. Brain Behav Immun 2021; 98:86-100. [PMID: 34390803 PMCID: PMC8591773 DOI: 10.1016/j.bbi.2021.08.207] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 07/10/2021] [Accepted: 08/06/2021] [Indexed: 12/15/2022] Open
Abstract
Prenatal alcohol exposure (PAE) can result in neurobehavioral anomalies, that may be exacerbated by co-occurring metabolic and immune system deficits. To test the hypothesis that the peripheral inflammation in adult PAE offspring is linked to poor glucose metabolism and neurocognitive deficits, pregnant Sprague-Dawley rats were exposed to ethanol vapor or ambient air during the latter half of gestation. We assessed, in adult offspring of both sexes, performance on a battery of neurocognitive behaviors, glucose tolerance, circulating and splenic immune cells by flow-cytometry, and circulating and tissue (liver, mesenteric adipose, and spleen) cytokines by multiplexed assays. PAE reduced both the ratio of spleen to body weight and splenic regulatory T-cell (Treg) numbers. PAE males, but not females exhibited an increase in circulating monocytes. Overall, PAE males exhibited a suppression of cytokine levels, while PAE females exhibited elevated cytokines in mesenteric adipose tissue (IL-6 and IL1α) and liver (IFN-γ, IL-1β, IL-13, IL-18, IL-12p70, and MCP-1), along with increased glucose intolerance. Behavioral analysis also showed sex-dependent PAE effects. PAE-males exhibited increased anxiety-like behavior while PAE-females showed decreased social interaction. PAE offspring of both sexes exhibited impaired recognition of novel objects. Multilinear regression modeling to predict the association between peripheral immune status, glucose intolerance and behavioral outcomes, showed that in PAE offspring, higher levels of adipose leptin and liver TNF- α predicted higher circulating glucose levels. Lower liver IL-1 α and higher plasma fractalkine predicted more time spent in the center of an open-field with sex being an additional predictor. Higher circulating and splenic Tregs predicted better social interaction in the PAE-offspring. Collectively, our data show that peripheral immune status is a persistent, sex-dependent predictor of glucose intolerance and neurobehavioral function in adult PAE offspring.
Collapse
Affiliation(s)
- Shameena Bake
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Marisa R Pinson
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Sivani Pandey
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Joanna P Chambers
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Roxanna Mota
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Ashlyn E Fairchild
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Rajesh C Miranda
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA.
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA.
| |
Collapse
|
37
|
Keirns BH, Sciarrillo CM, Koemel NA, Emerson SR. Fasting, non-fasting and postprandial triglycerides for screening cardiometabolic risk. J Nutr Sci 2021; 10:e75. [PMID: 34589207 PMCID: PMC8453457 DOI: 10.1017/jns.2021.73] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/23/2022] Open
Abstract
Fasting triacylglycerols have long been associated with cardiovascular disease (CVD) and other cardiometabolic conditions. Evidence suggests that non-fasting triglycerides (i.e. measured within 8 h of eating) better predict CVD than fasting triglycerides, which has led several organisations to recommend non-fasting lipid panels as the new clinical standard. However, unstandardised assessment protocols associated with non-fasting triglyceride measurement may lead to misclassification, with at-risk individuals being overlooked. A third type of triglyceride assessment, postprandial testing, is more controlled, yet historically has been difficult to implement due to the time and effort required to execute it. Here, we review differences in assessment, the underlying physiology and the pathophysiological relevance of elevated fasting, non-fasting and postprandial triglycerides. We also present data suggesting that there may be a distinct advantage of postprandial triglycerides, even over non-fasting triglycerides, for early detection of CVD risk and offer suggestions to make postprandial protocols more clinically feasible.
Collapse
Affiliation(s)
- Bryant H. Keirns
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK74075, USA
| | | | - Nicholas A. Koemel
- Boden Collaboration for Obesity, Nutrition, Exercise and Eating Disorders, The University of Sydney, Sydney, NSW2006, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW2006, Australia
| | - Sam R. Emerson
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK74075, USA
| |
Collapse
|
38
|
Zuccaro A, Zapatería B, Sánchez-Alonso MG, Haro M, Limones M, Terrados G, Izquierdo A, Corrales P, Medina-Gómez G, Herradón G, Sevillano J, Ramos-Álvarez MDP. Pleiotrophin Deficiency Induces Browning of Periovarian Adipose Tissue and Protects against High-Fat Diet-Induced Hepatic Steatosis. Int J Mol Sci 2021; 22:9261. [PMID: 34502170 PMCID: PMC8431550 DOI: 10.3390/ijms22179261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/14/2021] [Accepted: 08/20/2021] [Indexed: 01/14/2023] Open
Abstract
(1) Background: Pleiotrophin preserves insulin sensitivity, regulates adipose tissue lipid turnover and plasticity, energy metabolism and thermogenesis. The aim of this study was to determine the role of pleiotrophin in hepatic lipid metabolism and in the metabolic crosstalk between the liver and brown and white adipose tissue (AT) in a high-fat diet-induced (HFD) obesity mice model. (2) Methods: We analyzed circulating variables, lipid metabolism (hepatic lipid content and mRNA expression), brown AT thermogenesis (UCP-1 expression) and periovarian AT browning (brown adipocyte markers mRNA and immunodetection) in Ptn-/- mice either fed with standard-chow diet or with HFD and in their corresponding Ptn+/+ counterparts. (3) Results: HFD-Ptn-/- mice are protected against the development of HFD-induced insulin resistance, had lower liver lipid content and lower expression of the key enzymes involved in triacylglycerides and fatty acid synthesis in liver. HFD-Ptn-/- mice showed higher UCP-1 expression in brown AT. Moreover, Ptn deletion increased the expression of specific markers of brown/beige adipocytes and was associated with the immunodetection of UCP-1 enriched multilocular adipocytes in periovarian AT. (4) Conclusions: Ptn deletion protects against the development of HFD-induced insulin resistance and liver steatosis, by increasing UCP-1 expression in brown AT and promoting periovarian AT browning.
Collapse
Affiliation(s)
- Agata Zuccaro
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - Begoña Zapatería
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - María Gracia Sánchez-Alonso
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - María Haro
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - María Limones
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - Gloria Terrados
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - Adriana Izquierdo
- Department of Basic Sciences of Health, Facultad Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (A.I.); (P.C.); (G.M.-G.)
| | - Patricia Corrales
- Department of Basic Sciences of Health, Facultad Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (A.I.); (P.C.); (G.M.-G.)
| | - Gema Medina-Gómez
- Department of Basic Sciences of Health, Facultad Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (A.I.); (P.C.); (G.M.-G.)
| | - Gonzalo Herradón
- Department of Pharmaceutical and Health Sciences, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain;
| | - Julio Sevillano
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - María del Pilar Ramos-Álvarez
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| |
Collapse
|
39
|
Matli B, Schulz A, Koeck T, Falter T, Lotz J, Rossmann H, Pfeiffer N, Beutel M, Münzel T, Strauch K, Wild PS, Lackner KJ. Distribution of HOMA-IR in a population-based cohort and proposal for reference intervals. Clin Chem Lab Med 2021; 59:1844-1851. [PMID: 34380182 DOI: 10.1515/cclm-2021-0643] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/26/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Insulin resistance (IR) is a hallmark of type 2 diabetes mellitus (DM). The homeostatic model assessment of insulin resistance (HOMA-IR) provides an estimate for IR from fasting glucose and insulin serum concentrations. The aim of this study was to obtain a reference interval for HOMA-IR for a specific insulin immunoassay. METHODS The Gutenberg Health Study (GHS) is a population-based, prospective, single-center cohort study in Germany with 15,030 participants aged 35-74 years. Fasting glucose, insulin, and C-peptide were available in 10,340 participants. HOMA-IR was calculated in this group and three reference subgroups with increasingly more stringent inclusion criteria. Age- and sex-dependent distributions of HOMA-IR and reference intervals were obtained. In a substudy three insulin assays were compared and HOMA-IR estimated for each assay. RESULTS Among the 10,340 participants analyzed there were 6,590 non-diabetic, 2,901 prediabetic, and 849 diabetic individuals. Median (interquartile range [IQR]) HOMA-IR was 1.54 (1.13/2.19), 2.00 (1.39/2.99), and 4.00 (2.52/6.51), respectively. The most stringently selected reference group consisted of 1,065 persons. Median (IQR) HOMA-IR was 1.09 (0.85/1.42) with no significant difference between men and women. The 97.5th percentile was 2.35. There was a non-significant trend towards higher values with older age. Comparison of three immunoassays for insulin showed an unsatisfactory correlation among the assays and systematic differences in calculated HOMA-IR. CONCLUSIONS We present HOMA-IR reference intervals for adults derived by more or less stringent selection criteria for the reference cohort. In addition we show that assay specific reference intervals for HOMA-IR are required.
Collapse
Affiliation(s)
- Bassel Matli
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Andreas Schulz
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center, Mainz, Germany
| | - Thomas Koeck
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center, Mainz, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| | - Tanja Falter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Johannes Lotz
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Heidi Rossmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center Mainz, Mainz, Germany
| | - Manfred Beutel
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center, Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology - Cardiology I, University Medical Center, Mainz, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| | - Konstantin Strauch
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center, MainzGermany
| | - Philipp S Wild
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center, Mainz, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Mainz, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| |
Collapse
|
40
|
Changes in abdominal subcutaneous adipose tissue phenotype following menopause is associated with increased visceral fat mass. Sci Rep 2021; 11:14750. [PMID: 34285301 PMCID: PMC8292317 DOI: 10.1038/s41598-021-94189-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/30/2021] [Indexed: 12/19/2022] Open
Abstract
Menopause is associated with a redistribution of adipose tissue towards central adiposity, known to cause insulin resistance. In this cross-sectional study of 33 women between 45 and 60 years, we assessed adipose tissue inflammation and morphology in subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) across menopause and related this to menopausal differences in adipose tissue distribution and insulin resistance. We collected paired SAT and VAT biopsies from all women and combined this with anthropometric measurements and estimated whole-body insulin sensitivity. We found that menopause was associated with changes in adipose tissue phenotype related to metabolic dysfunction. In SAT, postmenopausal women showed adipocyte hypertrophy, increased inflammation, hypoxia and fibrosis. The postmenopausal changes in SAT was associated with increased visceral fat accumulation. In VAT, menopause was associated with adipocyte hypertrophy, immune cell infiltration and fibrosis. The postmenopausal changes in VAT phenotype was associated with decreased insulin sensitivity. Based on these findings we suggest, that menopause is associated with changes in adipose tissue phenotype related to metabolic dysfunction in both SAT and VAT. Whereas increased SAT inflammation in the context of menopause is associated with VAT accumulation, VAT morphology is related to insulin resistance.
Collapse
|
41
|
Miah P, Mohona SBS, Rahman MM, Subhan N, Khan F, Hossain H, Sharker SM, Alam MA. Supplementation of cumin seed powder prevents oxidative stress, hyperlipidemia and non-alcoholic fatty liver in high fat diet fed rats. Biomed Pharmacother 2021; 141:111908. [PMID: 34328087 DOI: 10.1016/j.biopha.2021.111908] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/22/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
The present investigation was an attempt to evaluate the hypoglycemic, lipid-lowering, antioxidant and hepatoprotective effects of cumin (Cuminum cyminum family: Apiaceae) supplementation in high fat (HF) diet fed rats. Male Wistar rats were divided into four groups, such as control, control+ cumin, HF and HF+ cumin. Oral glucose tolerance test, plasma lipids, oxidative stress parameters, antioxidant enzymes activities, and liver dysfunction marker enzyme activities were evaluated. Additionally, histological staining of liver tissue was performed to evaluate the inflammatory cells infiltration, iron deposition and fibrosis. The current investigation demonstrated that 1% (w/w) supplementation of cumin powder significantly reduced HF diet-induced glucose intolerance, epididymal and mesenteric fat wet weights and lipid parameters like triglycerides, total cholesterol and low-density lipoproteins. Oxidative stress-related biomarkers including thiobarbituric acid reactive substances (TBARS), nitric oxide (NO) and advanced oxidation protein product (APOP) were also reduced by cumin supplementation. Moreover, HF-diet increased the activity of hepatic biomarker enzymes such as alanine transaminase (ALT) and alkaline phosphatase (ALP) activities which were significantly reduced by cumin powder supplementation. On the other hand, cumin powder supplementation was able to restore the reduced glutathione level with parallel augmentation of the antioxidant enzymes activities such as superoxide dismutase (SOD) and catalase in liver of HF diet-fed rats. Additionally, histological assessments confirmed that cumin powder supplementation also normalized the fat droplet deposition and inflammatory cells infiltration in the liver of HF diet-fed rats. This study suggests that cumin powder supplementation ameliorates dyslipidemia, oxidative stress and hepatic damage in HF diet-fed rats.
Collapse
Affiliation(s)
- Pintu Miah
- Department of Pharmaceutical Sciences, North South University Bangladesh, Bangladesh
| | | | - Md Mizanur Rahman
- Department of Pharmaceutical Sciences, North South University Bangladesh, Bangladesh
| | - Nusrat Subhan
- Department of Pharmaceutical Sciences, North South University Bangladesh, Bangladesh.
| | - Ferdous Khan
- Department of Pharmaceutical Sciences, North South University Bangladesh, Bangladesh
| | - Hemayet Hossain
- BCSIR Laboratories, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh
| | - Shazid Md Sharker
- Department of Pharmaceutical Sciences, North South University Bangladesh, Bangladesh
| | - Md Ashraful Alam
- Department of Pharmaceutical Sciences, North South University Bangladesh, Bangladesh.
| |
Collapse
|
42
|
Cifarelli V, Appak-Baskoy S, Peche VS, Kluzak A, Shew T, Narendran R, Pietka KM, Cella M, Walls CW, Czepielewski R, Ivanov S, Randolph GJ, Augustin HG, Abumrad NA. Visceral obesity and insulin resistance associate with CD36 deletion in lymphatic endothelial cells. Nat Commun 2021; 12:3350. [PMID: 34099721 PMCID: PMC8184948 DOI: 10.1038/s41467-021-23808-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 05/13/2021] [Indexed: 12/18/2022] Open
Abstract
Disruption of lymphatic lipid transport is linked to obesity and type 2 diabetes (T2D), but regulation of lymphatic vessel function and its link to disease remain unclear. Here we show that intestinal lymphatic endothelial cells (LECs) have an increasing CD36 expression from lymphatic capillaries (lacteals) to collecting vessels, and that LEC CD36 regulates lymphatic integrity and optimizes lipid transport. Inducible deletion of CD36 in LECs in adult mice (Cd36ΔLEC) increases discontinuity of LEC VE-cadherin junctions in lacteals and collecting vessels. Cd36ΔLEC mice display slower transport of absorbed lipid, more permeable mesenteric lymphatics, accumulation of inflamed visceral fat and impaired glucose disposal. CD36 silencing in cultured LECs suppresses cell respiration, reduces VEGF-C-mediated VEGFR2/AKT phosphorylation and destabilizes VE-cadherin junctions. Thus, LEC CD36 optimizes lymphatic junctions and integrity of lymphatic lipid transport, and its loss in mice causes lymph leakage, visceral adiposity and glucose intolerance, phenotypes that increase risk of T2D. Genetic variants in CD36 have been associated with metabolic syndrome. Here, the authors found that lymphatic vessel integrity and lipid transport are influenced by CD36 expression, and lymphatic endothelial cell CD36 deficiency causes visceral obesity and insulin resistance, which are risk factors for metabolic syndrome and diabetes.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, USA.
| | - Sila Appak-Baskoy
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.,Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Vivek S Peche
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, USA
| | - Andrew Kluzak
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, USA
| | - Trevor Shew
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, USA
| | - Ramkumar Narendran
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, USA
| | - Kathryn M Pietka
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, USA
| | - Marina Cella
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Curtis W Walls
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, USA
| | - Rafael Czepielewski
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Stoyan Ivanov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Hellmut G Augustin
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.,Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Nada A Abumrad
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, USA. .,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, USA.
| |
Collapse
|
43
|
Tóth ME, Dukay B, Péter M, Balogh G, Szűcs G, Zvara Á, Szebeni GJ, Hajdu P, Sárközy M, Puskás LG, Török Z, Csont T, Vígh L, Sántha M. Male and Female Animals Respond Differently to High-Fat Diet and Regular Exercise Training in a Mouse Model of Hyperlipidemia. Int J Mol Sci 2021; 22:ijms22084198. [PMID: 33919597 PMCID: PMC8073713 DOI: 10.3390/ijms22084198] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 01/18/2023] Open
Abstract
Inappropriate nutrition and a sedentary lifestyle can lead to obesity, one of the most common risk factors for several chronic diseases. Although regular physical exercise is an efficient approach to improve cardiometabolic health, the exact cellular processes are still not fully understood. We aimed to analyze the morphological, gene expression, and lipidomic patterns in the liver and adipose tissues in response to regular exercise. Healthy (wild type on a normal diet) and hyperlipidemic, high-fat diet-fed (HFD-fed) apolipoprotein B-100 (APOB-100)-overexpressing mice were trained by treadmill running for 7 months. The serum concentrations of triglyceride and tumor necrosis factor α (TNFα), as well as the level of lipid accumulation in the liver, were significantly higher in HFD-fed APOB-100 males compared to females. However, regular exercise almost completely abolished lipid accumulation in the liver of hyperlipidemic animals. The expression level of the thermogenesis marker, uncoupling protein-1 (Ucp1), was significantly higher in the subcutaneous white adipose tissue of healthy females, as well as in the brown adipose tissue of HFD-fed APOB-100 females, compared to males. Lipidomic analyses revealed that hyperlipidemia essentially remodeled the lipidome of brown adipose tissue, affecting both the membrane and storage lipid fractions, which was partially restored by exercise in both sexes. Our results revealed more severe metabolic disturbances in HFD-fed APOB-100 males compared to females. However, exercise efficiently reduced the body weight, serum triglyceride levels, expression of pro-inflammatory factors, and hepatic lipid accumulation in our model.
Collapse
Affiliation(s)
- Melinda E. Tóth
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
- Correspondence: ; Tel.: +36-62-599-635
| | - Brigitta Dukay
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
- Doctoral School in Biology, University of Szeged, H-6726 Szeged, Hungary
| | - Mária Péter
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Gábor Balogh
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Gergő Szűcs
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, University of Szeged, H-6720 Szeged, Hungary; (G.S.); (M.S.); (T.C.)
| | - Ágnes Zvara
- Laboratory of Functional Genomics, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (Á.Z.); (G.J.S.); (L.G.P.)
| | - Gábor J. Szebeni
- Laboratory of Functional Genomics, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (Á.Z.); (G.J.S.); (L.G.P.)
| | - Petra Hajdu
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Márta Sárközy
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, University of Szeged, H-6720 Szeged, Hungary; (G.S.); (M.S.); (T.C.)
| | - László G. Puskás
- Laboratory of Functional Genomics, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (Á.Z.); (G.J.S.); (L.G.P.)
| | - Zsolt Török
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Tamás Csont
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, University of Szeged, H-6720 Szeged, Hungary; (G.S.); (M.S.); (T.C.)
| | - László Vígh
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Miklós Sántha
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| |
Collapse
|
44
|
Franczyk MP, He M, Yoshino J. Removal of Epididymal Visceral Adipose Tissue Prevents Obesity-Induced Multi-organ Insulin Resistance in Male Mice. J Endocr Soc 2021; 5:bvab024. [PMID: 33869980 PMCID: PMC8041347 DOI: 10.1210/jendso/bvab024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity is associated with insulin resistance, an important risk factor of type 2 diabetes, atherogenic dyslipidemia, and nonalcoholic fatty liver disease. The major purpose of this study was to test hypothesize that prophylactic removal of epididymal visceral adipose tissue (VAT) prevents obesity-induced multi-organ (liver, skeletal muscle, adipose tissue) insulin resistance. Accordingly, we surgically removed epididymal VAT pads from adult C57BL/6J mice and evaluated in vivo and cellular metabolic pathways involved in glucose and lipid metabolism following chronic high-fat diet (HFD) feeding. We found that VAT removal decreases HFD-induced body weight gain while increasing subcutaneous adipose tissue (SAT) mass. Strikingly, VAT removal prevents obesity-induced insulin resistance and hyperinsulinemia and markedly enhances insulin-stimulated AKT-phosphorylation at serine-473 (Ser473) and threonine-308 (Thr308) sites in SAT, liver, and skeletal muscle. VAT removal leads to decreases in plasma lipid concentrations and hepatic triglyceride (TG) content. In addition, VAT removal increases circulating adiponectin, a key insulin-sensitizing adipokine, whereas it decreases circulating interleukin 6, a pro-inflammatory adipokine. Consistent with these findings, VAT removal increases adenosine monophosphate-activated protein kinase C phosphorylation, a major downstream target of adiponectin signaling. Data obtained from RNA sequencing suggest that VAT removal prevents obesity-induced oxidative stress and inflammation in liver and SAT, respectively. Taken together, these findings highlight the metabolic benefits and possible action mechanisms of prophylactic VAT removal on obesity-induced insulin resistance and hepatosteatosis. Our results also provide important insight into understanding the extraordinary capability of adipose tissue to influence whole-body glucose and lipid metabolism as an active endocrine organ.
Collapse
Affiliation(s)
- Michael P Franczyk
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Mai He
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jun Yoshino
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
45
|
Keshavjee SH, Schwenger KJP, Yadav J, Jackson TD, Okrainec A, Allard JP. Factors Affecting Metabolic Outcomes Post Bariatric Surgery: Role of Adipose Tissue. J Clin Med 2021; 10:714. [PMID: 33670215 PMCID: PMC7916950 DOI: 10.3390/jcm10040714] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is an ever-growing public health crisis, and bariatric surgery (BS) has become a valuable tool in ameliorating obesity, along with comorbid conditions such as diabetes, dyslipidemia and hypertension. BS techniques have come a long way, leading to impressive improvements in the health of the majority of patients. Unfortunately, not every patient responds optimally to BS and there is no method that is sufficient to pre-operatively predict who will receive maximum benefit from this surgical intervention. This review focuses on the adipose tissue characteristics and related parameters that may affect outcomes, as well as the potential influences of insulin resistance, BMI, age, psychologic and genetic factors. Understanding the role of these factors may help predict who will benefit the most from BS.
Collapse
Affiliation(s)
- Sara H. Keshavjee
- Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA;
| | - Katherine J. P. Schwenger
- Division of Gastroenterology, Toronto General Hospital, University Health Network, Toronto, ON M5G 2N2, Canada;
| | - Jitender Yadav
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Timothy D. Jackson
- Division of General Surgery, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; (T.D.J.); (A.O.)
| | - Allan Okrainec
- Division of General Surgery, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; (T.D.J.); (A.O.)
| | - Johane P. Allard
- Division of Gastroenterology, Toronto General Hospital, University Health Network, Toronto, ON M5G 2N2, Canada;
| |
Collapse
|
46
|
Kabir F, Nahar K, Rahman MM, Mamun F, Lasker S, Khan F, Yasmin T, Akter KA, Subhan N, Alam MA. Etoricoxib treatment prevented body weight gain and ameliorated oxidative stress in the liver of high-fat diet-fed rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:33-47. [PMID: 32780227 DOI: 10.1007/s00210-020-01960-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/03/2020] [Indexed: 12/19/2022]
Abstract
The main focus of this study was to determine the role of etoricoxib in counterbalancing the oxidative stress, metabolic disturbances, and inflammation in high-fat (HF) diet-induced obese rats. To conduct this study, 28 male Wistar rats (weighing 190-210 g) were distributed randomly into four groups: control, control + etoricoxib, HF, and HF + etoricoxib. After 8 weeks of treatment with etoricoxib (200 mg/kg), all the animals were sacrificed followed by the collection of blood and tissue samples in order to perform biochemical tests along with histological staining on hepatic tissues. According to this study, etoricoxib treatment prevented the body weight gain in HF diet-fed rats. Furthermore, rats of HF + etoricoxib group exhibited better blood glucose tolerance than the rats of HF diet-fed group. In addition, etoricoxib also markedly normalized HF diet-mediated rise of hepatic enzyme activity. Etoricoxib treatment lowered the level of oxidative stress indicators significantly with a parallel augmentation of antioxidant enzyme activities. Furthermore, etoricoxib administration helped in preventing inflammatory cell invasion, collagen accumulation, and fibrotic catastrophe in HF diet-fed rats. The findings of the present work are suggestive of the helpful role of etoricoxib in deterring the metabolic syndrome as well as other deleterious pathological changes afflicting the HF diet-fed rats.
Collapse
Affiliation(s)
- Fariha Kabir
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Kamrun Nahar
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Md Mizanur Rahman
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Fariha Mamun
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Shoumen Lasker
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Ferdous Khan
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Tahmina Yasmin
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Khondker Ayesha Akter
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Nusrat Subhan
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Md Ashraful Alam
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh.
| |
Collapse
|
47
|
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: 905] [Impact Index Per Article: 181.0] [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
|
48
|
Abstract
Adipose, or fat, tissue (AT) was once considered an inert tissue that primarily existed to store lipids, and was not historically recognized as an important organ in the regulation and maintenance of health. With the rise of obesity and more rigorous research, AT is now recognized as a highly complex metabolic organ involved in a host of important physiological functions, including glucose homeostasis and a multitude of endocrine capabilities. AT dysfunction has been implicated in several disease states, most notably obesity, metabolic syndrome and type 2 diabetes. The study of AT has provided useful insight in developing strategies to combat these highly prevalent metabolic diseases. This review highlights the major functions of adipose tissue and the consequences that can occur when disruption of these functions leads to systemic metabolic dysfunction.
Collapse
Affiliation(s)
- Innocence Harvey
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Anik Boudreau
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Jacqueline M Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.,Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
49
|
Vila-Bedmar R, Cruces-Sande M, Arcones AC, Willemen HLDM, Prieto P, Moreno-Indias I, Díaz-Rodríguez D, Francisco S, Jaén RI, Gutiérrez-Repiso C, Heijnen CJ, Boscá L, Fresno M, Kavelaars A, Mayor F, Murga C. GRK2 levels in myeloid cells modulate adipose-liver crosstalk in high fat diet-induced obesity. Cell Mol Life Sci 2020; 77:4957-4976. [PMID: 31927610 PMCID: PMC11105060 DOI: 10.1007/s00018-019-03442-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023]
Abstract
Macrophages are key effector cells in obesity-associated inflammation. G protein-coupled receptor kinase 2 (GRK2) is highly expressed in different immune cell types. Using LysM-GRK2+/- mice, we uncover that a reduction of GRK2 levels in myeloid cells prevents the development of glucose intolerance and hyperglycemia after a high fat diet (HFD) through modulation of the macrophage pro-inflammatory profile. Low levels of myeloid GRK2 confer protection against hepatic insulin resistance, steatosis and inflammation. In adipose tissue, pro-inflammatory cytokines are reduced and insulin signaling is preserved. Macrophages from LysM-GRK2+/- mice secrete less pro-inflammatory cytokines when stimulated with lipopolysaccharide (LPS) and their conditioned media has a reduced pathological influence in cultured adipocytes or naïve bone marrow-derived macrophages. Our data indicate that reducing GRK2 levels in myeloid cells, by attenuating pro-inflammatory features of macrophages, has a relevant impact in adipose-liver crosstalk, thus preventing high fat diet-induced metabolic alterations.
Collapse
Affiliation(s)
- Rocío Vila-Bedmar
- Departamento de ciencias básicas de la salud, área de Bioquímica y Biología Molecular, Universidad Rey Juan Carlos (URJC), Madrid, Spain
| | - Marta Cruces-Sande
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (CSIC/UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Alba C Arcones
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (CSIC/UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Hanneke L D M Willemen
- Laboratory of Translational Immunology (LTI), University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Patricia Prieto
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Isabel Moreno-Indias
- CIBER de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Unidad de Endocrinología y Nutrición, Hospital Universitario Virgen de Victoria de Malaga, Universidad de Málaga, Málaga, Spain
| | - Daniel Díaz-Rodríguez
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (CSIC/UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain
| | - Sara Francisco
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (CSIC/UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain
| | - Rafael I Jaén
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Carolina Gutiérrez-Repiso
- CIBER de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Unidad de Endocrinología y Nutrición, Hospital Universitario Virgen de Victoria de Malaga, Universidad de Málaga, Málaga, Spain
| | - Cobi J Heijnen
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lisardo Boscá
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Manuel Fresno
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (CSIC/UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | | | - Federico Mayor
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (CSIC/UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain.
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Cristina Murga
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (CSIC/UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain.
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
50
|
Tanaka M, Okada H, Hashimoto Y, Kumagai M, Nishimura H, Fukui M. Intraperitoneal, but not retroperitoneal, visceral adipose tissue is associated with diabetes mellitus: a cross-sectional, retrospective pilot analysis. Diabetol Metab Syndr 2020; 12:103. [PMID: 33292449 PMCID: PMC7691054 DOI: 10.1186/s13098-020-00612-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
AIM Diabetes mellitus (DM) is associated with adverse outcomes, and visceral adipose tissue (VAT), classified into intraperitoneal VAT (IVAT) and retroperitoneal VAT (RVAT), is associated with insulin resistance. This study aimed to evaluate the association of IVAT and RVAT with the prevalence or incidence of DM. METHODS In this cross-sectional, retrospective, cohort study, the prevalence and incidence of DM was analyzed in 803 and 624 middle-aged Japanese participants, respectively. The cross-sectional area of the abdominal adipose tissue was evaluated from an unenhanced computed tomography scan at the third lumbar vertebrae, and the IVAT or RVAT was analyzed using specialized software. The areas were normalized for the square value of the participants' height in meters and described as the IVAT or RVAT area index. RESULTS The IVAT area index (adjusted odds ratio [OR], 1.04; 95% confidence intervals [CI], 1.02-1.07, per 1.0 cm2/m2) or IVAT/RVAT area ratio (1.89; 1.23-2.85, per 1.0) was independently associated with the prevalence of DM, whereas the RVAT area index was not. During a follow-up (mean) of 3.7 years, 30 participants were diagnosed with DM. The IVAT area index (adjusted hazards ratio [HR], 1.02; 95% CI 1.003-1.04, per 1.0 cm2/m2) or IVAT/RVAT area ratio (2.25; 1.40-3.43, per 1.0) was independently associated with the incidence of DM, whereas the RVAT area index was not. CONCLUSIONS IVAT, but not RVAT, is associated with the prevalence or incidence of DM.
Collapse
Affiliation(s)
- Muhei Tanaka
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Hiroshi Okada
- Department of Internal Medicine, Matsushita Memorial Hospital, Osaka, Japan
| | - Yoshitaka Hashimoto
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Muneaki Kumagai
- Medical Corporation Soukenkai, Nishimura Clinic, Kyoto, Japan
| | | | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| |
Collapse
|