1
|
Oliveira IOD, Correa TL, Ferreira GD, Nunes BP, Gonçalves H, Wehrmeister FC, Menezes AMB. Association of inflammatory markers and multimorbidity in young adults: cross-sectional findings from the Pelotas (Brazil) birth cohort, 1993. CAD SAUDE PUBLICA 2025; 41:e00191623. [PMID: 39936780 PMCID: PMC11805519 DOI: 10.1590/0102-311xen191623] [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: 04/08/2024] [Revised: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 02/13/2025] Open
Abstract
Early diagnosis can be a beneficial factor for minimizing health risks related to multimorbidity. This study aims to assess the association of multimorbidity with interleukin 6 (IL-6), C-reactive protein (CRP), and adiponectin in 22-year-old participants of the Pelotas (Brazil) birth cohort. A total of 3,578 subjects had serum measurements of IL-6, CRP, and adiponectin at the 22-year-old visit. For multimorbidity evaluation, a list of 15 morbidities was used and divided into subgroups (cardiometabolic, pulmonary, allergic diseases, and mental disorders). The occurrence of ≥ 2 morbidities was higher in females (55.1%) than in males (45.2%). A negative association between multimorbidity and adiponectin was found in females, whereas positive associations between the number of diseases and IL-6 and CRP were observed in males. For both sexes, cardiometabolic problem was the morbidity most associated with the markers. The analysis for isolated diseases identified dyslipidemia was the only cardiometabolic condition associated with physiological markers. Our findings suggest an inverse association between multimorbidity and adiponectin in females, as well as a direct cumulative association between the number of diseases and IL-6 and CRP in males at a young age.
Collapse
Affiliation(s)
| | - Tulio L Correa
- Faculdade de Medicina, Universidade Federal de Pelotas, Pelotas, Brasil
| | - Gustavo Dias Ferreira
- Escola Superior de Educação Física e Fisioterapia, Universidade Federal de Pelotas, Pelotas, Brasil
| | | | - Helen Gonçalves
- Faculdade de Medicina, Universidade Federal de Pelotas, Pelotas, Brasil
| | | | | |
Collapse
|
2
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [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: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
3
|
Ahmed B, Farb MG, Gokce N. Cardiometabolic implications of adipose tissue aging. Obes Rev 2024; 25:e13806. [PMID: 39076025 DOI: 10.1111/obr.13806] [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: 08/21/2023] [Revised: 03/14/2024] [Accepted: 07/05/2024] [Indexed: 07/31/2024]
Abstract
Adipose tissue is a large endocrine organ that serves numerous physiological functions. As we age, adipose tissue remodels and can develop functional changes that alters its phenotype, potentially contributing to metabolic and cardiovascular disorders. Aging adipose tissue is characterized by regional redistribution of fat, accumulation of senescent cells, fibrosis, and decline in adipocyte differentiation capacities, which collectively impact adipose tissue function and whole body health. A notable transformation involves increased accumulation of intra-abdominal visceral adipose tissue and ectopic fat around internal organs such as the heart, blood vessels, liver, and kidneys that alter their functions. Other changes associated with aging include alterations in adipokine secretion and changes in adipocyte size and numbers. Aging adipocytes play a role in mediating chronic inflammation, metabolic dysfunction, and insulin resistance. Visceral adipose tissue, which increases in volume with aging, is in particular associated with inflammation, angiogenic dysfunction, and microvascular abnormalities, and mediators released by visceral fat may have adverse consequences systemically in multiple target organs, including the cardiovascular system. Understanding mechanisms underlying adipose tissue aging and its impact on cardiovascular health are important for developing interventions and treatments to promote healthy aging and reduce cardiometabolic disease risk.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Melissa G Farb
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Dronkers J, van Veldhuisen DJ, van der Meer P, Meems LMG. Heart Failure and Obesity: Unraveling Molecular Mechanisms of Excess Adipose Tissue. J Am Coll Cardiol 2024; 84:1666-1677. [PMID: 39415402 DOI: 10.1016/j.jacc.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
Obesity is an ongoing pandemic and is associated with the development of heart failure (HF), and especially HF with preserved ejection fraction. The definition of obesity is currently based on anthropometric measurements but neglects the location and molecular properties of excess fat. Important depots associated with HF development are subcutaneous adipose tissue and visceral adipose tissue, both located in the abdominal region, and epicardial adipose tissue (EAT) surrounding the myocardium. However, mechanisms linking these different adipose tissue depots to HF development are incompletely understood. EAT in particular is of great interest because of its close proximity to the heart. In this review, we therefore focus on the characteristics of different adipose tissue depots and their response to obesity. In addition, we evaluate how different mechanisms associated with EAT expansion potentially contribute to HF and in particular HF with preserved ejection fraction development.
Collapse
Affiliation(s)
- Just Dronkers
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Peter van der Meer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Laura M G Meems
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands.
| |
Collapse
|
5
|
Hird EJ, Slanina-Davies A, Lewis G, Hamer M, Roiser JP. From movement to motivation: a proposed framework to understand the antidepressant effect of exercise. Transl Psychiatry 2024; 14:273. [PMID: 38961071 PMCID: PMC11222551 DOI: 10.1038/s41398-024-02922-y] [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: 04/05/2023] [Revised: 03/28/2024] [Accepted: 05/10/2024] [Indexed: 07/05/2024] Open
Abstract
Depression is the leading cause of disability worldwide, exerting a profound negative impact on quality of life in those who experience it. Depression is associated with disruptions to several closely related neural and cognitive processes, including dopamine transmission, fronto-striatal brain activity and connectivity, reward processing and motivation. Physical activity, especially aerobic exercise, reduces depressive symptoms, but the mechanisms driving its antidepressant effects are poorly understood. Here we propose a novel hypothesis for understanding the antidepressant effects of exercise, centred on motivation, across different levels of explanation. There is robust evidence that aerobic exercise decreases systemic inflammation. Inflammation is known to reduce dopamine transmission, which in turn is strongly implicated in effort-based decision making for reward. Drawing on a broad range of research in humans and animals, we propose that by reducing inflammation and boosting dopamine transmission, with consequent effects on effort-based decision making for reward, exercise initially specifically improves 'interest-activity' symptoms of depression-namely anhedonia, fatigue and subjective cognitive impairment - by increasing propensity to exert effort. Extending this framework to the topic of cognitive control, we explain how cognitive impairment in depression may also be conceptualised through an effort-based decision-making framework, which may help to explain the impact of exercise on cognitive impairment. Understanding the mechanisms underlying the antidepressant effects of exercise could inform the development of novel intervention strategies, in particular personalised interventions and boost social prescribing.
Collapse
Affiliation(s)
- E J Hird
- Institute of Cognitive Neuroscience, University College London, London, UK.
| | - A Slanina-Davies
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - G Lewis
- Division of Psychiatry, University College London, London, UK
| | - M Hamer
- Institute of Sport, Exercise and Health, University College London, London, UK
| | - J P Roiser
- Institute of Cognitive Neuroscience, University College London, London, UK
| |
Collapse
|
6
|
Reed JN, Huang J, Li Y, Ma L, Banka D, Wabitsch M, Wang T, Ding W, Björkegren JL, Civelek M. Systems genetics analysis of human body fat distribution genes identifies adipocyte processes. Life Sci Alliance 2024; 7:e202402603. [PMID: 38702075 PMCID: PMC11068934 DOI: 10.26508/lsa.202402603] [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: 01/18/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
Excess abdominal fat is a sexually dimorphic risk factor for cardio-metabolic disease and is approximated by the waist-to-hip ratio adjusted for body mass index (WHRadjBMI). Whereas this trait is highly heritable, few causal genes are known. We aimed to identify novel drivers of WHRadjBMI using systems genetics. We used two independent cohorts of adipose tissue gene expression and constructed sex- and depot-specific Bayesian networks to model gene-gene interactions from 8,492 genes. Using key driver analysis, we identified genes that, in silico and putatively in vitro, regulate many others. 51-119 key drivers in each network were replicated in both cohorts. In other cell types, 23 of these genes are found in crucial adipocyte pathways: Wnt signaling or mitochondrial function. We overexpressed or down-regulated seven key driver genes in human subcutaneous pre-adipocytes. Key driver genes ANAPC2 and RSPO1 inhibited adipogenesis, whereas PSME3 increased adipogenesis. RSPO1 increased Wnt signaling activity. In differentiated adipocytes, MIGA1 and UBR1 down-regulation led to mitochondrial dysfunction. These five genes regulate adipocyte function, and we hypothesize that they regulate fat distribution.
Collapse
Affiliation(s)
- Jordan N Reed
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jiansheng Huang
- Novo Nordisk Research Center China, Novo Nordisk A/S, Beijing, China
| | - Yong Li
- Novo Nordisk Research Center China, Novo Nordisk A/S, Beijing, China
| | - Lijiang Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dhanush Banka
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Martin Wabitsch
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Centre, Ulm, Germany
| | - Tianfang Wang
- Novo Nordisk Research Center China, Novo Nordisk A/S, Beijing, China
| | - Wen Ding
- Novo Nordisk Research Center China, Novo Nordisk A/S, Beijing, China
| | - Johan Lm Björkegren
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Mete Civelek
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
7
|
Shen H, He Y, Lu F, Lu X, Yang B, Liu Y, Guo Q. Association of ratios of visceral fat area/subcutaneous fat area and muscle area/standard body weight at T12 CT level with the prognosis of acute respiratory distress syndrome. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:106-118. [PMID: 39169930 PMCID: PMC11332858 DOI: 10.1016/j.pccm.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Indexed: 08/23/2024]
Abstract
Background It is well-known that body composition metrics can influence the prognosis of various diseases. This study investigated how body composition metrics predict acute respiratory distress syndrome (ARDS) prognosis, focusing on the ratio of visceral fat area (VFA) to subcutaneous fat area (SFA), SFA to standard body weight (SBW), VFA to SBW, and muscle area (MA) to SBW. These metrics were assessed at the level of the twelfth thoracic vertebra (T12 computed tomography [CT] level) to determine their correlation with the outcomes of ARDS. The goal was to utilize these findings to refine and personalize treatment strategies for ARDS. Methods Patients with ARDS admitted to the intensive care units (ICUs) of three hospitals from January 2016 to July 2023 were enrolled in this study. Within 24 hours of ARDS onset, we obtained chest CT scans to measure subcutaneous fat, visceral fat, and muscle area at the T12 level. We then compared these ratios between survivors and non-survivors. Logistic regression was employed to identify prognostic risk factors. Receiver operating characteristic (ROC) curve analysis was utilized to determine the optimal cutoff for predictors of in-hospital mortality. Based on this cutoff, patients with ARDS were stratified. To reduce confounding factors, 1:1 propensity score matching (PSM) was applied. We conducted analyses of clinical feature and prognostic differences pre- and post-PSM between the stratified groups. Additionally, Kaplan-Meier survival curves were generated to compare the survival outcomes of these groups. Results Of 258 patients with ARDS, 150 survived and 108 did not. Non-survivors had a higher VFA/SFA ratio (P <0.001) and lower SFA/SBW and MA/SBW ratios (both P <0.001). Key risk factors were high VFA/SFA ratio (OR=2.081; P=0.008), age, acute physiology and chronic health evaluation (APACHE) II score, and lactate levels, while MA/SBW and albumin were protective. Patients with a VFA/SFA ratio ≥0.73 were associated with increased mortality, while those with an MA/SBW ratio >1.55 cm²/kg had lower mortality, both pre- and post-PSM (P=0.001 and P <0.001, respectively). Among 170 patients with pulmonary-origin ARDS, 87 survived and 83 did not. The non-survivor group showed a higher VFA/SFA ratio (P <0.001) and lower SFA/SBW and MA/SBW (P=0.003, P <0.001, respectively). Similar risk and protective factors were observed in this cohort. For VFA/SFA, a value above the cutoff of 1.01 predicted higher mortality, while an MA/SBW value below the cutoff of 1.48 cm²/kg was associated with increased mortality (both P <0.001 pre-/post-PSM). Conclusions Among all patients with ARDS, the VFA to SFA ratio, MA to SBW ratio at the T12 level, age, APACHE II score, and lactate levels emerged as independent risk factors for mortality.
Collapse
Affiliation(s)
- Hui Shen
- Department of Emergency, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu 215000, China
| | - Ying He
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu 215000, China
| | - Fan Lu
- Department of Emergency, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu 215000, China
| | - Xiaoting Lu
- Department of Emergency, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu 215000, China
| | - Bining Yang
- Department of Emergency, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu 215000, China
| | - Yi Liu
- Department of Emergency, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu 215000, China
| | - Qiang Guo
- Department of Emergency, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu 215000, China
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University (Suzhou Dushu Lake Hospital), Suzhou, Jiangsu 215000, China
- Institute of Critical Care Medicine, Soochow University, Suzhou, Jiangsu 215000, China
- Medical Center of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| |
Collapse
|
8
|
Chand S, Tripathi AS, Dewani AP, Sheikh NWA. Molecular targets for management of diabetes: Remodelling of white adipose to brown adipose tissue. Life Sci 2024; 345:122607. [PMID: 38583857 DOI: 10.1016/j.lfs.2024.122607] [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: 12/24/2023] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/09/2024]
Abstract
Diabetes mellitus is a disorder characterised metabolic dysfunction that results in elevated glucose level in the bloodstream. Diabetes is of two types, type1 and type 2 diabetes. Obesity is considered as one of the major reasons intended for incidence of diabetes hence it turns out to be essential to study about the adipose tissue which is responsible for fat storage in body. Adipose tissues play significant role in maintaining the balance between energy stabilization and homeostasis. The three forms of adipose tissue are - White adipose tissue (WAT), Brown adipose tissue (BAT) and Beige adipose tissue (intermediate form). The amount of BAT gets reduced, and WAT starts to increase with the age. WAT when exposed to certain stimuli gets converted to BAT by the help of certain transcriptional regulators. The browning of WAT has been a matter of study to treat the metabolic disorders and to initiate the expenditure of energy. The three main regulators responsible for the browning of WAT are PRDM16, PPARγ and PGC-1α via various cellular and molecular mechanism. Presented review article includes the detailed elaborative aspect of genes and proteins involved in conversion of WAT to BAT.
Collapse
Affiliation(s)
- Shushmita Chand
- Amity Institute of Pharmacy, Amity University, Sector 125, Noida, Uttar Pradesh, India
| | - Alok Shiomurti Tripathi
- Department of Pharmacology, ERA College of Pharmacy, ERA University, Lucknow, Uttar Pradesh, India.
| | - Anil P Dewani
- Department of Pharmacology, P. Wadhwani College of Pharmacy, Yavatmal, Maharashtra, India
| | | |
Collapse
|
9
|
Lewis MY, Yonemori K, Ross A, Wilkens LR, Shepherd J, Cassel K, Stenger A, Rettenmeier C, Lim U, Boushey C, Le Marchand L. Effect of Intermittent vs. Continuous Energy Restriction on Visceral Fat: Protocol for The Healthy Diet and Lifestyle Study 2 (HDLS2). Nutrients 2024; 16:1478. [PMID: 38794715 PMCID: PMC11123735 DOI: 10.3390/nu16101478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Obesity in the United States and Western countries represents a major health challenge associated with an increased risk of metabolic diseases, including cardiovascular disease, hypertension, diabetes, and certain cancers. Our past work revealed a more pronounced obesity-cancer link in certain ethnic groups, motivating us to develop a tailored dietary intervention called the Healthy Diet and Lifestyle 2 (HDLS2). The study protocol is described herein for this randomized six-month trial examining the effects of intermittent energy restriction (5:2 Diet) plus the Mediterranean dietary pattern (IER + MED) on visceral adipose tissue (VAT), liver fat, and metabolic biomarkers, compared to a standard MED with daily energy restriction (DER + MED), in a diverse participant group. Using MRI and DXA scans for body composition analysis, as well as metabolic profiling, this research aims to contribute to nutritional guidelines and strategies for visceral obesity reduction. The potential benefits of IER + MED, particularly regarding VAT reduction and metabolic health improvement, could be pivotal in mitigating the obesity epidemic and its metabolic sequelae. The ongoing study will provide essential insights into the efficacy of these energy restriction approaches across varied racial/ethnic backgrounds, addressing an urgent need in nutrition and metabolic health research. Registered Trial, National Institutes of Health, ClinicalTrials.gov (NCT05132686).
Collapse
Affiliation(s)
- Michelle Y. Lewis
- Population Sciences in the Pacific Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| | - Kim Yonemori
- Population Sciences in the Pacific Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| | - Alison Ross
- Population Sciences in the Pacific Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| | - Lynne R. Wilkens
- Population Sciences in the Pacific Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| | - John Shepherd
- Population Sciences in the Pacific Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| | - Kevin Cassel
- Population Sciences in the Pacific Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| | - Andrew Stenger
- MRI Research Center, John A. Burns School of Medicine, University of Hawai’i, Honolulu, Hi 96813, USA
| | - Christoph Rettenmeier
- MRI Research Center, John A. Burns School of Medicine, University of Hawai’i, Honolulu, Hi 96813, USA
| | - Unhee Lim
- Population Sciences in the Pacific Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| | - Carol Boushey
- Population Sciences in the Pacific Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| | - Loïc Le Marchand
- Population Sciences in the Pacific Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| |
Collapse
|
10
|
Subramanian P, Prabhu V, Nehru M, Palanirasu R, Janardhanan R. Association of indoleamine 2,3 dioxygenase, brain derived neurotrophic factor and cellular senescence in type 2 diabetes mellitus with depression: a clinical approach. Mol Biol Rep 2024; 51:481. [PMID: 38578530 DOI: 10.1007/s11033-024-09435-3] [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: 12/04/2023] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND AND AIM Type 2 diabetes mellitus (T2DM) and depression are often linked. Several studies have reported the role of molecular markers either in diabetes or depression. The present study aimed at molecular level profiling of Indoleamine-2,3-dioxygenase (IDO), brain-derived neurotrophic factor (BDNF) and cellular senescence in patients with type 2 diabetes with and without depression compared to individuals with healthy controls. METHODS A total of 120 individuals diagnosed with T2DM were enlisted for the study, with a subset of participants with and without exhibiting depression. The gene expression analysis was done using quantitative real-time PCR. RESULTS Indoleamine 2,3 dioxygenase (p < 0.001) and senescence genes (p < 0.001) were significantly upregulated, while brain derived neurotrophic factor (p < 0.01) was significantly downregulated in T2DM patients comorbid with and without depression when compared to healthy controls. CONCLUSION Indoleamine 2,3 dioxygenase, Brain derived neurotrophic factor and cellular senescence may play a role in the progression of the disease. The aforementioned discoveries offer significant contributions to our understanding of the molecular mechanisms that underlie T2DM with depression, potentially aiding in the advancement of prediction and diagnostic methods for this particular ailment.
Collapse
Affiliation(s)
- Prasanth Subramanian
- Department of Medical Research, SRM Medical College Hospital & Research Centre, SRMIST, Kattankulathur, Chennai, Tamil Nadu, India
| | - Venkataraman Prabhu
- Department of Medical Research, SRM Medical College Hospital & Research Centre, SRMIST, Kattankulathur, Chennai, Tamil Nadu, India.
| | - Mohanraj Nehru
- Department of Medical Research, SRM Medical College Hospital & Research Centre, SRMIST, Kattankulathur, Chennai, Tamil Nadu, India
| | - Rajapriya Palanirasu
- Department of Transfusion Medicine, HLA and Transport Immunology, Dr Rela Institute and Medical Centre, Chromepet, Chennai, Tamil Nadu, India
| | - Rajiv Janardhanan
- Department of Medical Research, SRM Medical College Hospital & Research Centre, SRMIST, Kattankulathur, Chennai, Tamil Nadu, India
| |
Collapse
|
11
|
Menezes CCD, Barbirato DDS, Fogacci MF, Marañón-Vásquez GA, Carneiro JRI, Maia LC, Barros MCMD. Systemic benefits of periodontal therapy in patients with obesity and periodontitis: a systematic review. Braz Oral Res 2024; 38:e031. [PMID: 38597549 PMCID: PMC11376685 DOI: 10.1590/1807-3107bor-2024.vol38.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/09/2023] [Indexed: 04/11/2024] Open
Abstract
This systematic review aimed to answer the focused question: "What are the benefits of subgingival periodontal therapy on blood hematological and biochemical index, biomarkers of inflammation and oxidative stress, quality of life, and periodontal pathogen counts in patients with obesity and periodontitis?". A systematic literature search was performed in six databases: PubMed, Embase, LILACS, Web of Science, Cochrane and SCOPUS and other sources, and a manual search was conducted as well. Inclusion criteria were randomized and non-randomized clinical trials, and before-and-after studies on patients with obesity subjected to periodontal therapy. The results were synthesized qualitatively. Risk of bias within studies was assessed using RoB 2 and ROBINS-I tools. The certainty of evidence was evaluated following the GRADE approach. Three randomized controlled trials and 15 before-and-after studies were included. Randomized controlled trials were considered to have a low risk of bias, as compared to before-and-after studies assessed as having low, serious, and critical risks of bias. Non-surgical periodontal therapy plus azithromycin, chlorhexidine, and cetylpyridinium chloride reduced blood pressure and decreased serum levels of HbA1c, hsCRP, IL-1β, and TNF-α. Salivary resistin level also decreased in patients with obesity and periodontitis after therapy and chlorhexidine mouth rinse. Before-and-after data suggest an improvement in total cholesterol, LDL, triglycerides, insulin resistance, C3, GCF levels of TNF-α, chemerin, vaspin, omentin-1, visfatin, 8-OHdG, and periodontal pathogen counts after therapy.
Collapse
Affiliation(s)
- Cláudia Callegaro de Menezes
- Universidade Federal do Rio de Janeiro - UFRJ, Dental School, Division of Periodontics, Rio de Janeiro, RJ, Brazil
| | - Davi da Silva Barbirato
- Universidade Federal do Rio de Janeiro - UFRJ, Dental School, Division of Periodontics, Rio de Janeiro, RJ, Brazil
| | - Mariana Fampa Fogacci
- Universidade Federal de Pernambuco - UFPE, Department of Clinical and Preventive Dentistry, Recife, PE, Brazil
| | | | - João Régis Ivar Carneiro
- Universidade Federal do Rio de Janeiro - UFRJ, Clementino Fraga Filho Hospital University, Department of Nutrology/Bariatric Surgery, Rio de Janeiro, RJ, Brazil
| | - Lucianne Copple Maia
- Universidade Federal do Rio de Janeiro - UFRJ, Department of Pediatric Dentistry and Orthodontics, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
12
|
Gutowska K, Koźniewski K, Wąsowski M, Jonas MI, Bartoszewicz Z, Lisik W, Jonas M, Binda A, Jaworski P, Tarnowski W, Noszczyk B, Puzianowska-Kuźnicka M, Czajkowski K, Kuryłowicz A. AGER-1 Long Non-Coding RNA Levels Correlate with the Expression of the Advanced Glycosylation End-Product Receptor, a Regulator of the Inflammatory Response in Visceral Adipose Tissue of Women with Obesity and Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:17447. [PMID: 38139276 PMCID: PMC10743952 DOI: 10.3390/ijms242417447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
The advanced glycosylation end-product receptor (AGER) is involved in the development of metabolic inflammation and related complications in type 2 diabetes mellitus (T2DM). Tissue expression of the AGER gene (AGER) is regulated by epigenetic mediators, including a long non-coding RNA AGER-1 (lncAGER-1). This study aimed to investigate whether human obesity and T2DM are associated with an altered expression of AGER and lncAGER-1 in adipose tissue and, if so, whether these changes affect the local inflammatory milieu. The expression of genes encoding AGER, selected adipokines, and lncAGER-1 was assessed using real-time PCR in visceral (VAT) and subcutaneous (SAT) adipose tissue. VAT and SAT samples were obtained from 62 obese (BMI > 40 kg/m2; N = 24 diabetic) and 20 normal weight (BMI = 20-24.9 kg/m2) women, while a further 15 SAT samples were obtained from patients who were 18 to 24 months post-bariatric surgery. Tissue concentrations of adipokines were measured at the protein level using an ELISA-based method. Obesity was associated with increased AGER mRNA levels in SAT compared to normal weight status (p = 0.04) and surgical weight loss led to their significant decrease compared to pre-surgery levels (p = 0.01). Stratification by diabetic status revealed that AGER mRNA levels in VAT were higher in diabetic compared to non-diabetic women (p = 0.018). Elevated AGER mRNA levels in VAT of obese diabetic patients correlated with lncAGER-1 (p = 0.04, rs = 0.487) and with interleukin 1β (p = 0.008, rs = 0.525) and resistin (p = 0.004, rs = 0.6) mRNA concentrations. In conclusion, obesity in women is associated with increased expression of AGER in SAT, while T2DM is associated with increased AGER mRNA levels and pro-inflammatory adipokines in VAT.
Collapse
Affiliation(s)
- Klaudia Gutowska
- II Department of Obstetrics and Gynecology, Warsaw Medical University, 00-315 Warsaw, Poland; (K.G.); (K.C.)
| | - Krzysztof Koźniewski
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (K.K.); (M.I.J.); (M.P.-K.)
| | - Michał Wąsowski
- Department of General Medicine and Geriatric Cardiology, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland;
| | - Marta Izabela Jonas
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (K.K.); (M.I.J.); (M.P.-K.)
| | - Zbigniew Bartoszewicz
- Department of Internal Medicine and Endocrinology, The Medical University of Warsaw, 02- 097 Warsaw, Poland;
| | - Wojciech Lisik
- Department of General and Transplantation Surgery, The Medical University of Warsaw, 02-005 Warsaw, Poland;
| | - Maurycy Jonas
- Department of General Surgery, Barska Hospital, 02-315 Warsaw, Poland;
| | - Artur Binda
- Department of General, Oncological and Bariatric Surgery, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland; (A.B.); (P.J.); (W.T.)
| | - Paweł Jaworski
- Department of General, Oncological and Bariatric Surgery, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland; (A.B.); (P.J.); (W.T.)
| | - Wiesław Tarnowski
- Department of General, Oncological and Bariatric Surgery, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland; (A.B.); (P.J.); (W.T.)
| | - Bartłomiej Noszczyk
- Department of Plastic Surgery, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland;
| | - Monika Puzianowska-Kuźnicka
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (K.K.); (M.I.J.); (M.P.-K.)
- Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, 01-826 Warsaw, Poland
| | - Krzysztof Czajkowski
- II Department of Obstetrics and Gynecology, Warsaw Medical University, 00-315 Warsaw, Poland; (K.G.); (K.C.)
| | - Alina Kuryłowicz
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (K.K.); (M.I.J.); (M.P.-K.)
- Department of General Medicine and Geriatric Cardiology, Medical Centre of Postgraduate Education, 00-401 Warsaw, Poland;
| |
Collapse
|
13
|
Hruska P, Kucera J, Kuruczova D, Buzga M, Pekar M, Holeczy P, Potesil D, Zdrahal Z, Bienertova-Vasku J. Unraveling adipose tissue proteomic landscapes in severe obesity: insights into metabolic complications and potential biomarkers. Am J Physiol Endocrinol Metab 2023; 325:E562-E580. [PMID: 37792298 PMCID: PMC10864023 DOI: 10.1152/ajpendo.00153.2023] [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: 05/17/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Abstract
In this study, we aimed to comprehensively characterize the proteomic landscapes of subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) in patients with severe obesity, to establish their associations with clinical characteristics, and to identify potential serum protein biomarkers indicative of tissue-specific alterations or metabolic states. We conducted a cross-sectional analysis of 32 patients with severe obesity (16 males and 16 females) of Central European descent who underwent bariatric surgery. Clinical parameters and body composition were assessed using dual-energy X-ray absorptiometry (DXA) and bioelectrical impedance, with 15 patients diagnosed with type 2 diabetes (T2D) and 17 with hypertension. Paired SAT and VAT samples, along with serum samples, were subjected to state-of-the-art proteomics liquid chromatography-mass spectrometry (LC-MS). Our analysis identified 7,284 proteins across SAT and VAT, with 1,249 differentially expressed proteins between the tissues and 1,206 proteins identified in serum. Correlation analyses between differential protein expression and clinical traits suggest a significant role of SAT in the pathogenesis of obesity and related metabolic complications. Specifically, the SAT proteomic profile revealed marked alterations in metabolic pathways and processes contributing to tissue fibrosis and inflammation. Although we do not establish a definitive causal relationship, it appears that VAT might respond to SAT metabolic dysfunction by potentially enhancing mitochondrial activity and expanding its capacity. However, when this adaptive response is exceeded, it could possibly contribute to insulin resistance (IR) and in some cases, it may be associated with the progression to T2D. Our findings provide critical insights into the molecular foundations of SAT and VAT in obesity and may inform the development of targeted therapeutic strategies.NEW & NOTEWORTHY This study provides insights into distinct proteomic profiles of subcutaneous adipose tissue (SAT), visceral adipose tissue (VAT), and serum in patients with severe obesity and their associations with clinical traits and body composition. It underscores SAT's crucial role in obesity development and related complications, such as insulin resistance (IR) and type 2 diabetes (T2D). Our findings emphasize the importance of understanding the SAT and VAT balance in energy homeostasis, proteostasis, and the potential role of SAT capacity in the development of metabolic disorders.
Collapse
Affiliation(s)
- Pavel Hruska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Jan Kucera
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Physical Activities and Health Sciences, Faculty of Sports Studies, Masaryk University, Brno, Czech Republic
| | - Daniela Kuruczova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Marek Buzga
- Department of Laboratory Medicine, University hospital Ostrava, Ostrava, Czech Republic
- Department of Physiology and Pathophysiology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Matej Pekar
- Vascular and Miniinvasive Surgery Center, Hospital AGEL Trinec-Podlesi, Trinec, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavol Holeczy
- Department of Surgery, Vitkovice Hospital, Ostrava, Czech Republic
- Department of Surgical Disciplines, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - David Potesil
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zbynek Zdrahal
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Julie Bienertova-Vasku
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Physical Activities and Health Sciences, Faculty of Sports Studies, Masaryk University, Brno, Czech Republic
| |
Collapse
|
14
|
Reed JN, Huang J, Li Y, Ma L, Banka D, Wabitsch M, Wang T, Ding W, Björkegren JLM, Civelek M. Systems genetics analysis of human body fat distribution genes identifies Wnt signaling and mitochondrial activity in adipocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.06.556534. [PMID: 37732278 PMCID: PMC10508754 DOI: 10.1101/2023.09.06.556534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
BACKGROUND Excess fat in the abdomen is a sexually dimorphic risk factor for cardio-metabolic disease. The relative storage between abdominal and lower-body subcutaneous adipose tissue depots is approximated by the waist-to-hip ratio adjusted for body mass index (WHRadjBMI). Genome-wide association studies (GWAS) identified 346 loci near 495 genes associated with WHRadjBMI. Most of these genes have unknown roles in fat distribution, but many are expressed and putatively act in adipose tissue. We aimed to identify novel sex- and depot-specific drivers of WHRadjBMI using a systems genetics approach. METHODS We used two independent cohorts of adipose tissue gene expression with 362 - 444 males and 147 - 219 females, primarily of European ancestry. We constructed sex- and depot- specific Bayesian networks to model the gene-gene interactions from 8,492 adipose tissue genes. Key driver analysis identified genes that, in silico and putatively in vitro, regulate many others, including the 495 WHRadjBMI GWAS genes. Key driver gene function was determined by perturbing their expression in human subcutaneous pre-adipocytes using lenti-virus or siRNA. RESULTS 51 - 119 key drivers in each network were replicated in both cohorts. We used single-cell expression data to select replicated key drivers expressed in adipocyte precursors and mature adipocytes, prioritized genes which have not been previously studied in adipose tissue, and used public human and mouse data to nominate 53 novel key driver genes (10 - 21 from each network) that may regulate fat distribution by altering adipocyte function. In other cell types, 23 of these genes are found in crucial adipocyte pathways: Wnt signaling or mitochondrial function. We selected seven genes whose expression is highly correlated with WHRadjBMI to further study their effects on adipogenesis/Wnt signaling (ANAPC2, PSME3, RSPO1, TYRO3) or mitochondrial function (C1QTNF3, MIGA1, PSME3, UBR1).Adipogenesis was inhibited in cells overexpressing ANAPC2 and RSPO1 compared to controls. RSPO1 results are consistent with a positive correlation between gene expression in the subcutaneous depot and WHRadjBMI, therefore lower relative storage in the subcutaneous depot. RSPO1 inhibited adipogenesis by increasing β-catenin activation and Wnt-related transcription, thus repressing PPARG and CEBPA. PSME3 overexpression led to more adipogenesis than controls. In differentiated adipocytes, MIGA1 and UBR1 downregulation led to mitochondrial dysfunction, with lower oxygen consumption than controls; MIGA1 knockdown also lowered UCP1 expression. SUMMARY ANAPC2, MIGA1, PSME3, RSPO1, and UBR1 affect adipocyte function and may drive body fat distribution.
Collapse
|
15
|
Chen VY, Siegfried LG, Tomic-Canic M, Stone RC, Pastar I. Cutaneous changes in diabetic patients: Primed for aberrant healing? Wound Repair Regen 2023; 31:700-712. [PMID: 37365017 PMCID: PMC10966665 DOI: 10.1111/wrr.13108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 06/28/2023]
Abstract
Cutaneous manifestations affect most patients with diabetes mellitus, clinically presenting with numerous dermatologic diseases from xerosis to diabetic foot ulcers (DFUs). Skin conditions not only impose a significantly impaired quality of life on individuals with diabetes but also predispose patients to further complications. Knowledge of cutaneous biology and the wound healing process under diabetic conditions is largely limited to animal models, and studies focusing on biology of the human condition of DFUs remain limited. In this review, we discuss the critical molecular, cellular, and structural changes to the skin in the hyperglycaemic and insulin-resistant environment of diabetes with a focus specifically on human-derived data. Elucidating the breadth of the cutaneous manifestations coupled with effective diabetes management is important for improving patient quality of life and averting future complications including wound healing disorders.
Collapse
Affiliation(s)
- Vivien Y Chen
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Lindsey G Siegfried
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Rivka C Stone
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
16
|
Song MK, Kim JE, Kim JT, Kang YE, Han SJ, Kim SH, Kim HJ, Ku BJ, Lee JH. GDF10 is related to obesity as an adipokine derived from subcutaneous adipose tissue. Front Endocrinol (Lausanne) 2023; 14:1159515. [PMID: 37529611 PMCID: PMC10390302 DOI: 10.3389/fendo.2023.1159515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/04/2023] [Indexed: 08/03/2023] Open
Abstract
Introduction Adipokines are proteins that are secreted by the adipose tissue. Although they are associated with obesity-related metabolic disorders, most studies have focused on adipokines expressed by visceral adipose tissue (VAT). This study aimed to identify the adipokine potentially derived from subcutaneous adipose tissue (SAT) and its clinical significance. Methods Samples of SAT and VAT were obtained from six adult male patients who underwent laparoscopic surgery for benign gall bladder disease. Differentially expressed genes were analyzed by subjecting the samples to RNA sequencing. The serum concentration of selected proteins according to body mass index (BMI) was analyzed in 58 individuals. Results GDF10 showed significantly higher expression in the SAT, as per RNA sequencing (fold change = 5.8, adjusted P value = 0.009). Genes related to insulin response, glucose homeostasis, lipid homeostasis, and fatty acid metabolism were suppressed when GDF10 expression was high in SAT, as per genotype-tissue expression data. The serum GDF10 concentration was higher in participants with BMI ≥ 25 kg/m2 (n = 35, 2674 ± 441 pg/mL) than in those with BMI < 25 kg/m2 (n = 23, 2339 ± 639 pg/mL; P = 0.022). There was a positive correlation between BMI and serum GDF10 concentration (r = 0.308, P = 0.019). Conclusions GDF10 expression was higher in SAT than in VAT. Serum GDF10 concentration was high in patients with obesity. Therefore, GDF10 could be a SAT-derived protein related to obesity.
Collapse
Affiliation(s)
- Mi Kyung Song
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Ji Eun Kim
- Research Center for Endocrine and Metabolic Disease, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jung Tae Kim
- Research Center for Endocrine and Metabolic Disease, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Yea Eun Kang
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Research Center for Endocrine and Metabolic Disease, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Sun Jong Han
- Department of General Surgery, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Seok Hwan Kim
- Department of General Surgery, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Hyun Jin Kim
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Research Center for Endocrine and Metabolic Disease, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Bon Jeong Ku
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Research Center for Endocrine and Metabolic Disease, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Ju Hee Lee
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Research Center for Endocrine and Metabolic Disease, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
17
|
den Hartigh LJ, May KS, Zhang XS, Chait A, Blaser MJ. Serum amyloid A and metabolic disease: evidence for a critical role in chronic inflammatory conditions. Front Cardiovasc Med 2023; 10:1197432. [PMID: 37396595 PMCID: PMC10311072 DOI: 10.3389/fcvm.2023.1197432] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/15/2023] [Indexed: 07/04/2023] Open
Abstract
Serum amyloid A (SAA) subtypes 1-3 are well-described acute phase reactants that are elevated in acute inflammatory conditions such as infection, tissue injury, and trauma, while SAA4 is constitutively expressed. SAA subtypes also have been implicated as playing roles in chronic metabolic diseases including obesity, diabetes, and cardiovascular disease, and possibly in autoimmune diseases such as systemic lupus erythematosis, rheumatoid arthritis, and inflammatory bowel disease. Distinctions between the expression kinetics of SAA in acute inflammatory responses and chronic disease states suggest the potential for differentiating SAA functions. Although circulating SAA levels can rise up to 1,000-fold during an acute inflammatory event, elevations are more modest (∼5-fold) in chronic metabolic conditions. The majority of acute-phase SAA derives from the liver, while in chronic inflammatory conditions SAA also derives from adipose tissue, the intestine, and elsewhere. In this review, roles for SAA subtypes in chronic metabolic disease states are contrasted to current knowledge about acute phase SAA. Investigations show distinct differences between SAA expression and function in human and animal models of metabolic disease, as well as sexual dimorphism of SAA subtype responses.
Collapse
Affiliation(s)
- Laura J. den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Karolline S. May
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| | - Alan Chait
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
18
|
Vasamsetti SB, Natarajan N, Sadaf S, Florentin J, Dutta P. Regulation of cardiovascular health and disease by visceral adipose tissue-derived metabolic hormones. J Physiol 2023; 601:2099-2120. [PMID: 35661362 PMCID: PMC9722993 DOI: 10.1113/jp282728] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/04/2022] [Indexed: 11/08/2022] Open
Abstract
Visceral adipose tissue (VAT) is a metabolic organ known to regulate fat mass, and glucose and nutrient homeostasis. VAT is an active endocrine gland that synthesizes and secretes numerous bioactive mediators called 'adipocytokines/adipokines' into systemic circulation. These adipocytokines act on organs of metabolic importance like the liver and skeletal muscle. Multiple preclinical and in vitro studies showed strong evidence of the roles of adipocytokines in the regulation of metabolic disorders like diabetes, obesity and insulin resistance. Adipocytokines, such as adiponectin and omentin, are anti-inflammatory and have been shown to prevent atherogenesis by increasing nitric oxide (NO) production by the endothelium, suppressing endothelium-derived inflammation and decreasing foam cell formation. By inhibiting differentiation of vascular smooth muscle cells (VSMC) into osteoblasts, adiponectin and omentin prevent vascular calcification. On the other hand, adipocytokines like leptin and resistin induce inflammation and endothelial dysfunction that leads to vasoconstriction. By promoting VSMC migration and proliferation, extracellular matrix degradation and inflammatory polarization of macrophages, leptin and resistin increase the risk of atherosclerotic plaque vulnerability and rupture. Additionally, the plasma concentrations of these adipocytokines alter in ageing, rendering older humans vulnerable to cardiovascular disease. The disturbances in the normal physiological concentrations of these adipocytokines secreted by VAT under pathological conditions impede the normal functions of various organs and affect cardiovascular health. These adipokines could be used for both diagnostic and therapeutic purposes in cardiovascular disease.
Collapse
Affiliation(s)
- Sathish Babu Vasamsetti
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, USA
| | - Niranjana Natarajan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
| | - Samreen Sadaf
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, USA
| | - Jonathan Florentin
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA 15213
- Pittsburgh VA Medical Center-University Drive, University Drive C, Pittsburgh, PA, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA, 15213
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA, 15213
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
19
|
Wang SY, Zhang WS, Jiang CQ, Jin YL, Zhu T, Zhu F, Xu L. Association of novel and conventional obesity indices with colorectal cancer risk in older Chinese: a 14-year follow-up of the Guangzhou Biobank Cohort Study. BMC Cancer 2023; 23:286. [PMID: 36991401 DOI: 10.1186/s12885-023-10762-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Abstract
Background
Visceral adiposity index (VAI) and a body shape index (ABSI) were newly developed indices for visceral fat mass. Whether they are superior to conventional obesity indices in predicting colorectal cancer (CRC) remains unclear. We examined the associations of VAI and ABSI with CRC risk, and investigated their performance in discriminating CRC risk compared with conventional obesity indices in the Guangzhou Biobank Cohort Study.
Methods
A total of 28,359 participants aged 50 + years without cancer history at baseline (2003-8) were included. CRC were identified from the Guangzhou Cancer Registry. Cox proportional hazards regression was used to assess the association of obesity indices with the CRC risk. Discriminative abilities of obesity indices were assessed using Harrell’s C-statistic.
Results
During an average follow-up of 13.9 (standard deviation = 3.6) years, 630 incident CRC cases were recorded. After adjusting for potential confounders, the hazard ratio (95% confidence interval) of incident CRC for per standard deviation increment in VAI, ABSI, body mass index (BMI), waist circumference (WC), waist-to-hip ratio (WHR) and waist-to-height ratio (WHtR) was 1.04 (0.96, 1.12), 1.13 (1.04, 1.22), 1.08 (1.00, 1.17), 1.15 (1.06, 1.24), 1.16 (1.08, 1.25)and 1.13 (1.04, 1.22), respectively. Similar results for colon cancer were found. However, the associations of obesity indices with risk of rectal cancer were non-significant. All obesity indices showed similar discriminative abilities (C-statistics from 0.640 to 0.645), with WHR showing the highest whilst VAI and BMI the lowest.
Conclusions
ABSI, but not VAI, was positively associated with a higher risk of CRC. However, ABSI was not superior to the conventional abdominal obesity indices in predicting CRC.
Collapse
|
20
|
Urlacher SS. The energetics of childhood: Current knowledge and insights into human variation, evolution, and health. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2023. [PMID: 36866969 DOI: 10.1002/ajpa.24719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/22/2022] [Accepted: 02/10/2023] [Indexed: 03/04/2023]
Abstract
How organisms capture and ultimately use metabolic energy-a limiting resource of life-has profound implications for understanding evolutionary legacies and current patterns of phenotypic variation, adaptation, and health. Energetics research among humans has a rich history in biological anthropology and beyond. The energetics of childhood, however, remains relatively underexplored. This shortcoming is notable given the accepted importance of childhood in the evolution of the unique human life history pattern as well as the known sensitivity of childhood development to local environments and lived experiences. In this review, I have three objectives: (1) To overview current knowledge regarding how children acquire and use energy, highlighting work among diverse human populations and pointing to recent advances and remaining areas of uncertainty; (2) To discuss key applications of this knowledge for understanding human variation, evolution, and health; (3) To recommend future avenues for research. A growing body of evidence supports a model of trade-offs and constraint in childhood energy expenditure. This model, combined with advancements on topics such as the energetics of immune activity, the brain, and the gut, provides insights into the evolution of extended human subadulthood and the nature of variation in childhood development, lifetime phenotype, and health.
Collapse
Affiliation(s)
- Samuel S Urlacher
- Department of Anthropology, Baylor University, Waco, Texas, USA
- Child and Brain Development Program, CIFAR, Toronto, Canada
| |
Collapse
|
21
|
García-Pérez R, Ramirez JM, Ripoll-Cladellas A, Chazarra-Gil R, Oliveros W, Soldatkina O, Bosio M, Rognon PJ, Capella-Gutierrez S, Calvo M, Reverter F, Guigó R, Aguet F, Ferreira PG, Ardlie KG, Melé M. The landscape of expression and alternative splicing variation across human traits. CELL GENOMICS 2023; 3:100244. [PMID: 36777183 PMCID: PMC9903719 DOI: 10.1016/j.xgen.2022.100244] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/08/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022]
Abstract
Understanding the consequences of individual transcriptome variation is fundamental to deciphering human biology and disease. We implement a statistical framework to quantify the contributions of 21 individual traits as drivers of gene expression and alternative splicing variation across 46 human tissues and 781 individuals from the Genotype-Tissue Expression project. We demonstrate that ancestry, sex, age, and BMI make additive and tissue-specific contributions to expression variability, whereas interactions are rare. Variation in splicing is dominated by ancestry and is under genetic control in most tissues, with ribosomal proteins showing a strong enrichment of tissue-shared splicing events. Our analyses reveal a systemic contribution of types 1 and 2 diabetes to tissue transcriptome variation with the strongest signal in the nerve, where histopathology image analysis identifies novel genes related to diabetic neuropathy. Our multi-tissue and multi-trait approach provides an extensive characterization of the main drivers of human transcriptome variation in health and disease.
Collapse
Affiliation(s)
- Raquel García-Pérez
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Jose Miguel Ramirez
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Aida Ripoll-Cladellas
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Ruben Chazarra-Gil
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Winona Oliveros
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Oleksandra Soldatkina
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Mattia Bosio
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Paul Joris Rognon
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
- Department of Economics and Business, Universitat Pompeu Fabra, Barcelona, Catalonia 08005, Spain
- Department of Statistics and Operations Research, Universitat Politècnica de Catalunya, Barcelona, Catalonia 08034, Spain
| | - Salvador Capella-Gutierrez
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| | - Miquel Calvo
- Statistics Section, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Catalonia 08028, Spain
| | - Ferran Reverter
- Statistics Section, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Catalonia 08028, Spain
| | - Roderic Guigó
- Bioinformatics and Genomics, Center for Genomic Regulation, Barcelona, Catalonia 08003, Spain
| | | | - Pedro G. Ferreira
- Department of Computer Science, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal
- Laboratory of Artificial Intelligence and Decision Support, INESC TEC, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto, Institute for Research and Innovation in Health (i3s), R. Alfredo Allen 208, 4200-135 Porto, Portugal
| | | | - Marta Melé
- Department of Life Sciences, Barcelona Supercomputing Center (BCN-CNS), Barcelona, Catalonia 08034, Spain
| |
Collapse
|
22
|
Flikweert AW, Kobold ACM, van der Sar-van der Brugge S, Heeringa P, Rodenhuis-Zybert IA, Bijzet J, Tami A, van der Gun BTF, Wold KI, Huckriede A, Franke H, Emmen JMA, Emous M, Grootenboers MJJH, van Meurs M, van der Voort PHJ, Moser J. Circulating adipokine levels and COVID-19 severity in hospitalized patients. Int J Obes (Lond) 2023; 47:126-137. [PMID: 36509969 PMCID: PMC9742670 DOI: 10.1038/s41366-022-01246-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Obesity is a risk factor for adverse outcomes in COVID-19, potentially driven by chronic inflammatory state due to dysregulated secretion of adipokines and cytokines. We investigated the association between plasma adipokines and COVID-19 severity, systemic inflammation, clinical parameters, and outcome of COVID-19 patients. METHODS In this multi-centre prospective cross-sectional study, we collected blood samples and clinical data from COVID-19 patients. The severity of COVID-19 was classified as mild (no hospital admission), severe (ward admission), and critical (ICU admission). ICU non-COVID-19 patients were also included and plasma from healthy age, sex, and BMI-matched individuals obtained from Lifelines. Multi-analyte profiling of plasma adipokines (Leptin, Adiponectin, Resistin, Visfatin) and inflammatory markers (IL-6, TNFα, IL-10) were determined using Luminex multiplex assays. RESULTS Between March and December 2020, 260 SARS-CoV-2 infected individuals (age: 65 [56-74] BMI 27.0 [24.4-30.6]) were included: 30 mild, 159 severe, and 71 critical patients. Circulating leptin levels were reduced in critically ill patients with a high BMI yet this decrease was absent in patients that were administered dexamethasone. Visfatin levels were higher in critical COVID-19 patients compared to non-COVID-ICU, mild and severe patients (4.7 vs 3.4, 3.0, and 3.72 ng/mL respectively, p < 0.05). Lower Adiponectin levels, but higher Resistin levels were found in severe and critical patients, compared to those that did not require hospitalization (3.65, 2.7 vs 7.9 µg/mL, p < 0.001, and 18.2, 22.0 vs 11.0 ng/mL p < 0.001). CONCLUSION Circulating adipokine levels are associated with COVID-19 hospitalization, i.e., the need for oxygen support (general ward), or the need for mechanical ventilation and other organ support in the ICU, but not mortality.
Collapse
Affiliation(s)
- Antine W. Flikweert
- grid.4494.d0000 0000 9558 4598Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,grid.413711.10000 0004 4687 1426Department of Pulmonary Medicine, Amphia Hospital, Breda, The Netherlands
| | - Anneke C. Muller Kobold
- grid.4494.d0000 0000 9558 4598Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Peter Heeringa
- grid.4494.d0000 0000 9558 4598Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Izabela A. Rodenhuis-Zybert
- grid.4494.d0000 0000 9558 4598Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan Bijzet
- grid.4494.d0000 0000 9558 4598Department of Rheumatology & Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Adriana Tami
- grid.4494.d0000 0000 9558 4598Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bernardina T. F. van der Gun
- grid.4494.d0000 0000 9558 4598Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Karin I. Wold
- grid.4494.d0000 0000 9558 4598Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anke Huckriede
- grid.4494.d0000 0000 9558 4598Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hildegard Franke
- grid.4494.d0000 0000 9558 4598Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Judith M. A. Emmen
- grid.413711.10000 0004 4687 1426Result Laboratory, Amphia Hospital, Breda, The Netherlands
| | - Marloes Emous
- grid.414846.b0000 0004 0419 3743Center Obesity Northern Netherlands (CON), Department of Surgery, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | | | - Matijs van Meurs
- grid.4494.d0000 0000 9558 4598Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter H. J. van der Voort
- grid.4494.d0000 0000 9558 4598Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jill Moser
- Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. .,Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
23
|
Milano W, Carizzone F, Foia M, Marchese M, Milano M, Saetta B, Capasso A. Obesity and Its Multiple Clinical Implications between Inflammatory States and Gut Microbiotic Alterations. Diseases 2022; 11:7. [PMID: 36648872 PMCID: PMC9844347 DOI: 10.3390/diseases11010007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
Obesity is a chronic multifactorial disease that has become a serious health problem and is currently widespread over the world. It is, in fact, strongly associated with many other conditions, including insulin resistance, type 2 diabetes, cardiovascular and neurodegenerative diseases, the onset of different types of malignant tumors and alterations in reproductive function. According to the literature, obesity is characterized by a state of low-grade chronic inflammation, with a substantial increase in immune cells, specifically macrophage infiltrates in the adipose tissue which, in turn, secrete a succession of pro-inflammatory mediators. Furthermore, recent studies on microbiota have postulated new possible mechanisms of interaction between obesity and unbalanced nutrition with inflammation. This intestinal "superorganism" complex seems to influence not only the metabolic balance of the host but also the immune response, favoring a state of systemic inflammation and insulin resistance. This review summarizes the major evidence on the interactions between the gut microbiota, energetic metabolism and host immune system, all leading to a convergence of the fields of immunology, nutrients physiology and microbiota in the context of obesity and its possible clinical complications. Finally, possible therapeutic approaches aiming to rebalance the intestinal microbial ecosystem are evaluated to improve the alteration of inflammatory and metabolic states in obesity and related diseases.
Collapse
Affiliation(s)
- Walter Milano
- UOSD Eating Disorder Unit, Mental Health Department, ASL Napoli 2 Nord, 80027 Napoli, Italy
| | - Francesca Carizzone
- UOSD Eating Disorder Unit, Mental Health Department, ASL Napoli 2 Nord, 80027 Napoli, Italy
| | | | - Magda Marchese
- Clinical Pathology Services, Santa Maria Delle Grazie Hospital Pozzuoli, Asl Napoli 2 Nord, 80027 Napoli, Italy
| | - Mariafrancesca Milano
- UOSD Eating Disorder Unit, Mental Health Department, ASL Napoli 2 Nord, 80027 Napoli, Italy
| | - Biancamaria Saetta
- UOSD Eating Disorder Unit, Mental Health Department, ASL Napoli 2 Nord, 80027 Napoli, Italy
| | - Anna Capasso
- Department of Pharmacy, University of Salerno, Fisciano, 84084 Salerno, Italy
| |
Collapse
|
24
|
Transcriptomics and Lipid Metabolomics Analysis of Subcutaneous, Visceral, and Abdominal Adipose Tissues of Beef Cattle. Genes (Basel) 2022; 14:genes14010037. [PMID: 36672778 PMCID: PMC9858949 DOI: 10.3390/genes14010037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Fat deposition traits are influenced by genetics and environment, which affect meat quality, growth rate, and energy metabolism of domestic animals. However, at present, the molecular mechanism of fat deposition is not entirely understood in beef cattle. Therefore, the current study conducted transcriptomics and lipid metabolomics analysis of subcutaneous, visceral, and abdominal adipose tissue (SAT, VAT, and AAT) of Huaxi cattle to investigate the differences among these adipose tissues and systematically explore how candidate genes interact with metabolites to affect fat deposition. These results demonstrated that compared with SAT, the gene expression patterns and metabolite contents of VAT and AAT were more consistent. Particularly, SCD expression, monounsaturated fatty acid (MUFA) and triglyceride (TG) content were higher in SAT, whereas PCK1 expression and the contents of saturated fatty acid (SFA), diacylglycerol (DG), and lysoglycerophosphocholine (LPC) were higher in VAT. Notably, in contrast to PCK1, 10 candidates including SCD, ELOVL6, ACACA, and FABP7 were identified to affect fat deposition through positively regulating MUFA and TG, and negatively regulating SFA, DG, and LPC. These findings uncovered novel gene resources and offered a theoretical basis for future investigation of fat deposition in beef cattle.
Collapse
|
25
|
Attia SM, Das SC, Varadharajan K, Al-Naemi HA. White adipose tissue as a target for cadmium toxicity. Front Pharmacol 2022; 13:1010817. [PMID: 36278208 PMCID: PMC9582776 DOI: 10.3389/fphar.2022.1010817] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
Cadmium (Cd) is a widespread heavy metal known as a toxic environmental pollutant. Cd exposure is threatening due to its bioaccumulation trait in living systems that exceeds 35 years without a beneficial biological role. Acute exposure to high Cd doses was reported to impact adipose tissue (AT) function adversely. The main aim of this study is to investigate the effect of low-dose chronic Cd exposure on the genes involved in adipose tissue (AT) functions. Adult male Sprague-Dawley rats were exposed to a low Cd dose (15 mg/kg B.W./day) for 10 weeks. Then, three AT depots-subcutaneous AT (SUB-AT), abdominal AT (AB-AT), and retroperitoneal AT (REtrop-AT) were excised for Cd accumulation measures and gene expression analysis. Adiponectin and leptin gene expression levels were investigated as markers for adipocytes function and homeostasis. Our results showed that Cd accumulated in all the tested adipose depots, but SUB-AT was found to be the depot to most accumulate Cd. Also, it was exhibited that chronic exposure to low Cd doses altered the gene expression of adipocytokines. The levels of adiponectin and leptin mRNA expression were downregulated in all tested AT-depots after Cd exposure. The significant adverse effect on SUB-AT compared to other depots indicates different responses based on AT depots location toward Cd exposure. Collectively, these results suggest a toxic effect of Cd that influenced adipocyte function.
Collapse
Affiliation(s)
- Sarra Mohammed Attia
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
- Department of Biological and Environmental Science, Qatar University, Doha, Qatar
| | - Sandra Concepcion Das
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
- Department of Biological and Environmental Science, Qatar University, Doha, Qatar
| | | | - Hamda A. Al-Naemi
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
- Department of Biological and Environmental Science, Qatar University, Doha, Qatar
- *Correspondence: Hamda A. Al-Naemi,
| |
Collapse
|
26
|
Yığman M, Tangal S. Effects of body fat components on early renal functions of individuals following kidney donation. JOURNAL OF CLINICAL UROLOGY 2022. [DOI: 10.1177/20514158221109411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: Obesity stands as a risk factor for the chronic kidney disease. The objective of this study was to investigate the relationship between early renal function following kidney donation and the measurements of body fat components. Methods: In total, 86 donors followed up for at least 6 months postoperatively were included. Height and weight measurements and results of laboratory analysis of all donors were recorded retrospectively. Visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT), hepatic fat (HF), pancreatic fat (PF) and splenic fat (SF) measurements were performed, and pancreatic splenic fat fraction difference (P−S) and pancreatic splenic fat fraction ratio (P/S) were calculated by a radiologist using the records of preoperative computed tomography scans of donors. Results: The estimated glomerular filtration rate (eGFR), serum creatinine and spot urinary microalbumin/creatinine ratio values of the donors at the sixth month postoperatively were statistically different from those of the preoperative values ( p < 0.001). In addition, the individuals were divided into two categories based on the postoperative eGFR: ⩾ 60 mL/min/1.73 m2 and < 60 mL/min/1.73 m2. Age, low-density lipoprotein (LDL) level and VAT/SAT ratio were lower in group eGFR: ⩾ 60 ( p < 0.001, p = 0.03, p = 0.007, respectively). Age and VAT/SAT ratio were the parameters found to be affecting the eGFR significantly, and VAT/SAT ratio (0.729, 95% CI: 0.602–0.856, p = 0.007) had higher predictive value in receiver operating characteristic curve (ROC). Conclusion: Preoperative measurements of body fat components may provide significant information to predict postoperative renal functions of kidney donor candidates. Level of evidence: Not applicable.
Collapse
Affiliation(s)
- Metin Yığman
- Department of Urology, Faculty of Medicine, Dr. Ridvan Ege Hospital, Ufuk University, Turkey
| | - Semih Tangal
- Department of Urology, Faculty of Medicine, Dr. Ridvan Ege Hospital, Ufuk University, Turkey
| |
Collapse
|
27
|
Mela V, Ruiz-Limón P, Balongo M, Motahari Rad H, Subiri-Verdugo A, Gonzalez-Jimenez A, Soler R, Ocaña L, el Azzouzi H, Tinahones FJ, Valdivielso P, Murri M. Mitochondrial Homeostasis in Obesity-related Hypertriglyceridemia. J Clin Endocrinol Metab 2022; 107:2203-2215. [PMID: 35608825 PMCID: PMC9282366 DOI: 10.1210/clinem/dgac332] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Indexed: 12/04/2022]
Abstract
CONTEXT The prevalence of obesity and hypertriglyceridemia is an alarming worldwide health issue. Mitochondria play a central role in these disorders as they control cell metabolism. OBJECTIVE The aim of the present study was to characterize mitochondrial homeostasis in subcutaneous and visceral adipose tissue (SAT and VAT) in grade III obese patients with and without hypertriglyceridemia. Moreover, this study presents the evaluation of mitochondrial fitness as a marker for hypertriglyceridemia improvement. PATIENTS Eight control and 12 hypertriglyceridemic (HTG) grade III obese subjects undergoing bariatric surgery were included. MAIN OUTCOME MEASURES Anthropometric and biochemical data were obtained before and 3 months after surgery. Mitochondrial homeostasis was evaluated by mitochondrial DNA (mtDNA), gene expression and protein abundance in SAT and VAT. RESULTS Mitophagy-related gene expression was increased in HTG SAT and VAT, while mitochondrial marker gene expression and mtDNA were decreased, indicating an altered mitochondrial homeostasis in HTG. Mitophagy protein abundance was increased in VAT of those subjects that did not improve their levels of triglycerides after bariatric surgery, whereas mitochondrial protein was decreased in the same tissue. Indeed, triglyceride levels positively correlated with mitophagy-related genes and negatively with mitochondrial content markers. Moreover, mitochondria content and mitophagy markers seem to be significant predictors of hypertriglyceridemia and hypertriglyceridemia remission. CONCLUSIONS Mitochondrial homeostasis of adipose tissue is altered in hypertriglyceridemic patients. At the protein level, mitochondria content and mitophagy are potential markers of hypertriglyceridemia remission in obese patients after bariatric surgery. These results may contribute to the implementation of a clinical approach for personalized medicine.
Collapse
Affiliation(s)
- Virginia Mela
- Correspondence: Virginia Mela, PhD, Endocrine Diseases Research Group, Biomedical Research Institute of Malaga (IBIMA), University Hospital of Malaga (Virgen de la Victoria), Malaga, Campus de Teatinos s/n 29010 Málaga, Spain.
| | | | - Manuel Balongo
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010, Málaga, Spain
| | - Hanieh Motahari Rad
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010, Málaga, Spain
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154, Tehran, Iran
| | - Alba Subiri-Verdugo
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010, Málaga, Spain
| | | | - Rocio Soler
- Unidad de Gestión Clínica de Cirugía General y Digestiva, Hospital Clínico Virgen de la Victoria, 29010, Málaga, Spain
| | - Luis Ocaña
- Unidad de Gestión Clínica de Cirugía General y Digestiva, Hospital Clínico Virgen de la Victoria, 29010, Málaga, Spain
| | - Hamid el Azzouzi
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, 3436 HR, Netherlands
| | - Francisco J Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clínico Virgen de la Victoria, 29010, Málaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 29010, Málaga, Spain
- Department of Medicine and Dermatology, Faculty of Medicine, University ofMalaga, 29010, Malaga, Spain
| | | | - Mora Murri
- Mora Murri, PhD, Endocrine Diseases Research Group, Biomedical Research Institute of Malaga (IBIMA), University Hospital of Malaga (Virgen de la Victoria), Malaga, Campus de Teatinos s/n 29010 Málaga, Spain.
| |
Collapse
|
28
|
Wang T. Searching for the link between inflammaging and sarcopenia. Ageing Res Rev 2022; 77:101611. [PMID: 35307560 DOI: 10.1016/j.arr.2022.101611] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Tiantian Wang
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
29
|
Steele C, Nowak K. Obesity, Weight Loss, Lifestyle Interventions, and Autosomal Dominant Polycystic Kidney Disease. KIDNEY AND DIALYSIS 2022; 2:106-122. [PMID: 35350649 PMCID: PMC8959086 DOI: 10.3390/kidneydial2010013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Obesity remains a growing public health concern in industrialized countries around the world. The prevalence of obesity has also continued to rise in those with chronic kidney disease. Epidemiological data suggests those with overweight and obesity, measured by body mass index, have an increased risk for rapid kidney disease progression. Autosomal dominant polycystic kidney disease causes growth and proliferation of kidney cysts resulting in a reduction in kidney function in the majority of adults. An accumulation of adipose tissue may further exacerbate the metabolic defects that have been associated with ADPKD by affecting various cell signaling pathways. Lifestyle interventions inducing weight loss might help delay disease progression by reducing adipose tissue and systematic inflammation. Further research is needed to determine the mechanistic influence of adipose tissue on disease progression.
Collapse
Affiliation(s)
- Cortney Steele
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Kristen Nowak
- Division of Renal Diseases and Hypertension, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| |
Collapse
|
30
|
Xie Y, Zhang Y, Qin P, Ping Z, Wang C, Peng X, Chen H, Zhao D, Xu S, Wang L, Wei P, Hu F, Lou Y, Hu D. The association between Chinese Visceral Adipose Index and coronary heart disease: A cohort study in China. Nutr Metab Cardiovasc Dis 2022; 32:550-559. [PMID: 34961647 DOI: 10.1016/j.numecd.2021.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 10/22/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS The purpose of this study is to explore the relationship between Chinese visceral adipose index (CVAI) and the risk of coronary heart disease (CHD) in Chinese through a large cohort study. METHODS AND RESULTS This study included 42,165 adults who were without CHD at baseline and who completed at least one annual follow-up between 2009 and 2016. We used the Cox proportional hazards model to estimate Hazard Ratios (HRs) and 95% Confidence Intervals (CIs) for the association between CVAI and risk of CHD. During the median follow-up of 3.36 years (154,808 person years), 520 participants developed CHD, including 374 males and 146 females. Compared with the first quartile of CVAI, the risk of CHD was significantly increased in the fourth quartile of CVAI in multivariate model (HR [95% CI]: 9.92 [5.45, 18.04], P < 0.001). Sensitivity analysis by excluding incident CHD developed in the first two years of follow-up reinforced our results. Gender stratification analyses showed that the relationship between CVAI and CHD risk was higher in males than that in females. The restricted cubic spline showed a non-linear dose-response relationship between CVAI and CHD risk. In addition, CVAI was associated with CHD risk in the subgroups of participants without T2DM, without hypertension, and without fatty liver. CONCLUSION CVAI was significantly associated with the risk of CHD. Individuals should keep CVAI at normal level to prevent CHD.
Collapse
Affiliation(s)
- Yilin Xie
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China
| | - Yanyan Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China
| | - Pei Qin
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China
| | - Zhao Ping
- Department of Health Management, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping, Beijing 102200, People's Republic of China
| | - Changyi Wang
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Xiaolin Peng
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Hongen Chen
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Dan Zhao
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Shan Xu
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Li Wang
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Pengfei Wei
- Department of Endocrine, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, People's Republic of China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China.
| | - Yanmei Lou
- Department of Health Management, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping, Beijing 102200, People's Republic of China.
| | - Dongsheng Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China.
| |
Collapse
|
31
|
Abedpoor N, Taghian F, Hajibabaie F. Physical activity ameliorates the function of organs via adipose tissue in metabolic diseases. Acta Histochem 2022; 124:151844. [PMID: 35045377 DOI: 10.1016/j.acthis.2022.151844] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 12/15/2022]
Abstract
Adipose tissue is a dynamic organ in the endocrine system that can connect organs by secreting molecules and bioactive. Hence, adipose tissue really plays a pivotal role in regulating metabolism, inflammation, energy homeostasis, and thermogenesis. Disruption of hub bioactive molecules secretion such as adipokines leads to dysregulate metabolic communication between adipose tissue and other organs in non-communicable disorders. Moreover, a sedentary lifestyle may be a risk factor for adipose tissue function. Physical inactivity leads to fat tissue accumulation and promotes obesity, Type 2 diabetes, cardiovascular disease, neurodegenerative disease, fatty liver, osteoporosis, and inflammatory bowel disease. On the other hand, physical activity may ameliorate and protect the body against metabolic disorders, triggering thermogenesis, metabolism, mitochondrial biogenesis, β-oxidation, and glucose uptake. Furthermore, physical activity provides an inter-organ association and cross-talk between different tissues by improving adipose tissue function, reprogramming gene expression, modulating molecules and bioactive factors. Also, physical activity decreases chronic inflammation, oxidative stress and improves metabolic features in adipose tissue. The current review focuses on the beneficial effect of physical activity on the cardiovascular, locomotor, digestive, and nervous systems. In addition, we visualize protein-protein interactions networks between hub proteins involved in dysregulating metabolic induced by adipose tissue.
Collapse
Affiliation(s)
- Navid Abedpoor
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Farzaneh Taghian
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Fatemeh Hajibabaie
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| |
Collapse
|
32
|
Parent MB, Higgs S, Cheke LG, Kanoski SE. Memory and eating: A bidirectional relationship implicated in obesity. Neurosci Biobehav Rev 2022; 132:110-129. [PMID: 34813827 PMCID: PMC8816841 DOI: 10.1016/j.neubiorev.2021.10.051] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/17/2021] [Accepted: 10/28/2021] [Indexed: 01/03/2023]
Abstract
This paper reviews evidence demonstrating a bidirectional relationship between memory and eating in humans and rodents. In humans, amnesia is associated with impaired processing of hunger and satiety cues, disrupted memory of recent meals, and overconsumption. In healthy participants, meal-related memory limits subsequent ingestive behavior and obesity is associated with impaired memory and disturbances in the hippocampus. Evidence from rodents suggests that dorsal hippocampal neural activity contributes to the ability of meal-related memory to control future intake, that endocrine and neuropeptide systems act in the ventral hippocampus to provide cues regarding energy status and regulate learned aspects of eating, and that consumption of hypercaloric diets and obesity disrupt these processes. Collectively, this evidence indicates that diet-induced obesity may be caused and/or maintained, at least in part, by a vicious cycle wherein excess intake disrupts hippocampal functioning, which further increases intake. This perspective may advance our understanding of how the brain controls eating, the neural mechanisms that contribute to eating-related disorders, and identify how to treat diet-induced obesity.
Collapse
Affiliation(s)
- Marise B Parent
- Neuroscience Institute & Department of Psychology, Georgia State University, Box 5030, Atlanta, GA 30303-5030, United States.
| | - Suzanne Higgs
- School of Psychology, University of Birmingham, Edgbaston, Birmingham, BI5 2TT, United Kingdom.
| | - Lucy G Cheke
- Department of Psychology, University of Cambridge, Downing Street, Cambridge, CB2 3EB, United Kingdom.
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, 90089-0371, United States.
| |
Collapse
|
33
|
Xintong L, Dongmei X, Li Z, Ruimin C, Yide H, Lingling C, Tingting C, Yingying G, Jiaxin L. Correlation of body composition in early pregnancy on gestational diabetes mellitus under different body weights before pregnancy. Front Endocrinol (Lausanne) 2022; 13:916883. [PMID: 36387861 PMCID: PMC9649916 DOI: 10.3389/fendo.2022.916883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 10/13/2022] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE The prediction of gestational diabetes mellitus (GDM) by body composition-related indicators in the first trimester was analyzed under different body mass index (BMI) values before pregnancy. METHODS This was a retrospective analysis of pregnant women who were treated, had documented data, and received regular perinatal care at the Third Affiliated Hospital of Zhengzhou University from January 1, 2021, to December 31, 2021. Women with singleton pregnancies who did not have diabetes before pregnancy were included. In the first trimester (before the 14th week of pregnancy), bioelectric impedance assessment (BIA) was used to analyze body composition-related indicators such as protein levels, mineral levels, fat volume, and the waist-hip fat ratio. The Pearman's correlation coefficient was used to evaluate the linear relationship between the continuous variables and pre-pregnancy body mass index (BMI). In the univariate body composition analysis, the association with the risk of developing GDM was included in a multivariate analysis using the relative risk and 95% confidence interval obtained from logarithmic binomial regression, and generalized linear regression was used for multivariate regression analysis. Furthermore, the area under the curve (AUC) was calculated by receiver operating characteristic (ROC) curves. The optimal cutoff value of each risk factor was calculated according to the Youden Index. RESULTS In a retrospective study consisting of 6698 pregnant women, we collected 1109 cases of gestational diabetes. Total body water (TBW), protein levels, mineral levels, bone mineral content (BMC), body fat mass (BFM), soft lean mass (SLM), fat-free mass (FMM), skeletal muscle mass (SMM), percent body fat (PBF), the waist-hip ratio (WHR), the visceral fat level (VFL), and the basal metabolic rate (BMR) were significantly higher in the GDM group than in the normal group (P<0.05). Under the pre-pregnancy BMI groupings, out of 4157 pregnant women with a BMI <24 kg/m2, 456 (10.97%) were diagnosed with GDM, and out of 2541 pregnant women with a BMI ≥24 kg/m2, 653 (25.70%) were diagnosed with GDM. In the generalized linear regression model, it was found that in all groups of pregnant women, pre-pregnancy BMI, age, gestational weight gain (GWG) in the first trimester, and weight at the time of the BIA had a certain risk for the onset of GDM. In Model 1, without adjusting for confounders, the body composition indicators were all positively correlated with the risk of GDM. In Model 3, total body water, protein levels, mineral levels, bone mineral content, soft lean mass, fat-free mass, skeletal muscle mass, and the basal metabolic rate were protective factors for GDM. After Model 4 was adjusted for confounders, only the waist-hip ratio was positively associated with GDM onset. Among pregnant women with a pre-pregnancy BMI <24 kg/m2, the body composition-related indicators in Model 2 were all related to the onset of GDM. In Model 3, total body water, soft lean mass, fat-free mass, and the basal metabolic rate were negatively correlated with GDM onset. In the body composition analysis of among women with a pre-pregnancy BMI ≥ 24 kg/m2, only Model 1 and Model 2 were found to show positive associations with GDM onset. In the prediction model, in the basic data of pregnant women, the area under the receiver operating characteristic curve predicted by gestational weight gain for GDM was the largest (0.795), and its cutoff value was 1.415 kg. In the body composition results, the area under the receiver operating characteristic curve of body fat mass for predicting GDM risk was larger (0.663) in all pregnant women. CONCLUSIONS Through this retrospective study, it was found that the body composition-related indicators were independently associated with the onset of GDM in both the pre-pregnancy BMI <24 kg/m2 and pre-pregnancy BMI ≥24 kg/m2 groups. Body fat mass, the visceral fat level, and the waist-hip ratio had a higher correlation with pre-pregnancy BMI. Total body water, protein levels, mineral levels, bone mineral content, soft lean mass, fat-free mass, skeletal muscle mass, and the basal metabolic rate were protective factors for GDM after adjusting for some confounders. In all pregnant women, the waist-hip ratio was found to be up to 4.562 times the risk of GDM development, and gestational weight gain had the best predictive power for GDM. Gestational weight gain in early pregnancy, body fat mass, and the waist-hip ratio can assess the risk of GDM in pregnant women, which can allow clinicians to predict the occurrence of GDM in pregnant women as early as possible and implement interventions to reduce adverse perinatal outcomes.
Collapse
Affiliation(s)
- Li Xintong
- Department of Obstetrics and Gynecology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Dongmei
- Department of Obstetrics and Gynecology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Perinatal Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Xu Dongmei,
| | - Zhang Li
- Department of Obstetrics and Gynecology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cao Ruimin
- Department of Obstetrics and Gynecology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hao Yide
- Anesthesiology, Xinxiang Medical University, Xinxiang, China
| | - Cui Lingling
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Chen Tingting
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Guo Yingying
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Li Jiaxin
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
34
|
Amaral MA, Mundstock E, Scarpatto CH, Cañon-Montañez W, Mattiello R. Reference percentiles for bioimpedance body composition parameters of healthy individuals: A cross-sectional study. Clinics (Sao Paulo) 2022; 77:100078. [PMID: 36087566 PMCID: PMC9464854 DOI: 10.1016/j.clinsp.2022.100078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/30/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES The aim of this study was to estimate the percentage distribution of body composition parameters for healthy people at different ages from the assessment of electrical bioimpedance. METHODS A cross-sectional study of healthy Brazilian aged 5 years and older. Were evaluated: total body fat; percent body fat; fat-free mass; percent lean mass; fat mass index; and fat-free mass index. RESULTS Of 1240 participants, with a median age of 27.0 years, 52.5% were female, and 73.7% were Caucasian. Most of the body composition variables were associated with age. The fat-free mass increased from youth to adult and decreased in the elderly in both sexes, with higher values in males than in females. In males, the percentage of lean mass has higher values in adolescence compared to childhood, and in adults compared to the elderly, when analyzed from the 50th percentile. In women, fat-free mass compared to adulthood, values were higher in childhood and lower in older ages. CONCLUSIONS The study is the first to describe the Brazilian reference values for most clinical parameters of bioimpedance in percentiles stratified by different life cycles and sex. These findings can be very useful in clinical practice for health promotion and monitoring the nutritional status of the individual.
Collapse
Affiliation(s)
- Marina Azambuja Amaral
- Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Centro Universitário Ritter dos Reis (UniRitter), Porto Alegre, RS, Brazil
| | - Eduardo Mundstock
- Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Secretaria da Educação, Esporte e Lazer de Canela, Canela, RS, Brazil
| | - Camila H Scarpatto
- Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Rita Mattiello
- Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
35
|
Jin N, Lee HM, Hou Y, Yu ACS, Li JW, Kong APS, Lam CCH, Wong SKH, Ng EKW, Ma RCW, Chan JCN, Chan TF. Integratome analysis of adipose tissues reveals abnormal epigenetic regulation of adipogenesis, inflammation, and insulin signaling in obese individuals with type 2 diabetes. Clin Transl Med 2021; 11:e596. [PMID: 34923751 PMCID: PMC8684766 DOI: 10.1002/ctm2.596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022] Open
Affiliation(s)
- Nana Jin
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Heung-Man Lee
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Yong Hou
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Allen C S Yu
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jing-Woei Li
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alice P S Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China.,Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Candice C H Lam
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Simon K H Wong
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Enders K W Ng
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China.,Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China.,Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Ting-Fung Chan
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
36
|
Verdú E, Homs J, Boadas-Vaello P. Physiological Changes and Pathological Pain Associated with Sedentary Lifestyle-Induced Body Systems Fat Accumulation and Their Modulation by Physical Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:13333. [PMID: 34948944 PMCID: PMC8705491 DOI: 10.3390/ijerph182413333] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
A sedentary lifestyle is associated with overweight/obesity, which involves excessive fat body accumulation, triggering structural and functional changes in tissues, organs, and body systems. Research shows that this fat accumulation is responsible for several comorbidities, including cardiovascular, gastrointestinal, and metabolic dysfunctions, as well as pathological pain behaviors. These health concerns are related to the crosstalk between adipose tissue and body systems, leading to pathophysiological changes to the latter. To deal with these health issues, it has been suggested that physical exercise may reverse part of these obesity-related pathologies by modulating the cross talk between the adipose tissue and body systems. In this context, this review was carried out to provide knowledge about (i) the structural and functional changes in tissues, organs, and body systems from accumulation of fat in obesity, emphasizing the crosstalk between fat and body tissues; (ii) the crosstalk between fat and body tissues triggering pain; and (iii) the effects of physical exercise on body tissues and organs in obese and non-obese subjects, and their impact on pathological pain. This information may help one to better understand this crosstalk and the factors involved, and it could be useful in designing more specific training interventions (according to the nature of the comorbidity).
Collapse
Affiliation(s)
- Enrique Verdú
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| | - Judit Homs
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
- Department of Physical Therapy, EUSES-University of Girona, 17190 Salt, Spain
| | - Pere Boadas-Vaello
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| |
Collapse
|
37
|
Age and Sex: Impact on adipose tissue metabolism and inflammation. Mech Ageing Dev 2021; 199:111563. [PMID: 34474078 DOI: 10.1016/j.mad.2021.111563] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023]
Abstract
Age associated chronic inflammation is a major contributor to diseases with advancing age. Adipose tissue function is at the nexus of processes contributing to age-related metabolic disease and mediating longevity. Hormonal fluctuations in aging potentially regulate age-associated visceral adiposity and metabolic dysfunction. Visceral adiposity in aging is linked to aberrant adipogenesis, insulin resistance, lipotoxicity and altered adipokine secretion. Age-related inflammatory phenomena depict sex differences in macrophage polarization, changes in T and B cell numbers, and types of dendritic cells. Sex differences are also observed in adipose tissue remodeling and cellular senescence suggesting a role for sex steroid hormones in the regulation of the adipose tissue microenvironment. It is crucial to investigate sex differences in aging clinical outcomes to identify and better understand physiology in at-risk individuals. Early interventions aimed at targets involved in adipose tissue adipogenesis, remodeling and inflammation in aging could facilitate a profound impact on health span and overcome age-related functional decline.
Collapse
|
38
|
Santos D, Carvalho E. Adipose-related microRNAs as modulators of the cardiovascular system: the role of epicardial adipose tissue. J Physiol 2021; 600:1171-1187. [PMID: 34455587 DOI: 10.1113/jp280917] [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: 08/24/2021] [Indexed: 11/08/2022] Open
Abstract
Adipose tissue expansion and subsequent metabolic dysfunction has been considered one of the major risk factors for development of cardiometabolic disease. Epicardial adipose tissue (EAT) in particular is a unique subtype of visceral adipose tissue located on the surface of the heart, around the coronary arteries. Due to its proximity, EAT can modulate the local metabolic and immune function of cardiomyocytes and coronary arteries. Several microRNAs have been described as key players in both cardiac and vascular function that when dysregulated will contribute to dysfunction. Here we review the influence of obesity in the crosstalk between specific adipose tissue types, in particular the EAT-secreted microRNAs, as key modulators of cardiac disease progression, not only as early biomarkers but also as therapeutic targets for cardiometabolic disease.
Collapse
Affiliation(s)
- Diana Santos
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Eugenia Carvalho
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal.,Portuguese Diabetes Association (APDP), Lisbon, Portugal
| |
Collapse
|
39
|
Molani Gol R, Rafraf M. Association between abdominal obesity and pulmonary function in apparently healthy adults: A systematic review. Obes Res Clin Pract 2021; 15:415-424. [PMID: 34261619 DOI: 10.1016/j.orcp.2021.06.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 06/06/2021] [Accepted: 06/27/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Obesity, especially abdominal obesity as a chronic disorder is associated with a high risk of developing non-communicable diseases such as respiratory diseases. Impaired lung function is a sign of early respiratory injury. This review summarizes the current knowledge of the effects of abdominal obesity on pulmonary function in apparently healthy adults. METHODS Google Scholar, PubMed, Science Direct, and Scopus databases were searched from 2014 up to August 2020 using relevant keywords. All original articles written in English evaluating the effects of abdominal obesity on pulmonary function in apparently healthy adults were eligible for this review. RESULTS A total of 26 studies (23 cross-sectional and three cohort) involving 68,024 participants were included in this review. More than 88% of the included studies reported that abdominal obesity significantly inversely was associated with pulmonary function. CONCLUSION The findings indicate that in subjects with abdominal obesity respiratory function decline possibly due to mechanical compression and obesity-induced airway inflammation. Therefore, nutrition and lifestyle interventions are required for the reduction of abdominal obesity that leads to improving pulmonary function and metabolic disease.
Collapse
Affiliation(s)
- Roghayeh Molani Gol
- Student Research Committee, Tabriz university of Medical Sciences, Tabriz, Iran; Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Maryam Rafraf
- Nutrition Research Center, Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
40
|
Donia T, Khamis A. Management of oxidative stress and inflammation in cardiovascular diseases: mechanisms and challenges. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:34121-34153. [PMID: 33963999 DOI: 10.1007/s11356-021-14109-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/21/2021] [Indexed: 06/12/2023]
Abstract
Cardiovascular diseases (CVDs) have diverse physiopathological mechanisms with interconnected oxidative stress and inflammation as one of the common etiologies which result in the onset and development of atherosclerotic plaques. In this review, we illustrate this strong crosstalk between oxidative stress, inflammation, and CVD. Also, mitochondrial functions underlying this crosstalk, and various approaches for the prevention of redox/inflammatory biological impacts will be illustrated. In part, we focus on the laboratory biomarkers and physiological tests for the evaluation of oxidative stress status and inflammatory processes. The impact of a healthy lifestyle on CVD onset and development is displayed as well. Furthermore, the differences in oxidative stress and inflammation are related to genetic susceptibility to cardiovascular diseases and the variability in the assessment of CVDs risk between individuals; Omics technologies for measuring oxidative stress and inflammation will be explored. Finally, we display the oxidative stress-related microRNA and the functions of the redox basis of epigenetic modifications.
Collapse
Affiliation(s)
- Thoria Donia
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Abeer Khamis
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| |
Collapse
|
41
|
Wróblewski A, Strycharz J, Świderska E, Balcerczyk A, Szemraj J, Drzewoski J, Śliwińska A. Chronic and Transient Hyperglycemia Induces Changes in the Expression Patterns of IL6 and ADIPOQ Genes and Their Associated Epigenetic Modifications in Differentiating Human Visceral Adipocytes. Int J Mol Sci 2021; 22:ijms22136964. [PMID: 34203452 PMCID: PMC8268546 DOI: 10.3390/ijms22136964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Adipokines secreted by hypertrophic visceral adipose tissue (VAT) instigate low-grade inflammation, followed by hyperglycemia (HG)-related metabolic disorders. The latter may develop with the participation of epigenetic modifications. Our aim was to assess how HG influences selected epigenetic modifications and the expression of interleukin 6 (IL-6) and adiponectin (APN; gene symbol ADIPOQ) during the adipogenesis of human visceral preadipocytes (HPA-v). Adipocytes (Ads) were chronically or transiently HG-treated during three stages of adipogenesis (proliferation, differentiation, maturation). We measured adipokine mRNA, protein, proven or predicted microRNA expression (RT-qPCR and ELISA), and enrichment of H3K9/14ac, H3K4me3, and H3K9me3 at gene promoter regions (chromatin immunoprecipitation). In chronic HG, we detected different expression patterns of the studied adipokines at the mRNA and protein levels. Chronic and transient HG-induced changes in miRNA (miR-26a-5p, miR-26b-5p, let-7d-5p, let-7e-5p, miR-365a-3p, miR-146a-5p) were mostly convergent to altered IL-6 transcription. Alterations in histone marks at the IL6 promoter were also in agreement with IL-6 mRNA. The open chromatin marks at the ADIPOQ promoter mostly reflected the APN transcription during NG adipogenesis, while, in the differentiation stage, HG-induced changes in all studied marks were in line with APN mRNA levels. In summary, HG dysregulated adipokine expression, promoting inflammation. Epigenetic changes coexisted with altered expression of adipokines, especially for IL-6; therefore, epigenetic marks induced by transient HG may act as epi-memory in Ads. Such changes in the epigenome and expression of adipokines could be instrumental in the development of inflammation and metabolic deregulation of VAT.
Collapse
Affiliation(s)
- Adam Wróblewski
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (J.S.); (E.Ś.); (J.S.)
- Correspondence: (A.W.); (A.Ś.)
| | - Justyna Strycharz
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (J.S.); (E.Ś.); (J.S.)
| | - Ewa Świderska
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (J.S.); (E.Ś.); (J.S.)
| | - Aneta Balcerczyk
- Department of Molecular Biophysics, University of Lodz, 90-236 Lodz, Poland;
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (J.S.); (E.Ś.); (J.S.)
| | - Józef Drzewoski
- Central Teaching Hospital of the Medical University of Lodz, 92-213 Lodz, Poland;
| | - Agnieszka Śliwińska
- Department of Nucleic Acids Biochemistry, Medical University of Lodz, 92-213 Lodz, Poland
- Correspondence: (A.W.); (A.Ś.)
| |
Collapse
|
42
|
Monk JM, Liddle DM, Hutchinson AL, Burns JL, Wellings H, Cartwright NM, Muller WJ, Power KA, Robinson LE, Ma DWL. Fish oil supplementation increases expression of mammary tumor apoptosis mediators and reduces inflammation in an obesity-associated HER-2 breast cancer model. J Nutr Biochem 2021; 95:108763. [PMID: 33965532 DOI: 10.1016/j.jnutbio.2021.108763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 03/10/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022]
Abstract
Obesity is associated with inflammation and has been shown to increase breast cancer severity. The objective of this study was to examine the effect of fish oil (FO) supplementation in obesity-associated mammary tumorigenesis in the MMTV-neu(ndl)-YD5 mouse model of human epidermal growth factor receptor-2 positive BC. Female mice were fed one of three diets for 16 weeks: i) high fat diet [HF, % kacl: 41.2% lard, 18.7% corn oil (CO)], ii) an isocaloric HF plus menhaden FO diet (HF+FO, % kcal: 41.2 lard, 13.4% CO, 5.3% FO), iii) low fat diet (LF, % kcal: 4.7% lard, 6% CO). HF mice had increased body weight, visceral adipose weight and serum hormone concentrations (increased leptin and resistin; decreased adiponectin) versus LF, which was attenuated in the HF+FO group versus HF (P<.05). Compared to HF, tumor onset was delayed in HF+FO and LF mice (P<0.05). Compared to HF, HF+FO reduced mammary tumor multiplicity (-27%), tumor weight (-46%) and total tumor volume (-50%) (P<0.05). Additionally, HF+FO reduced mammary tumor multiplicity (-33%), tumor weight (-39%) and total tumor volume (-60%) versus LF. HF+FO improved mammary tumor apoptosis status with increased expression of pro-apoptotic Bad and decreased expression of anti-apoptotic Bcl-xLmediators versus HF (P<0.05). Additionally, HF+FO decreased tumor protein expression of activated Akt, NFκB p65 and STAT3, versus HF (P<0.05). Tumor mRNA expression of inflammatory mediators TNFα, IL-6 and leptin were reduced in HF+FO, whereas IL-10 expression was increased compared to HF (P<0.05). Collectively these results demonstrate the efficacy of FO supplementation for improving obesity-associated breast cancer outcomes.
Collapse
Affiliation(s)
- Jennifer M Monk
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1.
| | - Danyelle M Liddle
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - Amber L Hutchinson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - Jessie L Burns
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - Hannah Wellings
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - Nadia M Cartwright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - William J Muller
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research, Montreal, QC, Canada
| | - Krista A Power
- School of Nutrition Sciences, University of Ottawa, Ottawa ON, Canada, K1H 8L1
| | - Lindsay E Robinson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - David W L Ma
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1.
| |
Collapse
|
43
|
Nakamura S, Yonekura S, Shimosato T, Takaya T. Myogenetic Oligodeoxynucleotide (myoDN) Recovers the Differentiation of Skeletal Muscle Myoblasts Deteriorated by Diabetes Mellitus. Front Physiol 2021; 12:679152. [PMID: 34108889 PMCID: PMC8181739 DOI: 10.3389/fphys.2021.679152] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle wasting in patients with diabetes mellitus (DM) is a complication of decreased muscle mass and strength, and is a serious risk factor that may result in mortality. Deteriorated differentiation of muscle precursor cells, called myoblasts, in DM patients is considered to be one of the causes of muscle wasting. We recently developed myogenetic oligodeoxynucleotides (myoDNs), which are 18-base single-strand DNAs that promote myoblast differentiation by targeting nucleolin. Herein, we report the applicability of a myoDN, iSN04, to myoblasts isolated from patients with type 1 and type 2 DM. Myogenesis of DM myoblasts was exacerbated concordantly with a delayed shift of myogenic transcription and induction of interleukins. Analogous phenotypes were reproduced in healthy myoblasts cultured with excessive glucose or palmitic acid, mimicking hyperglycemia or hyperlipidemia. iSN04 treatment recovered the deteriorated differentiation of plural DM myoblasts by downregulating myostatin and interleukin-8 (IL-8). iSN04 also ameliorated the impaired myogenic differentiation induced by glucose or palmitic acid. These results demonstrate that myoDNs can directly facilitate myoblast differentiation in DM patients, making them novel candidates for nucleic acid drugs to treat muscle wasting in patients with DM.
Collapse
Affiliation(s)
- Shunichi Nakamura
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Shinichi Yonekura
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takeshi Shimosato
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Tomohide Takaya
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
44
|
Lopez-Perez D, Redruello-Romero A, Garcia-Rubio J, Arana C, Garcia-Escudero LA, Tamayo F, Puentes-Pardo JD, Moreno-SanJuan S, Salmeron J, Blanco A, Galvez J, Leon J, Carazo Á. In Patients With Obesity, the Number of Adipose Tissue Mast Cells Is Significantly Lower in Subjects With Type 2 Diabetes. Front Immunol 2021; 12:664576. [PMID: 34093556 PMCID: PMC8177010 DOI: 10.3389/fimmu.2021.664576] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes (T2D) is a rising global health problem mainly caused by obesity and a sedentary lifestyle. In healthy individuals, white adipose tissue (WAT) has a relevant homeostatic role in glucose metabolism, energy storage, and endocrine signaling. Mast cells contribute to these functions promoting WAT angiogenesis and adipogenesis. In patients with T2D, inflammation dramatically impacts WAT functioning, which results in the recruitment of several leukocytes, including monocytes, that enhance this inflammation. Accordingly, the macrophages population rises as the WAT inflammation increases during the T2D status worsening. Since mast cell progenitors cannot arrive at WAT, the amount of WAT mast cells depends on how the new microenvironment affects progenitor and differentiated mast cells. Here, we employed a flow cytometry-based approach to analyze the number of mast cells from omental white adipose tissue (o-WAT) and subcutaneous white adipose tissue (s-WAT) in a cohort of 100 patients with obesity. Additionally, we measured the number of mast cell progenitors in a subcohort of 15 patients. The cohort was divided in three groups: non-T2D, pre-T2D, and T2D. Importantly, patients with T2D have a mild condition (HbA1c <7%). The number of mast cells and mast cell progenitors was lower in patients with T2D in both o-WAT and s-WAT in comparison to subjects from the pre-T2D and non-T2D groups. In the case of mast cells in o-WAT, there were statistically significant differences between non-T2D and T2D groups (p = 0.0031), together with pre-T2D and T2D groups (p=0.0097). However, in s-WAT, the differences are only between non-T2D and T2D groups (p=0.047). These differences have been obtained with patients with a mild T2D condition. Therefore, little changes in T2D status have a huge impact on the number of mast cells in WAT, especially in o-WAT. Due to the importance of mast cells in WAT physiology, their decrease can reduce the capacity of WAT, especially o-WAT, to store lipids and cause hypoxic cell deaths that will trigger inflammation.
Collapse
Affiliation(s)
- David Lopez-Perez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain.,Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Anaïs Redruello-Romero
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | | | - Carlos Arana
- Endocrinology and Nutrition Unit, Virgen de las Nieves University Hospital, Granada, Spain
| | - Luis A Garcia-Escudero
- Department of Statistics and Operative Research, Faculty of Sciences, University of Valladolid, Valladolid, Spain
| | | | - Jose D Puentes-Pardo
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain.,Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Sara Moreno-SanJuan
- Cytometry and Microscopy Research Service, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Javier Salmeron
- Gastroenterology Unit, San Cecilio University Hospital, Granada, Spain
| | - Armando Blanco
- Department of Computer Science and Artificial Intelligence, University of Granada, Granada, Spain
| | - Julio Galvez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain.,Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.,Centro de Investigación Biomédica En Red para Enfermedades Hepáticas y Digestivas (CIBER-EHD), Center for Biomedical Research, University of Granada, Granada, Spain
| | - Josefa Leon
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Ángel Carazo
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.,Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, Granada, Spain
| |
Collapse
|
45
|
Masi S, Ambrosini S, Mohammed SA, Sciarretta S, Lüscher TF, Paneni F, Costantino S. Epigenetic Remodeling in Obesity-Related Vascular Disease. Antioxid Redox Signal 2021; 34:1165-1199. [PMID: 32808539 DOI: 10.1089/ars.2020.8040] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The prevalence of obesity and cardiometabolic phenotypes is alarmingly increasing across the globe and is associated with atherosclerotic vascular complications and high mortality. In spite of multifactorial interventions, vascular residual risk remains high in this patient population, suggesting the need for breakthrough therapies. The mechanisms underpinning obesity-related vascular disease remain elusive and represent an intense area of investigation. Recent Advances: Epigenetic modifications-defined as environmentally induced chemical changes of DNA and histones that do not affect DNA sequence-are emerging as a potent modulator of gene transcription in the vasculature and might significantly contribute to the development of obesity-induced endothelial dysfunction. DNA methylation and histone post-translational modifications cooperate to build complex epigenetic signals, altering transcriptional networks that are implicated in redox homeostasis, mitochondrial function, vascular inflammation, and perivascular fat homeostasis in patients with cardiometabolic disturbances. Critical Issues: Deciphering the epigenetic landscape in the vasculature is extremely challenging due to the complexity of epigenetic signals and their function in regulating transcription. An overview of the most important epigenetic pathways is required to identify potential molecular targets to treat or prevent obesity-related endothelial dysfunction and atherosclerotic disease. This would enable the employment of precision medicine approaches in this setting. Future Directions: Current and future research efforts in this field entail a better definition of the vascular epigenome in obese patients as well as the unveiling of novel, cell-specific chromatin-modifying drugs that are able to erase specific epigenetic signals that are responsible for maladaptive transcriptional alterations and vascular dysfunction in obese patients. Antioxid. Redox Signal. 34, 1165-1199.
Collapse
Affiliation(s)
- Stefano Masi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Sebastiano Sciarretta
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Heart Division, Royal Brompton and Harefield Hospital Trust, National Heart & Lung Institute, Imperial College, London, United Kingdom
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| |
Collapse
|
46
|
Pan X, Kaminga AC, Wen SW, Liu A. Chemokines in Prediabetes and Type 2 Diabetes: A Meta-Analysis. Front Immunol 2021; 12:622438. [PMID: 34054797 PMCID: PMC8161229 DOI: 10.3389/fimmu.2021.622438] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Background A growing number of studies found inconsistent results on the role of chemokines in the progression of type 2 diabetes (T2DM) and prediabetes (PDM). The purpose of this meta-analysis was to summarize the results of previous studies on the association between the chemokines system and T2DM/PDM. Methods We searched in the databases, PubMed, Web of Science, Embase and Cochrane Library, for eligible studies published not later than March 1, 2020. Data extraction was performed independently by 2 reviewers, on a standardized, prepiloted form. Group differences in chemokines concentrations were summarized using the standardized mean difference (SMD) with a 95% confidence interval (CI), calculated by performing a meta-analysis using the random-effects model. Results We identified 98 relevant studies that investigated the association between 32 different chemokines and T2DM/PDM. Altogether, these studies involved 14,708 patients and 14,574 controls. Results showed that the concentrations of CCL1, CCL2, CCL4, CCL5, CCL11, CXCL8, CXCL10 and CX3CL1 in the T2DM patients were significantly higher than that in the controls, while no difference in these concentrations was found between the PDM patients and controls. Conclusion Progression of T2DM may be associated with elevated concentrations of chemokines. Meta-Analysis Registration PROSPERO, identifier CRD42019148305.
Collapse
Affiliation(s)
- Xiongfeng Pan
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Atipatsa C Kaminga
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.,Department of Mathematics and Statistics, Mzuzu University, Mzuzu, Malawi
| | - Shi Wu Wen
- OMNI Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Obstetrics and Gynaecology and School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
| | - Aizhong Liu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
47
|
Baker JF, England BR, George M, Cannon G, Sauer B, Ogdie A, Hamilton BC, Hunter C, Duryee MJ, Thiele G, Mikuls TR. Disease activity, cytokines, chemokines and the risk of incident diabetes in rheumatoid arthritis. Ann Rheum Dis 2021; 80:566-572. [PMID: 33397733 PMCID: PMC8928597 DOI: 10.1136/annrheumdis-2020-219140] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/30/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE Rheumatoid arthritis (RA) is associated with a higher risk of diabetes mellitus (DM). Our aim was to determine associations between inflammatory disease activity (including evaluation of specific cytokines and chemokines) and incident DM. METHODS Participants were adults with physician-confirmed RA from Veteran's Affairs Rheumatoid Arthritis Registry. Disease activity and clinical assessments occur longitudinally as part of clinical care. Thirty cytokines and chemokines were measured in banked serum obtained at the time of enrolment. Cytokine/chemokine values were log-adjusted and standardised (per SD). Incident DM was defined based on validated algorithms using diagnostic codes and medications. Multivariable Cox proportional hazard models evaluated associations between clinical factors and incident DM. Independent associations between cytokines/chemokines and incident DM were assessed adjusting for age, sex, race, smoking, body mass index (BMI) and medication use at baseline. RESULTS Among 1866 patients with RA without prevalent DM at enrolment, there were 130 incident cases over 9223 person-years of follow-up. High Disease Activity Score (DAS28)-C reactive protein (CRP), obese BMI, older age and male sex were associated with greater risk for incident DM while current smoking and methotrexate use were protective. Patients using methotrexate were at lower risk. Several cytokines/chemokines evaluated were independently associated (per 1 SD) with DM incidence including interleukin(IL)-1, IL-6 and select macrophage-derived cytokines/chemokines (HR range 1.11-1.26). These associations were independent of the DAS28-CRP. CONCLUSIONS Higher disease activity and elevated levels of cytokines/chemokines are associated with a higher risk of incident DM in patients with RA. Future study may help to determine if targeted treatments in at-risk individuals could prevent the development of DM.
Collapse
Affiliation(s)
- Joshua F Baker
- Rheumatology, Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA, USA
- Departments of Medicine/Rheumatology and Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Bryant R England
- Rheumatology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Rheumatology, VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Michael George
- Departments of Medicine/Rheumatology and Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Grant Cannon
- Rheumatology, VA Salt Lake City Health Care System, Salt Lake City, Utah, USA
| | - Brian Sauer
- VA Salt Lake City Health Care System, Salt Lake City, Utah, USA
- University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Alexis Ogdie
- Departments of Medicine/Rheumatology and Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Bartlett C Hamilton
- University of Nebraska Medical Center and Omaha VA Medical Center, University of Nebraska, Omaha, Nebraska, USA
| | - Carlos Hunter
- University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Michael J Duryee
- Internal Medicine Division of Rheumatology, University of Nebraska System, Lincoln, Nebraska, USA
| | - Geoffrey Thiele
- Internal Medicine, University of Nebraska System, Lincoln, Nebraska, USA
- Research Service, 151, VAMC Omaha, Omaha, Nebraska, USA
| | - Ted R Mikuls
- Department of Medicine, University of Nebraska System, Lincoln, Nebraska, USA
| |
Collapse
|
48
|
Ahmad R, Singh JK, Wunnava A, Al-Obeed O, Abdulla M, Srivastava SK. Emerging trends in colorectal cancer: Dysregulated signaling pathways (Review). Int J Mol Med 2021; 47:14. [PMID: 33655327 PMCID: PMC7834960 DOI: 10.3892/ijmm.2021.4847] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequently detected type of cancer, and the second most common cause of cancer‑related mortality globally. The American Cancer Society predicted that approximately 147,950 individuals would be diagnosed with CRC, out of which 53,200 individuals would succumb to the disease in the USA alone in 2020. CRC‑related mortality ranks third among both males and females in the USA. CRC arises from 3 major pathways: i) The adenoma‑carcinoma sequence; ii) serrated pathway; and iii) the inflammatory pathway. The majority of cases of CRC are sporadic and result from risk factors, such as a sedentary lifestyle, obesity, processed diets, alcohol consumption and smoking. CRC is also a common preventable cancer. With widespread CRC screening, the incidence and mortality from CRC have decreased in developed countries. However, over the past few decades, CRC cases and mortality have been on the rise in young adults (age, <50 years). In addition, CRC cases are increasing in developing countries with a low gross domestic product (GDP) due to lifestyle changes. CRC is an etiologically heterogeneous disease classified by tumor location and alterations in global gene expression. Accumulating genetic and epigenetic perturbations and aberrations over time in tumor suppressor genes, oncogenes and DNA mismatch repair genes could be a precursor to the onset of colorectal cancer. CRC can be divided as sporadic, familial, and inherited depending on the origin of the mutation. Germline mutations in APC and MLH1 have been proven to play an etiological role, resulting in the predisposition of individuals to CRC. Genetic alterations cause the dysregulation of signaling pathways leading to drug resistance, the inhibition of apoptosis and the induction of proliferation, invasion and migration, resulting in CRC development and metastasis. Timely detection and effective precision therapies based on the present knowledge of CRC is essential for successful treatment and patient survival. The present review presents the CRC incidence, risk factors, dysregulated signaling pathways and targeted therapies.
Collapse
Affiliation(s)
- Rehan Ahmad
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh 11472, Saudi Arabia
| | - Jaikee Kumar Singh
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan 303007, India
| | - Amoolya Wunnava
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan 303007, India
| | - Omar Al-Obeed
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh 11472, Saudi Arabia
| | - Maha Abdulla
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh 11472, Saudi Arabia
| | | |
Collapse
|
49
|
Visceral Adipose Tissue of Prediabetic and Diabetic Females Shares a Set of Similarly Upregulated microRNAs Functionally Annotated to Inflammation, Oxidative Stress and Insulin Signaling. Antioxidants (Basel) 2021; 10:antiox10010101. [PMID: 33445738 PMCID: PMC7828194 DOI: 10.3390/antiox10010101] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/09/2021] [Accepted: 01/09/2021] [Indexed: 12/18/2022] Open
Abstract
Hypertrophic and hypoxic visceral adipose tissue (VAT) secretes proinflammatory cytokines promoting insulin resistance (IR), prediabetes and type 2 diabetes (T2DM) microRNAs (miRNAs) are markers of metabolic disorders regulating genes critical for e.g., inflammation, glucose metabolism, and antioxidant defense, with raising diagnostic value. The aim of the current study was to evaluate whether hyperglycemia is able to affect the expression of selected miRNAs in VAT of prediabetic (IFG) and diabetic (T2DM) patients vs. normoglycemic (NG) subjects using qPCR. Statistical analyses suggested that miRNAs expression could be sex-dependent. Thus, we determined 15 miRNAs as differentially expressed (DE) among NG, T2DM, IFG females (miR-10a-5p, let-7d-5p, miR-532-5p, miR-127-3p, miR-125b-5p, let-7a-5p, let-7e-5p, miR-199a-3p, miR-365a-3p, miR-99a-5p, miR-100-5p, miR-342-3p, miR-146b-5p, miR-204-5p, miR-409-3p). Majority of significantly changed miRNAs was similarly upregulated in VAT of female T2DM and IFG patients in comparison to NG subjects, positively correlated with FPG and HbA1c, yet, uncorrelated with WHR/BMI. Enrichment analyses indicated involvement of 11 top DE miRNAs in oxidative stress, inflammation and insulin signaling. Those miRNAs expression changes could be possibly associated with low-grade chronic inflammation and oxidative stress in VAT of hyperglycemic subjects.
Collapse
|
50
|
Goossens GH, Jocken JWE, Blaak EE. Sexual dimorphism in cardiometabolic health: the role of adipose tissue, muscle and liver. Nat Rev Endocrinol 2021; 17:47-66. [PMID: 33173188 DOI: 10.1038/s41574-020-00431-8] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/02/2020] [Indexed: 12/11/2022]
Abstract
Obesity is associated with many adverse health effects, such as an increased cardiometabolic risk. Despite higher adiposity for a given BMI, premenopausal women are at lower risk of cardiometabolic disease than men of the same age. This cardiometabolic advantage in women seems to disappear after the menopause or when type 2 diabetes mellitus develops. Sexual dimorphism in substrate supply and utilization, deposition of excess lipids and mobilization of stored lipids in various key metabolic organs (such as adipose tissue, skeletal muscle and the liver) are associated with differences in tissue-specific insulin sensitivity and cardiometabolic risk profiles between men and women. Moreover, lifestyle-related factors and epigenetic and genetic mechanisms seem to affect metabolic complications and disease risk in a sex-specific manner. This Review provides insight into sexual dimorphism in adipose tissue distribution, adipose tissue, skeletal muscle and liver substrate metabolism and tissue-specific insulin sensitivity in humans, as well as the underlying mechanisms, and addresses the effect of these sex differences on cardiometabolic health. Additionally, this Review highlights the implications of sexual dimorphism in the pathophysiology of obesity-related cardiometabolic risk for the development of sex-specific prevention and treatment strategies.
Collapse
Affiliation(s)
- Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands.
| | - Johan W E Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands.
| |
Collapse
|