1
|
Karlinsey K, Zhou B. How does obesity potentiate autoimmune disease in women? OBESITY MEDICINE 2024; 52:100570. [PMID: 39713554 PMCID: PMC11661832 DOI: 10.1016/j.obmed.2024.100570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Affiliation(s)
- Keaton Karlinsey
- Department of Immunology, School of Medicine, University of Connecticut, UConn Health, Farmington, CT 06030, USA
| | - Beiyan Zhou
- Department of Immunology, School of Medicine, University of Connecticut, UConn Health, Farmington, CT 06030, USA
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
2
|
He J, Yang Y, Zhang T, Wu C, Bao Y, Wang J, Jiang F. Mendelian randomization study reveals a causal relationship between body mass index in children and risk of autoimmune diseases. Medicine (Baltimore) 2024; 103:e40094. [PMID: 39465791 PMCID: PMC11479409 DOI: 10.1097/md.0000000000040094] [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: 12/20/2023] [Accepted: 09/26/2024] [Indexed: 10/29/2024] Open
Abstract
Newly emerging evidence indicates that body mass index (BMI) is a potential risk factor for autoimmune diseases (ADs). Nevertheless, the exact causal connection between ADs and BMI in children remains uncertain. To investigate the relationship between BMI in children and ADs, a 2-sample Mendelian randomization (MR) analysis was conducted. In this analysis, several regression methods were utilized, including the inverse-variance weighted (IVW), weighted mode, weighted median, and MR-Egger regression. Publicly available summary statistics datasets from meta-analyses of genome-wide association studies (GWAS) were employed, specifically focusing on BMI in children of European descent (n = 39,620) from the UK Biobank (ebi-a-GCST90002409) as the exposure group. The outcomes were derived from individuals included in the Finnish biobank study FinnGen, with 42,202 cases and 176,590 controls representing the ADs group (finngen_R5_AUTOIMMUNE). For instrumental variables, we carefully selected 16 single nucleotide polymorphisms (SNPs) from GWAS on BMI in children. Our analysis implemented the IVW method, which demonstrated supporting evidence for a causal association between BMI in children and ADs. The results indicated a significant effect with a beta coefficient of 0.22, standard error (SE) of 0.05, odds ratio (OR) of 1.25, and a 95% confidence interval (CI) ranging from 1.13 to 1.38, with a P-value of <.05. We also utilized the weighted median method, which yielded similar findings to the IVW method. The OR estimates from the weighted median analysis showed a beta coefficient of 0.20, SE of 0.06, OR of 1.22, and a 95% CI ranging from 1.08 to 1.36, with a P-value of <.05. No significant association was observed in the MR-Egger and Weighted mode analyses. The findings from the MR analysis suggest that there is evidence supporting a potential causal link between BMI in children and an increased susceptibility to ADs.
Collapse
Affiliation(s)
- Jiahui He
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yun Yang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Ting Zhang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chuyan Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunlei Bao
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jimei Wang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
3
|
Zhou T, Zhang B, Zhang D, Wu Q, Chen J, Li L, Lu Y, Becich MJ, Blecker S, Chilukuri N, Chrischilles EA, Chu H, Corsino L, Geary CR, Hornig M, Hornig-Rohan MM, Kim S, Liebovitz DM, Lorman V, Luo C, Morizono H, Mosa ASM, Pajor NM, Rao S, Razzaghi H, Suresh S, Tedla YG, Utset LV, Wang Y, Williams DA, Witvliet MG, Mangarelli C, Jhaveri R, Forrest CB, Chen Y. Body Mass Index and Postacute Sequelae of SARS-CoV-2 Infection in Children and Young Adults. JAMA Netw Open 2024; 7:e2441970. [PMID: 39466241 PMCID: PMC11581483 DOI: 10.1001/jamanetworkopen.2024.41970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/29/2024] [Indexed: 10/29/2024] Open
Abstract
Importance Obesity is associated with increased severity of COVID-19. Whether obesity is associated with an increased risk of post-acute sequelae of SARS-CoV-2 infection (PASC) among pediatric populations, independent of its association with acute infection severity, is unclear. Objective To quantify the association of body mass index (BMI) status before SARS-CoV-2 infection with pediatric PASC risk, controlling for acute infection severity. Design, Setting, and Participants This retrospective cohort study occurred at 26 US children's hospitals from March 2020 to May 2023 with a minimum follow-up of 179 days. Eligible participants included children and young adults aged 5 to 20 years with SARS-CoV-2 infection. Data analysis was conducted from October 2023 to January 2024. Exposures BMI status assessed within 18 months before infection; the measure closest to the index date was selected. The BMI categories included healthy weight (≥5th to <85th percentile for those aged 5-19 years or ≥18.5 to <25 for those aged >19 years), overweight (≥85th to <95th percentile for those aged 5-19 years or ≥25 to <30 for for those aged >19 years), obesity (≥95th percentile to <120% of the 95th percentile for for those aged 5-19 years or ≥30 to <40 for those aged >19 years), and severe obesity (≥120% of the 95th percentile for those aged 5-19 years or ≥40 for those aged >19 years). Main Outcomes And Measures To identify PASC, a diagnostic code specific for post-COVID-19 conditions was used and a second approach used clusters of symptoms and conditions that constitute the PASC phenotype. Relative risk (RR) for the association of BMI with PASC was quantified by Poisson regression models, adjusting for sociodemographic, acute COVID severity, and other clinical factors. Results A total of 172 136 participants (mean [SD] age at BMI assessment 12.6 [4.4] years; mean [SD] age at cohort entry, 13.1 [4.4] years; 90 187 female [52.4%]) were included. Compared with participants with healthy weight, those with obesity had a 25.4% increased risk of PASC (RR, 1.25; 95% CI, 1.06-1.48) and those with severe obesity had a 42.1% increased risk of PASC (RR, 1.42; 95% CI, 1.25-1.61) when identified using the diagnostic code. Compared with those with healthy weight, there was an increased risk for any occurrences of PASC symptoms and conditions among those with obesity (RR, 1.11; 95% CI, 1.06-1.15) and severe obesity (RR, 1.17; 95% CI, 1.14-1.21), and the association held when assessing total incident occurrences among those with overweight (RR, 1.05; 95% CI, 1.00-1.11), obesity (RR, 1.13; 95% CI, 1.09-1.19), and severe obesity (RR, 1.18; 95% CI, 1.14-1.22). Conclusions And Relevance In this cohort study, elevated BMI was associated with a significantly increased PASC risk in a dose-dependent manner, highlighting the need for targeted care to prevent chronic conditions in at-risk children and young adults.
Collapse
Affiliation(s)
- Ting Zhou
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Bingyu Zhang
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia
- The Graduate Group in Applied Mathematics and Computational Science, School of Arts and Sciences, University of Pennsylvania, Philadelphia
| | - Dazheng Zhang
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Qiong Wu
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Biostatistics and Health Data Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jiajie Chen
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Lu Li
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia
- The Graduate Group in Applied Mathematics and Computational Science, School of Arts and Sciences, University of Pennsylvania, Philadelphia
| | - Yiwen Lu
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia
- The Graduate Group in Applied Mathematics and Computational Science, School of Arts and Sciences, University of Pennsylvania, Philadelphia
| | - Michael J. Becich
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Saul Blecker
- Department of Population Health, New York University Grossman School of Medicine, New York
| | - Nymisha Chilukuri
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | | | - Haitao Chu
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis
- Statistical Research and Innovation, Global Biometrics and Data Management, Pfizer Inc, New York, New York
| | - Leonor Corsino
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Carol R. Geary
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha
| | - Mady Hornig
- RECOVER Patient, Caregiver, or Community Advocate Representative, New York, New York
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| | | | - Susan Kim
- Department of Pediatrics, Division of Rheumatology, University of California San Francisco Benioff Children’s Hospital, San Francisco
| | - David M. Liebovitz
- Division of General Internal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Vitaly Lorman
- Applied Clinical Research Center, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Chongliang Luo
- Division of Public Health Sciences, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Hiroki Morizono
- Center for Genetic Medicine Research, Children’s Research Institute, Children’s National Hospital, Washington DC
| | - Abu S. M. Mosa
- Department of Biomedical Informatics, Biostatistics, and Medical Epidemiology, University of Missouri School of Medicine, Columbia
| | - Nathan M. Pajor
- Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Suchitra Rao
- Department of Pediatrics, University of Colorado School of Medicine, Aurora
| | - Hanieh Razzaghi
- Applied Clinical Research Center, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Srinivasan Suresh
- Divisions of Health Informatics & Emergency Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yacob G. Tedla
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leah Vance Utset
- Division of Primary Care Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio
| | - Youfa Wang
- Global Health Institute, Xi’an Jiaotong University, Xi’an, China
| | | | - Margot Gage Witvliet
- RECOVER Patient, Caregiver, or Community Advocate Representative, New York, New York
- Department of Sociology, Social Work and Criminal Justice, Lamar University, Beaumont, Texas
| | - Caren Mangarelli
- Division of Advanced General Pediatrics and Primary Care, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - Ravi Jhaveri
- Division of Pediatric Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - Christopher B. Forrest
- Applied Clinical Research Center, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Yong Chen
- The Center for Health AI and Synthesis of Evidence (CHASE), University of Pennsylvania, Philadelphia
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
- The Graduate Group in Applied Mathematics and Computational Science, School of Arts and Sciences, University of Pennsylvania, Philadelphia
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia
- Penn Medicine Center for Evidence-Based Practice (CEP), University of Pennsylvania, Philadelphia
- Penn Institute for Biomedical Informatics (IBI), University of Pennsylvania, Philadelphia
| |
Collapse
|
4
|
Klobučar S, Kenđel Jovanović G, Kryczyk-Kozioł J, Cigrovski Berković M, Vučak Lončar J, Morić N, Peljhan K, Rahelić D, Mudri D, Bilić-Ćurčić I, Bogović Crnčić T. Association of Dietary Inflammatory Index and Thyroid Function in Patients with Hashimoto's Thyroiditis: An Observational Cross-Sectional Multicenter Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1454. [PMID: 39336495 PMCID: PMC11434592 DOI: 10.3390/medicina60091454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024]
Abstract
Background and Objectives: The available research suggests that dietary patterns with high inflammatory potential, as indicated by a high DII score, may exacerbate inflammation and potentially influence thyroid function. Therefore, the aim of this study was to investigate the associations between the inflammatory potential of a diet and thyroid function in adults with Hashimoto's thyroiditis (HT). Materials and Methods: A total of 149 adults diagnosed with Hashimoto's thyroiditis were enrolled in this observational, cross-sectional, multicenter study. The Dietary Inflammatory Index (DII®) was calculated using a 141-item food frequency questionnaire (FFQ). The serum levels of the thyroid-stimulating hormone (TSH), free thyroxine (fT4), thyroid peroxidase antibodies (TPO-Ab), and high-sensitivity C-reactive protein (hsCRP) were determined. Results: The DII® scores ranged from -3.49 (most anti-inflammatory) to +4.68 (most pro-inflammatory), whereas three DII® tertile ranges were defined as <-1.4, -1.39 to +1.20, and >+1.21, respectively. Participants in tertile 1 (more anti-inflammatory diet) had significantly higher levels of fT4 than those adhering to a more pro-inflammatory diet (p = 0.007). The levels of hsCRP and TSH appeared to increase with increasing the DII® score, but without statistical significance. A significant association was found between the DII® and TSH (β = 0.42, p < 0.001) and between DII® and free thyroxine (β = 0.19, p < 0.001). After adjustment for age, gender, energy intake, and physical activity, a significant positive correlation remained between the DII® and TSH (β = 0.33, p = 0.002) and between the DII® and body mass index (BMI) (β = 0.14, p = 0.04). Conclusions: Adherence to an anti-inflammatory diet appears to be beneficial in patients with Hashimoto's thyroiditis, suggesting that dietary modification aimed at lowering DII® levels may be a valuable strategy to improve clinical outcomes in these patients.
Collapse
Affiliation(s)
- Sanja Klobučar
- Department of Endocrinology, Diabetes and Metabolic Diseases, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia;
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| | - Gordana Kenđel Jovanović
- Department of Health Ecology, Teaching Institute of Public Health of Primorje—Gorski Kotar County, 51000 Rijeka, Croatia;
| | - Jadwiga Kryczyk-Kozioł
- Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, 31-008 Kraków, Poland;
| | - Maja Cigrovski Berković
- Department for Sport and Exercise Medicine, University of Zagreb, Faculty of Kinesiology, 10000 Zagreb, Croatia;
| | - Jelena Vučak Lončar
- Department of Health Studies, University of Zadar, 23000 Zadar, Croatia;
- Department of Endocrinology, Zadar General Hospital, 23000 Zadar, Croatia
| | - Nikolina Morić
- Health Center of Primorje—Gorski Kotar County, 51000 Rijeka, Croatia;
| | - Katarina Peljhan
- Department of Dermatology, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia;
| | - Dario Rahelić
- Faculty of Medicine, J.J. Strossmayer University Osijek, 31000 Osijek, Croatia; (D.R.)
- Endocrinology and Metabolic Diseases, Vuk Vrhovac University Clinic for Diabetes, Merkur University Hospital, 10000 Zagreb, Croatia
- School of Medicine, Catholic University of Croatia, 10000 Zagreb, Croatia
| | - Dunja Mudri
- Faculty of Medicine, J.J. Strossmayer University Osijek, 31000 Osijek, Croatia; (D.R.)
- Clinical Institute for Nuclear Medicine and Radiation Protection, Clinical Hospital Center Osijek, 31000 Osijek, Croatia
| | - Ines Bilić-Ćurčić
- Faculty of Medicine, J.J. Strossmayer University Osijek, 31000 Osijek, Croatia; (D.R.)
- Department of Endocrinology, Clinical Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tatjana Bogović Crnčić
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
- Clinical Department of Nuclear Medicine, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
5
|
Rajeev D, MacIver NJ. Metformin as a Therapeutic Agent for Obesity-Associated Immune Dysfunction. J Nutr 2024; 154:2534-2542. [PMID: 38972391 DOI: 10.1016/j.tjnut.2024.07.001] [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: 03/15/2024] [Revised: 05/16/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
Obesity is associated with impaired immune function, characterized by inflammation, and leading to poor response to infection, impaired vaccine response, increased susceptibility to autoimmune disease, and increased risk of cancer and cancer mortality. Worse, there is evidence that weight loss alone may be insufficient to reverse the immune dysfunction caused by obesity. It is therefore critically important to identify alternative therapeutic approaches to decrease the negative effects of obesity-associated inflammation. In this article, we will review evidence that the antidiabetic drug metformin may be considered as a therapeutic agent for obesity-associated immune dysfunction. Metformin has immunomodulatory effects, stimulating or suppressing the immune response in both a cell-specific and disease-specific manner. Although the mechanism of action of metformin on the immune system remains to be fully elucidated, there is strong evidence that metformin enters select immune cells and disrupts electron transport, leading to both AMP-activated protein kinase (AMPK)-dependent and AMPK-independent effects on immune cell differentiation and cytokine production. These effects of metformin on immune cells have been shown to improve immune responses to infection, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Devika Rajeev
- Department of Nutrition, University of North Carolina at Chapel Hill, NC, United States
| | - Nancie J MacIver
- Department of Nutrition, University of North Carolina at Chapel Hill, NC, United States; Department of Pediatrics, University of North Carolina at Chapel Hill, NC, United States; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, NC, United States.
| |
Collapse
|
6
|
Ma H, Liang X, Li SS, Li W, Li TF. The role of anti-citrullinated protein antibody in pathogenesis of RA. Clin Exp Med 2024; 24:153. [PMID: 38972923 PMCID: PMC11228005 DOI: 10.1007/s10238-024-01359-3] [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: 12/23/2023] [Accepted: 04/21/2024] [Indexed: 07/09/2024]
Abstract
Rheumatoid arthritis (RA) is a common autoimmune rheumatic disease that causes chronic synovitis, bone erosion, and joint destruction. The autoantigens in RA include a wide array of posttranslational modified proteins, such as citrullinated proteins catalyzed by peptidyl arginine deiminase4a. Pathogenic anti-citrullinated protein antibodies (ACPAs) directed against a variety of citrullinated epitopes are abundant both in plasma and synovial fluid of RA patients. ACPAs play an important role in the onset and progression of RA. Intensive and extensive studies are being conducted to unveil the mechanisms of RA pathogenesis and evaluate the efficacy of some investigative drugs. In this review, we focus on the formation and pathogenic function of ACPAs.
Collapse
Affiliation(s)
- Hang Ma
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xu Liang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shan-Shan Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Wei Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Tian-Fang Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
7
|
Corona-Meraz FI, Vázquez-Del Mercado M, Sandoval-García F, Robles-De Anda JA, Tovar-Cuevas AJ, Rosales-Gómez RC, Guzmán-Ornelas MO, González-Inostroz D, Peña-Nava M, Martín-Márquez BT. Biomarkers in Systemic Lupus Erythematosus along with Metabolic Syndrome. J Clin Med 2024; 13:1988. [PMID: 38610754 PMCID: PMC11012563 DOI: 10.3390/jcm13071988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Metabolic syndrome (MetS) is a group of physiological abnormalities characterized by obesity, insulin resistance (IR), and hypertriglyceridemia, which carry the risk of developing cardiovascular disease (CVD) and type 2 diabetes (T2D). Immune and metabolic alterations have been observed in MetS and are associated with autoimmune development. Systemic lupus erythematosus (SLE) is an autoimmune disease caused by a complex interaction of environmental, hormonal, and genetic factors and hyperactivation of immune cells. Patients with SLE have a high prevalence of MetS, in which elevated CVD is observed. Among the efforts of multidisciplinary healthcare teams to make an early diagnosis, a wide variety of factors have been considered and associated with the generation of biomarkers. This review aimed to elucidate some primary biomarkers and propose a set of assessments to improve the projection of the diagnosis and evolution of patients. These biomarkers include metabolic profiles, cytokines, cardiovascular tests, and microRNAs (miRs), which have been observed to be dysregulated in these patients and associated with outcomes.
Collapse
Affiliation(s)
- Fernanda Isadora Corona-Meraz
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Mónica Vázquez-Del Mercado
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Rheumatology Service, Internal Medicine Division, Civil Hospital of Guadalajara “Dr. Juan I. Menchaca”, Guadalajara 44340, Jalisco, Mexico
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Flavio Sandoval-García
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Jesus-Aureliano Robles-De Anda
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Alvaro-Jovanny Tovar-Cuevas
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Roberto-Carlos Rosales-Gómez
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Milton-Omar Guzmán-Ornelas
- Multidisciplinary Health Research Center, Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico; (A.-J.T.-C.); (R.-C.R.-G.); (M.-O.G.-O.)
| | - Daniel González-Inostroz
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Miguel Peña-Nava
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
| | - Beatriz-Teresita Martín-Márquez
- Department of Molecular Biology and Genomics, Institute of Rheumatology and Musculoskeletal System Research, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.V.-D.M.); (F.S.-G.); (J.-A.R.-D.A.); (D.G.-I.); (M.P.-N.)
- Academic Group UDG-CA-703, “Immunology and Rheumatology”, University Center of Health Sciences, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
8
|
Peng Y, Zhang L, Bao X, Qian X, Dong W, Jiang M. Palmitoleic acid-rich oleaginous yeast Scheffersomyces segobiensis DSM 27193 exerts anti-obesity effects by ameliorating hepatic steatosis and adipose tissue hypertrophy. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:2156-2164. [PMID: 37926439 DOI: 10.1002/jsfa.13100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/13/2023] [Accepted: 11/06/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Yeast biomass, encompassing fatty acids, terpenoids, vitamins, antioxidants, enzymes, and other bioactive compounds have been extensively utilized in food-related fields. The safety and potential bioactivities of Scheffersomyces segobiensis DSM 27193, an oleaginous yeast strain, are unclear. RESULTS Scheffersomyces segobiensis DSM 27193 accumulated large palmitoleic acid (POA) levels (43.4 g kg-1 biomass) according to the results of whole-cell components. We annotated the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and predicted the categories and host of the pathogen-host interactions (PHI) genes in S. segobiensis DSM 27193. However, S. segobiensis DSM 27193 did not exert toxic effects in mice. Administration of S. segobiensis DSM 27193 led to substantial weight reduction by diminishing food intake in an obesity mouse model. Additionally, it reversed hepatic steatosis and adipose tissue hypertrophy, and improved abnormalities in serum biochemical profiles such as triglyceride, total cholesterol, low-density lipoprotein cholesterol, lipopolysaccharide, tumor necrosis factor-α, interleukin-1β, and interleukin-6. CONCLUSION This study is the first to illustrate the safety and effects of S. segobiensis DSM 27193 against obesity and offers a scientific rationale for its application in functional food supplements. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yujia Peng
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Lili Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Xinhui Bao
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Xiujuan Qian
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Weiliang Dong
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Min Jiang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing, China
| |
Collapse
|
9
|
Tsilingiris D, Vallianou NG, Spyrou N, Kounatidis D, Christodoulatos GS, Karampela I, Dalamaga M. Obesity and Leukemia: Biological Mechanisms, Perspectives, and Challenges. Curr Obes Rep 2024; 13:1-34. [PMID: 38159164 PMCID: PMC10933194 DOI: 10.1007/s13679-023-00542-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE OF REVIEW To examine the epidemiological data on obesity and leukemia; evaluate the effect of obesity on leukemia outcomes in childhood acute lymphoblastic leukemia (ALL) survivors; assess the potential mechanisms through which obesity may increase the risk of leukemia; and provide the effects of obesity management on leukemia. Preventive (diet, physical exercise, obesity pharmacotherapy, bariatric surgery) measures, repurposing drugs, candidate therapeutic agents targeting oncogenic pathways of obesity and insulin resistance in leukemia as well as challenges of the COVID-19 pandemic are also discussed. RECENT FINDINGS Obesity has been implicated in the development of 13 cancers, such as breast, endometrial, colon, renal, esophageal cancers, and multiple myeloma. Leukemia is estimated to account for approximately 2.5% and 3.1% of all new cancer incidence and mortality, respectively, while it represents the most frequent cancer in children younger than 5 years. Current evidence indicates that obesity may have an impact on the risk of leukemia. Increased birthweight may be associated with the development of childhood leukemia. Obesity is also associated with worse outcomes and increased mortality in leukemic patients. However, there are several limitations and challenges in meta-analyses and epidemiological studies. In addition, weight gain may occur in a substantial number of childhood ALL survivors while the majority of studies have documented an increased risk of relapse and mortality among patients with childhood ALL and obesity. The main pathophysiological pathways linking obesity to leukemia include bone marrow adipose tissue; hormones such as insulin and the insulin-like growth factor system as well as sex hormones; pro-inflammatory cytokines, such as IL-6 and TNF-α; adipocytokines, such as adiponectin, leptin, resistin, and visfatin; dyslipidemia and lipid signaling; chronic low-grade inflammation and oxidative stress; and other emerging mechanisms. Obesity represents a risk factor for leukemia, being among the only known risk factors that could be prevented or modified through weight loss, healthy diet, and physical exercise. Pharmacological interventions, repurposing drugs used for cardiometabolic comorbidities, and bariatric surgery may be recommended for leukemia and obesity-related cancer prevention.
Collapse
Affiliation(s)
- Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, 68100, Alexandroupolis, Greece
| | - Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Nikolaos Spyrou
- Tisch Cancer Institute Icahn School of Medicine at Mount Sinai, 1190 One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Dimitris Kounatidis
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | | | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini Str, 12462, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, 11527, Athens, Greece.
| |
Collapse
|
10
|
Liu S, He M, Jiang J, Duan X, Chai B, Zhang J, Tao Q, Chen H. Triggers for the onset and recurrence of psoriasis: a review and update. Cell Commun Signal 2024; 22:108. [PMID: 38347543 PMCID: PMC10860266 DOI: 10.1186/s12964-023-01381-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/02/2023] [Indexed: 02/15/2024] Open
Abstract
Psoriasis is an immune-mediated inflammatory skin disease, involving a complex interplay between genetic and environmental factors. Previous studies have demonstrated that genetic factors play a major role in the pathogenesis of psoriasis. However, non-genetic factors are also necessary to trigger the onset and recurrence of psoriasis in genetically predisposed individuals, which include infections, microbiota dysbiosis of the skin and gut, dysregulated lipid metabolism, dysregulated sex hormones, and mental illness. Psoriasis can also be induced by other environmental triggers, such as skin trauma, unhealthy lifestyles, and medications. Understanding how these triggers play a role in the onset and recurrence of psoriasis provides insights into psoriasis pathogenesis, as well as better clinical administration. In this review, we summarize the triggers for the onset and recurrence of psoriasis and update the current evidence on the underlying mechanism of how these factors elicit the disease. Video Abstract.
Collapse
Grants
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
Collapse
Affiliation(s)
- Suwen Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mengwen He
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Jian Jiang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoru Duan
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bao Chai
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Jingyu Zhang
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Qingxiao Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China.
| |
Collapse
|
11
|
Hu Y, Bao J, Gao Z, Ye L, Wang L. Sodium-Glucose Cotransporter Protein 2 Inhibitors: Novel Application for the Treatment of Obesity-Associated Hypertension. Diabetes Metab Syndr Obes 2024; 17:407-415. [PMID: 38292009 PMCID: PMC10826576 DOI: 10.2147/dmso.s446904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/13/2024] [Indexed: 02/01/2024] Open
Abstract
Obesity is becoming increasingly prevalent in China and worldwide and is closely related to the development of hypertension. The pathophysiology of obesity-associated hypertension is complex, including an overactive sympathetic nervous system (SNS), activation of the renin-angiotensin-aldosterone system (RAAS), insulin resistance, hyperleptinemia, renal dysfunction, inflammatory responses, and endothelial function, which complicates treatment. Sodium-glucose cotransporter protein 2 (SGLT-2) inhibitors, novel hypoglycemic agents, have been shown to reduce body weight and blood pressure and may serve as potential novel agents for the treatment of obesity-associated hypertension. This review discusses the beneficial mechanisms of SGLT-2 inhibitors for the treatment of obesity-associated hypertension. SGLT-2 inhibitors can inhibit SNS activity, reduce RAAS activation, ameliorate insulin resistance, reduce leptin secretion, improve renal function, and inhibit inflammatory responses. SGLT-2 inhibitors can, therefore, simultaneously target multiple mechanisms of obesity-associated hypertension and may serve as an effective treatment for obesity-associated hypertension.
Collapse
Affiliation(s)
- Yilan Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Jiaqi Bao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Zhicheng Gao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Lifang Ye
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Lihong Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, People’s Republic of China
| |
Collapse
|
12
|
Kwon OC, Park MC. Patients with systemic lupus erythematosus who are underweight have distinct disease characteristics. Lupus 2024; 33:68-74. [PMID: 38050807 DOI: 10.1177/09612033231220726] [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] [Indexed: 12/06/2023]
Abstract
OBJECTIVE This study aimed to detail the disease characteristics of systemic lupus erythematosus (SLE) in individuals who are underweight and assess whether underweight status is associated with SLE disease activity. METHODS This was a retrospective cohort study involving 218 patients newly diagnosed with SLE. Patients were categorized as underweight (body mass index [BMI] <18.5 kg/m2) or not underweight (BMI ≥18.5 kg/m2). We reviewed disease characteristics including the SLE Disease Activity Index 2000 (SLEDAI-2K) at diagnosis. High disease activity was defined as SLEDAI-2K ≥10. Disease characteristics were compared between those who were underweight and not underweight. We used multivariable logistic regression analysis to determine whether underweight status is associated with high disease activity. RESULTS Out of the 218 patients, 35 (16.1%) were underweight and 183 (83.9%) were not. Underweight patients had less renal involvement (5.7% vs 20.2%, p = .040), lower C-reactive protein levels (1.0 [0.3-2.3] mg/L vs 1.2 [0.8-5.0] mg/L, p = .028), and lower SLEDAI-2K scores (6.7 ± 4.6 vs 9.1 ± 5.7, p = .009), and were less likely to be at high disease activity status (22.9% vs 42.6%, p = .028), compared with those who were not underweight. Following adjustment for multiple covariates, being underweight was inversely associated with high disease activity status (adjusted odds ratio = 0.38, 95% confidence interval = 0.16 to 0.92, p = .031). CONCLUSION Patients with SLE who were underweight showed less renal involvement and lower SLEDAI-2K scores compared with those who were not underweight. Moreover, those with SLE who were underweight had a 60% lower risk of exhibiting high disease activity.
Collapse
Affiliation(s)
- Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Min-Chan Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Li Y, Zhang J, Wen J, Liu M, Liu W, Li Y. Large-scale genome-wide association study to identify causal relationships and potential mediators between education and autoimmune diseases. Front Immunol 2023; 14:1249017. [PMID: 38146362 PMCID: PMC10749315 DOI: 10.3389/fimmu.2023.1249017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/08/2023] [Indexed: 12/27/2023] Open
Abstract
Objectives Epidemiological studies suggested a potential connection between education and autoimmune disorders. This study investigated the possible cause-and-effect relationship using a Mendelian randomization approach. Methods We explored the causality between four education traits (n = 257,841~1,131,881) and 22 autoimmune diseases. The mediating role of smoking (632,802 individuals), BMI (681,275 individuals), alcohol (335,394 individuals), and income (397,751 individuals) was also investigated. Transcriptome-wide association study (TWAS) and enriched signaling pathways analysis were used to investigate the underlying biological mechanisms. Results Especially, higher cognitive performance was protective for psoriasis (odds ratio (OR) = 0.69, 95% confidence interval (CI) = 0.60-0.79, p = 6.12×10-8), rheumatoid arthritis (RA) (OR = 0.75, 95% CI = 0.67-0.83, p = 4.62×10-6), and hypothyroidism (OR = 0.83, 95% CI = 0.77-0.90, p = 9.82×10-6). Higher levels of educational attainment decreased risks of psoriasis (OR = 0.61, 95% CI = 0.52-0.72, p = 1.12×10-9), RA (OR = 0.68, 95% CI = 0.59-0.79, p = 1.56×10-7), and hypothyroidism (OR = 0.80, 95% CI = 0.72-0.88, p = 5.00×10-6). The completion of highest-level math class genetically downregulates the incidence of psoriasis (OR = 0.66, 95% CI = 0.58-0.76, p = 2.47×10-9), RA (OR = 0.71, 95% CI = 0.63-0.81, p = 5.28×10-8), and hypothyroidism (OR = 0.85, 95% CI = 0.79-0.92, p = 8.88×10-5). Higher self-reported math ability showed protective effects on Crohn's disease (CD) (OR = 0.67, 95% CI = 0.55-0.81, p = 4.96×10-5), RA (OR = 0.76, 95% CI = 0.67-0.87, p = 5.21×10-5), and psoriasis (OR = 0.76, 95% CI = 0.65-0.88, p = 4.08×10-4). Protein modification and localization, response to arsenic-containing substances may participate in the genetic association of cognitive performance on UC, RA, psoriasis, and hypothyroidism. According to mediation analyses, BMI, smoking, and income served as significant mediators in the causal connection between educational traits and autoimmune diseases. Conclusion Higher levels of education-related factors have a protective effect on the risk of several autoimmune disorders. Reducing smoking and BMI and promoting income equality can mitigate health risks associated with low education levels.
Collapse
Affiliation(s)
- Yingjie Li
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
- The Institution of Hepatology, Central South University, Changsha, China
| | - Jingwei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Mingren Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yongzhen Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Wang PC, Ho KC, Ko WC, Lin WN. Impact of prior bariatric surgery on outcomes of hospitalized patients with systemic lupus erythematosus: a propensity score-matched analysis of the U.S. nationwide inpatient sample. Surg Obes Relat Dis 2023; 19:1382-1390. [PMID: 37735043 DOI: 10.1016/j.soard.2023.06.006] [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: 01/19/2023] [Revised: 05/19/2023] [Accepted: 06/13/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disease that primarily affects young women, has many different manifestations, and the disease severity can wax and wane. OBJECTIVES This study aims to determine the association between prior bariatric surgery and SLE outcomes. SETTING A population-based, retrospective study using data from the U.S. Nationwide Inpatient Sample (NIS) database between 2005 and 2018. METHODS Data of hospitalized patients with SLE and morbid obesity were extracted. Exclusion criteria were age <18 years and missing information on mortality. Patients were separated into 2 groups: with or without prior bariatric surgery. One: 4 propensity score matching (PSM) were performed to balance the characteristics between the groups. Associations between prior bariatric surgery and inpatient outcomes were determined by logistic regressions. RESULTS A total of 38,286 hospitalized patients with SLE and morbid obesity were identified. After exclusions and PSM, there remained 9050 subjects in the study sample (with prior bariatric surgery: 1810; without prior bariatric surgery: 7240), representing 44,952 U.S. inpatients. After adjusting for confounders in multivariable analysis, prior bariatric surgery was significantly associated with lower odds for prolonged length of stay (LOS), unfavorable discharge, and SLE-related complications. Specifically, prior bariatric surgery was significantly associated with lower risks of acute myocardial infarction (AMI)/stroke (aOR = .44, 95% CI: .30-.65), venous thromboembolism (VTE) (aOR = .74, 95% CI: .57-.96), pneumonia (aOR = .60, 95% CI: .47-.76), sepsis (aOR = .62, 95% CI: .51-.75), acute and chronic glomerulonephritis (aOR = .53, 95% CI: .38-.76), acute kidney injury (AKI) (aOR = .64, 95% CI: .52-.77), and anemia (aOR = .77, 95% CI: .68-.87). CONCLUSIONS Prior bariatric surgery is associated with favorable in-hospital outcomes in hospitalized patients with SLE and decreased likelihood for several SLE-related complications.
Collapse
Affiliation(s)
- Po-Chun Wang
- PhD Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City, Taiwan; Department of General Surgery, Mackay Memorial Hospital, Taipei, Taiwan
| | - Kung-Chan Ho
- Department of General Surgery, Mackay Memorial Hospital, Taipei, Taiwan
| | - Wen-Ching Ko
- Department of General Surgery, Mackay Memorial Hospital, Taipei, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
15
|
Huo J, Xu Y, Yu J, Guo Y, Hu X, Ou D, Qu R, Zhao L. Causal association between body mass index and autoimmune thyroiditis: evidence from Mendelian randomization. Eur J Med Res 2023; 28:526. [PMID: 37974233 PMCID: PMC10652572 DOI: 10.1186/s40001-023-01480-1] [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: 06/18/2023] [Accepted: 10/28/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Recent studies have reported associations between body mass index (BMI) and various autoimmune disorders. However, it is still uncertain whether there exists a direct cause-and-effect relationship between BMI and autoimmune thyroiditis (AIT). The aim of our study is to investigate the causal association between BMI and AIT. METHODS We conducted a two-sample summary data Mendelian randomization (MR) analysis using genome-wide association studies (GWAS) summary statistics data related to BMI as exposure, and GWAS summary statistic data sets for AIT as the outcome. Robustly associated single-nucleotide polymorphisms (SNPs) for BMI were selected as instrumental variables (IVs). We used the inverse variance weighted (IVW) method as the primary method and performed other MR methods such as MR-Egger regression, weighted median, simple mode, and weighted mode analyses for further validation. The slope of MR-Egger regression was used to correct for pleiotropy and provide estimates of causality. The p-value for the intercept in MR-Egger was utilized to detect any directional pleiotropic effects. Heterogeneity and sensitivity analyses were performed to assess the robustness of our findings. RESULTS Seventy-eight SNPs were selected from GWAS on BMI as the IVs. Our MR analysis using the IVW method showed a potential causal association between BMI and AIT (OR = 3.071, 95% CI 1.324-7.118). Findings from other MR methods are non-significant, although the direction of effect is consistent. There was no evidence that the result was affected by genetic pleiotropy (MR-Egger regression intercept = 0.01, SE = 0.00025, p = 0.719). Heterogeneity and sensitivity analyses revealed no significant heterogeneity among SNPs, and no single SNP drove the observed associations. CONCLUSION Our findings suggest a potential causal association between BMI and AIT, which may provide a basis for further investigation into the relationship between BMI and AIT. Further studies are required as only the IVW method shows significant results, and the case sample size is small.
Collapse
Affiliation(s)
- Jinlong Huo
- Department of Breast and Thyroid Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yaxuan Xu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou, China
| | - Jie Yu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou, China
| | - Youming Guo
- Department of Breast and Thyroid Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Xiaochi Hu
- Department of Breast and Thyroid Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Dong Ou
- Department of Oncology, The First People's Hospital of Zunyi), The Third Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Rui Qu
- Department of Breast and Thyroid Surgery, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China.
| | - Lijin Zhao
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou, China.
| |
Collapse
|
16
|
Nitecki M, Gerstein HC, Balmakov Y, Tsur E, Babushkin V, Michaeli T, Afek A, Pinhas-Hamiel O, Cukierman-Yaffe T, Twig G. High BMI and the risk for incident type 1 Diabetes Mellitus: a systematic review and meta-analysis of aggregated cohort studies. Cardiovasc Diabetol 2023; 22:300. [PMID: 37919779 PMCID: PMC10623818 DOI: 10.1186/s12933-023-02007-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/25/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND There is uncertainty regarding the role of obesity in type 1 diabetes development. The aim of this systematic review and meta-analysis was to collect and synthesize evidence regarding BMI and the risk of developing type 1 diabetes. METHODS A systematic review and meta-analysis were conducted to assess the association between BMI and incident type 1 diabetes. Databases were searched up to June 2022. Cohort studies were included reporting the association between overweight and/or obesity, as measured by BMI after age 2 years, with incident type 1 diabetes. Independent reviewers extracted data and assessed study quality. Risk estimates were pooled using a random-effects model. RESULTS Ten cohort studies met the inclusion criteria. The seven studies that classified BMI into categories were of high quality and involved 1,690,660 individuals and 1979 incident type 1 diabetes cases. The pooled risk ratio (RR) for type 1 diabetes was 1.35 (95% CI 0.93-1.97) among people with overweight (3 studies); 2.17 (95% CI 1.75-2.69) among people with obesity (5 studies); and 1·87 (95% CI 1.52-2.29) among people with overweight/obesity (two studies merged the categories). These point estimates persisted in sensitivity analyses that addressed the duration of follow-up, variability in baseline risk for incident type 1 diabetes, and potential misclassifications related to exposure or outcome definitions. People with overweight/obesity had a 2.55 (95% CI 1.11-5.86) greater risk for incident type 1 diabetes with positive islet autoantibodies. CONCLUSION This systematic review and meta-analysis of high-quality observational cohort studies indicated an association between high BMI and the risk of type 1 diabetes, in a graded manner.
Collapse
Affiliation(s)
- Maya Nitecki
- Israel Defense Forces, Medical Corps, Ramat Gan, Israel
- Department of Military Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Yulia Balmakov
- Department of Military Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eyal Tsur
- Department of Military Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vladislav Babushkin
- Department of Military Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tomer Michaeli
- Department of Military Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Arnon Afek
- Central Management, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Pinhas-Hamiel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Tali Cukierman-Yaffe
- Department of Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Endocrinology Diabetes and Metabolism, Sheba Medical Center, Ramat Gan, Israel
| | - Gilad Twig
- Department of Military Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- Department of Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Institute of Endocrinology Diabetes and Metabolism, Sheba Medical Center, Ramat Gan, Israel.
- The Gertner Institute for Epidemiology & Health Policy Research, Sheba Medical Center, Ramat Gan, Israel.
| |
Collapse
|
17
|
Vallianou NG, Kounatidis D, Panagopoulos F, Evangelopoulos A, Stamatopoulos V, Papagiorgos A, Geladari E, Dalamaga M. Gut Microbiota and Its Role in the Brain-Gut-Kidney Axis in Hypertension. Curr Hypertens Rep 2023; 25:367-376. [PMID: 37632662 DOI: 10.1007/s11906-023-01263-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE OF REVIEW The role of the gut microbiota in modulating blood pressure is increasingly being recognized, currently. The purpose of this review is to summarize recent findings about the mechanisms involved in hypertension with regard to the phenomenon of "gut dysbiosis." RECENT FINDINGS Gut dysbiosis, i.e., the imbalance between the gut microbiota and the host, is characterized by a disruption of the tight junction proteins, such as occludins, claudins, and JAMs (junctional adhesion molecules), resulting in increased gut permeability or the so called "leaky gut." Due to the influence of genetic as well as environmental factors, various metabolites produced by the gut microbiota, such as indole and p-cresol, are increased. Thereby, uremic toxins, such as indoxyl sulfates and p-cresol sulfates, accumulate in the blood and the urine, causing damage in the podocytes and the tubular cells. In addition, immunological mechanisms are implicated as well. In particular, a switch from M2 macrophages to M1 macrophages, which produce pro-inflammatory cytokines, occurs. Moreover, a higher level of Th17 cells, releasing large amounts of interleukin-17 (IL-17), has been reported, when a diet rich in salt is consumed. Therefore, apart from the aggravation of uremic toxins, which may account for direct harmful effects on the kidney, there is inflammation not only in the gut, but in the kidneys as well. This crosstalk between the gut and the kidney is suggested to play a crucial role in hypertension. Notably, the brain is also implicated, with an increasing sympathetic output. The brain-gut-kidney axis seems to be deeply involved in the development of hypertension and chronic kidney disease (CKD). The notion that, by modulating the gut microbiota, we could regulate blood pressure is strongly supported by the current evidence. A healthy diet, low in animal protein and fat, and low in salt, together with the utilization of probiotics, prebiotics, synbiotics, or postbiotics, may contribute to our fight against hypertension.
Collapse
Affiliation(s)
| | | | - Fotis Panagopoulos
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | | | | | | | - Eleni Geladari
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece
| |
Collapse
|
18
|
Tsilingiris D, Vallianou NG, Karampela I, Christodoulatos GS, Papavasileiou G, Petropoulou D, Magkos F, Dalamaga M. Laboratory Findings and Biomarkers in Long COVID: What Do We Know So Far? Insights into Epidemiology, Pathogenesis, Therapeutic Perspectives and Challenges. Int J Mol Sci 2023; 24:10458. [PMID: 37445634 PMCID: PMC10341908 DOI: 10.3390/ijms241310458] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Long COVID (LC) encompasses a constellation of long-term symptoms experienced by at least 10% of people after the initial SARS-CoV-2 infection, and so far it has affected about 65 million people. The etiology of LC remains unclear; however, many pathophysiological pathways may be involved, including viral persistence; a chronic, low-grade inflammatory response; immune dysregulation and a defective immune response; the reactivation of latent viruses; autoimmunity; persistent endothelial dysfunction and coagulopathy; gut dysbiosis; hormonal and metabolic dysregulation; mitochondrial dysfunction; and autonomic nervous system dysfunction. There are no specific tests for the diagnosis of LC, and clinical features including laboratory findings and biomarkers may not specifically relate to LC. Therefore, it is of paramount importance to develop and validate biomarkers that can be employed for the prediction, diagnosis and prognosis of LC and its therapeutic response, although this effort may be hampered by challenges pertaining to the non-specific nature of the majority of clinical manifestations in the LC spectrum, small sample sizes of relevant studies and other methodological issues. Promising candidate biomarkers that are found in some patients are markers of systemic inflammation, including acute phase proteins, cytokines and chemokines; biomarkers reflecting SARS-CoV-2 persistence, the reactivation of herpesviruses and immune dysregulation; biomarkers of endotheliopathy, coagulation and fibrinolysis; microbiota alterations; diverse proteins and metabolites; hormonal and metabolic biomarkers; and cerebrospinal fluid biomarkers. At present, there are only two reviews summarizing relevant biomarkers; however, they do not cover the entire umbrella of current biomarkers, their link to etiopathogenetic mechanisms or the diagnostic work-up in a comprehensive manner. Herein, we aim to appraise and synopsize the available evidence on the typical laboratory manifestations and candidate biomarkers of LC, their classification based on pathogenetic mechanisms and the main LC symptomatology in the frame of the epidemiological and clinical aspects of the syndrome and furthermore assess limitations and challenges as well as potential implications in candidate therapeutic interventions.
Collapse
Affiliation(s)
- Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece;
| | - Natalia G. Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou Street, 10676 Athens, Greece;
| | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini Street, 12462 Athens, Greece;
| | | | - Georgios Papavasileiou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (G.P.); (D.P.)
| | - Dimitra Petropoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (G.P.); (D.P.)
| | - Faidon Magkos
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, DK-2200 Frederiksberg, Denmark;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (G.P.); (D.P.)
| |
Collapse
|
19
|
Kounatidis D, Vallianou N, Evangelopoulos A, Vlahodimitris I, Grivakou E, Kotsi E, Dimitriou K, Skourtis A, Mourouzis I. SGLT-2 Inhibitors and the Inflammasome: What's Next in the 21st Century? Nutrients 2023; 15:nu15102294. [PMID: 37242177 DOI: 10.3390/nu15102294] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
The nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome in the kidney and the heart is increasingly being suggested to play a key role in mediating inflammation. In the kidney, NLRP3 activation was associated with the progression of diabetic kidney disease. In the heart, activation of the NLRP3 inflammasome was related to the enhanced release of interleukin-1β (IL-1β) and the subsequent induction of atherosclerosis and heart failure. Apart from their glucose-lowering effects, SGLT-2 inhibitors were documented to attenuate activation of the NLRP3, thus resulting in the constellation of an anti-inflammatory milieu. In this review, we focus on the interplay between SGLT-2 inhibitors and the inflammasome in the kidney, the heart and the neurons in the context of diabetes mellitus and its complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Iordanis Mourouzis
- Faculty of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|
20
|
Abdallah HY, Faisal S, Tawfik NZ, Soliman NH, Kishk RM, Ellawindy A. Expression Signature of Immune-Related MicroRNAs in Autoimmune Skin Disease: Psoriasis and Vitiligo Insights. Mol Diagn Ther 2023; 27:405-423. [PMID: 37016095 PMCID: PMC10151313 DOI: 10.1007/s40291-023-00646-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND Psoriasis and vitiligo are both chronic, skin-specific diseases classified as autoimmune diseases due to the involvement of several biochemical pathways in their pathogenesis, similar to those altered in other autoimmune diseases. The role of miRNAs in regulating skin autoimmune function has yet to be fully characterized. AIM The aim of this study was to assess the expression profile of a panel of 11 circulating immune-related miRNAs in patients with autoimmune skin diseases, specifically psoriasis and vitiligo, and correlate their expression signature with the clinicopathological features of the diseases. SUBJECTS AND METHODS Relative gene expression quantification for 11 immune-related circulating miRNAs in plasma was done for 300 subjects-100 patients with psoriasis, 100 patients with vitiligo and 100 normal healthy volunteers-followed by different modalities of bioinformatics analysis for the results. RESULTS The expression levels of all the studied immune-related miRNAs were elevated in both autoimmune skin disorders, with much higher levels of expression in psoriasis than in vitiligo patients. There was a significant correlation between most of the studied miRNAs, suggesting shared target genes and/or pathways. Moreover, all the studied miRNAs showed significant results as biomarkers for autoimmune skin disease, with miRNA-145 being the best candidate. Regarding the clinicopathological data, miRNA-7, miRNA-9, miRNA-145, miRNA-148a, and miRNA-148b were positively correlated with age. All the miRNAs were inversely correlated with obesity and disease duration. CONCLUSION This study highlights the critical role of miRNAs in skin-specific autoimmune diseases that proved to be potential biomarkers for autoimmune skin disorders, warranting their exploration as therapeutic targets.
Collapse
Affiliation(s)
- Hoda Y Abdallah
- Medical Genetics Unit, Histology & Cell Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
- Faculty of Medicine, Center of Excellence in Molecular and Cellular Medicine, Suez Canal University, Ismailia, Egypt.
| | - Salwa Faisal
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Noha Z Tawfik
- Dermatology, Venereology, and Andrology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Nourhan Hassan Soliman
- Clinical Pathology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Rania M Kishk
- Microbiology and Immunology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Alia Ellawindy
- Medical Genetics Unit, Histology & Cell Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
21
|
Yaskolka Meir A, Huang W, Cao T, Hong X, Wang G, Pearson C, Adams WG, Wang X, Liang L. Umbilical cord DNA methylation is associated with body mass index trajectories from birth to adolescence. EBioMedicine 2023; 91:104550. [PMID: 37088033 PMCID: PMC10141503 DOI: 10.1016/j.ebiom.2023.104550] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 02/27/2023] [Accepted: 03/15/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND DNA methylation (DNAm) in cord blood has been associated with various prenatal factors and birth outcomes. This study sought to fill an important knowledge gap: the link of cord DNAm with child postnatal growth trajectories from birth to age 18 years (y). METHODS Using data from a US predominantly urban, low-income, multi-ethnic birth cohort (N = 831), we first applied non-parametric methods to identify body-mass-index percentile (BMIPCT) trajectories from birth to age 18 y (the outcome); then, conducted epigenome-wide association study (EWAS) of the outcome, interrogating over 700,000 CpG sites profiled by the Illumina Infinium MethylationEPIC BeadChip. Multivariate linear regression models and likelihood ratio tests (LRT) were applied to examine the DNAm-outcome association in the overall sample and sex strata. FINDINGS We identified four distinct patterns of BMIPCT trajectories: normal weight (NW), Early overweight or obesity (OWO), Late OWO, and normal to very late OWO. DNAm at CpG18582997 annotated to TPGS1, CpG15241084 of TLR7, and cg24350936 of RAB31 were associated with BMIPCT at birth-to-3 y, 10 y, and 14 y, respectively (LRT FDR < 0.05 for all). INTERPRETATION In this prospective birth cohort study, we identified 4 distinct and robust patterns of growth trajectories from birth to 18 y, which were associated with variations in cord blood DNAm at genes implicated in inflammation induction pathways. These findings, if further replicated, raise the possibility that these DNAm markers along with early assessment of BMIPCT trajectories may help identify young children at high-risk for obesity later in life. FUNDING Detailed in the Acknowledgements section.
Collapse
Affiliation(s)
- Anat Yaskolka Meir
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II, 2nd Floor, Boston, MA 02115, USA
| | - Wanyu Huang
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, John Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Civil and Systems Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Tingyi Cao
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II, 4th Floor, Boston, MA 02115, USA
| | - Xiumei Hong
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, John Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Guoying Wang
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, John Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Colleen Pearson
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, 1 Boston Medical Center Pl, Boston, MA 02118, USA
| | - William G Adams
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, 1 Boston Medical Center Pl, Boston, MA 02118, USA
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, John Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II, 2nd Floor, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Building II, 4th Floor, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Vallianou NG, Kounatidis D, Tsilingiris D, Panagopoulos F, Christodoulatos GS, Evangelopoulos A, Karampela I, Dalamaga M. The Role of Next-Generation Probiotics in Obesity and Obesity-Associated Disorders: Current Knowledge and Future Perspectives. Int J Mol Sci 2023; 24:ijms24076755. [PMID: 37047729 PMCID: PMC10095285 DOI: 10.3390/ijms24076755] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Obesity and obesity-associated disorders pose a major public health issue worldwide. Apart from conventional weight loss drugs, next-generation probiotics (NGPs) seem to be very promising as potential preventive and therapeutic agents against obesity. Candidate NGPs such as Akkermansia muciniphila, Faecalibacterium prausnitzii, Anaerobutyricum hallii, Bacteroides uniformis, Bacteroides coprocola, Parabacteroides distasonis, Parabacteroides goldsteinii, Hafnia alvei, Odoribacter laneus and Christensenella minuta have shown promise in preclinical models of obesity and obesity-associated disorders. Proposed mechanisms include the modulation of gut flora and amelioration of intestinal dysbiosis, improvement of intestinal barrier function, reduction in chronic low-grade inflammation and modulation of gut peptide secretion. Akkermansia muciniphila and Hafnia alvei have already been administered in overweight/obese patients with encouraging results. However, safety issues and strict regulations should be constantly implemented and updated. In this review, we aim to explore (1) current knowledge regarding NGPs; (2) their utility in obesity and obesity-associated disorders; (3) their safety profile; and (4) their therapeutic potential in individuals with overweight/obesity. More large-scale, multicentric and longitudinal studies are mandatory to explore their preventive and therapeutic potential against obesity and its related disorders.
Collapse
Affiliation(s)
- Natalia G. Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou Street, 10676 Athens, Greece
| | - Dimitris Kounatidis
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou Street, 10676 Athens, Greece
| | - Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Fotis Panagopoulos
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou Street, 10676 Athens, Greece
| | - Gerasimos Socrates Christodoulatos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
- Department of Microbiology, Sismanogleio General Hospital, 1 Sismanogleiou Street, 15126 Athens, Greece
| | - Angelos Evangelopoulos
- Roche Hellas Diagnostics S.A., 18-20 Amarousiou-Chalandriou Street, 15125 Athens, Greece
| | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini Street, 12462 Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece
| |
Collapse
|
23
|
Dalamaga M, Nasiri-Ansari N, Spyrou N. Perspectives and Challenges of COVID-19 with Obesity-Related Cancers. Cancers (Basel) 2023; 15:cancers15061771. [PMID: 36980657 PMCID: PMC10046880 DOI: 10.3390/cancers15061771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
The emergence of COVID-19 has created an unprecedented threat worldwide, involving overwhelmed health-care systems in the majority of countries [...]
Collapse
Affiliation(s)
- Maria Dalamaga
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
- Correspondence:
| | - Narjes Nasiri-Ansari
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
| | - Nikolaos Spyrou
- Tisch Cancer Institute Icahn School of Medicine at Mount Sinai, 1190 One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
24
|
Obesity and main urologic cancers: Current systematic evidence, novel biological mechanisms, perspectives and challenges. Semin Cancer Biol 2023; 91:70-98. [PMID: 36893965 DOI: 10.1016/j.semcancer.2023.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/06/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
Urologic cancers (UC) account for 13.1% of all new cancer cases and 7.9% of all cancer-related deaths. A growing body of evidence has indicated a potential causal link between obesity and UC. The aim of the present review is to appraise in a critical and integrative manner evidence from meta-analyses and mechanistic studies on the role of obesity in four prevalent UC (kidney-KC, prostate-PC, urinary bladder-UBC, and testicular cancer-TC). Special emphasis is given on Mendelian Randomization Studies (MRS) corroborating a genetic causal association between obesity and UC, as well as on the role of classical and novel adipocytokines. Furthermore, the molecular pathways that link obesity to the development and progression of these cancers are reviewed. Available evidence indicates that obesity confers increased risk for KC, UBC, and advanced PC (20-82%, 10-19%, and 6-14%, respectively), whereas for TC adult height (5-cm increase) may increase the risk by 13%. Obese females tend to be more susceptible to UBC and KC than obese males. MRS have shown that a higher genetic-predicted BMI may be causally linked to KC and UBC but not PC and TC. Biological mechanisms that are involved in the association between excess body weight and UC include the Insulin-like Growth Factor axis, altered availability of sex hormones, chronic inflammation and oxidative stress, abnormal secretion of adipocytokines, ectopic fat deposition, dysbiosis of the gastrointestinal and urinary tract microbiomes and circadian rhythm dysregulation. Anti-hyperglycemic and non-steroidal anti-inflammatory drugs, statins, and adipokine receptor agonists/antagonists show potential as adjuvant cancer therapies. Identifying obesity as a modifiable risk factor for UC may have significant public health implications, allowing clinicians to tailor individualized prevention strategies for patients with excess body weight.
Collapse
|
25
|
Bieber K, Hundt JE, Yu X, Ehlers M, Petersen F, Karsten CM, Köhl J, Kridin K, Kalies K, Kasprick A, Goletz S, Humrich JY, Manz RA, Künstner A, Hammers CM, Akbarzadeh R, Busch H, Sadik CD, Lange T, Grasshoff H, Hackel AM, Erdmann J, König I, Raasch W, Becker M, Kerstein-Stähle A, Lamprecht P, Riemekasten G, Schmidt E, Ludwig RJ. Autoimmune pre-disease. Autoimmun Rev 2023; 22:103236. [PMID: 36436750 DOI: 10.1016/j.autrev.2022.103236] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022]
Abstract
Approximately 5% of the world-wide population is affected by autoimmune diseases. Overall, autoimmune diseases are still difficult to treat, impose a high burden on patients, and have a significant economic impact. Like other complex diseases, e.g., cancer, autoimmune diseases develop over several years. Decisive steps in the development of autoimmune diseases are (i) the development of autoantigen-specific lymphocytes and (often) autoantibodies and (ii) potentially clinical disease manifestation at a later stage. However, not all healthy individuals with autoantibodies develop disease manifestations. Identifying autoantibody-positive healthy individuals and monitoring and inhibiting their switch to inflammatory autoimmune disease conditions are currently in their infancy. The switch from harmless to inflammatory autoantigen-specific T and B-cell and autoantibody responses seems to be the hallmark for the decisive factor in inflammatory autoimmune disease conditions. Accordingly, biomarkers allowing us to predict this progression would have a significant impact. Several factors, such as genetics and the environment, especially diet, smoking, exposure to pollutants, infections, stress, and shift work, might influence the progression from harmless to inflammatory autoimmune conditions. To inspire research directed at defining and ultimately targeting autoimmune predisease, here, we review published evidence underlying the progression from health to autoimmune predisease and ultimately to clinically manifest inflammatory autoimmune disease, addressing the following 3 questions: (i) what is the current status, (ii) what is missing, (iii) and what are the future perspectives for defining and modulating autoimmune predisease.
Collapse
Affiliation(s)
- Katja Bieber
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Jennifer E Hundt
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Xinhua Yu
- Priority Area Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Marc Ehlers
- Institute of Nutritional Medicine, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Frank Petersen
- Priority Area Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany; Division of Immunobiology, Cincinnati Children's Hospital and University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Khalaf Kridin
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel; Unit of Dermatology and Skin Research Laboratory, Baruch Padeh Medical Center, Poriya, Israel
| | - Kathrin Kalies
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Anika Kasprick
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Stephanie Goletz
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Jens Y Humrich
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Rudolf A Manz
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Axel Künstner
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Christoph M Hammers
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | - Reza Akbarzadeh
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany
| | | | - Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Hanna Grasshoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Alexander M Hackel
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Jeanette Erdmann
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Inke König
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Mareike Becker
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Anja Kerstein-Stähle
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Peter Lamprecht
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany; Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Germany.
| |
Collapse
|
26
|
Avgerinos KI, Liu J, Dalamaga M. Could exercise hormone irisin be a therapeutic agent against Parkinson's and other neurodegenerative diseases? Metabol Open 2023; 17:100233. [PMID: 36785617 PMCID: PMC9918419 DOI: 10.1016/j.metop.2023.100233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 01/29/2023] [Indexed: 02/01/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease (AD). The pathologic hallmarks of the disease are the loss of dopaminergic neurons of substantia nigra pars compacta and the presence of intraneuronal alpha synuclein (a-syn) aggregates. Clinical features of PD include motor symptoms such as bradykinesia, rigidity, tremors, postural instability, and gait impairment, and non-motor symptoms such as constipation, orthostatic hypotension, REM sleep disorder, depression and dementia. Currently, there is no disease-modifying therapy for PD. Several human studies have shown that exercise reduces progression of motor symptoms, improves performance on cognitive tasks, and slows functional deterioration. However, regular exercise may not always be feasible in PD patients. Irisin is an exercise-induced myokine involved in metabolism modulation and body fat reduction, but it also crosses the blood-brain barrier and may mediate some of the benefits of exercise in brain function. Recent evidence has shown that irisin could be therapeutically promising in PD as an "exercise-mimicking" intervention. Exogenous irisin administration decreases brain a-syn pathology and loss of dopaminergic neurons, while it improves motor outcomes in preclinical models. Several other neurodegenerative disorders such as AD share common underlying pathogenetic mechanisms with PD such as protein misfolding and aggregation, neuroinflammation, brain metabolic abnormalities, and neuronal loss. Therefore, investigation of irisin as a disease-modifying therapy could be promising for PD and other neurodegenerative disorders including AD.
Collapse
|
27
|
Tabandeh MR, Taha AS, Addai Ali H, Razijalali M, Mohammadtaghvaei N. Type 2 Diabetes Mellitus Coincident with Clinical and Subclinical Thyroid Dysfunctions Results in Dysregulation of Circulating Chemerin, Resistin and Visfatin. Biomedicines 2023; 11:biomedicines11020346. [PMID: 36830883 PMCID: PMC9952980 DOI: 10.3390/biomedicines11020346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/17/2022] [Accepted: 12/28/2022] [Indexed: 01/27/2023] Open
Abstract
The alterations of circulating adipocytokines have been reported in thyroid diseases or type 2 diabetes mellitus (T2DM), but such data in T2DM coincident with clinical and subclinical thyroid-dysfunctions are limited, and remain to be investigated. We studied the changes in serum chemerin, resisitin and visfatin in T2DM patients with thyroid dysfunctions, and their association with inflammatory and insulin resistance-markers. A total of 272 female and male Iranian participants were selected and divided into six groups: the euthyroid group, T2DM, T2DM coincident with clinical and sub clinical hypothyroidism (SC-HO, and C-HO), and T2DM coincident with clinical and sub clinical hyperthyroidism (SC-HR, C-HR).Demographic characteristics, serum levels of adipocytokines, thyroid hormones, inflammatory factors (IL1-β, IL-6 and CRP) and insulin resistance-markers were determined in all participants. T2DM patients with clinical thyroid dysfunctions showed higher levels of circulating resistin, visfatin, chemerin and inflammatory factors, compared with the T2DM group and T2DM coexisted with subclinical thyroid diseases. No significant differences were observed in circulating adipocytokines and inflammatory markers between T2DM coexisting with subclinical thyroid diseases and those without thyroid dysfunctions. Our results revealed that clinical thyroid dysfunction in T2DM patients was associated with elevated levels of circulating resistin, chemerin, visfatin and inflammatory factors, while no such alteration was detected in T2DM coincident with subclinical thyroid dysfunction.
Collapse
Affiliation(s)
- Mohammad Reza Tabandeh
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran
- Correspondence: ; Tel.: +98-61-33226601
| | - Amal Sattar Taha
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran
| | - Hanaa Addai Ali
- Department of Chemistry, Faculty of Science, University of Kufa, Najaf 54001, Iraq
| | - Mohammad Razijalali
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran
| | - Narges Mohammadtaghvaei
- Department of Laboratory Sciences, School of Paramedical Sciences, Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135783151, Iran
| |
Collapse
|
28
|
Zi C, Wang D, Gao Y, He L. The role of Th17 cells in endocrine organs: Involvement of the gut, adipose tissue, liver and bone. Front Immunol 2023; 13:1104943. [PMID: 36726994 PMCID: PMC9884980 DOI: 10.3389/fimmu.2022.1104943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
T Helper 17 (Th17) cells are adaptive immune cells that play myriad roles in the body. Immune-endocrine interactions are vital in endocrine organs during pathological states. Th17 cells are known to take part in multiple autoimmune diseases over the years. Current evidence has moved from minimal to substantial that Th17 cells are closely related to endocrine organs. Diverse tissue Th17 cells have been discovered within endocrine organs, including gut, adipose tissue, liver and bone, and these cells are modulated by various secretions from endocrine organs. Th17 cells in these endocrine organs are key players in the process of an array of metabolic disorders and inflammatory conditions, including obesity, insulin resistance, nonalcoholic fatty liver disease (NAFLD), primary sclerosing cholangitis (PSC), osteoporosis and inflammatory bowel disease (IBD). We reviewed the pathogenetic or protective functions played by Th17 cells in various endocrine tissues and identified potential regulators for plasticity of it. Furthermore, we discussed the roles of Th17 cells in crosstalk of gut-organs axis.
Collapse
Affiliation(s)
- Changyan Zi
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Die Wang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongxiang Gao
- School of International Education, Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Yongxiang Gao, ; Lisha He,
| | - Lisha He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Yongxiang Gao, ; Lisha He,
| |
Collapse
|
29
|
Zhou L, Song K, Luo W. Association between circulating resistin levels and thyroid dysfunction: A systematic review and meta-analysis. Front Endocrinol (Lausanne) 2023; 13:1071922. [PMID: 36686437 PMCID: PMC9845899 DOI: 10.3389/fendo.2022.1071922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Background As a product of adipose tissue, resistin exceeds other adipokines in its role in regulating appetite, energy expenditure, insulin sensitivity, inflammation, and immunity, similar to thyroid hormones. This study aimed to evaluate the association between resistin levels and thyroid dysfunction and to explore variations in circulating resistin levels before and after treatment for thyroid dysfunction. Methods This study was conducted according to the Preferred Reporting Items for Systematic Review and Meta-Analysis statement. A comprehensive search of PubMed, Embase, and Cochrane databases was conducted until June 15, 2022, with no start date restriction, according to the preregistered protocol (PROSPERO-CRD42022336617). RevMan version 5.4 and R software package version 4.2.0 were used for statistical analyses. Results Fourteen studies with 1716 participants were included in this study. The findings of the meta-analysis confirmed that the resistin levels of patients with thyroid dysfunction were significantly higher than those of the euthyroid function control group (mean difference [MD] = 2.11, 95% confidence interval [CI] = 1.11-3.11, P < 0.00001). Furthermore, the resistin levels of patients with hyperthyroidism (MD = 3.23, 95% CI = 0.68-5.79, P = 0.01) and subclinical hypoidism (MD = 1.37, 95% CI = 0.31-2.42, P = 0.01) were significantly higher than those of euthyroid controls. The resistin levels of patients with thyroid dysfunction after treatment were significantly lower than those before treatment (MD = 1.00, 95% CI = 0.34-1.65, P = 0.003), especially in patients with hyperthyroidism (MD = 2.16, 95% CI = 1.00-3.32, P = 0.0003). Correlation analysis confirmed a positive correlation between resistin levels and free triiodothyronine (FT3) levels in patients with thyroid dysfunction (r = 0.27578, P = 0.001). Conclusions Our meta-analysis demonstrates that resistin levels are significantly higher in patients with thyroid dysfunction, and the resistin levels after treatment in patients with thyroid dysfunction are significantly lower than those before treatment. Correlation analysis shows a positive correlation between resistin levels and FT3 levels in patients with thyroid dysfunction. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022336617.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Postgraduate, Qinghai University, Xining, China
- Department of Endocrinology, Qinghai Provincial People’s Hospital, Xining, China
| | - Kang Song
- Department of Endocrinology, Qinghai Provincial People’s Hospital, Xining, China
| | - Wei Luo
- Department of Endocrinology, Qinghai Provincial People’s Hospital, Xining, China
| |
Collapse
|
30
|
Oboza P, Ogarek N, Olszanecka-Glinianowicz M, Kocelak P. Can type 1 diabetes be an unexpected complication of obesity? Front Endocrinol (Lausanne) 2023; 14:1121303. [PMID: 37065759 PMCID: PMC10102381 DOI: 10.3389/fendo.2023.1121303] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Type 1 diabetes (T1D) is one of the most common chronic autoimmune diseases, characterized by absolute insulin deficiency caused via inflammatory destruction of the pancreatic β-cell. Genetic, epigenetic, and environmental factors play a role in the development of diseases. Almost ⅕ of cases involve people under the age of 20. In recent years, the incidence of both T1D and obesity has been increasing, especially among children, adolescents, and young people. In addition, according to the latest study, the prevalence of overweight or obesity in people with T1D has increased significantly. The risk factors of weight gain included using exogenous insulin, intensifying insulin therapy, fear of hypoglycemia and related decrease in physical activity, and psychological factors, such as emotional eating and binge eating. It has also been suggested that T1D may be a complication of obesity. The relationship between body size in childhood, increase in body mass index values in late adolescence and the development of T1D in young adulthood is considered. Moreover, the coexistence of T1D and T2D is increasingly observed, this situation is called double or hybrid diabetes. This is associated with an increased risk of the earlier development of dyslipidemia, cardiovascular diseases, cancer, and consequently a shortening of life. Thus, the purpose of this review was to summarize the relationships between overweight or obesity and T1D.
Collapse
Affiliation(s)
- Paulina Oboza
- Students’ Scientific Society at the Pathophysiology Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, The Medical University of Silesia, Katowice, Poland
| | - Natalia Ogarek
- Pathophysiology Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, The Medical University of Silesia, Katowice, Poland
| | - Magdalena Olszanecka-Glinianowicz
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, The Medical University of Silesia, Katowice, Poland
| | - Piotr Kocelak
- Pathophysiology Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, The Medical University of Silesia, Katowice, Poland
- *Correspondence: Piotr Kocelak,
| |
Collapse
|
31
|
Rivera-Gonzalez O, Case CT, Wilson NA, Speed JS, Taylor EB. Endothelin receptor antagonism improves glucose tolerance and adipose tissue inflammation in an experimental model of systemic lupus erythematosus. Am J Physiol Endocrinol Metab 2023; 324:E73-E84. [PMID: 36476039 PMCID: PMC9870584 DOI: 10.1152/ajpendo.00274.2022] [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: 10/17/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Endothelin-1 (ET-1) is elevated in patients with systemic lupus erythematosus (SLE), an autoimmune disease characterized by high rates of hypertension, renal injury, and cardiovascular disease. SLE is also associated with an increased prevalence of obesity and insulin resistance compared to the general population. In the present study, we tested the hypothesis that elevated ET-1 in SLE contributes to obesity and insulin resistance. For these studies, we used the NZBWF1 mouse model of SLE, which develops obesity and insulin resistance on a normal chow diet. To test this hypothesis, we treated control (NZW) and SLE (NZBWF1) mice with vehicle, atrasentan (ETA receptor antagonist, 10 mg/kg/day), or bosentan (ETA/ETB receptor antagonist, 100 mg/kg/day) for 4 wk. Neither treatment impacted circulating immunoglobulin levels, but treatment with bosentan lowered anti-dsDNA IgG levels, a marker of SLE disease activity. Treatment with atrasentan and bosentan decreased glomerulosclerosis, and atrasentan lowered renal T-cell infiltration. Body weight was lower in SLE mice treated with atrasentan or bosentan. Endothelin receptor antagonism also improved hyperinsulinemia, homeostatic model assessment for insulin resistance, and glucose tolerance in SLE mice. Adipose tissue inflammation was also improved by endothelin receptor blockade. Taken together, these data suggest a potential therapeutic benefit for SLE patients with obesity and insulin resistance.NEW & NOTEWORTHY SLE is an autoimmune disease that is associated with obesity, insulin resistance, and elevated endothelin-1. The present study demonstrated that pharmacological inhibition of endothelin receptors decreased body weight, insulin resistance, and adipose tissue inflammation in a murine model of SLE. The therapeutic potential of endothelin receptor antagonists to treat obesity-related diseases and pathophysiological conditions, such as autoimmune diseases and insulin resistance, has become increasingly clear.
Collapse
Affiliation(s)
- Osvaldo Rivera-Gonzalez
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Clinton T Case
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Natalie A Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joshua S Speed
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
32
|
Karampela I. Perspectives in vaccines, immune response, therapeutic interventions and COVID-19. Metabol Open 2022; 17:100223. [PMID: 36570684 PMCID: PMC9758070 DOI: 10.1016/j.metop.2022.100223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
33
|
Stiebel-Kalish H, Rubarth K, Shouchane-Blum K, Tiosano A, Lotan I, Hellmann MA, Wilf-Yarkoni A, Bialer O, Flanagan EP, Pittock SJ, Bhatti MT, Schmitz-Hübsch T, Paul F, Asseyer S, Chen JJ. Obesity is associated with myelin oligodendrocyte glycoprotein antibody-associated disease in acute optic neuritis. Sci Rep 2022; 12:21312. [PMID: 36494385 PMCID: PMC9734097 DOI: 10.1038/s41598-022-21592-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/29/2022] [Indexed: 12/13/2022] Open
Abstract
Optic neuritis (ON) is a frequent presentation at onset of multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), and myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD). The pathophysiology underlying these diseases, especially MOGAD, is still being elucidated. While obesity has been reported to potentially be a risk factor for MS, this has not been explored in NMOSD or MOGAD. We aimed to investigate a possible association between obesity (body mass index [BMI] > 30 kg/m2) in patients with MOGAD, aquaporin 4-IgG positive NMOSD (AQP4-IgG+ NMOSD) or MS. In this multicenter non-interventional retrospective study, data was collected from patients with a first ever demyelinating attack of ON subsequently diagnosed with MOGAD (n = 44), AQP4-IgG+ NMOSD (n = 49) or MS (n = 90) between 2005 and 2020. The following data was collected: age, sex, ethnicity, BMI (documented before corticosteroid treatment), and the ON etiology after diagnostic work-up. A mixed model analysis was performed to assess the potential of obesity or BMI to predict MOGAD-ON, and to distinguish MOGAD-ON from AQP4-IgG+ NMOSD-ON and MS-ON. Main outcome measures included BMI in patients with acute ON and subsequent diagnosis of MOGAD, AQP4-IgG+ NMOSD or MS. A higher BMI was significantly associated with a diagnosis of MOGAD-ON (p < 0.001); in MOGAD patients the mean BMI was 31.6 kg/m2 (standard deviation (SD) 7.2), while the mean BMI was 24.7 kg/m2 (SD 5.3) in AQP4-IgG+ NMOSD patients, and 26.9 kg/m2 (SD 6.2) in MS patients. Mixed-effects multinomial logistic regression, adjusted for age and sex, with obesity as a binary variable, revealed that obesity was associated with a higher odds ratio (OR) of a subsequent MOGAD diagnosis (OR 5.466, 95% CI [2.039, 14.650], p = 0.001) in contradistinction with AQP4-IgG+ NMOSD. This study suggests an association between obesity and MOGAD. Our findings require further exploration, but could have significant pathophysiologic implications if confirmed in larger prospective studies.
Collapse
Affiliation(s)
- Hadas Stiebel-Kalish
- grid.12136.370000 0004 1937 0546Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel ,grid.12136.370000 0004 1937 0546Felsenstein Medical Research Center, Petach Tikva, Israel ,grid.413156.40000 0004 0575 344XDivision of Neuro-Ophthalmology, Department of Neuro-Ophthalmology, Rabin Medical Center, Beilinson Hospital, 4941492 Petach Tikva, Israel
| | - Kerstin Rubarth
- grid.6363.00000 0001 2218 4662Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin, Germany ,grid.484013.a0000 0004 6879 971XBerlin Institute of Health (BIH), Berlin, Germany ,grid.6363.00000 0001 2218 4662Institute of Medical Informatics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin, Germany
| | - Karny Shouchane-Blum
- grid.12136.370000 0004 1937 0546Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel ,grid.12136.370000 0004 1937 0546Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Alon Tiosano
- grid.12136.370000 0004 1937 0546Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel ,grid.12136.370000 0004 1937 0546Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Itay Lotan
- grid.12136.370000 0004 1937 0546Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel ,grid.413156.40000 0004 0575 344XDepartment of Neurology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Mark A. Hellmann
- grid.413156.40000 0004 0575 344XDepartment of Neurology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Adi Wilf-Yarkoni
- grid.413156.40000 0004 0575 344XDepartment of Neurology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
| | - Omer Bialer
- grid.12136.370000 0004 1937 0546Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel ,grid.12136.370000 0004 1937 0546Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Eoin P. Flanagan
- grid.66875.3a0000 0004 0459 167XDepartments of Ophthalmology and Neurology, Mayo Clinic, Rochester, MN USA
| | - Sean J. Pittock
- grid.66875.3a0000 0004 0459 167XDepartments of Ophthalmology and Neurology, Mayo Clinic, Rochester, MN USA
| | - M. Tariq Bhatti
- grid.66875.3a0000 0004 0459 167XDepartment of Neurology, Laboratory Medicine and Pathology, Center for MS and Autoimmune Neurology, Mayo Clinic, Rochester, MN USA ,grid.280062.e0000 0000 9957 7758Present Address: The Permanente Medical Group, Oakland, NC USA
| | - Tanja Schmitz-Hübsch
- grid.6363.00000 0001 2218 4662Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Friedemann Paul
- grid.6363.00000 0001 2218 4662Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Susanna Asseyer
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.
| | - John J. Chen
- grid.66875.3a0000 0004 0459 167XDepartments of Ophthalmology and Neurology, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
34
|
The catcher in the gut: Tirzepatide, a dual incretin analog for the treatment of type 2 diabetes mellitus and obesity. Metabol Open 2022; 16:100220. [DOI: 10.1016/j.metop.2022.100220] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/11/2022] Open
|
35
|
Ibrahim HY, Sulaiman GM, Al‐shammaa MS, Ad'hiah AH. Evaluation of interleukins 37 and 38 and vitamin D status in the serum of women with Graves' disease. J Clin Lab Anal 2022; 36:e24776. [PMID: 36397279 PMCID: PMC9756989 DOI: 10.1002/jcla.24776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Graves' disease (GD) is an autoimmune thyroid disorder and recent studies have proposed a role for interleukin (IL)-37, IL-38, and vitamin D (VitD) in the pathophysiology of disease. Therefore, this study investigated the expression of IL-37, IL-38, and VitD in the serum of GD patients and correlations of their levels with some demographic and clinical characteristics. METHODS Serum IL-37, IL-38, and VitD levels were evaluated in 90 women with GD and 93 control women using enzyme-linked immunosorbent assay kits. Depending on therapy, six patients were newly diagnosed (ND; untreated), and 50 patients were receiving only carbimazole (CMZ), while 34 patients were also on CMZ but also received one (31 patients), two (one patient), or three (two patients) doses of radioactive iodine (RAI). RESULTS IL-37 levels were significantly higher in GD patients than in controls, while IL-38 and VitD levels were significantly decreased. As indicated by the area under the curve (AUC), receiver operating characteristic curve analysis demonstrated the potential of IL-37, IL-38, and VitD as biomarkers to distinguish GD patients from controls (AUC = 0.953, 0.959, and 0.793, respectively). Multinomial logistic regression analysis showed that altered levels of IL-37, IL-38, and VitD were most likely associated with the pathogenesis of GD. IL-37 was negatively correlated with IL-38 and VitD, while IL-38 and VitD were positively correlated. CONCLUSION Serum Il-37 levels were upregulated in women with GD, while IL-38 and VitD levels showed downregulated levels. The latter two were positively correlated while they showed a negative correlation with IL-37.
Collapse
Affiliation(s)
- Hiba Y. Ibrahim
- Division of Biotechnology, Department of Applied SciencesUniversity of TechnologyBaghdadIraq
| | - Ghassan M. Sulaiman
- Division of Biotechnology, Department of Applied SciencesUniversity of TechnologyBaghdadIraq
| | | | - Ali H. Ad'hiah
- Tropical‐Biological Research Unit, College of ScienceUniversity of BaghdadBaghdadIraq
| |
Collapse
|
36
|
Zucker I, Zloof Y, Bardugo A, Tsur AM, Lutski M, Cohen Y, Cukierman-Yaffe T, Minsky N, Derazne E, Tzur D, Melzer Cohen C, Pinhas-Hamiel O, Chodick G, Raz I, Afek A, Gerstein HC, Tirosh A, Twig G. Obesity in late adolescence and incident type 1 diabetes in young adulthood. Diabetologia 2022; 65:1473-1482. [PMID: 35665825 DOI: 10.1007/s00125-022-05722-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/08/2022] [Indexed: 01/25/2023]
Abstract
AIMS/HYPOTHESIS Studies in children have reported an association between increased BMI and risk for developing type 1 diabetes, but evidence in late adolescence is limited. We studied the association between BMI in late adolescence and incident type 1 diabetes in young adulthood. METHODS All Israeli adolescents, ages 16-19 years, undergoing medical evaluation in preparation for mandatory military conscription between January 1996 and December 2016 were included for analysis unless they had a history of dysglycaemia. Data were linked with information about adult onset of type 1 diabetes in the Israeli National Diabetes Registry. Weight and height were measured at study entry. Cox proportional models were applied, with BMI being analysed both as a categorical and as a continuous variable. RESULTS There were 777 incident cases of type 1 diabetes during 15,819,750 person-years (mean age at diagnosis 25.2±3.9 years). BMI was associated with incident type 1 diabetes. In a multivariable model adjusted for age, sex and sociodemographic variables, the HRs for type 1 diabetes were 1.05 (95% CI 0.87, 1.27) for the 50th-74th BMI percentiles, 1.41 (95% CI 1.11, 1.78) for the 75th-84th BMI percentiles, 1.54 (95% CI 1.23, 1.94) for adolescents who were overweight (85th-94th percentiles), and 2.05 (95% CI 1.58, 2.66) for adolescents with obesity (≥95th percentile) (reference group: 5th-49th BMI percentiles). One increment in BMI SD was associated with a 25% greater risk for incidence of type 1 diabetes (HR 1.25, 95% CI 1.17, 1.32). CONCLUSIONS Excessively high BMI in otherwise healthy adolescents is associated with increased risk for incident type 1 diabetes in early adulthood.
Collapse
Affiliation(s)
- Inbar Zucker
- Department of Preventive Medicine and Epidemiology, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Israel Center for Disease Control, Ministry of Health, Ramat Gan, Israel
| | - Yair Zloof
- Department of Preventive Medicine and Epidemiology, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Israel Defense Forces Medical Corps, Ramat Gan, Israel
| | - Aya Bardugo
- Israel Defense Forces Medical Corps, Ramat Gan, Israel
- Department of Military Medicine, Hebrew University, Jerusalem, Israel
| | - Avishai M Tsur
- Department of Preventive Medicine and Epidemiology, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Israel Defense Forces Medical Corps, Ramat Gan, Israel
- Department of Medicine, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Miri Lutski
- Department of Preventive Medicine and Epidemiology, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Israel Center for Disease Control, Ministry of Health, Ramat Gan, Israel
| | - Yaron Cohen
- Israel Defense Forces Medical Corps, Ramat Gan, Israel
- Department of Military Medicine, Hebrew University, Jerusalem, Israel
| | - Tali Cukierman-Yaffe
- Department of Preventive Medicine and Epidemiology, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Noga Minsky
- Department of Preventive Medicine and Epidemiology, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Estela Derazne
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Tzur
- Israel Defense Forces Medical Corps, Ramat Gan, Israel
- Department of Military Medicine, Hebrew University, Jerusalem, Israel
| | - Cheli Melzer Cohen
- Department of Preventive Medicine and Epidemiology, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- MaccabiTech, Maccabi Health Services, Tel Aviv, Israel
| | - Orit Pinhas-Hamiel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- MaccabiTech, Maccabi Health Services, Tel Aviv, Israel
- Department of Pediatric Endocrinology, Edmond and Lilly Safra Children Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Gabriel Chodick
- Department of Preventive Medicine and Epidemiology, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- MaccabiTech, Maccabi Health Services, Tel Aviv, Israel
| | - Itamar Raz
- The Diabetes Unit, Department of Internal Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Arnon Afek
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Central Management, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Hertzel C Gerstein
- Department of Medicine, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Amir Tirosh
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gilad Twig
- Department of Preventive Medicine and Epidemiology, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Israel Defense Forces Medical Corps, Ramat Gan, Israel.
- Department of Military Medicine, Hebrew University, Jerusalem, Israel.
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.
| |
Collapse
|
37
|
Jennings M, Burova M, Hamilton LG, Hunter E, Morden C, Pandya D, Beecham R, Moyses H, Saeed K, Afolabi PR, Calder PC, Dushianthan A. Body mass index and clinical outcome of severe COVID-19 patients with acute hypoxic respiratory failure: Unravelling the “obesity paradox” phenomenon. Clin Nutr ESPEN 2022; 51:377-384. [PMID: 36184231 PMCID: PMC9356629 DOI: 10.1016/j.clnesp.2022.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022]
Abstract
Background and aims Although obesity have been generally shown to be an independent risk factor for poor outcomes in COVID-19 infection, some studies demonstrate a paradoxical protective effect (“obesity paradox”). This study examines the influence of obesity categories on clinical outcomes of severe COVID-19 patients admitted to an intensive care unit with acute hypoxic respiratory failure requiring either non-invasive or invasive mechanical ventilation. Methods This is a single centre, retrospective study of consecutive COVID-19 patients admitted to the intensive care unit between 03/2020 to 03/2021. Patients were grouped according to the NICE Body Mass Index (BMI) category. Admission variables including age, sex, comorbidities, and ICU severity indices (APACHE-II, SOFA and PaO2/FiO2) were collected. Data were compared between BMI groups for outcomes such as need for invasive mechanical ventilation (IMV), renal replacement therapy (RRT) and 28-day and overall hospital mortality. Results 340 patients were identified and of those 333 patients had their BMI documented. Just over half of patients (53%) had obesity. Those with extreme obesity (obesity groups II and III) were younger with fewer comorbidities, but were more hypoxaemic at presentation, than the healthy BMI group. Although non-significant, obesity groups II and III paradoxically showed a lower in-hospital mortality than the healthy weight group. However, adjusted (age, sex, APACHE-II and CCI) competing risk regression analysis showed three-times higher mortality in obese category I (sub-distribution hazard ratio = 3.32 (95% CI 1.30–8.46), p = 0.01) and a trend to higher mortality across all obesity groups compared to the healthy weight group. Conclusions In this cohort, those with obesity were at higher risk of mortality after adjustment for confounders. We did not identify an “obesity paradox” in this cohort. The obesity paradox may be explained by confounding factors such as younger age, fewer comorbidities, and less severe organ failures. The impact of obesity on indicators of morbidity including likelihood of requirement for organ support measures was not conclusively demonstrated and requires further scrutiny.
Collapse
Affiliation(s)
- Michael Jennings
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK; NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Maria Burova
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| | - Laura G Hamilton
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| | - Elsie Hunter
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| | - Clare Morden
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| | - Darshni Pandya
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| | - Ryan Beecham
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK; NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Helen Moyses
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Kordo Saeed
- Department of Infection, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, SO16 6YD, UK; Faculty of Medicine, University of Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Paul R Afolabi
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Tremona Road, Southampton SO16 6YD, UK; Faculty of Medicine, University of Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Philip C Calder
- NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Tremona Road, Southampton SO16 6YD, UK; Faculty of Medicine, University of Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Ahilanandan Dushianthan
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK; NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Tremona Road, Southampton SO16 6YD, UK; Faculty of Medicine, University of Southampton, Tremona Road, Southampton SO16 6YD, UK.
| |
Collapse
|
38
|
Arias-de la Rosa I, Escudero-Contreras A, Ruiz-Ponce M, Cuesta-López L, Román-Rodríguez C, Pérez-Sánchez C, Ruiz-Limón P, Ruiz RG, Leiva-Cepas F, Alcaide J, Segui P, Plasencia C, Martinez-Feito A, Font P, Ábalos MC, Ortega R, Malagón MM, Tinahones FJ, Collantes-Estévez E, López-Pedrera C, Barbarroja N. Pathogenic mechanisms involving the interplay between adipose tissue and autoantibodies in Rheumatoid arthritis. iScience 2022; 25:104893. [PMID: 36046189 PMCID: PMC9421387 DOI: 10.1016/j.isci.2022.104893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/04/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022] Open
Abstract
We aimed to evaluate the association between adipose tissue (AT) dysfunction, autoimmunity, and disease activity in rheumatoid arthritis (RA). A cross-sectional study including 150 RA patients and 50 healthy donors and longitudinal study with 122 RA patients treated with anti-tumor necrosis factor (TNF)-α, anti-interleukin 6 receptor (IL6R) or anti-CD20 therapies for 6 months were carried out. In vitro experiments with human AT and adipocyte and macrophage cell lines were performed. A collagen-induced arthritis mouse model was developed. The insulin resistance and the altered adipocytokine profile were associated with disease activity, the presence of anti-citrullinated proteins anti-bodies (ACPAs), and worse response to therapy in RA. AT in the context of arthritis is characterized by an inflammatory state alongside the infiltration of macrophages and B/plasmatic cells, where ACPAs can have a direct impact, inducing inflammation and insulin resistance in macrophages and promoting a defective adipocyte differentiation, partially restored by biologicals. IR is related to disease activity, inflammation, and autoimmunity in RA patients IR state and adipocytokines might be associated with a worse response to biologics Visfatin could be used as a potential biomarker of subclinical atherosclerosis ACPAs might directly impact adipose tissue
Collapse
|
39
|
Kakalij RM, Dsouza DL, Boesen EI. Development of High Fat Diet-Induced Hyperinsulinemia in Mice Is Enhanced by Co-treatment With a TLR7 Agonist. Front Physiol 2022; 13:930353. [PMID: 35874527 PMCID: PMC9298857 DOI: 10.3389/fphys.2022.930353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/17/2022] [Indexed: 12/20/2022] Open
Abstract
Metabolic syndrome (MetS) is common in Systemic Lupus Erythematosus (SLE) patients and is associated with increased cardio-renal risk. Toll-like receptor 7 (TLR7) stimulation promotes the development of SLE through mechanisms including activating type I Interferon (IFN) and autoreactive B cells. The current study tested whether combined TLR7 agonist treatment and exposure to a high fat, high sucrose “Western diet” intervention affects the early-stage development of SLE or MetS features. Female C57BL/6 mice were untreated or treated with the TLR7 agonist imiquimod (IMQ) and fed a high-fat diet (HFD; fat 42% kcal, sucrose 34% kcal) or control diet (fat 12.6% kcal, sucrose 34% kcal) for 6 weeks. Supporting early-stage induction of autoimmunity, spleen weights were significantly increased and anti-nuclear antibody (ANA) positivity was detected in IMQ-treated mice. Increased body weight, gonadal fat pad mass, and plasma leptin levels were observed between HFD and control animals for both IMQ and untreated mice. However, the increase in these parameters with HFD was slightly but significantly diminished in IMQ-treated mice. Both the HFD and IMQ treatments significantly increased fasting blood glucose levels. Notably, IMQ treatment affected fasting insulin concentrations in a diet-dependent manner, with hyperinsulinemia observed in IMQ-HFD treated mice. Together, this indicates that the IMQ model of SLE is associated with metabolic alterations, impaired glycemic control, and hyperinsulinemia under HFD conditions. This model may be helpful in further investigating the relationship between MetS and SLE, and supports a role of TLR7 signaling in promoting or accelerating the development of dysglycemia and hyperinsulinemia.
Collapse
Affiliation(s)
- Rahul M Kakalij
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Del L Dsouza
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Erika I Boesen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
40
|
Liao Z, Kong Y, Zeng L, Wan Q, Hu J, Cai Y. Effects of high-fat diet on thyroid autoimmunity in the female rat. BMC Endocr Disord 2022; 22:179. [PMID: 35840950 PMCID: PMC9287994 DOI: 10.1186/s12902-022-01093-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While contributions of dyslipidemia to autoimmune diseases have been described, its impact on thyroid autoimmunity (TA) is less clear. Programmed cell death 1(PD-1)/PD-ligand 1 (PD-L1) immune checkpoint is crucial in preventing autoimmune attack while its blockade exacerbates TA. We thus unveiled the effect of high-fat diet (HFD) on TA, focusing on the contribution of PD-1/PD-L1. METHODS Female Sprague Dawley (SD) rats were randomly fed with a regular diet or HFD (60% calories from fat) for 24 weeks. Then, thyroid ultrasonography was performed and samples were collected for lipid and thyroid-related parameter measure. RESULTS HFD rats exhibited hyperlipemia and abnormal biosynthesis of the unsaturated fatty acid in serum detected by lipidomics. These rats displayed a relatively lower echogenicity and increased inflammatory infiltration in thyroid accompanied by rising serum thyroid autoantibody levels and hypothyroidism, mimicking human Hashimoto's thyroiditis. These alterations were concurrent with decreased mRNA and immunostaining of intrathyroidal PD-1 and also serum PD-1 levels but not the PD-L1 expression, suggesting a role of a PD-1 pathway. Meanwhile, the infiltration of B and T cell, a key cellular event inhibited by the PD-1 signals, was enhanced in the thyroid of HFD rats, along with thyroid fibrosis and apoptosis. CONCLUSIONS Our data suggest that HFD triggers TA through a mechanism possibly involving downregulation of PD-1-related immunosuppression, providing a novel insight into the link between dyslipidemia and autoimmune toxicities.
Collapse
Affiliation(s)
- Zhengzheng Liao
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, People's Republic of China
| | - Ying Kong
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, People's Republic of China
| | - Liang Zeng
- Department of Otorhinolaryngology, Head & Neck Surgery, the First Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, People's Republic of China
| | - Qing Wan
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, People's Republic of China
| | - Jinfang Hu
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, People's Republic of China.
| | - Yaojun Cai
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, People's Republic of China.
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Jiangxi, 330006, Nanchang, People's Republic of China.
- Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Jiangxi, 330006, Nanchang, People's Republic of China.
| |
Collapse
|
41
|
Alsomali N, Alsharif R, Albalawi B, Alharthi R, Junaidallah W, Alshammari S, Alhawiti F, Alenezi A, Alarieh R, Alsaeed W, AlTowaijri G. Lack of association between B 12 and Body Mass Index among Saudi multiple sclerosis patients. Metabol Open 2022; 14:100181. [PMID: 35392379 PMCID: PMC8980604 DOI: 10.1016/j.metop.2022.100181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/16/2022] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
Background Multiple sclerosis (MS) is a chronic inflammatory demyelinating disorder of the central nervous system. It is an autoimmune disease of multifactorial etiology, linked to a variety of genetic and well-defined environmental factors. It typically affects more women than men and more frequently affects adults aged 20-45 years. Besides, vitamin B12 deficiency and obesity are associated with exacerbating central nervous system inflammation and a higher clinical disability. Objective The study aims to determine the association of the vitamin B12 serum concentration with the Body Mass Index BMI, thyroid-stimulating hormone serum levels and MS clinical features in Saudi MS patients. Methods and results This is a retrospective cohort study, and data were collected from the MS database at the King Fahad Medical City Multiple Sclerosis Clinic, from December 2015 to December 2019. Data were entered and analyzed using the Statistical Package for Social Sciences (SPSS ver. 20, Chicago, IL, USA). Cobalamin, also known as vitamin B12, has a reference concentration that ranges from 138 to 652 Pmol/L in adults. The patient's BMI was calculated by dividing the weight (in kilograms) by the square of the height (in square meters), expressed in kg/m2.Data for 169 MS subjects were collected. A total 83 of them, with a mean age of 36.2 ± 9.57 years, had vitamin B12 results. Of all patients, 16.6% had vitamin B12 deficiency (<138 pmol/L) and 9.52% of them were overweight, BMI kg/m2 = (25-29.9). The mean vitamin B12 level in all MS subjects was 240 ± 117 pmol/L. Moreover, 58.33% of the MS patients had high BMIs (BMI >25). However, no significant correlation was found between vitamin B12 deficiency neither with the BMI nor TSH concentration in MS cases (r = 0.03, p = 0.64), (r = 0.00, P = 0.9) respectively. Conclusion These findings revealed no association between serum vitamin B12 concentration and TSH, BMI in MS clinical parameters, however, further studies are required to validate these results.
Collapse
Affiliation(s)
- N. Alsomali
- Research Centre, Neuroscience Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - R. Alsharif
- Research Centre, Neuroscience Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | - R. Alarieh
- National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - W. Alsaeed
- National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - G. AlTowaijri
- National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
42
|
Choi MY, Hahn J, Malspeis S, Stevens EF, Karlson EW, Sparks JA, Yoshida K, Kubzansky L, Costenbader KH. Association of a Combination of Healthy Lifestyle Behaviors With Reduced Risk of Incident Systemic Lupus Erythematosus. Arthritis Rheumatol 2022; 74:274-283. [PMID: 34313398 PMCID: PMC8792100 DOI: 10.1002/art.41935] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/10/2021] [Accepted: 07/22/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE While previous studies have demonstrated an association between individual factors related to lifestyle and the risk of systemic lupus erythematosus (SLE), it is unclear how the combination of these factors might affect the risk of incident SLE. This study was undertaken to prospectively evaluate whether a combination of healthy lifestyle factors is associated with a lower risk of incident SLE and its subtypes (anti-double-stranded DNA [anti-dsDNA]-positive and anti-dsDNA-negative SLE). METHODS The study included 185,962 women from the Nurses' Health Study (NHS) and NHSII cohorts, among whom there were 203 incident cases of SLE (96 with anti-dsDNA-positive SLE, 107 with anti-dsDNA-negative SLE) during 4,649,477 person-years of follow-up. The Healthy Lifestyle Index Score (HLIS) was calculated at baseline and approximately every 2 years during follow-up, with scores assigned for 5 healthy lifestyle factors: alcohol consumption, body mass index, smoking, diet, and exercise. A time-varying Cox proportional hazards regression model was used to estimate the adjusted hazard ratios (HRs) with 95% confidence intervals (95% CIs) for the risk of SLE. In addition, the percentage of partial population attributable risk (PAR%) of SLE development was calculated. RESULTS A higher HLIS was associated with a lower risk of SLE overall (HR 0.81 [95% CI 0.71-0.94]) and a lower risk of anti-dsDNA-positive SLE (HR 0.78 [95% CI 0.63-0.95]). Women with ≥4 healthy lifestyle factors had the lowest risk of SLE overall (HR 0.42, 95% CI 0.25-0.70) and lowest risk of anti-dsDNA-positive SLE (HR 0.35, 95% CI 0.17-0.75) as compared to women with only 1 healthy behavior or no healthy behaviors. The PAR% of SLE development was 47.7% (95% CI 23.1-66.6%), assuming that the entire population had adhered to at least 4 healthy lifestyle behaviors. CONCLUSION These results indicate that the risk of developing SLE, a disease in which significant evidence of genetic involvement has been established, might be reduced by nearly 50% with adherence to modifiable healthy lifestyle behaviors.
Collapse
Affiliation(s)
- May Y Choi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, and University of Calgary, Calgary, Alberta, Canada
| | - Jill Hahn
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Malspeis
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Emma F Stevens
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth W Karlson
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey A Sparks
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kazuki Yoshida
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura Kubzansky
- Harvard University T.H. Chan School of Public Health, Social and Behavioral Sciences, Boston, Massachusetts, USA
| | - Karen H Costenbader
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
43
|
Yang H, Xia Q, Shen Y, Chen TL, Wang J, Lu YY. Gender-Specific Impact of Metabolic Obesity Phenotypes on the Risk of Hashimoto’s Thyroiditis: A Retrospective Data Analysis Using a Health Check-Up Database. J Inflamm Res 2022; 15:827-837. [PMID: 35173456 PMCID: PMC8835981 DOI: 10.2147/jir.s353384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/28/2022] [Indexed: 11/23/2022] Open
Abstract
Background Hashimoto’s thyroiditis (HT) is recognized as the most common autoimmune thyroid disease, often accompanied by the diffuse enlargement of thyroid with abundant blood flow and elevated level of thyroid autoantibodies. As obesity had a positive association with the risk of HT. Thus, this retrospective study was established to further explore the gender relationship between metabolic obesity phenotypes and the risk of Hashimoto’s thyroiditis (HT). Materials and Methods Data for 3697 subjects aged ≥18 years were randomly collected from a Health check-up database from April to December 2019. Obesity was defined by general obesity (GO; body mass index [BMI] ≥28 kg/m2) and abdominal obesity (AO; waist circumstance, male ≥90 cm, female ≥85 cm). Metabolic unhealthy was defined as having at least one metabolic syndrome component and a homeostasis model assessment of insulin resistance ≥2.5. Obesity phenotypes were divided into three groups: GO, AO, compound obesity (GO+AO). After adjustment for potential confounding factors, multivariate logistic regression was used to assess the association between metabolic obesity phenotypes and risk of HT by sex and explore the correlation between different obesity patterns and HT risk by metabolic health status. Results The incidence of HT was 23.5% and significantly higher among females than males with different metabolic phenotypes (26.2% vs 20.5%, p<0.05), except metabolically healthy AO. Compared with non-obese subjects, different metabolic obesity phenotypes were independent risk factors among males (p<0.05). Among females, unhealthy metabolic status with GO (adjusted odds ratio [OR]=2.62) or AO (adjusted OR=2.87) and metabolically healthy non-GO (adjusted OR=2.05) were risk factors of HT (p<0.05). Increasing BMI categories and waist circumstance quartiles were positively correlated with HT risk (p for trend <0.05). Subgroup analyses indicated that GO+AO (adjusted OR=2.52) or only AO (adjusted OR=2.41) were risk factors for HT for those with unhealthy metabolic status. Moreover, GO+AO (adjusted OR=2.37) was an independent risk factor for HT under healthy metabolic status. Conclusion GO+AO was associated with an increased risk of HT, identifying higher BMI/WC as a significant risk factor for HT. Males with unhealthy metabolic state or obesity and metabolically unhealthy females with obesity are high-risk group for HT. Additionally, only AO and GO+AO conferred increased risk of HT for individuals with metabolic abnormalities.
Collapse
Affiliation(s)
- Hao Yang
- Department of Stomatology, Huadong Sanatorium, Wuxi, 214065, People’s Republic of China
| | - Qing Xia
- Health Examination Center, Huadong Sanatorium, Wuxi, 214065, People’s Republic of China
| | - Yan Shen
- Health Examination Center, Huadong Sanatorium, Wuxi, 214065, People’s Republic of China
| | - Ting-Li Chen
- Department of Ophthalmology, Huadong Sanatorium, Wuxi, 214065, People’s Republic of China
| | - Jing Wang
- Department of Ophthalmology, Huadong Sanatorium, Wuxi, 214065, People’s Republic of China
| | - Ya-Yun Lu
- Health Examination Center, Huadong Sanatorium, Wuxi, 214065, People’s Republic of China
- Correspondence: Ya-Yun Lu, Health Examination Center, Huadong Sanatorium, Wuxi, 214065, People’s Republic of China, Tel +86 13763384379, Email
| |
Collapse
|
44
|
Ciężki S, Kurpiewska E, Bossowski A, Głowińska-Olszewska B. Multi-Faceted Influence of Obesity on Type 1 Diabetes in Children - From Disease Pathogenesis to Complications. Front Endocrinol (Lausanne) 2022; 13:890833. [PMID: 35784568 PMCID: PMC9243381 DOI: 10.3389/fendo.2022.890833] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/13/2022] [Indexed: 01/08/2023] Open
Abstract
The prevalence of overweight and obesity among youth patients with diabetes type 1 is increasing. It is estimated, that even up to 35% of young patients with this type of diabetes, considered so far to be characteristic for slim figure, are overweight or even obese. General increase of obesity in children's population complicates differential diagnosis of the type of diabetes in youths. Coexistence of obesity has clinical implications for all stages of diabetes course. It is confirmed that obesity is the risk factor for autoimmune diabetes, and is connected with the earlier onset of diabetes in predisposed patients. Many diabetic patients with obesity present additional risk factors for macroangiopathy, and are recognised to present metabolic syndrome, insulin resistance, and typical for diabetes type 2 - polycystic ovary syndrome, or non-alcoholic fatty liver disease. The prevalence of obesity rises dramatically in adolescence of diabetic child, more often in girls. It has negative impact on metabolic control, glycaemic variability and insulin demand. The risk for microangiopathic complications increases as well. The treatment is difficult and includes not only insulinotherapy and non-pharmacological trials. Recently treatment of insulin resistance with biguanids, and treatment with typical for type 2 new diabetes drugs like GLP-1 analogues, SGLT-2 receptor inhibitors, or even cases of bariatric surgery also has been reported.
Collapse
|
45
|
Kumar V, Kiran S, Kumar S, Singh UP. Extracellular vesicles in obesity and its associated inflammation. Int Rev Immunol 2022; 41:30-44. [PMID: 34423733 PMCID: PMC8770589 DOI: 10.1080/08830185.2021.1964497] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Obesity is characterized by low-grade, chronic inflammation, which promotes insulin resistance and diabetes. Obesity can lead to the development and progression of many autoimmune diseases, including inflammatory bowel disease, psoriasis, psoriatic arthritis, rheumatoid arthritis, thyroid autoimmunity, and type 1 diabetes mellitus (T1DM). These diseases result from an alteration of self-tolerance by promoting pro-inflammatory immune response by lowering numbers of regulatory T cells (Tregs), increasing Th1 and Th17 immune responses, and inflammatory cytokine production. Therefore, understanding the immunological changes that lead to this low-grade inflammatory milieu becomes crucial for the development of therapies that suppress the risk of autoimmune diseases and other immunological conditions. Cells generate extracellular vesicles (EVs) to eliminate cellular waste as well as communicating the adjacent and distant cells through exchanging the components (genetic material [DNA or RNA], lipids, and proteins) between them. Immune cells and adipocytes from individuals with obesity and a high basal metabolic index (BMI) produce also release exosomes (EXOs) and microvesicles (MVs), which are collectively called EVs. These EVs play a crucial role in the development of autoimmune diseases. The current review discusses the immunological dysregulation that leads to inflammation, inflammatory diseases associated with obesity, and the role played by EXOs and MVs in the induction and progression of this devastating conditi8on.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee, 38103 USA
| | - Sonia Kiran
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee, 38103 USA
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee, 38103 USA
| | - Udai P. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee, 38103 USA,Correspondence: Udai P Singh, Ph.D., Associate Professor, Department of Pharmaceutical Sciences, College of Pharmacy, 881 Madison Avenue, The University of Tennessee Health Science Center Memphis, TN, 38163 USA,
| |
Collapse
|
46
|
Raicevic M, Samardzic M, Soldatovic I, Curovic Popovic N, Vukovic R. Trends in nationwide incidence of pediatric type 1 diabetes in Montenegro during the last 30 years. Front Endocrinol (Lausanne) 2022; 13:991533. [PMID: 36147568 PMCID: PMC9485557 DOI: 10.3389/fendo.2022.991533] [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: 07/11/2022] [Accepted: 08/18/2022] [Indexed: 12/01/2022] Open
Abstract
Significant and unexplained variations in type 1 diabetes (T1D) incidence through the years were observed all around the world. The update on this disorder's incidence is crucial for adequate healthcare resource planning and monitoring of the disease. The aim of this study was to give an update on the current incidence of pediatric T1D in Montenegro and to analyze incidence changes over time and how the exposure to different factors might have affected it. This retrospective cohort study included a total of 582 patients younger than 15 years who were newly diagnosed with T1D during the past 30 years. The average age at diagnosis was 8.4 ± 3.91 years. The mean annual incidence of T1D in the Montenegro population during the whole study period of 30 years was 15.2/100,000 person-years. Slightly higher incidence rates were observed in male compared to female individuals, and the incidence increased with age, with the highest incidence in the 10-14 age group. If the model is observed as one without jointpoints, the annual percentage change (APC) for the total population is 3.1 (1.8-4.4); for male individuals, 3.8 (2.1-5.5); and for female individuals, 2.1 (0.6-3.5). In 2020, the first year of the coronavirus disease of 2019 (COVID-19) pandemic, in comparison to 2019, the incidence rate increased from 19.7/100,000 to 21.5/100,000, with the highest increase in the age group of 5-9 years. This is the first nationwide report on a 30-year period of T1D incidence trend in Montenegro. It suggests that T1D incidence among Montenegrin children is rising again and that there is a short-term influence of COVID-19 on new-onset T1D.
Collapse
Affiliation(s)
- Maja Raicevic
- Department of Endocrinology, Institute for Children’s Diseases, Clinical Centre of Montenegro, Podgorica, Montenegro
- *Correspondence: Maja Raicevic,
| | - Mira Samardzic
- Department of Endocrinology, Institute for Children’s Diseases, Clinical Centre of Montenegro, Podgorica, Montenegro
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Ivan Soldatovic
- Institute of Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Natasa Curovic Popovic
- Department of Endocrinology, Institute for Children’s Diseases, Clinical Centre of Montenegro, Podgorica, Montenegro
| | - Rade Vukovic
- Department of Endocrinology, Mother and Child Health Care Institute of Serbia “Dr Vukan Cupic”, Belgrade, Serbia
- School of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
47
|
Chylińska-Frątczak A, Pietrzak I, Michalak A, Wyka K, Szadkowska A. Autoimmune reaction against pancreatic beta cells in children and adolescents with simple obesity. Front Endocrinol (Lausanne) 2022; 13:1061671. [PMID: 36589801 PMCID: PMC9794760 DOI: 10.3389/fendo.2022.1061671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION One of the most important complications of obesity is insulin resistance, which leads to carbohydrate metabolism disorders such as type 2 diabetes. However, obesity is also associated with development of an autoimmune response against various organs, including pancreatic beta cells. The prevalence of such autoimmune processes in children and their possible contribution to the increased incidence of type 1 diabetes is currently unclear. Therefore, the present study assessed the prevalence of autoantibodies against pancreatic islet beta cell's antigens in children and adolescents with simple obesity. MATERIAL AND METHODS This prospective observational study included pediatric patients (up to 18 years of age) with simple obesity hospitalized between 2011 and 2016 at the Department of Pediatrics, Diabetology, Endocrinology and Nephrology of the Medical University of Lodz. Children with acute or chronic conditions that might additionally affect insulin resistance or glucose metabolism were excluded. Collected clinical data included sex, age, sexual maturity ratings (Tanner`s scale), body height and weight, waist and hip circumference, amount of body fat and lean body mass. Each participant underwent a 2-hour oral glucose tolerance test with simultaneous measurements of glycaemia and insulinemia at 0`, 60` and 120`. In addition, glycated hemoglobin HbA1c, fasting and stimulated c-peptide, total cholesterol, as well as high- and low-density cholesterol and triglycerides were measured. Insulin resistance was assessed by calculating HOMA-IR index. The following autoantibodies against pancreatic islet beta cells were determined in each child: ICA - antibodies against cytoplasmic antigens of pancreatic islets, GAD - antibodies against glutamic acid decarboxylase, ZnT8 - antibodies against zinc transporter, IA2 - antibodies against tyrosine phosphatase, IAA - antibodies against insulin. RESULTS The study group included 161 children (57.4% boys, mean age 13.1 ± 2.9 years) with simple obesity (mean BMI z-score +2.2 ± 1.6). Among them, 28 (17.4%) were diagnosed with impaired glucose metabolism during OGTT [23 (82.2%) - isolated impaired glucose tolerance (IGT), 3 (10.7%) - isolated impaired fasting glucose (IFG), 2 (7.1%) - IFG and IGT]. Of the children tested, 28 (17.4%) were tested positive for at least one islet-specific autoantibody [with similar percentages in boys (15, 17.4%) and girls (13, 17.3%), p=0.9855], with ICA being the most common (positive in 18, 11.2%), followed by IAA (7, 4.3%), ZnT8 (5, 3.1%), GADA (3, 1.9%) and IA2 (1, 0.6%). There was no association between the presence of the tested antibodies and age, sex, stage of puberty, parameters assessing the degree of obesity, HbA1c, lipid levels and basal metabolic rate. However, autoantibody-positive subjects were more likely to present IFG or IGT in OGTT compared to those who tested completely negative (9, 32.1% vs 19, 14.3%, p=0.0280). Their HOMA-IR was also significantly higher (HOMA-IR: 4.3 ± 1.9 vs 3.4 ± 1.9, p=0.0203) and this difference remained statistically significant after adjusting for sex and age (p=0.0340). CONCLUSIONS Children and adolescents with simple obesity presented a higher prevalence of markers of autoimmune response against pancreatic beta cells than the general population. Most often, they had only one type of antibody - ICA. The presence of autoimmune response indicators against pancreatic islet antigens is more common in obese patients with impaired carbohydrate metabolism and is associated with lower insulin sensitivity.
Collapse
Affiliation(s)
- Aneta Chylińska-Frątczak
- Department of Pediatrics, Endocrinology, Diabetology and Nephrology, Maria Konopnicka University Pediatrics Center, Lodz, Poland
| | - Iwona Pietrzak
- Department of Pediatrics, Diabetology, Endocrinology and Nephrology, Medical University of Lodz, Lodz, Poland
- *Correspondence: Iwona Pietrzak,
| | - Arkadiusz Michalak
- Department of Pediatrics, Diabetology, Endocrinology and Nephrology, Medical University of Lodz, Lodz, Poland
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Krystyna Wyka
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| | - Agnieszka Szadkowska
- Department of Pediatrics, Diabetology, Endocrinology and Nephrology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
48
|
Diakomopoulos A, Dalamaga M, Papadavid E. Understanding the enigmatic association between mycosis fungoides and psoriasis: Report of two cases and review of the literature. Metabol Open 2021; 12:100148. [PMID: 34816115 PMCID: PMC8591362 DOI: 10.1016/j.metop.2021.100148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 01/04/2023] Open
Abstract
Psoriatic patients present an increased risk for developing lymphoma, particularly cutaneous T-cell lymphoma (CTCL). To what degree psoriasis itself through chronic immune stimulation, or the immunosuppressive medications used for its treatment or comorbidities (obesity, diabetes mellitus, etc), or lifestyle (smoking, alcohol, diet, etc) may play a role in the onset of MF is not yet clear. Psoriasis and Mycosis Fungoides (MF), the most common variant of CTCL, represent two distinct entities sharing common pathogenetic mechanisms and a wide spectrum of common clinical features associated with the abnormal activation of T-cells. The aim of this study is to explore the relationship between MF and psoriasis by presenting two cases with clinical and histopathologic features of both psoriasis and MF with a particular emphasis on the time of presentation of both disorders, the use of previous immunosuppressive drugs as well as the therapeutic management of patients. Biopsy of the cutaneous lesions before the introduction of biologics should be incorporated in clinical practice. Biopsy of the cutaneous lesion should also be performed in the case of appearance of psoriasiform lesions during biologic treatment for autoimmune disorders because this may represent an indolent form of MF. Psoriatic patients with poor or no-response to treatment should be examined thoroughly for MF using immunochemistry and, if necessary, molecular biology techniques. In concomitant MF and psoriasis, combination treatment may be beneficial for both entities. Finally, a large multicentric registry of MF patients who were treated for benign dermatoses (i.e. eczema, psoriasis) with classic immunosuppressive drugs and/or biologics is needed to collect data and further clarify the enigmatic relationship between psoriasis, MF and immunosuppressive treatment.
Collapse
Affiliation(s)
- Achilleas Diakomopoulos
- 2nd Department of Dermatology and Venereology, National and Kapodistrian University of Athens Medical School, Attikon Hospital, 1 Rimini Street, 12462, Athens, Chaidari, Greece
| | - Maria Dalamaga
- 2nd Department of Dermatology and Venereology, National and Kapodistrian University of Athens Medical School, Attikon Hospital, 1 Rimini Street, 12462, Athens, Chaidari, Greece.,Department of Biological Chemistry, Medical School, National & Kapodistrian University of Athens, 75 Mikras Asias Street, 11527, Athens, Greece
| | - Evangelia Papadavid
- 2nd Department of Dermatology and Venereology, National and Kapodistrian University of Athens Medical School, Attikon Hospital, 1 Rimini Street, 12462, Athens, Chaidari, Greece
| |
Collapse
|
49
|
Dalamaga M, Christodoulatos GS, Karampela I, Vallianou N, Apovian CM. Understanding the Co-Epidemic of Obesity and COVID-19: Current Evidence, Comparison with Previous Epidemics, Mechanisms, and Preventive and Therapeutic Perspectives. Curr Obes Rep 2021; 10:214-243. [PMID: 33909265 PMCID: PMC8080486 DOI: 10.1007/s13679-021-00436-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW A growing body of evidence suggests that obesity and increased visceral adiposity are strongly and independently linked to adverse outcomes and death due to COVID-19. This review summarizes current epidemiologic data, highlights pathogenetic mechanisms on the association between excess body weight and COVID-19, compares data from previous pandemics, discusses why COVID-19 challenges the "obesity paradox," and presents implications in prevention and treatment as well as future perspectives. RECENT FINDINGS Data from meta-analyses based on recent observational studies have indicated that obesity increases the risks of infection from SARS-CoV-2, severe infection and hospitalization, admission to the ICU and need of invasive mechanical ventilation (IMV), and the risk of mortality, particularly in severe obesity. The risks of IMV and mortality associated with obesity are accentuated in younger individuals (age ≤ 50 years old). The meta-inflammation in obesity intersects with and exacerbates underlying pathogenetic mechanisms in COVID-19 through the following mechanisms and factors: (i) impaired innate and adaptive immune responses; (ii) chronic inflammation and oxidative stress; (iii) endothelial dysfunction, hypercoagulability, and aberrant activation of the complement; (iv) overactivation of the renin-angiotensin-aldosterone system; (v) overexpression of the angiotensin-converting enzyme 2 receptor in the adipose tissue; (vi) associated cardiometabolic comorbidities; (vii) vitamin D deficiency; (viii) gut dysbiosis; and (ix) mechanical and psychological issues. Mechanistic and large epidemiologic studies using big data sources with omics data exploring genetic determinants of risk and disease severity as well as large randomized controlled trials (RCTs) are necessary to shed light on the pathways connecting chronic subclinical inflammation/meta-inflammation with adverse COVID-19 outcomes and establish the ideal preventive and therapeutic approaches for patients with obesity.
Collapse
Affiliation(s)
- Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527 Athens, Greece
| | - Gerasimos Socrates Christodoulatos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527 Athens, Greece
| | - Irene Karampela
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527 Athens, Greece
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 1 Rimini St, Haidari, 12462 Athens, Greece
| | - Natalia Vallianou
- Department of Internal Medicine and Endocrinology, Evangelismos General Hospital of Athens, 45-47 Ypsilantou street, 10676 Athens, Greece
| | - Caroline M. Apovian
- Section of Endocrinology, Diabetes, Nutrition, and Weight Management, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Doctor’s Office Building, 720 Harrison Avenue, Suite, Boston, MA 8100 USA
| |
Collapse
|
50
|
Spyrou N, Vallianou N, Kadillari J, Dalamaga M. The interplay of obesity, gut microbiome and diet in the immune check point inhibitors therapy era. Semin Cancer Biol 2021; 73:356-376. [PMID: 33989733 DOI: 10.1016/j.semcancer.2021.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/22/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
|