1
|
Monahan RC, Li-Gao R, Noordam R, van der Velde JHPM, Willems van Dijk K, de Koning E, Mook-Kanamori DO, le Cessie S, Rosendaal FR, de Mutsert R. Glycoprotein acetyls response to a mixed meal is associated with increased insulin resistance and risk of type 2 diabetes in a middle-aged population. Clin Nutr ESPEN 2025; 66:523-529. [PMID: 40020917 DOI: 10.1016/j.clnesp.2025.02.013] [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: 08/27/2024] [Revised: 01/17/2025] [Accepted: 02/15/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND & AIMS Systemic inflammation increases the risk of type 2 diabetes (T2D). It remains unclear whether the inflammatory meal response identifies other people at risk of T2D than fasting levels alone. We aimed to examine associations between the meal response of glycoprotein acetyls (GlycA) with insulin resistance and incident type 2 diabetes. METHODS In 5755 middle-aged participants (47 % men) without pre-existing diabetes GlycA was measured after an overnight fast and 150 min after a liquid mixed meal (400 mL, 600 kCal, 16 percent of energy (En%) derived from protein, 50 En% carbohydrates, and 34 En% fat). With linear regression, we examined associations between the GlycA meal response and measures of insulin resistance. With Cox regression analyses, we examined associations between the GlycA meal response and incident type 2 diabetes, adjusted for demography and lifestyle factors. RESULTS In our population, mean (SD) fasting GlycA was 1.28 (0.21) mmol/L and mean GlycA meal response was 0.16 (0.13) mmol/L, and these were weakly correlated (ρ = 0.19). After adjustment for confounding, insulin resistance (HOMA-IR) was 1.11-fold higher (95 % CI: 1.09; 1.13) per SD GlycA meal response. During a median follow-up of 6.7 years, 287 participants developed T2D. Per SD GlycA meal response, the adjusted hazard ratio of incident type 2 diabetes was 1.12 (95 % CI: 1.00; 1.26). CONCLUSION In this middle-aged population with overweight, those with a high GlycA meal response, including those with low fasting GlycA, had an increased risk of diabetes. A high inflammatory meal response shows promise to identify other people at risk of T2D than fasting levels alone. This risk group may benefit from prevention by precision nutrition to reduce the inflammatory meal response.
Collapse
Affiliation(s)
- Rory C Monahan
- Clinical Epidemiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands.
| | - Ruifang Li-Gao
- Clinical Epidemiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, LUMC, Leiden, the Netherlands
| | | | - Ko Willems van Dijk
- Department of Human Genetics, LUMC, Leiden, the Netherlands; Department of Internal Medicine, Division of Endocrinology, LUMC, Leiden, the Netherlands
| | - Eelco de Koning
- Department of Internal Medicine, Division of Endocrinology, LUMC, Leiden, the Netherlands; Department of Internal Medicine, Division of Nephrology and Transplantation, LUMC, Leiden, the Netherlands
| | - Dennis O Mook-Kanamori
- Clinical Epidemiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands; Department of Public Health and Primary Care, LUMC, Leiden, the Netherlands
| | - Saskia le Cessie
- Clinical Epidemiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Frits R Rosendaal
- Clinical Epidemiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Renée de Mutsert
- Clinical Epidemiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| |
Collapse
|
2
|
Shokri-Mashhadi N, Baechle C, Schiemann T, Schaefer E, Barbaresko J, Schlesinger S. Effects of carotenoid supplementation on glycemic control: a systematic review and meta-analysis of randomized clinical trials. Eur J Clin Nutr 2025; 79:113-125. [PMID: 39327454 DOI: 10.1038/s41430-024-01511-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/28/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVES We conducted a systematic review and meta-analysis to assess the effects of carotenoid supplementation on glycemic indices, and the certainty of evidence. METHODS A systematic literature search in PubMed, SCOPUS, ISI-Web of Science, and Cochrane Library was conducted from inception up to Jun 17, 2024. Randomized controlled trials (RCTs) investigating the effect of carotenoid supplementation on circulating glycemic parameters were included. Records were excluded when studies reported the effect of co-interventions with other nutrients, did not provide mean differences (MDs) and standard deviations (SD) for outcomes, or administered whole food rather than supplements of carotenoids. Summary mean differences (MDs) and 95% CI between intervention and control groups were estimated using a random-effects model. The risk of bias of the included studies was assessed using the Risk of Bias 2.0 (RoB 2.0) tool. RESULTS Overall, 36 publications with 45 estimated effect sizes were included in the meta-analyses. The overall findings showed an improvement in fasting blood glucose (FBG) (MD = -4.54 mg/dl; 95% CI: -5.9, -3.2; n = 45), and hemoglobin A1C (HbA1C) (MD = -0.25% (95% CI: -0.4, -0.11; n = 22) in the intervention group in comparison with the control group. Moreover, in individuals with type 2 diabetes (T2D), interventions with astaxanthin and fucoxanthin led to a reduction in FBG by 4.36 mg/dl (95% CI: -6.13, -2.6; n = 10). The findings also showed that the intervention with crocin reduced FBG levels by 13.5 mg/dl (95% CI: -15.5, -7.8; n = 5), and HbA1C by 0.55% (95% CI: -0.77, -0.34; n = 5) in individuals with T2D. However, the certainty of evidence was very low. CONCLUSION Carotenoid's supplementation improved glycemic parameters especially in people with T2D. However. the certainty of evidence was very low, mainly due to small sample size, and indirectness. Therefore, no specific recommendations can be provided at present and well-designed RCTs are required. REGISTRY URL: https://www.crd.york.ac.uk/PROSPERO/ REGISTRY NUMBER: CRD42021285084 REGISTRY AND REGISTRY NUMBER FOR SYSTEMATIC REVIEWS OR META-ANALYSES: PROSPERO ID: CRD42021285084.
Collapse
Affiliation(s)
- Nafiseh Shokri-Mashhadi
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Muenchen-Neuherberg, Düsseldorf, Germany.
| | - Christina Baechle
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Muenchen-Neuherberg, Düsseldorf, Germany
| | - Tim Schiemann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Nutritional and Food Sciences, University of Bonn, Bonn, Germany
| | - Edyta Schaefer
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Muenchen-Neuherberg, Düsseldorf, Germany
| | - Janett Barbaresko
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sabrina Schlesinger
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Muenchen-Neuherberg, Düsseldorf, Germany
| |
Collapse
|
3
|
Xu Y, Wang Z, Li S, Su J, Gao L, Ou J, Lin Z, Luo OJ, Xiao C, Chen G. An in-depth understanding of the role and mechanisms of T cells in immune organ aging and age-related diseases. SCIENCE CHINA. LIFE SCIENCES 2025; 68:328-353. [PMID: 39231902 DOI: 10.1007/s11427-024-2695-x] [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: 02/24/2024] [Accepted: 07/28/2024] [Indexed: 09/06/2024]
Abstract
T cells play a critical and irreplaceable role in maintaining overall health. However, their functions undergo alterations as individuals age. It is of utmost importance to comprehend the specific characteristics of T-cell aging, as this knowledge is crucial for gaining deeper insights into the pathogenesis of aging-related diseases and developing effective therapeutic strategies. In this review, we have thoroughly examined the existing studies on the characteristics of immune organ aging. Furthermore, we elucidated the changes and potential mechanisms that occur in T cells during the aging process. Additionally, we have discussed the latest research advancements pertaining to T-cell aging-related diseases. These findings provide a fresh perspective for the study of T cells in the context of aging.
Collapse
Affiliation(s)
- Yudai Xu
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zijian Wang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shumin Li
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jun Su
- First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Junwen Ou
- Anti Aging Medical Center, Clifford Hospital, Guangzhou, 511495, China
| | - Zhanyi Lin
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Chanchan Xiao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China.
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), Jinan University, Dongguan, 523000, China.
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai, 519070, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China.
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), Jinan University, Dongguan, 523000, China.
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai, 519070, China.
| |
Collapse
|
4
|
Habibi A, Letafatkar N, Sattari N, Nobakht S, Rafat Z, Soltani Moghadam S, Mirdamadi A, Javid M, Jamilian P, Hassanipour S, Keivanlou MH, Amini-Salehi E. Modulation of inflammatory markers in type 2 diabetes mellitus through gut microbiome-targeted interventions: An umbrella review on meta-analyses. Clin Nutr ESPEN 2025; 65:93-104. [PMID: 39551350 DOI: 10.1016/j.clnesp.2024.11.011] [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: 05/07/2024] [Revised: 09/23/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND & AIMS Type 2 diabetes mellitus (T2DM) poses a significant global health challenge due to various lifestyle factors contributing to its prevalence and associated complications. Chronic low-grade inflammation, characterized by elevated levels of inflammatory markers such as C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α), plays a pivotal role in the pathogenesis of T2DM. Modulation of the gut microbiota through microbiome-targeted therapy (MTT), including probiotics, prebiotics, and synbiotics, has emerged as a potential strategy to mitigate inflammation and improve metabolic outcomes in T2DM. METHODS A systematic review and meta-analysis were conducted following PRISMA guidelines to evaluate the impact of MTT on inflammatory markers in patients with T2DM. Searches were performed in PubMed, Scopus, and Web of Science databases up to June 2024, with inclusion criteria limited to English-language meta-analyses of randomized controlled trials (RCTs) assessing the effects of probiotics, prebiotics, or synbiotics on inflammatory markers in T2DM patients. RESULTS Ten meta-analyses met the inclusion criteria, comprising studies investigating the effects of various MTT interventions on CRP, IL-6, and TNF-α levels in T2DM patients. Meta-analysis results indicated significant reductions in CRP (SMD: -0.070; 95 % CI: -0.119 to -0.020) and TNF-α (SMD: -0.370; 95 % CI: -0.554 to -0.186) levels following MTT, while IL-6 reductions (SMD: -0.070; 95 % CI: -0.269 to 0.129) did not reach statistical significance. However, heterogeneity in study quality, intervention protocols, and participant demographics posed challenges in interpretation. CONCLUSIONS While improvements in inflammatory markers with MTT have been observed, significant limitations-such as heterogeneity in study quality and variation in intervention protocols-highlight the need for further research to confirm its efficacy and clarify underlying mechanisms. Future studies should aim to address these limitations by exploring variations in dosage, supplement formulations, and bacterial strains, which are crucial for improving the reliability and broader applicability of MTT in the management of T2DM.
Collapse
Affiliation(s)
- Arman Habibi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Negin Letafatkar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nazila Sattari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sara Nobakht
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Rafat
- Department of Medical Parasitology and Mycology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Arian Mirdamadi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mona Javid
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Mohammad-Hossein Keivanlou
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
5
|
Elfaki I, Mir R, Elnageeb ME, Hamadi A, Alharbi ZM, Bedaiwi RI, Javid J, Alrasheed T, Alatawi D, Alrohaf BM, Abunab MK, Muqri TA. Identification of Interactive Genetic Loci Linked to Insulin Resistance in Metabolic Syndrome-An Update. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:83. [PMID: 39859066 PMCID: PMC11767019 DOI: 10.3390/medicina61010083] [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: 11/26/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025]
Abstract
Metabolic syndrome is a metabolic disorder characterized by hypertension, dyslipidemia, impaired glucose tolerance, and abdominal obesity. Impaired insulin action or insulin resistance initiates metabolic syndrome. The prevalence of insulin resistance is increasing all over the world. Insulin resistance results in the defective metabolism of carbohydrates and lipids, in addition to low-grade chronic inflammation. Insulin resistance is associated with metabolic syndrome, which is a risk factor for a number of pathological conditions, such as Type 2 diabetes (T2D), cardiovascular disease (CVD), nonalcoholic fatty liver disease (NAFLD), and polycystic ovarian syndrome (PCOS). Genome-wide association studies have increased our understanding of many loci linked to these diseases and others. In this review, we discuss insulin resistance and its contribution to metabolic syndrome and these diseases. We also discuss the genetic loci associated with them. Genetic testing is invaluable in the identification and stratification of susceptible populations and/or individuals. After susceptible individuals and/or populations have been identified via genetic testing or screening, lifestyle modifications such as regular exercise, weight loss, a healthy diet, and smoking cessation can reduce or prevent metabolic syndrome and its associated pathologies.
Collapse
Affiliation(s)
- Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Rashid Mir
- Prince Fahd Bin Sultan Research Chair for Biomedical Research, Department of MLT, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (A.H.); (Z.M.A.); (R.I.B.); (J.J.)
| | - Mohamed E. Elnageeb
- Department of Basic Medical Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Abdullah Hamadi
- Prince Fahd Bin Sultan Research Chair for Biomedical Research, Department of MLT, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (A.H.); (Z.M.A.); (R.I.B.); (J.J.)
| | - Zeyad M. Alharbi
- Prince Fahd Bin Sultan Research Chair for Biomedical Research, Department of MLT, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (A.H.); (Z.M.A.); (R.I.B.); (J.J.)
| | - Ruqaiah I. Bedaiwi
- Prince Fahd Bin Sultan Research Chair for Biomedical Research, Department of MLT, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (A.H.); (Z.M.A.); (R.I.B.); (J.J.)
| | - Jamsheed Javid
- Prince Fahd Bin Sultan Research Chair for Biomedical Research, Department of MLT, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (A.H.); (Z.M.A.); (R.I.B.); (J.J.)
| | - Tariq Alrasheed
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Dalal Alatawi
- Haematology Laboratory, King Fahd Special Hospital, Ministry of Health, Tabuk 71491, Saudi Arabia;
| | | | | | - Turki Ahmed Muqri
- Asir Health Cluster, Tarj General Hospital, Bisha 67721, Saudi Arabia;
| |
Collapse
|
6
|
Velloso LA, Donato J. Growth Hormone, Hypothalamic Inflammation, and Aging. J Obes Metab Syndr 2024; 33:302-313. [PMID: 39639711 PMCID: PMC11704225 DOI: 10.7570/jomes24032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 09/26/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
While inflammation is a crucial response in injury repair and tissue regeneration, chronic inflammation is a prevalent feature in various chronic, non-communicable diseases such as obesity, diabetes, and cancer and in cardiovascular and neurodegenerative diseases. Long-term inflammation considerably affects disease prevalence, quality of life, and longevity. Our research indicates that the growth hormone/insulin-like growth factor 1 (GH/IGF-1) axis is a pivotal regulator of inflammation in some tissues, including the hypothalamus, which is a key player in systemic metabolism regulation. Moreover, the GH/IGF-1 axis is strongly linked to longevity, as GH- or GH receptor-deficient mice live approximately twice as long as wild-type animals and exhibit protection against aging-induced inflammation. Conversely, GH excess leads to increased neuroinflammation and reduced longevity. Our review studies the associations between the GH/IGF-1 axis, inflammation, and aging, with a particular focus on evidence suggesting that GH receptor signaling directly induces hypothalamic inflammation. This finding underscores the significant impact of changes in the GH axis on metabolism and on the predisposition to chronic, non-communicable diseases.
Collapse
Affiliation(s)
- Licio A. Velloso
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, Campinas, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
7
|
Zhang Y, Fu L. Evaluating the causal effects between Grave's disease and diabetes mellitus: a bidirectional Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1420499. [PMID: 39568808 PMCID: PMC11576183 DOI: 10.3389/fendo.2024.1420499] [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: 04/20/2024] [Accepted: 10/01/2024] [Indexed: 11/22/2024] Open
Abstract
Background Graves' disease (GD) is an autoimmune disease associated with an increased incidence of other autoimmune diseases. To investigate the causality between GD and Diabetes mellitus (DM), we designed bidirectional two-sample Mendelian randomization (MR) and multivariable MR (MVMR) studies. Methods Single-nucleotide polymorphisms (SNPs) associated with GD, thyroid peroxidase (TPO), thyroglobulin (Tg), thyroid-stimulating hormone (TSH), type 1 diabetes (T1D), and type 2 diabetes (T2D) were obtained from the IEU Open GWAS and FinnGen biobank databases. For the forward MR study, we used GD (sample size = 458,620) as the exposure and T1D (sample size = 520,580) and T2D (sample size = 211,766) as the outcomes. Next, high risk of T1D and T2D were used as exposure variables, and GD was used as the outcome variable for the reverse MR analysis. Finally, MVMR analysis was conducted to investigate the probable relationship between DM and indicators for thyroid function like TPO, Tg, and TSH. The inverse variance weighting (IVW) was used as the main method. Finally, the heterogeneity and sensitivity were assessed. Results There were 27, 88, and 55 SNPs associated with GD, T1D, and T2D, respectively. A significant causal connection between higher genetic liability of GD and the risk of T2D (OR [95% CI] = 1.059 [1.025-1.095], P = 5.53e-04) was found in the forward MR analysis. Comparatively, the significant causal relationship between higher genetic liability of GD and the risk of T1D was not demonstrated (OR [95% CI] = 0.998[0.927,1.074], P=0.949). However, reverse MR suggested that there was a genetic susceptibility to T1D that increased the likelihood of developing GD (OR [95% CI] = 1.173[1.117,1.231], P = 1.913e-10), while T2D did not (OR [95% CI] = 0.963 [0.870-1.066], P = 0.468). Furthermore, there was inadequate evidence to suggest that abnormal TSH, TPO, and Tg levels increase the risk of incident T1D or T2D in individuals with GD. MVMR revealed no causal relationship among Tg, TSH, TPO, T1D, or T2D. Conclusion There was no increased risk of T1D with an increase in genetic susceptibility to GD, although higher genetic susceptibility to T1D has been shown to be associated with increased risk of developing GD. A unidirectional causal relationship between the genetic liability for GD and increased risk of T2D was observed using MR analyses. MVMR analysis showed no statistically relevant causality between the genetic liability for TSH, TPO, or Tg and the risk of either T1D or T2D.
Collapse
Affiliation(s)
- Yuhan Zhang
- Emergency Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, China
- General Surgery Center, Department of Thyroid Surgery, The 1st Hospital of Jilin University, Chang Chun, China
- Department of General Surgery, Panzhihua Central Hospital, Panzhihua, China
| | - Liuxiang Fu
- Emergency Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, China
| |
Collapse
|
8
|
Shi W, Chen D, Chen W, Chen Y, Fu X, Xiao Y, Duan W, Zhang J. Relationship of thyroid parameters with chronic inflammation in patients with euthyroid type 2 diabetes. Front Endocrinol (Lausanne) 2024; 15:1433782. [PMID: 39525855 PMCID: PMC11543494 DOI: 10.3389/fendo.2024.1433782] [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: 05/16/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Aims We evaluated the association of multiple thyroid parameters with the levels interleukin 6 (IL6) and interleukin 8 (IL8) in patients with euthyroid type 2 diabetes (T2D). Methods A total of 166 adults with euthyroid T2D were examined. Serum IL6, IL8, triiodothyronine (FT3), free thyroxin (FT4), thyroid-stimulating hormone (TSH), five thyroid sensitivity indexes (FT3/FT4, TSH index [TSHI], thyrotroph T4 resistance index [TT4RI], thyroid feedback quantile-based index by FT3 [TFQIFT3], and TFQIFT4), and biochemical parameters were determined. Results The median age was 64 years (IQR: 54.75,70) and the median duration of diabetes was 10 years (IQR: 3,18). Patients with high levels of IL6 (≥2.38 pg/mL) had lower levels of FT3 and TFQIFT3 (P<0.05). Patients with high levels of IL8 (≥18.1 pg/mL) had lower levels of TSH and higher levels of FT4 (P<0.05). IL6 was negatively correlated with FT3 (r=-0.359, P<0.001), TFQIFT3 (r=-0.273, P=0.009), and FT3/FT4 (r=-0.22, P=0.037). IL8 was negatively correlated with TSH (r=-0.256, P=0.01), TSHI (r=-0.226, P=0.033), and TT4RI (r=-0.244, P=0.021). Binary logistic regression analysis with multiple adjusted models showed that a high level of IL6 was negatively associated with FT3 (aOR: 0.529; 95%CI: 0.302, 0.926, P=0.026), and that a high level of IL8 was negatively associated with TSH (aOR: 0.343; 95%CI: 0.155, 0.759; P=0.008) and TT4RI (aOR: 0.398;95%CI: 0.191, 0.831; P=0.014). Conclusion Patients with euthyroid T2D who had lower levels of FT3 had significantly higher levels of IL6, and those with lower levels of TSH and TT4RI had significantly higher levels of IL8.
Collapse
Affiliation(s)
- Wei Shi
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Hubei University of Chinese Medicine, Wuhan, China
| | | | | | | | | | | | | | - Jing Zhang
- Department of Endocrinology, Hubei Integrated Traditional Chinese and Western Medicine Hospital, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
9
|
He M, Niu J, Cheng H, Guo C. Identification and validation of diagnostic genes associated with neutrophil extracellular traps of type 2 diabetes mellitus. Front Genet 2024; 15:1373807. [PMID: 39296548 PMCID: PMC11408200 DOI: 10.3389/fgene.2024.1373807] [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: 02/12/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024] Open
Abstract
Background Neutrophil extracellular traps (NETs) cause delayed wound closed up in type 2 diabetes mellitus (T2DM), but the specific regulatory mechanism of NETs-related genes (NETs-RGs) in T2DM is unclear. Methods We acquired GSE21321 and GSE15932 datasets from gene expression omnibus (GEO) database. First, differentially expressed genes (DEGs) between T2DM and control samples of GSE21321 dataset were sifted out by differential expression analysis. NETs scores were calculated for all samples in GSE21321 dataset, and key module genes associated with NETs scores were screened by constructing co-expression network. Then, DEGs and key module genes were intersected to yield intersection genes, and candidate genes were identified by constructing a protein protein interaction (PPI) network. Least absolute shrinkage and selection operator (LASSO) regression analysis was implemented on candidate genes to screen out diagnostic genes, and they were subjected to single sample gene set enrichment analysis (ssGSEA). Finally, immune characteristic analysis was carried out, and we constructed the gene-drug and transcription factor (TF)-miRNA-mRNA networks. Besides, we validated the expression of diagnostic genes by quantitative real-time polymerase chain reaction (qRT-PCR). Results In total, 23 candidate genes were gained by PPI analysis. The 5 diagnostic genes, namely, inter-trypsin inhibitor heavy chain 3 (ITIH3), fibroblast growth factor 1 (FGF1), neuron cell adhesion molecule (NRCAM), advanced glycosylation end-product-specific receptor (AGER), and calcium voltage-gated channel subunit alpha1 C (CACNA1C), were identified via LASSO analysis, and they were involved in carboxylic acid transport, axonogenesis, etc. M2 Macrophage, Monocyte, Natural killer (NK) cell, and Myeloid dendritic cells (DC) were remarkably different between T2DM and control samples. Diagnostic genes had the strongest and the most significant positive correlation with B cells. The gene-drug network included CACNA1C-Isradipine, CACNA1C-Benidipine and other relationship pairs. Totally 76 nodes and 44 edges constituted the TF-miRNA-mRNA network, including signal transducer and activator of transcription 1(STAT1) -hsa-miR-3170-AGER, CCCTC-binding factor (CTCF)-hsa-miR-455-5p-CACNA1C, etc. Moreover, qRT-PCR suggested that the expression trends of FGF1 and AGER were in keeping with the results of bioinformatic analysis. FGF1 and AGER were markedly regulated downwards in the T2DM group. Conclusion Through bioinformatic analysis, we identified NETs-related diagnostic genes (ITIH3, FGF1, NRCAM, AGER, CACNA1C) in T2DM, and explored their mechanism of action from different aspects, providing new ideas for the studies related to diagnosis and treatment of T2DM.
Collapse
Affiliation(s)
- Meifang He
- Endocrinoloy Department, Peking University First Hospital Taiyuan Hospital (Taiyuan Central Hospital), Taiyuan, China
| | - Jin Niu
- Endocrinoloy Department, Peking University First Hospital Taiyuan Hospital (Taiyuan Central Hospital), Taiyuan, China
| | - Haihua Cheng
- Endocrinoloy Department, Peking University First Hospital Taiyuan Hospital (Taiyuan Central Hospital), Taiyuan, China
| | - Chaoying Guo
- Endocrinoloy Department, Peking University First Hospital Taiyuan Hospital (Taiyuan Central Hospital), Taiyuan, China
| |
Collapse
|
10
|
Galli A, Moretti S, Dule N, Di Cairano ES, Castagna M, Marciani P, Battaglia C, Bertuzzi F, Fiorina P, Pastore I, La Rosa S, Davalli A, Folli F, Perego C. Hyperglycemia impairs EAAT2 glutamate transporter trafficking and glutamate clearance in islets of Langerhans: implications for type 2 diabetes pathogenesis and treatment. Am J Physiol Endocrinol Metab 2024; 327:E27-E41. [PMID: 38690938 PMCID: PMC11390119 DOI: 10.1152/ajpendo.00069.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Abstract
Pancreatic endocrine cells employ a sophisticated system of paracrine and autocrine signals to synchronize their activities, including glutamate, which controls hormone release and β-cell viability by acting on glutamate receptors expressed by endocrine cells. We here investigate whether alteration of the excitatory amino acid transporter 2 (EAAT2), the major glutamate clearance system in the islet, may occur in type 2 diabetes mellitus and contribute to β-cell dysfunction. Increased EAAT2 intracellular localization was evident in islets of Langerhans from T2DM subjects as compared with healthy control subjects, despite similar expression levels. Chronic treatment of islets from healthy donors with high-glucose concentrations led to the transporter internalization in vesicular compartments and reduced [H3]-d-glutamate uptake (65 ± 5% inhibition), phenocopying the findings in T2DM pancreatic sections. The transporter relocalization was associated with decreased Akt phosphorylation protein levels, suggesting an involvement of the phosphoinositide 3-kinase (PI3K)/Akt pathway in the process. In line with this, PI3K inhibition by a 100-µM LY294002 treatment in human and clonal β-cells caused the transporter relocalization in intracellular compartments and significantly reduced the glutamate uptake compared to control conditions, suggesting that hyperglycemia changes the trafficking of the transporter to the plasma membrane. Upregulation of the glutamate transporter upon treatment with the antibiotic ceftriaxone rescued hyperglycemia-induced β-cells dysfunction and death. Our data underscore the significance of EAAT2 in regulating islet physiology and provide a rationale for potential therapeutic targeting of this transporter to preserve β-cell survival and function in diabetes.NEW & NOTEWORTHY The glutamate transporter SLC1A2/excitatory amino acid transporter 2 (EAAT2) is expressed on the plasma membrane of pancreatic β-cells and controls islet glutamate clearance and β-cells survival. We found that the EAAT2 membrane expression is lost in the islets of Langerhans from type 2 diabetes mellitus (T2DM) patients due to hyperglycemia-induced downregulation of the phosphoinositide 3-kinase/Akt pathway and modification of its intracellular trafficking. Pharmacological rescue of EAAT2 expression prevents β-cell dysfunction and death, suggesting EAAT2 as a new potential target of intervention in T2DM.
Collapse
Affiliation(s)
- Alessandra Galli
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Stefania Moretti
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nevia Dule
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Eliana Sara Di Cairano
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Michela Castagna
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paola Marciani
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Cristina Battaglia
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | | | - Paolo Fiorina
- Department of Biomedical and Clinical Sciences "L. Sacco,"Università degli Studi di Milano, Milan, Italy
- Endocrinology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Ida Pastore
- Endocrinology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Stefano La Rosa
- Unit of Pathology, Department of Oncology, ASST Sette Laghi, Varese, Italy
- Department of Medicine and Technological Innovation, Università degli Studi dell'Insubria, Varese, Italy
| | - Alberto Davalli
- Diabetes and Endocrinology Unit, Department of Internal Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Franco Folli
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Carla Perego
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
11
|
Lin Z, Zhang J, Duan T, Yang J, Yang Y. Trefoil factor 3 can stimulate Th17 cell response in the development of type 2 diabetes mellitus. Sci Rep 2024; 14:10340. [PMID: 38710764 PMCID: PMC11074263 DOI: 10.1038/s41598-024-60426-7] [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: 01/09/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024] Open
Abstract
This study aims to evaluate the role of trefoil factor 3 (TFF3) peptides in type 2 diabetes mellitus (T2DM) from an inflammatory perspective. The focus was on exploring how TFF3 affects the function of T cells. TFF3 overexpression model was constructed using lentivirus in Jurkat cell lines. We evaluated the impact of TFF3 on the proliferation, apoptosis, and IL-17A levels of Jurkat cells cultured in high glucose. The T2DM model was induced in TFF3 knockout (KO) mice through streptozotocin combined with high-fat diet. The measurements included glucose tolerance, insulin tolerance, inflammation markers, Th17 cell proportion, and pancreatic pathological changes. The T2DM modeling led to splenomegaly in mice, and increased expression of TFF3 in their spleens. Overexpression of TFF3 increased the proportion of IL-17+ T cells and the levels of Th17-related cytokines in Jurkat cells. There was no difference in body weight and blood glucose levels between wild-type and TFF3 KO mice. However, T2DM mice lacking the TFF3 gene showed improved glucose utilization, ameliorated pancreatic pathology, decreased inflammation levels, and reduced Th17 cell ratio. TFF3 may be involved in the chronic inflammatory immune response in T2DM. Its mechanism may be related to the regulation of the RORγt/IL-17 signaling pathway and its impact on T cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Ziyang Lin
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, The Institute of Chinese Medicinal Sciences, Science and Technology Building, Guangzhou Higher Education Mega Centre, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou, 510006, People's Republic of China
| | - Jinyuan Zhang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, The Institute of Chinese Medicinal Sciences, Science and Technology Building, Guangzhou Higher Education Mega Centre, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou, 510006, People's Republic of China
| | - Tingting Duan
- Guangdong Nephrotic Drug Engineering Technology Research Center, Institute of Consun Co. for Chinese Medicine in Kidney Diseases, Guangdong Consun Pharmaceutical Group, Guangzhou, People's Republic of China
| | - Junzheng Yang
- Guangdong Nephrotic Drug Engineering Technology Research Center, Institute of Consun Co. for Chinese Medicine in Kidney Diseases, Guangdong Consun Pharmaceutical Group, Guangzhou, People's Republic of China.
| | - Yiqi Yang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, The Institute of Chinese Medicinal Sciences, Science and Technology Building, Guangzhou Higher Education Mega Centre, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
12
|
Mahmoud M, Abdel-Rasheed M. Influence of type 2 diabetes and obesity on adipose mesenchymal stem/stromal cell immunoregulation. Cell Tissue Res 2023; 394:33-53. [PMID: 37462786 PMCID: PMC10558386 DOI: 10.1007/s00441-023-03801-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/21/2023] [Indexed: 10/07/2023]
Abstract
Type 2 diabetes (T2D), associated with obesity, represents a state of metabolic inflammation and oxidative stress leading to insulin resistance and progressive insulin deficiency. Adipose-derived stem cells (ASCs) are adult mesenchymal stem/stromal cells identified within the stromal vascular fraction of adipose tissue. These cells can regulate the immune system and possess anti-inflammatory properties. ASCs are a potential therapeutic modality for inflammatory diseases including T2D. Patient-derived (autologous) rather than allogeneic ASCs may be a relatively safer approach in clinical perspectives, to avoid occasional anti-donor immune responses. However, patient characteristics such as body mass index (BMI), inflammatory status, and disease duration and severity may limit the therapeutic utility of ASCs. The current review presents human ASC (hASC) immunoregulatory mechanisms with special emphasis on those related to T lymphocytes, hASC implications in T2D treatment, and the impact of T2D and obesity on hASC immunoregulatory potential. hASCs can modulate the proliferation, activation, and functions of diverse innate and adaptive immune cells via direct cell-to-cell contact and secretion of paracrine mediators and extracellular vesicles. Preclinical studies recommend the therapeutic potential of hASCs to improve inflammation and metabolic indices in a high-fat diet (HFD)-induced T2D disease model. Discordant data have been reported to unravel intact or detrimentally affected immunomodulatory functions of ASCs, isolated from patients with obesity and/or T2D patients, in vitro and in vivo. Numerous preconditioning strategies have been introduced to potentiate hASC immunomodulation; they are also discussed here as possible options to potentiate the immunoregulatory functions of hASCs isolated from patients with obesity and T2D.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt.
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
| | - Mazen Abdel-Rasheed
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt
- Department of Reproductive Health Research, National Research Centre, Cairo, Egypt
| |
Collapse
|
13
|
Nakandakari SCBR, Gaspar RC, Kuga GK, Ramos CDO, Vieira RF, Rios TDS, Muñoz VR, Sant'ana MR, Simabuco FM, da Silva ASR, Moura LP, Ropelle ER, Pauli JR, Cintra DE. Short-term flaxseed oil, rich in omega 3, protects mice against metabolic damage caused by high-fat diet, but not inflammation. J Nutr Biochem 2023; 114:109270. [PMID: 36706930 DOI: 10.1016/j.jnutbio.2023.109270] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 01/02/2023] [Accepted: 01/10/2023] [Indexed: 01/26/2023]
Abstract
It is known that long-term high-fat diet (HF) feeding drastically affects the adipose tissue, contributing to metabolic disorders. Recently, short-term HF consumption was shown to affect different neuronal signaling pathways. Thus, we aimed to evaluate the inflammatory effects of a short-term HF and whether a diet containing omega-3 fatty acid fats from flaxseed oil (FS) has protective effects. Mice were divided into three groups for 3 d, according to their diets: Control group (CT), HF, or FS for 3 d. Lipid profiles were assessed through mass spectrometry and inflammatory markers by RT-qPCR and Western blotting. After short-term HF, mice increased food intake, body weight, adiposity, and fasting glucose. Increased mRNA content of Ccl2 and Tnf was demonstrated in the HF compared to CT in mesenteric adipose tissue. In the liver, TNFα protein was higher in the HF group than in CT, followed by a decreased polyunsaturated fatty acids tissue incorporation in HF. On the other hand, the consumption of FS reduced food intake and fasting glucose, as well as increased omega-3 fatty acid incorporation in MAT and the liver. However, short-term FS was insufficient to control the early inflammation triggered by HF in MAT and the liver. These data demonstrated that a 3-d HF diet is enough to damage glucose homeostasis and trigger inflammation. In contrast, short-term FS protects against increased food intake and fasting glucose but not inflammation in mice.
Collapse
Affiliation(s)
- Susana Castelo Branco Ramos Nakandakari
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Nutrigenomics and Lipids Research Center (CELN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rafael Calais Gaspar
- Nutrigenomics and Lipids Research Center (CELN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Gabriel Keine Kuga
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Camila de Oliveira Ramos
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Renan Fudoli Vieira
- Nutrigenomics and Lipids Research Center (CELN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Thaiane da Silva Rios
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Nutrigenomics and Lipids Research Center (CELN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Vitor Rosetto Muñoz
- Nutrigenomics and Lipids Research Center (CELN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Marcella Ramos Sant'ana
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino Sanchez Ramos da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Leandro Pereira Moura
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Nutrigenomics and Lipids Research Center (CELN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil
| | - José Rodrigo Pauli
- Nutrigenomics and Lipids Research Center (CELN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Nutrigenomics and Lipids Research Center (CELN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
14
|
Chikoti S, Najiya U, Sumanlatha G, Jahan P. Cytokine gene variants of TNF-α and IL-10 in the propensity of type 2 diabetes in south Indian population. J Diabetes Complications 2022; 36:108304. [PMID: 36148706 DOI: 10.1016/j.jdiacomp.2022.108304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/14/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022]
Abstract
Chronic inflammation plays an important role in type 2 diabetes mellitus (T2DM), a common endocrinological pro-inflammatory disorder associated with insulin resistance. The objective of the present study is to see individual and combined effect of TNF-α (rs361525, rs1800629) and IL-10 (rs1800872, rs1800896) genes on T2DM susceptibility The genotyping was carried out in 200 T2DM patients and 200 healthy controls by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) using suitable primers. The results shown that TNF-α (GA of rs361525 & rs1800629) and IL-10 (AA of rs1800872 & GA of rs1800896) genes are significantly linked with T2DM development. The presence of AA-GA genotype combination for both TNF-α and IL-10 genes were elevating the risk of T2DM. Moreover, individuals bearing haplotypes AAAA, AACA and AAAG experience the increased risk of T2DM. Furthermore, gene-gene interaction analysis shown that TNF-α (GA of rs361525 & rs1800896) gene redundantly confer 3.4-fold elevated risk for T2DM. In gene-environment interaction, GA of TNF-α -1800896, W/H ratio and TG/HDL ratio were redundantly interacted each other and increase the risk of T2DM by 67-times. In conclusion, our results reveal that there is a significant association between foresaid TNF-α, IL-10 gene promoter polymorphisms and T2DM development. To the best of our knowledge this study is the first of its kind in the literature reporting the epistatic association of TNF-α (rs1800629G/A) gene with TG/HDL ratio and W/H ratio over IL-10 gene polymorphisms for T2DM susceptibility among south Indians.
Collapse
Affiliation(s)
- Swetha Chikoti
- Department of Genetics, Osmania University, Hyderabad, India
| | - Umme Najiya
- Department of Genetics, Osmania University, Hyderabad, India
| | | | - Parveen Jahan
- School of Sciences, Maulana Azad National Urdu University, Hyderabad, India.
| |
Collapse
|
15
|
Wang X, Sun X, Abulizi A, Xu J, He Y, Chen Q, Yan R. Effects of salvianolic acid A on intestinal microbiota and lipid metabolism disorders in Zucker diabetic fatty rats. Diabetol Metab Syndr 2022; 14:135. [PMID: 36127704 PMCID: PMC9490915 DOI: 10.1186/s13098-022-00868-z] [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: 02/18/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Salvianolic acid A (SalA) is the main water-soluble component isolated from Salvia miltiorrhiza. This study explored the influences of SalA on intestinal microbiota composition and lipid metabolism in Zucker diabetic fatty (ZDF) rats. The 6-week-old male ZDF rats were treated with distilled water (N = 10) and low dose (SalA 0.5 mg/kg/d, N = 10), medium dose (SalA 1 mg/kg/d, N = 10), and high dose (SalA 2 mg/kg/d, N = 10) of SalA, with the male Zucker lean normoglycemic rats of the same week age as controls (given distilled water, N = 10). The blood glucose, body weight, and food intake of rats were examined. After 7 and 8 weeks of continuous administration, oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) were performed, respectively. Serum fasting insulin (FINS), total cholesterol (TC), triglyceride (TG), and free fatty acid (FFA) were determined. Liver tissues were stained using hematoxylin-eosin (HE) and oil red O staining. Fecal samples were analyzed by 16S rRNA gene sequencing. Small intestinal tissues were stained using HE and immunohistochemistry. The tight junction proteins (ZO-1/Occludin/Claudin-1) and serum levels of LPS/TNF-α/IL-6 were evaluated. SalA reduced insulin resistance, liver injury, serum FFA, liver TC and TG levels in ZDF rats, and improved lipid metabolism. After SalA treatment, intestinal microbiota richness and diversity of ZDF rats were promoted. SalA retained the homeostasis of intestinal core microbiota. SalA reduced intestinal epithelial barrier damage, LPS, and inflammatory cytokines in ZDF rats. Overall, SalA can sustain intestinal microbiota balance and improve the lipid metabolism of ZDF rats.
Collapse
Affiliation(s)
- Xufeng Wang
- Department of Gastrointestinal Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, 856 Luoyu Rd, Hongshan District, Wuhan, 430061, Hubei, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, Hubei, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, Hubei, China
| | - Xiangjun Sun
- Department of Gastrointestinal Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, 856 Luoyu Rd, Hongshan District, Wuhan, 430061, Hubei, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, Hubei, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, Hubei, China
| | - Abulikemu Abulizi
- Department of Gastrointestinal Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, 856 Luoyu Rd, Hongshan District, Wuhan, 430061, Hubei, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, Hubei, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, Hubei, China
| | - Jinyao Xu
- Department of Gastrointestinal Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, 856 Luoyu Rd, Hongshan District, Wuhan, 430061, Hubei, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, Hubei, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, Hubei, China
| | - Yun He
- Department of Gastrointestinal Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, 856 Luoyu Rd, Hongshan District, Wuhan, 430061, Hubei, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, Hubei, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, Hubei, China
| | - Qian Chen
- Department of Gastrointestinal Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, 856 Luoyu Rd, Hongshan District, Wuhan, 430061, Hubei, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, Hubei, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, Hubei, China
| | - Ruicheng Yan
- Department of Gastrointestinal Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, 856 Luoyu Rd, Hongshan District, Wuhan, 430061, Hubei, China.
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, Hubei, China.
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, Hubei, China.
| |
Collapse
|
16
|
Li C, Gao Q, Jiang H, Liu C, Du Y, Li L. Changes of macrophage and CD4 + T cell in inflammatory response in type 1 diabetic mice. Sci Rep 2022; 12:14929. [PMID: 36056051 PMCID: PMC9440103 DOI: 10.1038/s41598-022-19031-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022] Open
Abstract
Immune cells play an important role in the development of inflammation in type 1 diabetes mellitus, so we want to explore the changes of CD4+ T cells and macrophages in vivo, which can provide an experimental basis for immunotherapy based on CD4+ T cells and macrophages. The intraperitoneal injection of streptozocin was used to induce a type 1 diabetes mellitus mouse model; the blood glucose, body weight, and the expression of inflammatory factors in the kidney were measured. Immunohistochemistry was applied to determine and analyze the infiltration of CD4+ T cells and macrophages in the spleen, pancreas, and kidney. The subtypes of macrophages in the kidney and CD4+ T cells in the spleen were analyzed by flow cytometry. Our study suggests that CD4+ T cells and macrophages increase, while the inflammatory immune response system is activated in the development of T1DM. CD4+ T cells positively correlated with macrophages in the pancreas and kidney of T1DM. CD4+ T cells turn to pro-inflammatory subtypes in the spleen of T1DM, while macrophages turn to pro-inflammatory subtypes in the kidney of T1DM. Therefore, regulation of CD4+ T cells and macrophages may be a potential target for T1DM and kidney complications.
Collapse
Affiliation(s)
- Chenhao Li
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Qingyuan Gao
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Hao Jiang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Chengrun Liu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
17
|
Hypoglycemic, Hypolipidemic, and Anti-Inflammatory Effects of Beta-Pinene in Diabetic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8173307. [PMID: 35620400 PMCID: PMC9129963 DOI: 10.1155/2022/8173307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 12/28/2022]
Abstract
Background Diabetes is a metabolic disease linked to multiple comorbidities, such as low-grade inflammation. β-pinene, a monoterpene commonly found in aromatic plants, is endowed with anti-inflammatory effect and this fact lead us to investigate the possible hypoglycemic, hypolipidemic and anti-inflammatory effects of the monoterpene in the alloxan-induced diabetes experimental model. Methods Male Wistar rats (200–250 g) were treated orally with β-pinene (25, 50, 100, and 200 mg/kg) or glibenclamide (5 mg/kg), for seven consecutive days. Diabetes was induced by alloxan (40 mg/kg) through the penile vein. On the seventh day of treatment, blood samples were collected for biochemical analysis. The anti-inflammatory effect of β-pinene was evaluated using the carrageenan-induced paw edema model, followed by the carrageenan-induced peritonitis. Results The treatment with β-pinene decreased plasma glucose, triglyceride, VLDL, LDL, and HDL levels, when compared to those of the control group. In addition, the association β-pinene 10 mg/kg + glibenclamide 2 mg/kg significantly decreased blood glucose, total cholesterol, and triglyceride level. Finally, oral treatment with β-pinene reduced carrageenan-induced paw edema and leukocyte migration in the peritoneum. Taken together, our results indicate that β-pinene shows hypoglycemic and hypolipemic effects, which may involve some common mechanisms of glibenclamide. Besides, the monoterpene presented an anti-inflammatory action in diabetic rats that needs further investigation in order to clarify such effect and its correlation with the alterations observed in plasma parameters of β-pinene-treated diabetic rats.
Collapse
|
18
|
[Two Case Reports of Type 2 Diabetes Induced by Immune Checkpoint Inhibitors
Combined with Chemotherapy]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:287-290. [PMID: 35477193 PMCID: PMC9051303 DOI: 10.3779/j.issn.1009-3419.2022.102.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have become an important means of cancer treatment, and their application in the clinic is becoming more and more widespread. The adverse reactions caused by ICIs are gradually recognized. Among them, immunotherapy-related diabetes is a rare adverse reaction and type 1 diabetes mellitusis common. With the wide application of ICIs combined with chemotherapy in lung cancer patients, patients with type 2 diabetes mellitus have gradually been discovered during the treatment. However, the effect of continued use of ICIs maintenance therapy on blood glucose and ICIs treatment process in these patients is still unclear. This article reports two cases of type 2 diabetes mellitus induced by immune checkpoint inhibitor combined with chemotherapy, one of whom converted to type 1 diabetes mellitus, in order to increase the understanding of immunotherapy-related diabetes.
.
Collapse
|
19
|
Ding LN, Ding WY, Ning J, Wang Y, Yan Y, Wang ZB. Effects of Probiotic Supplementation on Inflammatory Markers and Glucose Homeostasis in Adults With Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Front Pharmacol 2021; 12:770861. [PMID: 34955840 PMCID: PMC8706119 DOI: 10.3389/fphar.2021.770861] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Several studies have revealed the effect of probiotic supplementation in patients with type 2 diabetes (T2DM) on the amelioration of low-grade inflammation, which plays an important role in the pathogenesis of T2DM. However, the effects of the clinical application of probiotics on inflammation in individuals with T2DM remain inconsistent. This study aims to investigate the comprehensive effects of probiotics on inflammatory markers in adults with T2DM. Methods: PubMed, Embase, Cochrane Library, and the Web of Science were searched to identify randomized controlled trials (RCTs) exploring the effect of probiotic supplementation on inflammatory markers in individuals with T2DM through March 11, 2021. Two reviewers independently screened the literature, extracted data, and assessed the risk of bias of the included studies. We used a random-effects model to calculate the standardized mean difference (SMD) between the probiotic supplementation and control groups. Results: Seventeen eligible studies were selected with a total of 836 participants, including 423 participants in probiotic supplementation groups and 413 participants in control groups. Our study demonstrated that compared with the control condition, probiotic intake produced a beneficial effect in reducing the levels of plasma inflammation markers, including tumour necrosis factor-α (TNF-α) (SMD [95% CI]; −0.37 [−0.56, −0.19], p < 0.0001) and C-reactive protein (CRP) (SMD [95% CI]; −0.21 [−0.42, −0.01], p = 0.040), while it had no effect on the plasma interleukin-6 (IL-6) level (SMD [95% CI]; −0.07 [−0.27, 0.13], p = 0.520). In addition, our results support the notion that probiotic supplementation improves glycaemic control, as evidenced by a significant reduction in fasting blood glucose (FPG), HbA1c and HOMA-IR (SMD [95% CI]: −0.24 [−0.42, −0.05], p = 0.010; −0.19 [−0.37, −0.00], p = 0.040; −0.36 [−0.62, −0.10], p = 0.006, respectively). Conclusion: Our study revealed some beneficial effects of probiotic supplementation on improving inflammatory markers and glucose homeostasis in individuals with T2DM. Probiotics might be a potential adjuvant therapeutic approach for T2DM.
Collapse
Affiliation(s)
- Li-Na Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Shandong Institute of Endocrine and Metabolic Diseases, Jinan, China
| | - Wen-Yu Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Shandong Institute of Endocrine and Metabolic Diseases, Jinan, China
| | - Jie Ning
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Shandong Institute of Endocrine and Metabolic Diseases, Jinan, China
| | - Yao Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Shandong Institute of Endocrine and Metabolic Diseases, Jinan, China
| | - Yan Yan
- Department of Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Zhi-Bin Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Shandong Institute of Endocrine and Metabolic Diseases, Jinan, China
| |
Collapse
|
20
|
Afzal M, Al-Abbasi FA, Nadeem MS, Alshehri S, Ghoneim MM, Imam SS, Almalki WH, Kazmi I. Sodium-Glucose Cotransporter-2 Inhibitors Improve Cardiovascular Dysfunction in Type 2 Diabetic East Asians. Metabolites 2021; 11:794. [PMID: 34822452 PMCID: PMC8622829 DOI: 10.3390/metabo11110794] [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: 09/29/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 11/22/2022] Open
Abstract
In East Asians, the incidence of type 2 DM (T2DM) has increased as a result of major alterations in life. Cardiovascular problems are more likely in those with T2DM. Sodium-glucose cotransporter-2 (SGLT2) inhibitors are novel insulin-independent antihyperglycemic drugs that limit renal glucose reabsorption and thereby improve glycemic control. They are used alone or in combination with insulin and other antihyperglycemic medications to treat diabetes, and they are also helpful in protecting against the progression of complications. This review has evaluated the available evidence not only on the efficacy of SGLT2 inhibitors in T2DM, but also on their favourable cardiovascular events in East Asians. DM is an independent risk factor for cardiovascular diseases. As a result, in addition to glycemic control in diabetes management, the therapeutic goal in East Asian diabetic patients should be to improve adverse cardiovascular outcomes. Besides establishing antidiabetic effects, several studies have reported cardioprotective benefits of SGLT2 inhibitors via numerous pathways. SGLT2 inhibitors show promising antidiabetic drugs with potential cardiovascular advantages, given that a high number of diabetic patients in East Asia have co-existing cardiovascular disorders. Despite significant positive results in favour of SGLT2, more research is needed to determine how SGLT2 inhibitors exert these impressive cardiovascular effects.
Collapse
Affiliation(s)
- Muhammad Afzal
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia;
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.-A.); (M.S.N.)
| | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.-A.); (M.S.N.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Imran Kazmi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia;
| |
Collapse
|
21
|
Bülbül T, Baharlooie M, Safaeinejad Z, Gure AO, Ghaedi K. Hypothetical molecular interconnection between type 2 diabetes and dyslexia. BMC Neurosci 2021; 22:63. [PMID: 34674647 PMCID: PMC8529849 DOI: 10.1186/s12868-021-00666-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dyslexia is one of the most common learning disabilities, especially among children. Type 2 diabetes is a metabolic disorder that affects a large population globally, with metabolic disorders. There have been several genes that are identified as causes of Dyslexia, and in recent studies, it has been found out that some of those genes are also involved in several metabolic pathways. For several years, it has been known that type 2 diabetes causes several neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease. Furthermore, in several studies, it was suggested that type 2 diabetes also has some associations with learning disabilities. This raises the question of whether "Is there a connection between type 2 diabetes and dyslexia?". In this study, this question is elaborated by linking their developmental processes via bioinformatics analysis about these two diseases individually and collectively. RESULT The literature review for dyslexia and type two diabetes was completed. As the result of this literature review, the genes that are associated to type 2 diabetes and dyslexia were identified. The biological pathways of dyslexia, and dyslexia associated genes, type 2 diabetes, and type 2 diabetes associated genes were identified. The association of these genes, regarding to their association with pathways were analysed, and using STRING database the gene associations were analysed and identified. CONCLUSION The findings of this research included the interaction analysis via gene association, co-expression and protein-protein interaction. These findings clarified the interconnection between dyslexia and type 2 diabetes in molecular level and it will be the beginning of an answer regarding to the relationship between T2D and dyslexia. Finally, by improving the understanding this paper aims to open the way for the possible future approach to examine this hypothesis.
Collapse
Affiliation(s)
- Tuğba Bülbül
- Department of Biomedical Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, West Midlands, UK
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
| | - Maryam Baharlooie
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Avenue, Azadi Sq., P.O.Code 81746-73441, Isfahan, Iran
| | - Zahra Safaeinejad
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Ali Osmay Gure
- Department of Medical Biology, Acibadem University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Avenue, Azadi Sq., P.O.Code 81746-73441, Isfahan, Iran.
| |
Collapse
|
22
|
Insulin Signal Transduction Perturbations in Insulin Resistance. Int J Mol Sci 2021; 22:ijms22168590. [PMID: 34445300 PMCID: PMC8395322 DOI: 10.3390/ijms22168590] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus is a widespread medical condition, characterized by high blood glucose and inadequate insulin action, which leads to insulin resistance. Insulin resistance in insulin-responsive tissues precedes the onset of pancreatic β-cell dysfunction. Multiple molecular and pathophysiological mechanisms are involved in insulin resistance. Insulin resistance is a consequence of a complex combination of metabolic disorders, lipotoxicity, glucotoxicity, and inflammation. There is ample evidence linking different mechanistic approaches as the cause of insulin resistance, but no central mechanism is yet described as an underlying reason behind this condition. This review combines and interlinks the defects in the insulin signal transduction pathway of the insulin resistance state with special emphasis on the AGE-RAGE-NF-κB axis. Here, we describe important factors that play a crucial role in the pathogenesis of insulin resistance to provide directionality for the events. The interplay of inflammation and oxidative stress that leads to β-cell decline through the IAPP-RAGE induced β-cell toxicity is also addressed. Overall, by generating a comprehensive overview of the plethora of mechanisms involved in insulin resistance, we focus on the establishment of unifying mechanisms to provide new insights for the future interventions of type 2 diabetes mellitus.
Collapse
|
23
|
Dilworth L, Facey A, Omoruyi F. Diabetes Mellitus and Its Metabolic Complications: The Role of Adipose Tissues. Int J Mol Sci 2021; 22:ijms22147644. [PMID: 34299261 PMCID: PMC8305176 DOI: 10.3390/ijms22147644] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Many approaches have been used in the effective management of type 2 diabetes mellitus. A recent paradigm shift has focused on the role of adipose tissues in the development and treatment of the disease. Brown adipose tissues (BAT) and white adipose tissues (WAT) are the two main types of adipose tissues with beige subsets more recently identified. They play key roles in communication and insulin sensitivity. However, WAT has been shown to contribute significantly to endocrine function. WAT produces hormones and cytokines, collectively called adipocytokines, such as leptin and adiponectin. These adipocytokines have been proven to vary in conditions, such as metabolic dysfunction, type 2 diabetes, or inflammation. The regulation of fat storage, energy metabolism, satiety, and insulin release are all features of adipose tissues. As such, they are indicators that may provide insights on the development of metabolic dysfunction or type 2 diabetes and can be considered routes for therapeutic considerations. The essential roles of adipocytokines vis-a-vis satiety, appetite, regulation of fat storage and energy, glucose tolerance, and insulin release, solidifies adipose tissue role in the development and pathogenesis of diabetes mellitus and the complications associated with the disease.
Collapse
Affiliation(s)
- Lowell Dilworth
- Department of Pathology, Mona Campus, University of the West Indies, Kingston 7, Jamaica;
| | - Aldeam Facey
- Mona Academy of Sport, Mona Campus, University of the West Indies, Kingston 7, Jamaica;
| | - Felix Omoruyi
- Department of Life Sciences, Texas A&M University, Corpus Christi, TX 78412, USA
- Correspondence:
| |
Collapse
|
24
|
Sazonova MA, Ryzhkova AI, Sinyov VV, Sazonova MD, Kirichenko TV, Doroschuk NA, Karagodin VP, Orekhov AN, Sobenin IA. Mutations of mtDNA in some Vascular and Metabolic Diseases. Curr Pharm Des 2021; 27:177-184. [PMID: 32867647 DOI: 10.2174/1381612826999200820162154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The present review article considers some chronic diseases of vascular and metabolic genesis, the causes of which may be mitochondrial dysfunction. Very often, in the long course of the disease, complications may occur, leading to myocardial infarction or ischemic stroke and, as a result, death. In particular, a large percentage of human deaths nowadays belongs to cardiovascular diseases, such as coronary heart disease (CHD), arterial hypertension, cardiomyopathies, and type 2 diabetes mellitus. OBJECTIVE The aim of the present review was the analysis of literature sources, devoted to an investigation of a link of mitochondrial DNA mutations with chronic diseases of vascular and metabolic genesis. RESULTS The analysis of literature indicates the association of the mitochondrial genome mutations with coronary heart disease, type 2 diabetes mellitus, hypertension, and various types of cardiomyopathies. CONCLUSION The detected mutations can be used to analyze the predisposition to chronic diseases of vascular and metabolic genesis. They can also be used to create molecular-cell models necessary to evaluate the effectiveness of drugs developed for the treatment of these pathologies. MtDNA mutations associated with the absence of diseases of vascular and metabolic genesis could be potential candidates for gene therapy of the said diseases.
Collapse
Affiliation(s)
- Margarita A Sazonova
- Laboratory of angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Anastasia I Ryzhkova
- Laboratory of angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Vasily V Sinyov
- Laboratory of angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Marina D Sazonova
- Laboratory of angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Tatiana V Kirichenko
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, Moscow, Russian Federation
| | - Natalya A Doroschuk
- Laboratory of angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Vasily P Karagodin
- Laboratory of angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Alexander N Orekhov
- Laboratory of angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Igor A Sobenin
- Laboratory of angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, 0
| |
Collapse
|
25
|
Sen ZD, Danyeli LV, Woelfer M, Lamers F, Wagner G, Sobanski T, Walter M. Linking atypical depression and insulin resistance-related disorders via low-grade chronic inflammation: Integrating the phenotypic, molecular and neuroanatomical dimensions. Brain Behav Immun 2021; 93:335-352. [PMID: 33359233 DOI: 10.1016/j.bbi.2020.12.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Insulin resistance (IR) and related disorders, such as T2DM, increase the risk of major depressive disorder (MDD) and vice versa. Current evidence indicates that psychological stress and overeating can induce chronic low-grade inflammation that can interfere with glutamate metabolism in MDD as well as insulin signaling, particularly in the atypical subtype. Here we first review the interactive role of inflammatory processes in the development of MDD, IR and related metabolic disorders. Next, we describe the role of the anterior cingulate cortex in the pathophysiology of MDD and IR-related disorders. Furthermore, we outline how specific clinical features of atypical depression, such as hyperphagia, are more associated with inflammation and IR-related disorders. Finally, we examine the regional specificity of the effects of inflammation on the brain that show an overlap with the functional and morphometric brain patterns activated in MDD and IR-related disorders.
Collapse
Affiliation(s)
- Zümrüt Duygu Sen
- Department of Psychiatry and Psychotherapy, University Tuebingen, Calwerstraße 14, 72076 Tuebingen, Germany; Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany
| | - Lena Vera Danyeli
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Leipziger Str. 44, Building 65, 39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Marie Woelfer
- Clinical Affective Neuroimaging Laboratory (CANLAB), Leipziger Str. 44, Building 65, 39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Femke Lamers
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Oldenaller 1, 1081 HJ Amsterdam, the Netherlands
| | - Gerd Wagner
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany
| | - Thomas Sobanski
- Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, Thueringen-Kliniken "Georgius Agricola" GmbH, Rainweg 68, 07318 Saalfeld, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, University Tuebingen, Calwerstraße 14, 72076 Tuebingen, Germany; Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Leipziger Str. 44, Building 65, 39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany.
| |
Collapse
|
26
|
Sadras T, Chan LN, Xiao G, Müschen M. Metabolic Gatekeepers of Pathological B Cell Activation. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 16:323-349. [DOI: 10.1146/annurev-pathol-061020-050135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Unlike other cell types, B cells undergo multiple rounds of V(D)J recombination and hypermutation to evolve high-affinity antibodies. Reflecting high frequencies of DNA double-strand breaks, adaptive immune protection by B cells comes with an increased risk of malignant transformation. In addition, the vast majority of newly generated B cells express an autoreactive B cell receptor (BCR). Thus, B cells are under intense selective pressure to remove autoreactive and premalignant clones. Despite stringent negative selection, B cells frequently give rise to autoimmune disease and B cell malignancies. In this review, we discuss mechanisms that we term metabolic gatekeepers to eliminate pathogenic B cell clones on the basis of energy depletion. Chronic activation signals from autoreactive BCRs or transforming oncogenes increase energy demands in autoreactive and premalignant B cells. Thus, metabolic gatekeepers limit energy supply to levels that are insufficient to fuel either a transforming oncogene or hyperactive signaling from an autoreactive BCR.
Collapse
Affiliation(s)
- Teresa Sadras
- Center of Molecular and Cellular Oncology, Yale Cancer Center, and Department of Immunobiology, Yale University, New Haven, Connecticut 06520, USA
| | - Lai N. Chan
- Center of Molecular and Cellular Oncology, Yale Cancer Center, and Department of Immunobiology, Yale University, New Haven, Connecticut 06520, USA
| | - Gang Xiao
- Current affiliation: Department of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Markus Müschen
- Center of Molecular and Cellular Oncology, Yale Cancer Center, and Department of Immunobiology, Yale University, New Haven, Connecticut 06520, USA
| |
Collapse
|
27
|
Longitudinal plasma protein profiling of newly diagnosed type 2 diabetes. EBioMedicine 2020; 63:103147. [PMID: 33279861 PMCID: PMC7718461 DOI: 10.1016/j.ebiom.2020.103147] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/05/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Comprehensive proteomics profiling may offer new insights into the dysregulated metabolic milieu of type 2 diabetes, and in the future, serve as a useful tool for personalized medicine. This calls for a better understanding of circulating protein patterns at the early stage of type 2 diabetes as well as the dynamics of protein patterns during changes in metabolic status. METHODS To elucidate the systemic alterations in early-stage diabetes and to investigate the effects on the proteome during metabolic improvement, we measured 974 circulating proteins in 52 newly diagnosed, treatment-naïve type 2 diabetes subjects at baseline and after 1 and 3 months of guideline-based diabetes treatment, while comparing their protein profiles to that of 94 subjects without diabetes. FINDINGS Early stage type 2 diabetes was associated with distinct protein patterns, reflecting key metabolic syndrome features including insulin resistance, adiposity, hyperglycemia and liver steatosis. The protein profiles at baseline were attenuated during guideline-based diabetes treatment and several plasma proteins associated with metformin medication independently of metabolic variables, such as circulating EPCAM. INTERPRETATION The results advance our knowledge about the biochemical manifestations of type 2 diabetes and suggest that comprehensive protein profiling may serve as a useful tool for metabolic phenotyping and for elucidating the biological effects of diabetes treatments. FUNDING This work was supported by the Swedish Heart and Lung Foundation, the Swedish Research Council, the Erling Persson Foundation, the Knut and Alice Wallenberg Foundation, and the Swedish state under the agreement between the Swedish government and the county councils (ALF-agreement).
Collapse
|
28
|
Sankar SB, Infante-Garcia C, Weinstock LD, Ramos-Rodriguez JJ, Hierro-Bujalance C, Fernandez-Ponce C, Wood LB, Garcia-Alloza M. Amyloid beta and diabetic pathology cooperatively stimulate cytokine expression in an Alzheimer's mouse model. J Neuroinflammation 2020; 17:38. [PMID: 31992349 PMCID: PMC6988295 DOI: 10.1186/s12974-020-1707-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/08/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Diabetes is a risk factor for developing Alzheimer's disease (AD); however, the mechanism by which diabetes can promote AD pathology remains unknown. Diabetes results in diverse molecular changes in the brain, including dysregulation of glucose metabolism and loss of cerebrovascular homeostasis. Although these changes have been associated with increased Aβ pathology and increased expression of glial activation markers in APPswe/PS1dE9 (APP/PS1) mice, there has been limited characterization, to date, of the neuroinflammatory changes associated with diabetic conditions. METHODS To more fully elucidate neuroinflammatory changes associated with diabetes that may drive AD pathology, we combined the APP/PS1 mouse model with either high-fat diet (HFD, a model of pre-diabetes), the genetic db/db model of type 2 diabetes, or the streptozotocin (STZ) model of type 1 diabetes. We then used a multiplexed immunoassay to quantify cortical changes in cytokine proteins. RESULTS Our analysis revealed that pathology associated with either db/db, HFD, or STZ models yielded upregulation of a broad profile of cytokines, including chemokines (e.g., MIP-1α, MIP-1β, and MCP-1) and pro-inflammatory cytokines, including IL-1α, IFN-γ, and IL-3. Moreover, multivariate partial least squares regression analysis showed that combined diabetic-APP/PS1 models yielded cooperatively enhanced expression of the cytokine profile associated with each diabetic model alone. Finally, in APP/PS1xdb/db mice, we found that circulating levels of Aβ1-40, Aβ1-42, glucose, and insulin all correlated with cytokine expression in the brain, suggesting a strong relationship between peripheral changes and brain pathology. CONCLUSIONS Altogether, our multiplexed analysis of cytokines shows that Alzheimer's and diabetic pathologies cooperate to enhance profiles of cytokines reported to be involved in both diseases. Moreover, since many of the identified cytokines promote neuronal injury, Aβ and tau pathology, and breakdown of the blood-brain barrier, our data suggest that neuroinflammation may mediate the effects of diabetes on AD pathogenesis. Therefore, strategies targeting neuroinflammatory signaling, as well as metabolic control, may provide a promising strategy for intervening in the development of diabetes-associated AD.
Collapse
Affiliation(s)
- Sitara B Sankar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Carmen Infante-Garcia
- Division of Physiology, School of Medicine, Universidad de Cadiz, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Cadiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Cádiz, Spain
| | - Laura D Weinstock
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Juan Jose Ramos-Rodriguez
- Division of Physiology, School of Medicine, Universidad de Cadiz, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Cadiz, Spain
- Departamento de Fisiología, Facultad de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Carmen Hierro-Bujalance
- Division of Physiology, School of Medicine, Universidad de Cadiz, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Cadiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Cádiz, Spain
| | - Cecilia Fernandez-Ponce
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Cádiz, Spain
- Área de Inmunología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
| | - Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Dr, Rm 3303, Atlanta, GA, 30332, USA.
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Cadiz, Spain.
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Cádiz, Spain.
| |
Collapse
|
29
|
Th17 and regulatory T cells in patients with different time of progression of type 2 diabetes mellitus. Cent Eur J Immunol 2020; 45:29-36. [PMID: 32425677 PMCID: PMC7226552 DOI: 10.5114/ceji.2020.94670] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/26/2018] [Indexed: 12/15/2022] Open
Abstract
Introduction Type 2 diabetes mellitus (T2DM) is characterized by chronic inflammation, in which different types of immune cells participate, such as TH17 cells and Treg cells. The aim of this study was to determine the relationship between Treg and Th17 in patients with different times of T2DM progression. Material and methods Nineteen control subjects and 40 patients with T2DM were included. T2DM patients were classified into two groups: the first group consisted of twenty patients with less than10 years of disease progression (T2DM < 10), and the second group included 20 patients with a disease progression of 10 years or more (T2DM ≥ 10). Additionally, an analysis was performed according to the metabolic control, depending on HbA1c levels. The peripheral blood ratio of both Th17 and Treg cells was measured by standard flow cytometry protocols. Results No significant difference was found in Th17 cells of patients with T2DM < 10 or T2DM ≥ 10 and controls. With respect to CD4+CD25+FoxP3+ and CD4+CD25h Treg cells, a significant decrease was observed in patients with T2DM ≥ 10, mainly in patients with poor or moderate metabolic control. Statistical analysis performed in all patients with T2DM revealed a decrease in three cell subsets as well a negative correlation between Th17 cells and total cholesterol, CD4+CD25h cells with glucose and HbA1c levels, while a positive correlation was observed between CD4+CD25h cells and BMI. Conclusions A decrease on both Treg and Th17 cell subsets in T2DM patients was observed suggesting that the metabolic decontrol and the progression time of T2DM could modify the proportions of Th17 and Treg cells.
Collapse
|
30
|
Chávez-Castillo M, Nava M, Ortega Á, Rojas M, Núñez V, Salazar J, Bermúdez V, Rojas-Quintero J. Depression as an Immunometabolic Disorder: Exploring Shared Pharmacotherapeutics with Cardiovascular Disease. Curr Neuropharmacol 2020; 18:1138-1153. [PMID: 32282306 PMCID: PMC7709154 DOI: 10.2174/1570159x18666200413144401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/04/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Modern times have seen depression and cardiovascular disease (CVD) become notorious public health concerns, corresponding to alarming proportions of morbidity, mortality, decreased quality of life, and economic costs. Expanding comprehension of the pathogenesis of depression as an immunometabolic disorder has identified numerous pathophysiologic phenomena in common with CVD, including chronic inflammation, insulin resistance, and oxidative stress. These shared components could be exploited to offer improved alternatives in the joint management of these conditions. Abundant preclinical and clinical data on the impact of established treatments for CVD in the management of depression have allowed for potential candidates to be proposed for the joint management of depression and CVD as immunometabolic disorders. However, a large proportion of the clinical investigation currently available exhibits marked methodological flaws which preclude the formulation of concrete recommendations in many cases. This situation may be a reflection of pervasive problems present in clinical research in psychiatry, especially pertaining to study homogeneity. Therefore, further high-quality research is essential in the future in this regard.
Collapse
Affiliation(s)
| | | | | | | | | | - Juan Salazar
- Address correspondence to this author at the Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 20th Avenue 4004, Venezuela; Tel/Fax: ++582617597279; E-mail:
| | | | | |
Collapse
|
31
|
Guest PC. The Impact of New Biomarkers and Drug Targets on Age-Related Disorders. Methods Mol Biol 2020; 2138:3-28. [PMID: 32219738 DOI: 10.1007/978-1-0716-0471-7_1] [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: 06/10/2023]
Abstract
The increase in the human lifespan has not been paralleled by an increase in healthy life. With the increase in the proportion of the aged population, there has been a natural increase in the prevalence of age-related disorders, such as Alzheimer's disease, type 2 diabetes mellitus, frailty, and various other disorders. A continuous rise in these conditions could lead to a widespread medical and social burden. There are now considerable efforts underway to address these deficits in preclinical and clinical studies, which include the use of better study cohorts, longitudinal designs, improved translation of data from preclinical models, multi-omics profiling, identification of new biomarker candidates and refinement of computational tools and databases containing relevant information. Such efforts will support future interdisciplinary studies and help to identify potential new targets that are amenable to therapeutic approaches such as pharmacological interventions to increase the human healthspan in parallel with the lifespan.
Collapse
Affiliation(s)
- Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
32
|
Brombacher EC, Everts B. Shaping of Dendritic Cell Function by the Metabolic Micro-Environment. Front Endocrinol (Lausanne) 2020; 11:555. [PMID: 33013685 PMCID: PMC7493661 DOI: 10.3389/fendo.2020.00555] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Nutrients are required for growth and survival of all cells, but are also crucially involved in cell fate determination of many cell types, including immune cells. There is a growing appreciation that the metabolic micro-environment also plays a major role in shaping the functional properties of dendritic cells (DCs). Under pathological conditions nutrient availability can range from a very restricted supply, such as seen in a tumor micro-environment, to an overabundance of nutrients found in for example obese adipose tissue. In this review we will discuss what is currently known about the metabolic requirements for DC differentiation and immunogenicity and compare that to how function and fate of DCs under pathological conditions can be affected by alterations in environmental levels of carbohydrates, lipids and amino acids as well as by other metabolic cues, including availability of oxygen, redox homeostasis and lactate levels. Many of these insights have been generated using in vitro model systems, which have revealed highly diverse effects of different metabolic cues on DC function. However, they also stress the importance of shifting toward more physiologically relevant experimental settings to be able to fully delineate the role of the metabolic surroundings in its full complexity in shaping the functional properties of DCs in health and disease.
Collapse
|
33
|
Nogueira PAS, Pereira MP, Soares JJG, de Assis Silva Gomes J, Ribeiro DL, Razolli DS, Velloso LA, Neto MB, Zanon RG. Swimming reduces fatty acids-associated hypothalamic damage in mice. J Chem Neuroanat 2019; 103:101713. [PMID: 31726089 DOI: 10.1016/j.jchemneu.2019.101713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/20/2019] [Accepted: 11/09/2019] [Indexed: 12/28/2022]
Abstract
The arcuate and the paraventricular and lateral hypothalamic nuclei, related to hunger and satiety control, are generally compromised by excess fatty acids. In this situation, fatty acids cause inflammation via TLR4 (toll like receptor 4) and the nuclei become less responsive to the hormones leptin and insulin, contributing to the development of obesity. In this work, these nuclei were analyzed in animals fed with high-fat diet and submitted to swimming without and with load for two months. For this, frontal sections of the hypothalamus were immunolabelled with GFAP (glial fibrillary acidic protein), synaptophysin, IL-6 (interleukin 6) and TLR4. Also, proteins extracted from the hypothalamus were analyzed using Western blotting (GFAP and synaptophysin), fluorometric analysis for caspases 3 and 7, and CBA (cytometric bead array) for Th1, Th2, and Th17 profiles. The high-fat diet significantly caused overweight and, in the hypothalamus, decreased synapses and increased astrocytic reactivity. The swimming with load, especially 80 % of the maximum load, reduced those consequences. The high-fat diet increased TLR4 in the arcuate nucleus and the swimming exercise with 80 % of the maximum load showed a tendency of reducing this expression. Swimming did not significantly influence the inflammatory or anti-inflammatory cytokines in the hypothalamus or in plasma. The high-fat diet in sedentary animals increased the expression of caspases 3 and 7 and swimming practice reduced this increment to levels compatible with animals fed on a normal diet. The set of results conclude that the impact of swimming on the damage caused in the hypothalamus by a high-fat diet is positive. The different aspects analyzed in here point to better cellular viability and conservation of the synapses in the hypothalamic nuclei of overweight animals that practiced swimming with a load.
Collapse
Affiliation(s)
- Pedro Augusto Silva Nogueira
- Institute of Biomedical Science, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil; Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil; Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo, Brazil
| | - Miriam Pimenta Pereira
- Institute of Biomedical Science, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | | | - Juliana de Assis Silva Gomes
- Laboratory of Cellular Interactions Biology, Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniele Lisboa Ribeiro
- Institute of Biomedical Science, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | - Daniela Soares Razolli
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo, Brazil; Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, São Paulo, Brazil
| | - Licio Augusto Velloso
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo, Brazil
| | - Morun Bernardino Neto
- Department of Basic and Environmental Sciences, University of São Paulo, Lorena, São Paulo, Brazil
| | - Renata Graciele Zanon
- Institute of Biomedical Science, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil.
| |
Collapse
|
34
|
Roshanzamir N, Hassan-Zadeh V. Methylation of Specific CpG Sites in IL-1β and IL1R1 Genes is Affected by Hyperglycaemia in Type 2 Diabetic Patients. Immunol Invest 2019; 49:287-298. [PMID: 31476928 DOI: 10.1080/08820139.2019.1656227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objective: Type 2 diabetes (T2D), which is the most common metabolic disorder in the world, results from insulin resistance of target tissues and reduced production of insulin from pancreatic β cells with genetic and environmental factors both playing roles in the pathogenesis. The aim of this study was to investigate the effect of blood glucose levels on DNA methylation of IL-1β and IL1R1 genes in the peripheral blood mononuclear cells (PBMCs) of non-diabetic, type 2 pre-diabetic and diabetic individuals.Methods: In this case-control study, 54 non-diabetic, pre-diabetic and type 2 diabetic individuals were enrolled and categorized based on their fasting plasma glucose (FPG) and glycated hemoglobin (A1C) levels. DNA was extracted from PBMCs and subjected to bisulfite treatment. The methylation status of two CpG sites in the IL-1β gene and three CpG sites in IL1R1 gene was then determined using Sanger sequencing.Results: Our results show that the methylation of IL-1β gene is decreased and the methylation of IL1R1 gene is increased in diabetic individuals with hyperglycemia. Further analysis revealed that both CpG sites in IL-1β gene are affected by hyperglycemia and display decreased methylation while only one CpG site in IL1R1 gene is affected by hyperglycemia.Conclusion: We propose that the DNA methylation status of the CpG sites cg18773937 and cg23149881 in IL-1β gene and the CpG site cg13399261 in IL1R1 gene could serve as an epigenetic marker of chronic inflammation and T2D development. These CpG sites can also be considered for studies on metabolic memory.
Collapse
Affiliation(s)
- Naeimeh Roshanzamir
- Department of Cell and Molecular Biology, Faculty of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Vahideh Hassan-Zadeh
- Department of Cell and Molecular Biology, Faculty of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
35
|
Clapp C, Diaz-Lezama N, Adan-Castro E, Ramirez-Hernandez G, Moreno-Carranza B, Sarti AC, Falzoni S, Solini A, Di Virgilio F. Pharmacological blockade of the P2X7 receptor reverses retinal damage in a rat model of type 1 diabetes. Acta Diabetol 2019; 56:1031-1036. [PMID: 30982154 DOI: 10.1007/s00592-019-01343-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/05/2019] [Indexed: 02/07/2023]
Abstract
AIMS Retinopathy is a leading cause of vision impairment in diabetes. Its pathogenesis involves inflammation, pathological angiogenesis, neuronal and glial dysfunction. The purinergic P2X7 receptor (P2X7R) has a leading role in inflammation and angiogenesis. Potent and selective P2X7R blockers have been synthesized and tested in Phase I/II clinical studies. We hypothesize that P2X7R blockade will ameliorate diabetes-related pathological retinal changes. METHODS Streptozotocin (STZ)-treated rats were intraperitoneally inoculated with either of two small molecule P2X7R receptor inhibitors, A740003 and AZ10606120, and after blood glucose levels increased to above 400 mg/dL, retinae were analyzed for P2X7R expression, vascular permeability, VEGF, and IL-6 expression. RESULTS STZ administration caused a near fourfold increase in blood glucose, a large increase in retinal microvasculature permeability, as well as in retinal P2X7R, VEGF, and IL-6 expression. P2X7R blockade fully reversed retinal vascular permeability increase, VEGF accumulation, and IL-6 expression, with no effect on blood glucose. CONCLUSION P2X7R blockade might be promising strategy for the treatment of microvascular changes observed in the early phases of diabetic retinopathy.
Collapse
Affiliation(s)
- Carmen Clapp
- Institute of Neurobiology, National University of Mexico [UNAM], Querétaro, Mexico
| | - Nundehui Diaz-Lezama
- Institute of Neurobiology, National University of Mexico [UNAM], Querétaro, Mexico
| | - Elva Adan-Castro
- Institute of Neurobiology, National University of Mexico [UNAM], Querétaro, Mexico
| | | | | | - Alba Clara Sarti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Borsari 46, 44121, Ferrara, Italy
| | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Borsari 46, 44121, Ferrara, Italy
| | - Anna Solini
- Department of Surgical, Medical, Molecular, and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Borsari 46, 44121, Ferrara, Italy.
| |
Collapse
|
36
|
Lin YC, Chen YC, Hsiao HP, Kuo CH, Chen BH, Chen YT, Wang SL, Tsai ML, Hung CH. The effects of acarbose on chemokine and cytokine production in human monocytic THP-1 cells. Hormones (Athens) 2019; 18:179-187. [PMID: 30827017 DOI: 10.1007/s42000-019-00101-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 02/15/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Chronic inflammation induced by proinflammatory cytokines and chemokines is postulated to be involved in insulin resistance and β-cell dysfunction in type 2 diabetes mellitus (T2DM). Acarbose, the α-glucosidase inhibitor, is an oral antidiabetic drug for T2DM. Acarbose suppresses inflammatory cytokine production in patients with T2DM, though the underlying mechanisms are unclear. In the present study, we aimed to investigate the anti-inflammatory effects and the exact mechanisms of acarbose in human monocytic THP-1 cells. METHODS THP-1 cells were pretreated with acarbose and then stimulated with lipopolysaccharide (LPS). The levels of Th1-related chemokines, including interferon-γ-inducible protein-10 (IP-10), monocyte chemoattractant protein-1 (MCP-1), Th2-related chemokine macrophage-derived chemokine (MDC), and proinflammatory cytokine tumor necrosis factor-α (TNF-α), were determined by enzyme-linked immunosorbent assay. Intracellular signaling pathways were explored by Western blot analysis and using a chromatin immunoprecipitation assay. RESULTS Acarbose suppressed the levels of IP-10, MCP-1, MDC, and TNF-α and downregulated phosphorylation of p38, c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), and nuclear factor-kappa B-p65 (NF-κB-p65) in LPS-stimulated THP-1 cells. Acarbose suppressed LPS-induced acetylation of histones H3 (H3) and H4 in the IP-10 and MCP-1 promoter regions. These findings revealed the suppressive effects of acarbose on IP-10, MCP-1, MDC, and TNF-α production in THP-1 cells via, at least partially, the p38, JNK, ERK, and NF-κB-p65 pathways, as well as through epigenetic regulation via histone H3 and H4 acetylation. CONCLUSION Our study points to the therapeutic anti-inflammatory potential of acarbose.
Collapse
Affiliation(s)
- Yi-Ching Lin
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Department of Laboratory Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Research Center for Environmental Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Yen-Chun Chen
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, No.482, Shanming Road, Siaogang District, Kaohsiung City, 812, Taiwan, Republic of China
| | - Hui-Pin Hsiao
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Chang-Hung Kuo
- Ta-Kuo Clinic, No.69, Ziqiang 2nd Road, Cianjin District, Kaohsiung City, 144, Taiwan, Republic of China
- Department of Pediatrics, Kaohsiung Municipal Ta-Tung Hospital, No.68, Jhonghua 3rd Road, Cianjin District, Kaohsiung City, 145, Taiwan, Republic of China
| | - Bai-Hsiun Chen
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Research Center for Environmental Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Yi-Ting Chen
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Shih-Ling Wang
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Mei-Lan Tsai
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China
| | - Chih-Hsing Hung
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No.100, Tzyou 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China.
- Research Center for Environmental Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China.
- Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, No.482, Shanming Road, Siaogang District, Kaohsiung City, 812, Taiwan, Republic of China.
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shihchuan 1st Road, Sanmin District, Kaohsiung City, 807, Taiwan, Republic of China.
| |
Collapse
|
37
|
C-reactive protein and blood pressure variability in type 2 hypertensive diabetic patients. Blood Press Monit 2019; 24:52-58. [PMID: 30640739 DOI: 10.1097/mbp.0000000000000362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Increased blood pressure (BP) variability and inflammation are included among the factors recognized as potential predictors of cardiovascular events in type 2 diabetes and hypertension. This study aimed to evaluate whether C-reactive protein (CRP) is associated with increased BP variability in diabetic-hypertensive patients. PATIENTS AND METHODS We carried out a cross-sectional study with 285 diabetic-hypertensive patients, evaluating laboratory characteristics and 24-h ambulatory BP monitoring. SD, coefficient of variation (CV%), time-rate index of 24-h systolic BP (SBP), and 24-h BP patterns were evaluated. Pearson's χ-test, Student's t-test, and the Mann-Whitney test were used to compare the groups. Groups were defined by CRP of up to 3 mg/l (low) and more than 3 mg/l (high). RESULTS The age of the patients was 59 (54-62) years; 101 (35%) were men. There was an increase in office SBP [137 (127-148) vs. 145 (130-157) mmHg] and DBP [79 (73-86) vs. 82 (76-91) mmHg] in the high CRP group. Blood pressure variability indexes were not different among groups [SD: 11.2 (9-15) vs. 12.2 (10-15) mmHg; CV%: 8.6 (7-11) vs. 9.4 (7-12); time rate: 0.55±0.12 vs.12.2 (10-15) mmHg/min]. In addition, BMI (29.3±3.8 vs. 30.9±3.6 kg/m), total cholesterol [166 (148-190) vs. 177 (156-210) mg/dl], and HbA1c [7.5% (6.6-8.9) vs.8.3% (7.1-9.9)] were higher in the high CRP group. CONCLUSION In patients with diabetes and hypertension, higher CRP levels are linked to cardiometabolic derangements, although they are not associated with increased BP variability.
Collapse
|
38
|
Mignogna MD, Leuci S. Interface Between Oral and Systemic Disease. CONTEMPORARY ORAL MEDICINE 2019:67-136. [DOI: 10.1007/978-3-319-72303-7_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
39
|
Deschênes SS, Burns RJ, Schmitz N. Comorbid depressive and anxiety symptoms and the risk of type 2 diabetes: Findings from the Lifelines Cohort Study. J Affect Disord 2018; 238:24-31. [PMID: 29852343 DOI: 10.1016/j.jad.2018.05.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/16/2018] [Accepted: 05/21/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Prior studies indicate that depression is a risk factor for type 2 diabetes (T2D), though there is considerable heterogeneity in reported estimates. Identifying homogeneous subgroups of depression strongly associated with T2D may be beneficial. This study examined associations between depression and anxiety comorbidity with the risk of T2D. METHODS 78,025 participants from the Lifelines Cohort Study (age range = 30-75 years) without diabetes at baseline were included in this study. The Mini-International Neuropsychiatric Interview assessed depressive and anxiety symptoms at baseline. Incident T2D was assessed by self-report or hemoglobin A1c levels during an approximately 3.8-year follow-up period. Risk of T2D was compared across four groups (no depressive or anxiety symptoms, depressive symptoms alone, anxiety symptoms alone, comorbid depressive/anxiety symptoms) using mixed effects logistic regression analyses adjusted for sociodemographic, lifestyle, and cardiometabolic characteristics. RESULTS 1,096 participants developed diabetes. Compared to those without depressive or anxiety symptoms (n = 74,467), those with comorbid depressive and anxiety symptoms (n = 743) were more likely to develop T2D (n = 28, OR = 2.12, 95% CI = 1.22-3.68). Depressive symptoms alone (n = 650) and anxiety symptoms alone (n = 2,165) were not significantly associated with T2D (n = 23 and n = 24, respectively, developed diabetes). Those with comorbid symptoms were also more likely to develop T2D compared to those with depressive symptoms alone (OR = 2.86, 95% CI = 1.25-6.54). LIMITATIONS Depressive and anxiety symptom assessments were based on a screening tool. Hemoglobin A1c data were only available for a subset of participants. CONCLUSIONS Depression with comorbid anxiety may be a subgroup of depression that is strongly associated with the risk of T2D.
Collapse
Affiliation(s)
- Sonya S Deschênes
- Department of Psychiatry, McGill University, Montreal, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| | - Rachel J Burns
- Department of Psychology, Carleton University, Ottawa, Ontario, Canada
| | - Norbert Schmitz
- Department of Psychiatry, McGill University, Montreal, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Epidemiology and Biostatistics, McGill University, Montreal, Canada
| |
Collapse
|
40
|
Development of a Predictive Model for Hyperglycemia in Nondiabetic Recipients After Liver Transplantation. Transplant Direct 2018; 4:e393. [PMID: 30498770 PMCID: PMC6233666 DOI: 10.1097/txd.0000000000000830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/03/2018] [Indexed: 12/20/2022] Open
Abstract
Background Posttransplant hyperglycemia has been associated with increased risks of transplant rejection, infections, length of stay, and mortality. Methods To establish a predictive model to identify nondiabetic recipients at risk for developing postliver transplant (LT) hyperglycemia, we performed this secondary, retrospective data analysis of a single-center, prospective, randomized, controlled trial of glycemic control among 107 adult LT recipients in the inpatient period. Hyperglycemia was defined as a posttransplant glucose level greater than 200 mg/dL after initial discharge up to 1 month following surgery. Candidate variables with P less than 0.10 in univariate analyses were used to build a multivariable logistic regression model using forward stepwise selection. The final model chosen was based on statistical significance and additive contribution to the model based on the Bayesian Information Criteria. Results Forty-three (40.2%) patients had at least 1 episode of hyperglycemia after transplant after the resolution of the initial postoperative hyperglycemia. Variables selected for inclusion in the model (using model optimization strategies) included length of hospital stay (odds ratio [OR], 0.83; P < 0.001), use of glucose-lowering medications at discharge (OR, 3.76; P = 0.03), donor female sex (OR, 3.18; P = 0.02) and donor white race (OR, 3.62; P = 0.01). The model had good calibration (Hosmer-Lemeshow goodness-of-fit test statistic = 9.74, P = 0.28) and discrimination (C-statistic = 0.78; 95% confidence interval, 0.65-0.81, bias-corrected C-statistic = 0.78). Conclusions Shorter hospital stay, use of glucose-lowering medications at discharge, donor female sex and donor white race are important determinants in predicting hyperglycemia in nondiabetic recipients after hospital discharge up to 1 month after liver transplantation.
Collapse
|
41
|
Noordam R, Oudt CH, Bos MM, Smit RAJ, van Heemst D. High-sensitivity C-reactive protein, low-grade systemic inflammation and type 2 diabetes mellitus: A two-sample Mendelian randomization study. Nutr Metab Cardiovasc Dis 2018; 28:795-802. [PMID: 29753585 DOI: 10.1016/j.numecd.2018.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND AIMS The role of inflammation in type 2 diabetes mellitus (T2D) remains unclear. We investigated the associations of high sensitivity C-reactive protein (hsCRP) concentration with T2D and glycemic traits using two-sample Mendelian Randomization. METHODS AND RESULTS We used publically available summary-statistics data from genome-wide association studies on T2D (DIAGRAM: 12 171 cases; 56 862 controls) and glycemic traits (MAGIC: 46 186 participants without diabetes mellitus). We combined the effects of the genetic instrumental variables through inverse-variance weighting (IVW), and MR-Egger regression and weighted-median estimation as sensitivity analyses which take into account potential violations (e.g., directional pleiotropy) of the assumptions of instrumental variable analyses. Analyses were conducted using 15 known hsCRP genetic instruments among which 6 instruments are hsCRP specific and not involved in inflammatory processes beyond hsCRP concentration regulation. Though we found no association between the combined effect of the genetic instrumental variables for hsCRP and T2D with IVW (odds ratio per 1 ln [hsCRP in mg/L]: 1.15; 95% confidence interval: 0.93, 1.42), we found associations for T2D with MR-Egger regression and weighted-median estimation (odds ratio with 95% confidence interval per 1 ln [hsCRP in mg/L], MR-Egger regression: 1.29; 1.08, 1.49; weighted-median estimator: 1.21; 1.02, 1.39). We found no association with T2D for the combination of hsCRP-specific genetic instruments nor did we found associations with glycemic traits in any of the analyses. CONCLUSION Evidence was provided for a potential causal association between hsCRP and T2D, but only after considering directional pleiotropy. However, hsCRP was not causally associated with glycemic traits.
Collapse
Affiliation(s)
- R Noordam
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands.
| | - C H Oudt
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - M M Bos
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - R A J Smit
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands; Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - D van Heemst
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
42
|
Meyerovich K, Ortis F, Cardozo AK. The non-canonical NF-κB pathway and its contribution to β-cell failure in diabetes. J Mol Endocrinol 2018; 61:F1-F6. [PMID: 29728424 DOI: 10.1530/jme-16-0183] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/04/2018] [Indexed: 12/20/2022]
Abstract
The prevalence of diabetes has reached 8.8% in worldwide population and is predicted to increase up to 10.4% by 2040. Thus, there is an urgent need for the development of means to treat or prevent this major disease. Due to its role in inflammatory responses, several studies demonstrated the importance of the transcription factor nuclear factor-κB (NF-κB) in both type 1 diabetes (T1D) and type 2 diabetes (T2D). The two major NF-κB pathways are the canonical and the non-canonical. The later pathway is activated by the NF-κB-inducing kinase (NIK) that triggers p100 processing into p52, which forms with RelB its main dimer. Cytokines mediating the activation of this pathway are present in the serum of T1D and T2D patients. Conversely, limited information is available regarding the role of the alternative pathway on diabetes development and β-cell fate. In the present review, we will briefly describe the involvement of NF-κB on diabetes pathology and discuss new studies indicating an important role for the non-canonical NF-κB activation in β-cell function and survival. The non-canonical NF-κB pathway is emerging as a novel potential target for the development of therapeutic strategies to treat or prevent diabetes.
Collapse
Affiliation(s)
- Kira Meyerovich
- ULB Center for Diabetes ResearchUniversité Libre de Bruxelles (ULB), Brussels, Belgium
| | - Fernanda Ortis
- Department of Cell and Developmental BiologyUniversidade de São Paulo, São Paulo, Brazil
| | - Alessandra K Cardozo
- ULB Center for Diabetes ResearchUniversité Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
43
|
Progress in Metabonomics of Type 2 Diabetes Mellitus. Molecules 2018; 23:molecules23071834. [PMID: 30041493 PMCID: PMC6100487 DOI: 10.3390/molecules23071834] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/20/2022] Open
Abstract
With the improvement of living standards and a change in lifestyle, the incidence of type 2 diabetes mellitus (T2DM) is increasing. Its etiology is too complex to be completely understand yet. Metabonomics techniques are used to study the changes of metabolites and metabolic pathways before and after the onset of diabetes and make it more possible to further understand the pathogenesis of T2DM and improve its prediction, early diagnosis, and treatment. In this review, we summarized the metabonomics study of T2DM in recent years and provided a theoretical basis for the study of pathogenesis and the effective prevention and treatment of T2DM.
Collapse
|
44
|
Malmström H, Walldius G, Carlsson S, Grill V, Jungner I, Gudbjörnsdottir S, Kosiborod M, Hammar N. Elevations of metabolic risk factors 20 years or more before diagnosis of type 2 diabetes: Experience from the AMORIS study. Diabetes Obes Metab 2018; 20:1419-1426. [PMID: 29400911 DOI: 10.1111/dom.13241] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 12/11/2022]
Abstract
AIMS To describe trajectories for metabolic risk factors for type 2 diabetes (T2D) up to 25 years prior to diagnosis and to estimate the absolute 20-year risk for T2D based on a simple set of commonly measured key risk factors. METHODS From the Swedish AMORIS cohort we included 296 428 individuals with data on fasting glucose obtained in health examinations during 1985-1996 (baseline period). All participants were followed until 2012 for development of incident T2D. The 20-year T2D risk based on age, sex, body mass index (BMI), fasting glucose and triglycerides was estimated. Trajectories for biomedical risk factors of T2D starting from >20 years before diagnosis and including fasting glucose, triglycerides and BMI were evaluated according to yearly means for cases and controls. RESULTS We identified 28 244 new T2D cases during the study period, with an average 20-year risk of 8.1%. This risk was substantially increased in overweight and obese participants and those with elevated fasting glucose and triglyceride levels, in both men and women. T2D cases had higher mean BMI and fasting glucose and triglyceride levels compared with controls >20 years before diagnosis and the difference in fasting glucose levels increased over time. CONCLUSIONS Development of T2D is associated with subtle elevations in glucose and lipid levels >20 years before diagnosis. This suggests that diabetogenic processes tied to chronic insulin resistance operate for decades prior to the development of T2D. A simple risk classification can help in early identification of individuals who are at increased risk.
Collapse
Affiliation(s)
- Håkan Malmström
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
- Swedish Orphan Biovitrum AB, Stockholm, Sweden
| | - Göran Walldius
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Sofia Carlsson
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Valdemar Grill
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Trondheim University Hospital, Trondheim, Norway
| | - Ingmar Jungner
- Swedish Orphan Biovitrum AB, Stockholm, Sweden
- CALAB Research, Stockholm, Sweden
| | - Soffia Gudbjörnsdottir
- Department of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mikhail Kosiborod
- Saint Luke's Mid America Heart Institute and University of Missouri-Kansas City, Kansas City, Missouri
| | - Niklas Hammar
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
- Medical Evidence and Observational Research, Global Medical Evidence and Observational Research, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
45
|
Liu T, Yang W, Pang S, Yu S, Yan B. Functional genetic variants within the SIRT2 gene promoter in type 2 diabetes mellitus. Diabetes Res Clin Pract 2018; 137:200-207. [PMID: 29371109 DOI: 10.1016/j.diabres.2018.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 01/17/2018] [Indexed: 02/06/2023]
Abstract
AIMS Type 2 diabetes mellitus (T2D) is a common and complex metabolic diseases caused by interactions between environmental and genetic factors. Genome-wide association studies have identified more than 80 common genetic variants for T2D, which account for only ∼10% of the heritability of T2D cases. SIRT2, a member of NAD(+)-dependent class III deacetylases, is involved in genomic stability, metabolism, inflammation, oxidative stress and autophagy. In maintaining metabolic homeostasis, SIRT2 regulates adipocyte differentiation, fatty acid oxidation, gluconeogenesis, and insulin sensitivity. Thus, we hypothesized that DNA sequence variants (DSVs) in SIRT2 gene promoter may change SIRT2 levels, contributing to T2D. METHODS SIRT2 gene promoter was genetically and functionally analyzed in large cohorts of T2D patients (n = 365) and ethnic-matched controls (n = 358). RESULTS A total of 18 DSVs, including 5 SNPs, were identified in this study. Four novel heterozygous DSVs (g.38900912G > T, g.38900561C > T, g.38900359C > T and g.38900237G > A) were identified in four T2D patients, three of which (g.38900912G > T, g.38900359C > T and g.38900237G > A) significantly increased the transcriptional activity of the SIRT2 gene promoter in cultured pancreatic beta cells (P < .01). Seven novel heterozygous DSVs were only found in controls, and one heterozygous deletion DSV and five SNPs were found in both T2D patients and controls, which did not significantly affect SIRT2 gene promoter activity (P > .05). CONCLUSIONS Our findings suggested that the DSVs may increase SIRT2 gene promoter activity and SIRT2 levels, contributing to T2D development as a risk factor.
Collapse
Affiliation(s)
- Tingting Liu
- College of Clinical Medicine, Jining Medical University, Jining, Shandong 272100, China
| | - Wentao Yang
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Shipeng Yu
- Division of Endocrinology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| |
Collapse
|
46
|
Gaspar RC, Botezelli JD, Kuga GK, Muñoz VR, Coope A, Pereira RM, da Silva ASR, Cintra DE, de Moura LP, Ropelle ER, Pauli JR. High Dosage of Vitamin D Regulates the Energy Metabolism and Increases Insulin Sensitivity, but are Associated with High Levels of Kidney Damage. Drug Dev Res 2017; 78:203-209. [PMID: 28718949 DOI: 10.1002/ddr.21394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 06/12/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Rafael Calais Gaspar
- Department of Nutrition, Metabolism and Exercise; University of Campinas (UNICAMP); Limeira SP Brazil
| | - José Diego Botezelli
- Department of Nutrition, Metabolism and Exercise; University of Campinas (UNICAMP); Limeira SP Brazil
| | - Gabriel Keine Kuga
- Post-Graduate Program in Movement Sciences, São Paulo State University (Unesp), Institute of Biosciences; Rio Claro SP Brazil
| | - Vitor Rosetto Muñoz
- Department of Nutrition, Metabolism and Exercise; University of Campinas (UNICAMP); Limeira SP Brazil
| | - Andressa Coope
- Medical Sciences University, University of Campinas (UNICAMP); Campinas SP Brazil
| | - Rodrigo Martins Pereira
- Department of Nutrition, Metabolism and Exercise; University of Campinas (UNICAMP); Limeira SP Brazil
| | | | - Dennys Esper Cintra
- Department of Nutrition, Metabolism and Exercise; University of Campinas (UNICAMP); Limeira SP Brazil
- Laboratory of Cell Signaling; Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP); Campinas SP Brazil
| | - Leandro Pereira de Moura
- Department of Nutrition, Metabolism and Exercise; University of Campinas (UNICAMP); Limeira SP Brazil
- Laboratory of Cell Signaling; Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP); Campinas SP Brazil
- CEPECE - Center of Research in Sport Sciences, School of Applied Sciences; University of Campinas (UNICAMP); Limeira SP Brazil
| | - Eduardo Rochete Ropelle
- Department of Nutrition, Metabolism and Exercise; University of Campinas (UNICAMP); Limeira SP Brazil
- Laboratory of Cell Signaling; Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP); Campinas SP Brazil
- CEPECE - Center of Research in Sport Sciences, School of Applied Sciences; University of Campinas (UNICAMP); Limeira SP Brazil
| | - José Rodrigo Pauli
- Department of Nutrition, Metabolism and Exercise; University of Campinas (UNICAMP); Limeira SP Brazil
- Laboratory of Cell Signaling; Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP); Campinas SP Brazil
- CEPECE - Center of Research in Sport Sciences, School of Applied Sciences; University of Campinas (UNICAMP); Limeira SP Brazil
| |
Collapse
|
47
|
Pereira RF, Cintra LTA, Tessarin GWL, Chiba FY, de Lima Coutinho Mattera MS, Scaramele NF, Tsosura TVS, Ervolino E, de Oliveira SHP, Sumida DH. Periapical Lesions Increase Macrophage Infiltration and Inflammatory Signaling in Muscle Tissue of Rats. J Endod 2017; 43:982-988. [DOI: 10.1016/j.joen.2017.01.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 01/25/2017] [Accepted: 01/29/2017] [Indexed: 12/20/2022]
|
48
|
Interaction effect of PGC-1α rs10517030 variants and energy intake in the risk of type 2 diabetes in middle-aged adults. Eur J Clin Nutr 2017; 71:1442-1448. [DOI: 10.1038/ejcn.2017.68] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/29/2017] [Accepted: 03/19/2017] [Indexed: 12/22/2022]
|
49
|
Santos IS, Bittencourt MS, Goulart AC, Schmidt MI, Diniz MDFH, Lotufo PA, Benseñor IM. Insulin resistance is associated with carotid intima-media thickness in non-diabetic subjects. A cross-sectional analysis of the ELSA-Brasil cohort baseline. Atherosclerosis 2017; 260:34-40. [DOI: 10.1016/j.atherosclerosis.2017.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/01/2017] [Accepted: 03/06/2017] [Indexed: 12/12/2022]
|
50
|
Bone and adipose tissue formation. Z Rheumatol 2017. [DOI: 10.1007/s00393-016-0143-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|