1
|
Yu Y, Tan T, Yang W, Xu Z, Liu Y. Association between the systemic immune-inflammation index and obesity among adults: Insights from the NHANES 2017-2018. PLoS One 2024; 19:e0308288. [PMID: 39116149 PMCID: PMC11309425 DOI: 10.1371/journal.pone.0308288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/21/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Inflammation is an important causative factor of obesity. This study aimed to explore the possible association between the systemic immune-inflammatory index, a novel indicator of inflammation, and obesity. METHODS Data were collected from 4395 participants of the National Health and Nutrition Examination Survey 2017-2018 aged ≥ 20 years. The systemic immune-inflammatory index was calculated by multiplying the platelet count by the neutrophil-to-lymphocyte ratio. Obesity was defined as a body mass index ≥ 30 kg/m2. RESULTS A significant positive correlation was observed between the systemic immune-inflammatory index and body mass index following multivariate linear regression analysis (β = 1.75; 95% confidence interval = 1.16-2.33), which was greatest in adults aged < 60 years without hypertension and diabetes. Smoothed curve fitting and threshold effect analysis were used to characterize the nonlinear association between the systemic immune-inflammatory index and body mass index, and the inflection point was found to be 729.3. CONCLUSIONS The systemic immune-inflammatory index is positively associated with body mass index among adults in the United States and has the potential to enhance efforts to prevent adult obesity.
Collapse
Affiliation(s)
- Yanmei Yu
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Tongcai Tan
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wei Yang
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhitao Xu
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yong Liu
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Sasso E, Baticic L, Sotosek V. Postprandial Dysmetabolism and Its Medical Implications. Life (Basel) 2023; 13:2317. [PMID: 38137918 PMCID: PMC10744591 DOI: 10.3390/life13122317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
An unbalanced diet increases the risk of developing a variety of chronic diseases and cancers, leading to higher morbidity and mortality rates worldwide. Low-grade systemic chronic inflammation mediated by the activation of the innate immune system is common to all these pathologies. Inflammation is a biological response of the body and a normal part of host defense to combat the effects of bacteria, viruses, toxins and macronutrients. However, when the innate immune system is constantly activated, it can promote the development of low-grade systemic chronic inflammation, which could play an important role in the development of chronic diseases and cancer. Since most chronic inflammatory diseases are associated with diet, a balanced healthy diet high in anti-inflammatory food components could prevent chronic diseases and cancer. The cells of the body's immune system produce chemokines and cytokines which can have pro-inflammatory and tumor-promoting as well as anti-inflammatory and tumor-fighting functions. A challenge in the future will be to assess whether polymorphisms in immune-related genes may play a role in promoting pro-inflammatory activity. Thanks to this duality, future research on immune regulation could focus on how innate immune cells can be modified to convert a pro-inflammatory and tumor-friendly microenvironment into an anti-inflammatory and anti-tumor one. This review describes inflammatory responses mediated by the innate immune system in various diseases such as hyperglycemia and/or hyperlipemia, obesity, type II diabetes, cardiovascular disease and cancer.
Collapse
Affiliation(s)
- Emanuel Sasso
- Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia
| | - Vlatka Sotosek
- Department of Anesthesiology, Reanimatology, Emergency and Intensive Care Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia;
| |
Collapse
|
3
|
Borja-Magno AI, Furuzawa-Carballeda J, Guevara-Cruz M, Arias C, Granados J, Bourges H, Tovar AR, Sears B, Noriega LG, Gómez FE. Supplementation with EPA and DHA omega-3 fatty acids improves peripheral immune cell mitochondrial dysfunction and inflammation in subjects with obesity. J Nutr Biochem 2023; 120:109415. [PMID: 37437746 DOI: 10.1016/j.jnutbio.2023.109415] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/01/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
Omega-3 fatty acids (w-3 FA) have anti-inflammatory effects and improve mitochondrial function. Nonetheless, little is known about their effect on mitochondrial bioenergetics of peripheral blood mononuclear cells (PBMCs) in individuals with obesity. Thus, this study aimed to determine the mitochondrial bioenergetics status and cell subset composition of PBMCs during obesity, before and after 1 month supplementation with w-3 FA. We performed a case-control study with twelve women with normal BMI (lean group) and 19 with grade 2 obesity (obese group), followed by a before-after prospective study where twelve subjects with obesity received a 1 month intervention with 5.25 g of w-3 FA (3.5 g eicosapentaenoic (EPA) and 1.75 g docosahexaenoic (DHA) acids), and obtained PBMCs from all participants. Mitochondrial bioenergetic markers, including basal and ATP-production associated respiration, proton leak, and nonmitochondrial respiration, were higher in PBMCs from the obese group vs. the lean group. The bioenergetic health index (BHI), a marker of mitochondrial function, was lower in the obese vs. the lean group. In addition, Th1, Th2, Th17, CD4+ Tregs, CD8+ Tregs, and Bregs, M1 monocytes and pDCreg cells were higher in PBMCs from the obese group vs. the lean group. The w-3 FA intervention improved mitochondrial function, mainly by decreasing nonmitochondrial respiration and increasing the reserve respiratory capacity and BHI. The intervention also reduced circulating pro-inflammatory and anti-inflammatory lymphocyte and monocytes subsets in individuals with obesity. The mitochondrial dysfunction of PBMCs and the higher proportion of peripheral pro-inflammatory and anti-inflammatory immune cells in subjects with obesity, improved with 1 month supplementation with EPA and DHA.
Collapse
Affiliation(s)
- Angélica I Borja-Magno
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Janette Furuzawa-Carballeda
- Department of Experimental Surgery, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Martha Guevara-Cruz
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Clorinda Arias
- Department of Genomics Medicine and Environmental Toxicology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacan, Ciudad de México, Mexico
| | - Julio Granados
- Department of Transplants, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Hector Bourges
- Divission of Nutrition, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Armando R Tovar
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico
| | - Barry Sears
- Inflammation Research Foundation, Peabody, Massachusetts, USA
| | - Lilia G Noriega
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico.
| | - Francisco Enrique Gómez
- Department of Nutritional Physiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México, Mexico.
| |
Collapse
|
4
|
Agarwal J, Awasthi NP, Singh S, Tiwari V. Sequential Shifting in T-helper and T-cytotoxic Subset Cell Population in Mild and Severe COVID-19 Patients Infected With Variant B.1.61. Cureus 2023; 15:e40556. [PMID: 37465793 PMCID: PMC10351332 DOI: 10.7759/cureus.40556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2023] [Indexed: 07/20/2023] Open
Abstract
AIM Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) modulates antiviral immunity via T cells, but whether these cells are active or abundant in coronavirus disease 2019 (COVID-19) patients is unknown. The present study aimed to investigate the temporal shifting in the T-cell population and their subsets, T-Helper (Th) cell (CD4) and T-Cytotoxic (Tc) cell (CD8) in COVID-19 patients. METHOD Thirty confirmed COVID-19 patients (nasal swab reverse transcription-polymerase chain reaction (RT-PCR) confirmed) were enrolled. On the basis of oxygen saturation (SpO2) levels, patients were stratified into two categories: (i) mild (n=11) having fever and SpO2 level >95%, and (ii) severe (n=19) on the ventilator, and in the intensive care unit (ICU) as per the Indian Council of Medical Research (ICMR) guidelines. Thirty age-sex-matched controls without infectious diseases unrelated to COVID-19 were also enrolled in the study. Patients with inflammatory diseases and severe comorbidities that compromise immunity were excluded from the study. Immunophenotyping flow cytometry assay was used to evaluate T-cell viability, Th, and Tc cells population in mild and severe COVID-19 patients on day 1 (at admission) and day 4 (decreasing the infection load) in the second COVID-19 wave (variant: B.1.61). Categorical variables were expressed as frequency and percentage and p-values were calculated by Chi-square test. All the variables were represented in median and Q1 (25 percentile) and Q3 (75 percentile). The Mann-Whitney test was used to compare the study groups. The Δ mean differences were calculated by using the Paired samples t-test. The statistically significant level was taken as p<0.05. RESULTS Hemoglobin, total leukocyte count (TLC), lymphocytes, monocytes, and eosinophils were significantly reduced in patients (p<0.05). A significant decrease of CD4 and CD8 cells in severe COVID-19 patients vs. controls (CD4, median 49; CD8, 40.12; p>0.05) was seen. Th-EM (effector memory)-Tim-3 (T-cell immunoglobulin domain and mucin domain 3)+ was significantly higher (p=0.002) however, Tc-EMRA (effector memory cells re-expressing)-Tim-3+, Tc-Naive-Tim-3+, Tc-EM-PD1+ and Tc-CM (central memory)-Tim-3+ significantly reduced (p<0.05) in mild COVID-19 patients than controls. Similarly, in severe COVID-19 patients, Th-EMRA-Tim-3+, Th-Naive-PD1+, Th-EM-PD1+, Th-EM-Tim 3+ and Th-CM-Tim-3+ showed a significant reduction (p<0.05) and Tc-EMRA-Tim-3+, Tc-Naive-Tim-3+, Tc-EM-PD1+, and Tc-CM-Tim-3+ showed similar results. In mild vs. severe group, decreased T-cells (p=0.001), Th-EMRA-Tim-3+ (p=0.024), and Th-Navie-Tim-3+ (p=0.005), and significantly increased (p<0.05) Tc-Naive-Tim3+ (p=0.001), Tc-EM-Tim-3+ (p=0.031), and Tc-CM-Tim-3+ (p=0.08) were observed. Severe COVID-19 patients showed a significant increase in Th-Naive-Tim3+ (day 4-day 1; δ43, p=0.019), Th-EM-Tim3+ (δ 16.24, p=0.033), and Th-CM-Tim3+ (δ 13.57, p=0.041). CONCLUSION T-cell populations and CD8 subset help to differentiate the mild and severe COVID-19 patients. Monitoring T cells, especially CD8 subset changes, has important implications for diagnosing and treating mild and severe patients being critically ill.
Collapse
Affiliation(s)
- Jyotsna Agarwal
- Microbiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, IND
| | - Namrata P Awasthi
- Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, IND
| | - Shivani Singh
- Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, IND
| | - Vandana Tiwari
- Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, IND
| |
Collapse
|
5
|
In obese hypertensives cholecalciferol inhibits circulating TH17 cells but not macrophage infiltration on adipose tissue. Clin Immunol 2023; 247:109244. [PMID: 36706826 DOI: 10.1016/j.clim.2023.109244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023]
Abstract
In arterial hypertension, increased Th17 cells and reduced Tregs are the hallmarks of immunological dysfunction and the basis for the investigation of immunomodulatory drugs. Although cholecalciferol is not a primary immunomodulator, it has recognized action on immune cells, leading us to hypothesise if cholecalciferol can induce a more tolerogenic phenotype in obese hypertensives. In a phase-2, single-centre, randomised, open, 24-week trial, we assigned adults with obesity-associated hypertension and vitamin D deficiency to receive usual therapy plus 50,000 IU/week of cholecalciferol or usual therapy alone. The primary endpoint was the percentual variation in T CD4+, T CD8+, Tregs, and Th17 cells. Secondary endpoints included the percentual variation in Th1, Tc1, Tc17, and monocytes and variation in the number of perivascular and non-perivascular macrophages, T CD4+ and T CD8+ lymphocytes in subcutaneous abdominal adipose tissue. A control group of 12 overweight normotensives was also evaluated for peripheral immune cells. A total of 36 obese hypertensives were randomised, 18 in each group. In comparison with normotensive controls, hypertensives presented higher percentages of T lymphocytes (p = 0.016), Tregs (p = 0.014), and non-classical monocytes (p < 0.001). At week 24, Th17 cells increased in control group (p = 0.017) but remained stable in cholecalciferol group. For Tregs, downregulation towards the values of normotensive controls was observed (p = 0.003), and in multivariate analysis, an increased loading in the setting of the cells of adaptive immunity observed (eigenvalue 1.78, p < 0.001). No changes were documented for monocytes. In adipose tissue, a baseline negative correlation between vitamin D and perivascular macrophages was observed (r = -0.387, p = 0.024) that persisted in the control group (r = -0.528, p = 0.024) but not in the cholecalciferol group, which presented an increase in non-perivascular macrophages (p = 0.029) at week 24. No serious adverse events were reported for all the participants. In this trial, we found that supplementation with cholecalciferol interfered with peripheral and adipose tissue immune cell profile, downregulating peripheral Th17 cells, but increasing the number of infiltrating subcutaneous adipose tissue macrophages. (Funded by Núcleo Estudos Hipertensão da Beira Interior; EudraCT number: 2015-003910-26).
Collapse
|
6
|
Zi C, Wang D, Gao Y, He L. The role of Th17 cells in endocrine organs: Involvement of the gut, adipose tissue, liver and bone. Front Immunol 2023; 13:1104943. [PMID: 36726994 PMCID: PMC9884980 DOI: 10.3389/fimmu.2022.1104943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
T Helper 17 (Th17) cells are adaptive immune cells that play myriad roles in the body. Immune-endocrine interactions are vital in endocrine organs during pathological states. Th17 cells are known to take part in multiple autoimmune diseases over the years. Current evidence has moved from minimal to substantial that Th17 cells are closely related to endocrine organs. Diverse tissue Th17 cells have been discovered within endocrine organs, including gut, adipose tissue, liver and bone, and these cells are modulated by various secretions from endocrine organs. Th17 cells in these endocrine organs are key players in the process of an array of metabolic disorders and inflammatory conditions, including obesity, insulin resistance, nonalcoholic fatty liver disease (NAFLD), primary sclerosing cholangitis (PSC), osteoporosis and inflammatory bowel disease (IBD). We reviewed the pathogenetic or protective functions played by Th17 cells in various endocrine tissues and identified potential regulators for plasticity of it. Furthermore, we discussed the roles of Th17 cells in crosstalk of gut-organs axis.
Collapse
Affiliation(s)
- Changyan Zi
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Die Wang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongxiang Gao
- School of International Education, Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Yongxiang Gao, ; Lisha He,
| | - Lisha He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Yongxiang Gao, ; Lisha He,
| |
Collapse
|
7
|
Li C, Li Y, Wang N, Ge Z, Shi Z, Wang J, Ding B, Bi Y, Wang Y, Wang Y, Hong Z. The Increased Risk of Hypertension Caused by Irrational Dietary Pattern May Be Associated with Th17 Cell in the Middle-Aged and Elderly Rural Residents of Beijing City, Northern China: A 1:1 Matched Case-Control Study. Nutrients 2023; 15:nu15020290. [PMID: 36678161 PMCID: PMC9863205 DOI: 10.3390/nu15020290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
An irrational diet has been widely considered as one of the vital risk factors of hypertension. Previous studies have indicated that immune dysfunction may be involved in the pathogenic process of hypertension, while fewer studies have mentioned whether CD4+ T cells are involved in the association between dietary pattern and hypertension. This present 1:1 matched case-control study was conducted to analyze the association among dietary pattern, CD4+ T cells and hypertension. A total of 56 patients with diagnosed hypertension and 56 subjects without diagnosed hypertension in the rural area of Beijing City, northern China, were matched by age and gender, and then classified into a case group and a control group, respectively. Compared with the control group, higher frequencies of pro-inflammatory CD4+ T cells, such as Th1, Th1(IFN-γ), Th17(IL-17A), and Th1/17 (IFN-γ/IL-17A), were found in the case group (p < 0.05). A significantly higher level of circulating IL-17A was also found in the case group (7.4 pg/mL vs. 8.2 pg/mL, p < 0.05). Five dietary patterns were identified using exploratory factor analysis. An irrational dietary pattern, characterized by high-factor loadings of refined wheat (0.65), meat (0.78), poultry (0.76), and alcoholic beverage (0.73), was positively associated with SBP (β = 5.38, 95%CI = 0.73~10.03, p < 0.05) in the multiple linear regression model with the adjustment of potential covariates. The other dietary patterns showed no significant association with blood pressure. Furthermore, meat, processed meat, and animal viscera were positively correlated with the peripheral Th17 or Th1/17. In conclusion, the irrational dietary pattern characterized by refined wheat, meat, poultry, and alcoholic beverage, was positively correlated with blood pressure, and may increase the risk of hypertension in the rural area of Beijing, northern China. Th17, a subset of the CD4+ T helper cells, may be involved in the association between irrational dietary pattern and hypertension.
Collapse
|
8
|
Zhang YX, Ou MY, Yang ZH, Sun Y, Li QF, Zhou SB. Adipose tissue aging is regulated by an altered immune system. Front Immunol 2023; 14:1125395. [PMID: 36875140 PMCID: PMC9981968 DOI: 10.3389/fimmu.2023.1125395] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023] Open
Abstract
Adipose tissue is a widely distributed organ that plays a critical role in age-related physiological dysfunctions as an important source of chronic sterile low-grade inflammation. Adipose tissue undergoes diverse changes during aging, including fat depot redistribution, brown and beige fat decrease, functional decline of adipose progenitor and stem cells, senescent cell accumulation, and immune cell dysregulation. Specifically, inflammaging is common in aged adipose tissue. Adipose tissue inflammaging reduces adipose plasticity and pathologically contributes to adipocyte hypertrophy, fibrosis, and ultimately, adipose tissue dysfunction. Adipose tissue inflammaging also contributes to age-related diseases, such as diabetes, cardiovascular disease and cancer. There is an increased infiltration of immune cells into adipose tissue, and these infiltrating immune cells secrete proinflammatory cytokines and chemokines. Several important molecular and signaling pathways mediate the process, including JAK/STAT, NFκB and JNK, etc. The roles of immune cells in aging adipose tissue are complex, and the underlying mechanisms remain largely unclear. In this review, we summarize the consequences and causes of inflammaging in adipose tissue. We further outline the cellular/molecular mechanisms of adipose tissue inflammaging and propose potential therapeutic targets to alleviate age-related problems.
Collapse
Affiliation(s)
- Yi-Xiang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min-Yi Ou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zi-Han Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang-Bai Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Li S, Zong X, Zhang L, Li L, Wu J. A chromatin accessibility landscape during early adipogenesis of human adipose-derived stem cells. Adipocyte 2022; 11:239-249. [PMID: 35435105 PMCID: PMC9037556 DOI: 10.1080/21623945.2022.2063015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Obesity has become a serious global public health problem; a deeper understanding of systemic change of chromatin accessibility during human adipogenesis contributes to conquering obesity and its related diseases. Here, we applied the ATAC-seq method to depict a high-quality genome‐wide time-resolved accessible chromatin atlas during adipogenesis of human adipose-derived stem cells (hASCs). Our data indicated that the chromatin accessibility drastic dynamically reformed during the adipogenesis of hASCs and 8 h may be the critical transition node of adipogenesis chromatin states from commitment phase to determination phase. Moreover, upon adipogenesis, we also found that the chromatin accessibility of regions related to anti-apoptotic, angiogenic and immunoregulatory gradually increased, which is beneficial to maintaining the health of adipose tissue (AT). Finally, the chromatin accessibility changed significantly in intronic regions of peroxisome proliferator‐activated receptor γ during adipogenesis, and these regions were rich in transcription factors binding motifs that were exposed for further regulation. Overall, we systematically analysed the complex change of chromatin accessibility occurring in the early stage of adipogenesis and deepened our understanding of human adipogenesis. Furthermore, we also provided a good reference data resource of genome‐wide chromatin accessibility for future studies on human adipogenesis.
Collapse
Affiliation(s)
- Sen Li
- Department of Biochemistry & Immunology, Capital Institute of Pediatrics, Beijing, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Xiaolin Zong
- Division of achievements transformation, Development Center for Medical Science & Technology National Health Commission of the People’s Republic of China, Beijing, China
| | - Liheng Zhang
- Shanghai Jiayin Biotechnology Co., Ltd, Shanghai, China
| | - Luya Li
- Department of Biochemistry & Immunology, Capital Institute of Pediatrics, Beijing, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Jianxin Wu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- Beijing TongRen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Wen J, Wang L. Identification of key genes and their association with immune infiltration in adipose tissue of obese patients: a bioinformatic analysis. Adipocyte 2022; 11:401-412. [PMID: 35894174 PMCID: PMC9336476 DOI: 10.1080/21623945.2022.2104512] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Immune cell-mediated adipose tissue (AT) inflammation contributes to obesity-related metabolic disorders, but the precise underlying mechanisms remain largely elusive. In this study, we used the R software to screen key differentially expressed genes (DEGs) in AT from lean and obese individuals and conducted function enrichment analysis. We then analysed their PPI network by using the STRING database. Hub genes were screened by cytohubba plugin. Subsequently, CIBERSORTx was used to predict the proportion of immune cells in AT from lean and obese subjects. Finally, the correlation between hub genes and immune cell proportions was analysed. These studies identified 290 DEGs in the AT between lean and obese subjects. Among them, IL6, CCL19, CXCL8, CXCL12, CCL2, CCL3, CCL4, CXCL2, IL1B, and CXCL1 were proved to be hub genes in regulating the protein-protein interaction (PPI) network. We also found that CXCL8 is positively correlated with resting NK cells, monocytes, activated mast cells, and eosinophils, but negatively correlated with CD8+ T cells and activated NK cells in obese individuals. Taken together, our study identified key genes in AT that are correlated with immune cell infiltration, uncovering potential new targets for the prevention and treatment of obesity and its related complications via regulating the immune microenvironment.
Collapse
Affiliation(s)
- Jie Wen
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liwen Wang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Zhang S, Sun Z, Jiang X, Lu Z, Ding L, Li C, Tian X, Wang Q. Ferroptosis increases obesity: Crosstalk between adipocytes and the neuroimmune system. Front Immunol 2022; 13:1049936. [PMID: 36479119 PMCID: PMC9720262 DOI: 10.3389/fimmu.2022.1049936] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
Ferroptosis requires not only the accumulation of iron ions, but also changes in many ferroptosis-related regulators, including a decrease in GPX4 and inhibition of SLC7A11 for classical ferroptosis, a deletion of FSP1 or GCH1. Surprisingly, adipose tissue (AT) in the obesity conditions is also accompanied by iron buildup, decreased GSH, and increased ROS. On the neurological side, the pro-inflammatory factor released by AT may have first caused ferroptosis in the vagus nerve by inhibiting of the NRF2-GPX4 pathway, resulting in disorders of the autonomic nervous system. On the immune side, obesity may cause M2 macrophages ferroptosis due to damage to iron-rich ATMs (MFehi) and antioxidant ATMs (Mox), and lead to Treg cells ferroptosis through reductions in NRF2, GPX4, and GCH1 levels. At the same time, the reduction in GPX4 may also trigger the ferroptosis of B1 cells. In addition, some studies have also found the role of GPX4 in neutrophil autophagy, which is also worth pondering whether there is a connection with ferroptosis. In conclusion, this review summarizes the associations between neuroimmune regulation associated with obesity and ferroptosis, and on the basis of this, highlights their potential molecular mechanisms, proposing that ferroptosis in one or more cells in a multicellular tissue changes the fate of that tissue.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xuewen Tian
- *Correspondence: Xuewen Tian, ; Qinglu Wang,
| | - Qinglu Wang
- *Correspondence: Xuewen Tian, ; Qinglu Wang,
| |
Collapse
|
12
|
Li G, Li X, Yang L, Wang S, Dai Y, Fekry B, Veillon L, Tan L, Berdeaux R, Eckel-Mahan K, Lorenzi PL, Zhao Z, Lehner R, Sun K. Adipose tissue-specific ablation of Ces1d causes metabolic dysregulation in mice. Life Sci Alliance 2022; 5:e202101209. [PMID: 35459739 PMCID: PMC9034061 DOI: 10.26508/lsa.202101209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 01/25/2023] Open
Abstract
Carboxylesterase 1d (Ces1d) is a crucial enzyme with a wide range of activities in multiple tissues. It has been reported to localize predominantly in ER. Here, we found that Ces1d levels are significantly increased in obese patients with type 2 diabetes. Intriguingly, a high level of Ces1d translocates onto lipid droplets where it digests the lipids to produce a unique set of fatty acids. We further revealed that adipose tissue-specific Ces1d knock-out (FKO) mice gained more body weight with increased fat mass during a high fat-diet challenge. The FKO mice exhibited impaired glucose and lipid metabolism and developed exacerbated liver steatosis. Mechanistically, deficiency of Ces1d induced abnormally large lipid droplet deposition in the adipocytes, causing ectopic accumulation of triglycerides in other peripheral tissues. Furthermore, loss of Ces1d diminished the circulating free fatty acids serving as signaling molecules to trigger the epigenetic regulations of energy metabolism via lipid-sensing transcriptional factors, such as HNF4α. The metabolic disorders induced an unhealthy microenvironment in the metabolically active tissues, ultimately leading to systemic insulin resistance.
Collapse
Affiliation(s)
- Gang Li
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Li Yang
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shuyue Wang
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Baharan Fekry
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lucas Veillon
- Metabolomic Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Metabolomic Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca Berdeaux
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center-UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Kristin Eckel-Mahan
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center-UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Philip L Lorenzi
- Metabolomic Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Richard Lehner
- Group on Molecular and Cell Biology of Lipids, Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center-UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
13
|
Wu H, Zhu L, Kinnear D, Triggs N, Quintanilla NM, Himes R. Clinical, Laboratory, and Histologic Correlates of Serum Antinuclear Antibody in Hispanic Pediatric Patients With Nonalcoholic Fatty Liver Disease. Am J Clin Pathol 2022; 158:221-227. [PMID: 35311933 DOI: 10.1093/ajcp/aqac028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 02/01/2022] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVES The significance of antinuclear antibody (ANA) positivity in pediatric Hispanic patients with nonalcoholic fatty liver disease (NAFLD) is unknown. METHODS ANA status was correlated with clinical, laboratory, and histologic parameters in Hispanic patients with a histologic diagnosis of NAFLD. RESULTS Thirty-eight Hispanic children (27 male and 11 female) underwent liver biopsy at a median age of 12.1 years. Twenty patients (53%) had positive ANAs. The ANA-positive patients had higher fasting insulin levels (median [interquartile range (IQR)], 32.4 [25.4] µU/mL) and higher insulin resistance (median [Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) IQR], 5.9 [3.1]) than the ANA-negative patients (fasting insulin: median [IQR], 17 [13.9] µU/mL and median [HOMA-IR IQR], 3.5 [2.6] µU/mL; P = .05 and .01, respectively). Serum high-density lipoprotein (HDL) cholesterol levels were higher in the ANA-negative patients (median [IQR], 47 [18] mg/dL) than the ANA-positive patients (38 [12] mg/dL) (P = .03). There were no statistical differences in a series of demographic, clinical, laboratory, and histologic parameters between the ANA-positive and the ANA-negative patients. At a median follow-up of 2.6 years, alanine aminotransferase was significantly lower than the baseline levels in both groups. In 1 patient undergoing ANA retesting, the titer had normalized from a baseline of 1:1,280 3.8 years earlier. CONCLUSIONS In pediatric Hispanic patients with NAFLD, a positive ANA result is associated with insulin resistance and lower HDL cholesterol levels.
Collapse
Affiliation(s)
- Hao Wu
- Department of Pathology, Yale New Haven Hospital and Yale School of Medicine, New Haven, CT, USA
| | - Liang Zhu
- Biostatistics & Epidemiology Research Design Core, Center for Clinical and Translational Sciences, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Darryl Kinnear
- Departments of Pathology, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Nicole Triggs
- Pediatrics (Section of Gastroenterology, Hepatology and Nutrition), Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Norma M Quintanilla
- Departments of Pathology, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Ryan Himes
- Pediatric Gastroenterology, Hepatology and Nutrition, Ochsner Hospital for Children, New Orleans, LA, USA
| |
Collapse
|
14
|
Liu L, Hu J, Wang Y, Lei H, Xu D. The role and research progress of the balance and interaction between regulatory T cells and other immune cells in obesity with insulin resistance. Adipocyte 2021; 10:66-79. [PMID: 33472506 PMCID: PMC7834085 DOI: 10.1080/21623945.2021.1876375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Metabolic homoeostasis in adipose tissue plays a major role in obesity-related insulin resistance (IR). Regulatory T (Treg) cells have been recorded to regulate metabolic homoeostasis in adipose tissue. However, their specific mechanism is not yet known. This review aims to present the role of Treg cells and other immune cells in obesity-associated IR, focusing on the balance of numbers and functions of Treg cells and other immune cells as well as the crucial role of their interactions in maintaining adipose tissue homoeostasis. Th1 cells, Th17 cells, CD8+ T cells, and pro-inflammatory macrophages mediate the occurrence of obesity and IR by antagonizing Treg cells, while anti-inflammatory dendritic cells, eosinophils and type 2 innate lymphoid cells (ILC2s) regulate the metabolic homoeostasis of adipose tissue by promoting the proliferation and differentiation of Treg cells. γ δ T cells and invariant natural killer T (iNKT) cells have complex effects on Treg cells, and their roles in obesity-associated IR are controversial. The balance of Treg cells and other immune cells can help maintain the metabolic homoeostasis of adipose tissue. Further research needs to explore more specific molecular mechanisms, thus providing more precise directions for the treatment of obesity with IR.
Collapse
Affiliation(s)
- Leiling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yating Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
15
|
Teklu M, Zhou W, Kapoor P, Patel N, Playford MP, Sorokin AV, Dey AK, Teague HL, Manyak GA, Rodante JA, Keel A, Chen MY, Bluemke DA, Khera AV, Mehta NN. Abdominal subcutaneous adipose tissue negatively associates with subclinical coronary artery disease in men with psoriasis. Am J Prev Cardiol 2021; 8:100231. [PMID: 34553185 PMCID: PMC8441148 DOI: 10.1016/j.ajpc.2021.100231] [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: 04/28/2021] [Revised: 07/13/2021] [Accepted: 08/02/2021] [Indexed: 11/25/2022] Open
Abstract
Objective Understand the relationship between abdominal subcutaneous adipose tissue (ASAT) and coronary atherosclerosis defined as noncalcified and lipid-rich necrotic core burden in psoriasis. Methods We performed a cross-sectional study of 232 participants (92 women) with psoriasis and without known cardiovascular disease. Participants underwent coronary computed tomography angiography to characterize coronary atherosclerosis burden and low dose abdominal computed tomography to quantify subcutaneous and visceral adipose tissue. Fat depot volumes were first adjusted for each participant's BMI (ASATadjBMI). Results In women, there was a positive correlation between ASATadjBMI and systemic inflammation as assessed by hs-C-reactive protein (r=0.30; p=.004) and GlycA (r=0.29; p=.007) as well as total cholesterol (r=0.24; p=.02) and low-density lipoprotein cholesterol (r=0.22; p=.04). In men, ASATadjBMI correlated with hs-C-reactive protein (r=0.18; p=.04) and insulin resistance (r=0.17; p=.04). In models fully adjusted for traditional cardiovascular risk factors, ASATadjBMI negatively associated with noncalcified and lipid-rich necrotic core burden in men (β= -0.17; p=.03, β= -0.20; p=.03, respectively), but not women (β= -0.06; p=.57, β= 0.09; p=.49, respectively) with psoriasis. Conclusions For a given BMI, ASAT negatively associated with coronary atherosclerosis burden in male participants with psoriasis. The observed sex-specific effects warrant further study of ASAT in states of chronic inflammation.
Collapse
Affiliation(s)
- Meron Teklu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wunan Zhou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Promita Kapoor
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nidhi Patel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin P Playford
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander V Sorokin
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amit K Dey
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heather L Teague
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grigory A Manyak
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Justin A Rodante
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrew Keel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marcus Y Chen
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - David A Bluemke
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Amit V Khera
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Alhamdan F, Marsh LM, Pedersen F, Alhamwe BA, Thölken C, Pfefferle PI, Bahmer T, Greulich T, Potaczek DP, Garn H. Differential Regulation of Interferon Signaling Pathways in CD4 + T Cells of the Low Type-2 Obesity-Associated Asthma Phenotype. Int J Mol Sci 2021; 22:ijms221810144. [PMID: 34576307 PMCID: PMC8469911 DOI: 10.3390/ijms221810144] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/14/2022] Open
Abstract
In the era of personalized medicine, insights into the molecular mechanisms that differentially contribute to disease phenotypes, such as asthma phenotypes including obesity-associated asthma, are urgently needed. Peripheral blood was drawn from 10 obese, non-atopic asthmatic adults with a high body mass index (BMI; 36.67 ± 6.90); 10 non-obese, non-atopic asthmatic adults with normal BMI (23.88 ± 2.73); and 10 healthy controls with normal BMI (23.62 ± 3.74). All asthmatic patients were considered to represent a low type-2 asthma phenotype according to selective clinical parameters. RNA sequencing (RNA-Seq) was conducted on peripheral blood CD4+ T cells. Thousands of differentially expressed genes were identified in both asthma groups compared with heathy controls. The expression of interferon (IFN)-stimulated genes associated with IFN-related signaling pathways was specifically affected in obese asthmatics, while the gap junction and G protein-coupled receptor (GPCR) ligand binding pathways were enriched in both asthma groups. Furthermore, obesity gene markers were also upregulated in CD4+ T cells from obese asthmatics compared with the two other groups. Additionally, the enriched genes of the three abovementioned pathways showed a unique correlation pattern with various laboratory and clinical parameters. The specific activation of IFN-related signaling and viral infection pathways might provide a novel view of the molecular mechanisms associated with the development of the low type-2 obesity-associated asthma phenotype, which is a step ahead in the development of new stratified therapeutic approaches.
Collapse
Affiliation(s)
- Fahd Alhamdan
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, Medical Faculty, Philipps University of Marburg, D-35043 Marburg, Germany; (F.A.); (D.P.P.)
| | - Leigh M. Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, A-8010 Graz, Austria;
| | - Frauke Pedersen
- Lungen Clinic Grosshansdorf GmbH, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), D-22927 Großhansdorf, Germany; (F.P.); (T.B.)
| | - Bilal Alashkar Alhamwe
- Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Institute of Tumor Immunology, Medical Faculty, Philipps University of Marburg, D-35043 Marburg, Germany;
- College of Pharmacy, International University for Science and Technology (IUST), Daraa 15, Syria
| | - Clemens Thölken
- Institute of Medical Bioinformatics and Biostatistics, Medical Faculty, Philipps University of Marburg, D-35037 Marburg, Germany;
| | - Petra Ina Pfefferle
- Comprehensive Biobank Marburg (CBBMR), Member of the German Biobank Alliance (GBA) and the German Center for Lung Research (DZL), Medical Faculty, Philipps University of Marburg, D-35043 Marburg, Germany;
| | - Thomas Bahmer
- Lungen Clinic Grosshansdorf GmbH, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), D-22927 Großhansdorf, Germany; (F.P.); (T.B.)
- Department for Internal Medicine I, Campus Kiel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), University Hospital Schleswig-Holstein, D-24105 Kiel, Germany
| | - Timm Greulich
- Pulmonary and Critical Care Medicine, Member of the German Center for Lung Research, University Medical Center Giessen and Marburg, Department of Medicine, D-35043 Marburg, Germany;
| | - Daniel P. Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, Medical Faculty, Philipps University of Marburg, D-35043 Marburg, Germany; (F.A.); (D.P.P.)
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, Medical Faculty, Philipps University of Marburg, D-35043 Marburg, Germany; (F.A.); (D.P.P.)
- Correspondence: ; Tel.: +49-6421-2866040
| |
Collapse
|
17
|
Teklu M, Parel PM, Mehta NN. Psoriasis and Cardiometabolic Diseases: The Impact of Inflammation on Vascular Health. PSORIASIS-TARGETS AND THERAPY 2021; 11:99-108. [PMID: 34322373 PMCID: PMC8312325 DOI: 10.2147/ptt.s320016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022]
Abstract
Psoriasis is a common chronic inflammatory condition associated with a higher risk of cardiovascular disease. Psoriasis confers a dose-dependent increase in risk for the metabolic syndrome and its components. The metabolic syndrome and its components have been associated with higher coronary atherosclerosis in psoriasis and cardiovascular events in the general population. In this review, we discuss the role of inflammation and psoriasis in cardiometabolic diseases with a focus on the metabolic syndrome and its components. We highlight the relationship between psoriasis and important cardiovascular risk factors encompassed by obesity, dyslipidemia, insulin resistance and hypertension. Furthermore, we briefly highlight literature on anti-inflammatory therapies and their impact on the components of the metabolic syndrome as well as directly quantified coronary atherosclerosis burden.
Collapse
Affiliation(s)
- Meron Teklu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Philip M Parel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
Kuperberg SJ, Navetta-Modrov B. The Role of Obesity in the Immunopathogenesis of COVID-19 Respiratory Disease and Critical Illness. Am J Respir Cell Mol Biol 2021; 65:13-21. [PMID: 33797351 PMCID: PMC8320126 DOI: 10.1165/rcmb.2020-0236tr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Coronavirus disease (COVID-19), the clinical syndrome caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is currently a global health pandemic with substantial morbidity and mortality. COVID-19 has cast a shadow on nearly every aspect of society, straining health systems and economies across the world. Although it is widely accepted that a close relationship exists between obesity, cardiovascular disease, and metabolic disorders on infection, we are only beginning to understand ways in which the immunological sequelae of obesity functions as a predisposing factor related to poor clinical outcomes in COVID-19. As both the innate and adaptive immune systems are each primed by obesity, the alteration of key pathways results in both an immunosuppressed and hyperinflammatory state. The present review will discuss the cellular and molecular immunology of obesity in the context of its role as a risk factor for severe COVID-19, discuss the role of cytokine storm, and draw parallels to prior viral epidemics such as severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and 2009 H1N1.
Collapse
Affiliation(s)
- Stephen J Kuperberg
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Brianne Navetta-Modrov
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Stony Brook University Hospital/Renaissance School of Medicine, Stony Brook, New York
| |
Collapse
|
19
|
Adipose Tissue Immunomodulation and Treg/Th17 Imbalance in the Impaired Glucose Metabolism of Children with Obesity. CHILDREN-BASEL 2021; 8:children8070554. [PMID: 34199040 PMCID: PMC8305706 DOI: 10.3390/children8070554] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
In the last few decades, obesity has increased dramatically in pediatric patients. Obesity is a chronic disease correlated with systemic inflammation, characterized by the presence of CD4 and CD8 T cell infiltration and modified immune response, which contributes to the development of obesity related diseases and metabolic disorders, including impaired glucose metabolism. In particular, Treg and Th17 cells are dynamically balanced under healthy conditions, but imbalance occurs in inflammatory and pathological states, such as obesity. Some studies demonstrated that peripheral Treg and Th17 cells exhibit increased imbalance with worsening of glucose metabolic dysfunction, already in children with obesity. In this review, we considered the role of adipose tissue immunomodulation and the potential role played by Treg/T17 imbalance on the impaired glucose metabolism in pediatric obesity. In the patient care, immune monitoring could play an important role to define preventive strategies of pediatric metabolic disease treatments.
Collapse
|
20
|
Zhang Y, Cao H, Chen J, Li Y, Xu A, Wang Y. Adiponectin-expressing Treg facilitate T lymphocyte development in thymic nurse cell complexes. Commun Biol 2021; 4:344. [PMID: 33727658 PMCID: PMC7966800 DOI: 10.1038/s42003-021-01877-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/19/2021] [Indexed: 02/06/2023] Open
Abstract
Adiponectin is a well-known insulin sensitizer and anti-inflammatory molecule, possessing therapeutic potentials in cardiovascular, metabolic and cancer diseases. Results of the present study demonstrate that adiponectin is expressed in a population of regulatory T-cells (Treg) resided within the thymic nurse cell (TNC) complexes. Adoptive transfer of adiponectin-expressing Treg precursors effectively attenuated obesity, improved glucose and insulin tolerance, prevented fatty liver injuries in wild-type mice fed a high-fat diet, and significantly inhibited breast cancer development in MMTV-PyVT transgenic mice. Within the TNC complexes, locally produced adiponectin bound to and regulated the expression as well as the distribution of CD100, a transmembrane lymphocyte semaphorin, in turn modulating the lymphoepithelial interactions to facilitate T-cell development and maturation. In summary, adiponectin plays an important role in the selection and development of T lymphocytes within the TNC complexes. Adiponectin-expressing Treg represent a promising candidate for adoptive cell immunotherapy against obesity-related metabolic and cancer diseases.
Collapse
MESH Headings
- Adiponectin/genetics
- Adiponectin/metabolism
- Adoptive Transfer
- Animals
- Antigens, CD/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/prevention & control
- Cell Differentiation
- Cell Line, Tumor
- Disease Models, Animal
- Female
- Glucose Intolerance/immunology
- Glucose Intolerance/metabolism
- Glucose Intolerance/prevention & control
- Humans
- Insulin Resistance
- Mammary Tumor Virus, Mouse/genetics
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Non-alcoholic Fatty Liver Disease/immunology
- Non-alcoholic Fatty Liver Disease/metabolism
- Non-alcoholic Fatty Liver Disease/prevention & control
- Obesity/immunology
- Obesity/metabolism
- Obesity/prevention & control
- Phenotype
- Semaphorins/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Thymocytes/immunology
- Thymocytes/metabolism
- Thymocytes/transplantation
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Mice
Collapse
Affiliation(s)
- Yiwei Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Handi Cao
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jie Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Henry Fok College of Biology and Agriculture, Shaoguan University, Shaoguan, Guangdong, China
| | - Yuanxin Li
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China.
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
21
|
Zhou H, Wang L, Liu F. Immunological Impact of Intestinal T Cells on Metabolic Diseases. Front Immunol 2021; 12:639902. [PMID: 33679800 PMCID: PMC7930072 DOI: 10.3389/fimmu.2021.639902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Emerging evidence accumulated over the past several years has uncovered intestinal CD4+ T cells as an essential mediator in modulating intestinal immunity in health and diseases. It has also been increasingly recognized that dietary and microbiota-derived factors play key roles in shaping the intestinal CD4+ T-cell compartment. This review aims to discuss the current understanding on how the intestinal T cell immune responses are disturbed by obesity and metabolic stress. In addition, we review how these changes influence systemic metabolic homeostasis and the T-cell-mediated crosstalk between gut and liver or brain in the progression of obesity and its related diseases. Lastly, we highlight the potential roles of some drugs that target intestinal T cells as a therapeutic treatment for metabolic diseases. A better understanding of the interaction among metabolites, bacterial signals, and T cell immune responses in the gut and their roles in systemic inflammation in metabolic tissues should shed new light on the development of effective treatment of obesity and related disorders.
Collapse
Affiliation(s)
- Haiyan Zhou
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liwen Wang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feng Liu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
22
|
Fedullo AL, Schiattarella A, Morlando M, Raguzzini A, Toti E, De Franciscis P, Peluso I. Mediterranean Diet for the Prevention of Gestational Diabetes in the Covid-19 Era: Implications of Il-6 In Diabesity. Int J Mol Sci 2021; 22:1213. [PMID: 33530554 PMCID: PMC7866163 DOI: 10.3390/ijms22031213] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
The aim of this review is to highlight the influence of the Mediterranean Diet (MedDiet) on Gestational Diabetes Mellitus (GDM) and Gestational Weight Gain (GWG) during the COVID-19 pandemic era and the specific role of interleukin (IL)-6 in diabesity. It is known that diabetes, high body mass index, high glycated hemoglobin and raised serum IL-6 levels are predictive of poor outcomes in coronavirus disease 2019 (COVID-19). The immunopathological mechanisms of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection include rising levels of several cytokines and in particular IL-6. The latter is associated with hyperglycemia and insulin resistance and could be useful for predicting the development of GDM. Rich in omega-3 polyunsaturated fatty acids, vitamins, and minerals, MedDiet improves the immune system and could modulate IL-6, C reactive protein and Nuclear Factor (NF)-κB. Moreover, polyphenols could modulate microbiota composition, inhibit the NF-κB pathway, lower IL-6, and upregulate antioxidant enzymes. Finally, adhering to the MedDiet prior to and during pregnancy could have a protective effect, reducing GWG and the risk of GDM, as well as improving the immune response to viral infections such as COVID-19.
Collapse
Affiliation(s)
- Anna Lucia Fedullo
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), 00178 Rome, Italy; (A.L.F.); (A.R.); (E.T.)
| | - Antonio Schiattarella
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.S.); (M.M.); (P.D.F.)
| | - Maddalena Morlando
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.S.); (M.M.); (P.D.F.)
| | - Anna Raguzzini
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), 00178 Rome, Italy; (A.L.F.); (A.R.); (E.T.)
| | - Elisabetta Toti
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), 00178 Rome, Italy; (A.L.F.); (A.R.); (E.T.)
| | - Pasquale De Franciscis
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.S.); (M.M.); (P.D.F.)
| | - Ilaria Peluso
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), 00178 Rome, Italy; (A.L.F.); (A.R.); (E.T.)
| |
Collapse
|
23
|
Zhou H, Peng X, Hu J, Wang L, Luo H, Zhang J, Zhang Y, Li G, Ji Y, Zhang J, Bai J, Liu M, Zhou Z, Liu F. DsbA-L deficiency in T cells promotes diet-induced thermogenesis through suppressing IFN-γ production. Nat Commun 2021; 12:326. [PMID: 33436607 PMCID: PMC7804451 DOI: 10.1038/s41467-020-20665-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 12/10/2020] [Indexed: 01/17/2023] Open
Abstract
Adipose tissue-resident T cells have been recognized as a critical regulator of thermogenesis and energy expenditure, yet the underlying mechanisms remain unclear. Here, we show that high-fat diet (HFD) feeding greatly suppresses the expression of disulfide-bond A oxidoreductase-like protein (DsbA-L), a mitochondria-localized chaperone protein, in adipose-resident T cells, which correlates with reduced T cell mitochondrial function. T cell-specific knockout of DsbA-L enhances diet-induced thermogenesis in brown adipose tissue (BAT) and protects mice from HFD-induced obesity, hepatosteatosis, and insulin resistance. Mechanistically, DsbA-L deficiency in T cells reduces IFN-γ production and activates protein kinase A by reducing phosphodiesterase-4D expression, leading to increased BAT thermogenesis. Taken together, our study uncovers a mechanism by which T cells communicate with brown adipocytes to regulate BAT thermogenesis and whole-body energy homeostasis. Our findings highlight a therapeutic potential of targeting T cells for the treatment of over nutrition-induced obesity and its associated metabolic diseases.
Collapse
MESH Headings
- Adipocytes, Brown/drug effects
- Adipocytes, Brown/metabolism
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Animals
- Diet, High-Fat
- Down-Regulation/drug effects
- Energy Metabolism/drug effects
- Feeding Behavior
- Glutathione Transferase/deficiency
- Glutathione Transferase/metabolism
- Insulin Resistance
- Interferon-gamma/administration & dosage
- Interferon-gamma/biosynthesis
- Interferon-gamma/pharmacology
- Male
- Mice, Knockout
- Mitochondria/drug effects
- Mitochondria/metabolism
- Obesity/genetics
- Obesity/pathology
- T-Lymphocytes/drug effects
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/metabolism
- Thermogenesis/drug effects
- Thermogenesis/genetics
- Uncoupling Protein 1/metabolism
- Mice
Collapse
Affiliation(s)
- Haiyan Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Xinyi Peng
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Jie Hu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Liwen Wang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Hairong Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Junyan Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Yacheng Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Guobao Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Yujiao Ji
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Juli Bai
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China
| | - Feng Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
24
|
Wen J, Liu Q, Liu M, Wang B, Li M, Wang M, Shi X, Liu H, Wu J. Increasing Imbalance of Treg/Th17 Indicates More Severe Glucose Metabolism Dysfunction in Overweight/obese Patients. Arch Med Res 2020; 52:339-347. [PMID: 33317842 DOI: 10.1016/j.arcmed.2020.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 11/15/2020] [Accepted: 11/27/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chronic low-grade inflammation and dysfunction of metabolism has been reported to be involved in obesity. Regulatory T cell (Treg) and helper T cell 17 (Th17) are involved in chronic inflammatory diseases. Impaired balance of Treg/Th17 is one of the major factors contributing to inflammatory status in obesity. METHODS Overweight/obese patients (n = 80) were recruited and classified into three subgroups: normal glucose tolerance group (NGT, n = 32), impaired glucose regulation group (IGR, n = 19) and type two diabetes mellitus group (T2DM, n = 29). Healthy individuals were paired as normal control group (NC, n = 37). We used flow cytometry to test the frequencies of circulating Treg and Th17 cells of all subjects. Serum IL-6, IL-10, TNF-α, IL-17A levels were detected by cytometric bead array and clinical information was extracted from medical records. RESULTS In group IGR and T2DM, we revealed a severe decrease in peripheral ratio of Treg/Th17 compared with NC, but no significant difference was seen in group NGT. The serum level of IL-6 in group NGT and T2DM was higher than healthy subjects. The FPG and HbA1c levels were negatively correlated with the ratio of Treg/Th17 in overweight/obese patients. ROC curve analysis revealed that peripheral Treg/Th17 ratio <1.255 was a risk factor for prediabetes and diabetes in overweight/obese patients. CONCLUSION Peripheral Treg/Th17 imbalance exists in overweight/obese patients with IGR or T2DM and peripheral Treg/Th17 imbalance might be a risk factor for prediabetes and diabetes in overweight/obese patients.
Collapse
Affiliation(s)
- Jie Wen
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qingjing Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengmeng Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bian Wang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mei Li
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Wang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiajie Shi
- Department of Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
25
|
Kwok S, Adam S, Ho JH, Iqbal Z, Turkington P, Razvi S, Le Roux CW, Soran H, Syed AA. Obesity: A critical risk factor in the COVID-19 pandemic. Clin Obes 2020; 10:e12403. [PMID: 32857454 PMCID: PMC7460880 DOI: 10.1111/cob.12403] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/04/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022]
Abstract
Obesity is an emerging independent risk factor for susceptibility to and severity of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Previous viral pandemics have shown that obesity, particularly severe obesity (BMI > 40 kg/m2 ), is associated with increased risk of hospitalization, critical care admission and fatalities. In this narrative review, we examine emerging evidence of the influence of obesity on COVID-19, the challenges to clinical management from pulmonary, endocrine and immune dysfunctions in individuals with obesity and identify potential areas for further research. We recommend that people with severe obesity be deemed a vulnerable group for COVID-19; clinical trials of pharmacotherapeutics, immunotherapies and vaccination should prioritize inclusion of people with obesity.
Collapse
Affiliation(s)
- See Kwok
- Cardiovascular Trials UnitManchester University NHS Foundation TrustManchesterUK
- Faculty of BiologyMedicine and Health, University of ManchesterManchesterUK
| | - Safwaan Adam
- Faculty of BiologyMedicine and Health, University of ManchesterManchesterUK
- Department of EndocrinologyChristie NHS Foundation TrustManchesterUK
| | - Jan Hoong Ho
- Cardiovascular Trials UnitManchester University NHS Foundation TrustManchesterUK
- Faculty of BiologyMedicine and Health, University of ManchesterManchesterUK
| | - Zohaib Iqbal
- Cardiovascular Trials UnitManchester University NHS Foundation TrustManchesterUK
- Faculty of BiologyMedicine and Health, University of ManchesterManchesterUK
| | - Peter Turkington
- Department of Respiratory MedicineSalford Royal NHS Foundation TrustSalfordUK
| | - Salman Razvi
- Cardiovascular Research CentreInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
| | - Carel W. Le Roux
- Diabetes Complications Research CentreUniversity College DublinDublinIreland
| | - Handrean Soran
- Cardiovascular Trials UnitManchester University NHS Foundation TrustManchesterUK
- Faculty of BiologyMedicine and Health, University of ManchesterManchesterUK
| | - Akheel A. Syed
- Faculty of BiologyMedicine and Health, University of ManchesterManchesterUK
- Department of DiabetesEndocrinology and Obesity Medicine, Salford Royal NHS Foundation TrustSalfordUK
| |
Collapse
|
26
|
Bahmad HF, Daouk R, Azar J, Sapudom J, Teo JCM, Abou-Kheir W, Al-Sayegh M. Modeling Adipogenesis: Current and Future Perspective. Cells 2020; 9:cells9102326. [PMID: 33092038 PMCID: PMC7590203 DOI: 10.3390/cells9102326] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is contemplated as a dynamic organ that plays key roles in the human body. Adipogenesis is the process by which adipocytes develop from adipose-derived stem cells to form the adipose tissue. Adipose-derived stem cells’ differentiation serves well beyond the simple goal of producing new adipocytes. Indeed, with the current immense biotechnological advances, the most critical role of adipose-derived stem cells remains their tremendous potential in the field of regenerative medicine. This review focuses on examining the physiological importance of adipogenesis, the current approaches that are employed to model this tightly controlled phenomenon, and the crucial role of adipogenesis in elucidating the pathophysiology and potential treatment modalities of human diseases. The future of adipogenesis is centered around its crucial role in regenerative and personalized medicine.
Collapse
Affiliation(s)
- Hisham F. Bahmad
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, 1107 2260 Beirut, Lebanon; (H.F.B.); (R.D.); (J.A.)
| | - Reem Daouk
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, 1107 2260 Beirut, Lebanon; (H.F.B.); (R.D.); (J.A.)
| | - Joseph Azar
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, 1107 2260 Beirut, Lebanon; (H.F.B.); (R.D.); (J.A.)
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, 2460 Abu Dhabi, UAE;
| | - Jeremy C. M. Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, 2460 Abu Dhabi, UAE;
- Correspondence: (J.C.M.T.); (W.A.-K.); (M.A.-S.); Tel.: +97126286689 (J.C.M.T.); +9611350000 (ext. 4778) (W.A.-K.); +97126284560 (M.A.-S.)
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, 1107 2260 Beirut, Lebanon; (H.F.B.); (R.D.); (J.A.)
- Correspondence: (J.C.M.T.); (W.A.-K.); (M.A.-S.); Tel.: +97126286689 (J.C.M.T.); +9611350000 (ext. 4778) (W.A.-K.); +97126284560 (M.A.-S.)
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, 2460 Abu Dhabi, UAE
- Correspondence: (J.C.M.T.); (W.A.-K.); (M.A.-S.); Tel.: +97126286689 (J.C.M.T.); +9611350000 (ext. 4778) (W.A.-K.); +97126284560 (M.A.-S.)
| |
Collapse
|
27
|
Liu R, Wang Y, Li J, Han H, Xia Z, Liu F, Wu K, Yang L, Liu X, Zhu C. Decreased T cell populations contribute to the increased severity of COVID-19. Clin Chim Acta 2020; 508:110-114. [PMID: 32405080 PMCID: PMC7219428 DOI: 10.1016/j.cca.2020.05.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND We observe changes of the main lymphocyte subsets (CD16+CD56、CD19、CD3、CD4、and CD8) in COVID-19-infected patients and explore whether the changes are associated with disease severity. METHODS One-hundred and fifty-four cases of COVID-19-infected patients were selected and divided into 3 groups (moderate group, severe group and critical group). The flow cytometry assay was performed to examine the numbers of lymphocyte subsets. RESULTS CD3+, CD4+ and CD8 + T lymphocyte subsets were decreased in COVID-19-infected patients. Compared with the moderate group and the sever group, CD3+, CD4+ and CD8+ T cells in the critical group decreased greatly (P < 0.001, P = 0.005 or P = 0.001). CONCLUSIONS Reduced CD3+, CD4+, CD8+ T lymphocyte counts may reflect the severity of the COVID-19. Monitoring T cell changes has important implications for the diagnosis and treatment of severe patients who may become critically ill.
Collapse
Affiliation(s)
- Rui Liu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Ying Wang
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Sino-French Cooperative Central Lab, Shanghai Pudong Gongli Hospital, Secondary Military Medical University, Shanghai 200135, PR China
| | - Jie Li
- Postgraduate training base in Shanghai Gongli Hospital, Ningxia medical university, Pudong New Area, Shanghai200135, PR China
| | - Huan Han
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Zunen Xia
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Fang Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, PR China; Wuhan Institute of Biotechnology, Wuhan, Hubei 430075, PR China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, PR China
| | - Lan Yang
- Department of Clinical Laboratory, Shanghai Gongli Hospital, the Second Military Medical University, Pudong New Area, Shanghai200135, PR China
| | - Xinghui Liu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, the Second Military Medical University, Pudong New Area, Shanghai200135, PR China
| | - Chengliang Zhu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China.
| |
Collapse
|
28
|
Xing Z, Peng Z, Wang X, Zhu Z, Pei J, Hu X, Chai X. Waist circumference is associated with major adverse cardiovascular events in male but not female patients with type-2 diabetes mellitus. Cardiovasc Diabetol 2020; 19:39. [PMID: 32213183 PMCID: PMC7093979 DOI: 10.1186/s12933-020-01007-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Although studies have shown that waist circumference (WC) is positively associated with an increased risk of cardiovascular diseases among the normal population, few studies have investigated WC in patients with type-2 diabetes mellitus (T2DM). METHODS This was a post hoc analysis of the Action to Control Cardiovascular Risk in Diabetes (ACCORD) study. The Cox proportional hazards models was used to investigate the relationship between WC and major adverse cardiovascular events (MACEs) in T2DM patients with cardiovascular disease (CVD) or high risk factors of CVD. RESULTS A total of 10,251 T2DM patients (6299 men [61.4%], 3952 women [38.6%]) were included in our analysis. The mean age was 64.0 ± 7.53 years. After a mean follow-up at 9.2 ± 2.4 years later, 1804 patients (event rate of 23 per 1000 person-years) had developed MACEs. MACEs rates in men and women were 18.0 and 26.0 events per 1000 person-years, respectively. After multivariable adjustment, each increase in WC of 1 SD increased the risk of MACEs (HR: 1.10, 95% CI 1.04-1.17; P < 0.01) in men, with a non-significant increase in MACEs (HR: 1.04, 95% CI 0.95-1.13; P = 0.40) in women. Compared with those in the first quartile of WC, male patients in the fourth quartile of WC had a hazard ratio (HR) of 1.24 (95% CI 1.05-1.46) for MACEs; female patients in the fourth quartile of WC had an HR of 1.22 (95% CI 0.96-1.56) for MACEs. CONCLUSIONS Higher WC is associated with increased risks of MACEs in male but not female T2DM patients. Trial registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT00000620).
Collapse
Affiliation(s)
- Zhenhua Xing
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China.,Emergency Medicine and Difficult Diseases Institute,Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhenyu Peng
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China.,Emergency Medicine and Difficult Diseases Institute,Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Xiaopu Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Zhaowei Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Junyu Pei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinqun Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xiangping Chai
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China. .,Emergency Medicine and Difficult Diseases Institute,Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
29
|
Chirumbolo S. Oxidative Stress, Nutrition and Cancer: Friends or Foes? World J Mens Health 2020; 39:19-30. [PMID: 32202081 PMCID: PMC7752511 DOI: 10.5534/wjmh.190167] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022] Open
Abstract
The relationship between cancer and nutrition, as well as nutrition and oxidative stress, shares puzzling aspects that current research is investigating as the possible components of an intriguing regulating mechanism involving the complex interplay between adipose tissue and other compartments. Along the very recent biological evolution, humans underwent a rapid change in their lifestyles and henceforth the role of the adipocytes earned a much more complex task in the fine tuning of the tissue microenvironment. A lipidic signaling language probably evolved in association with the signaling role of reactive oxygen species, which gained a fundamental part in the regulation of cell stem and plasticity. The possible relationship with cancer onset might have some causative mechanism in the impairment of this complex task, usually deregulated by drastic changes in one's own lifestyle and dietary habit. This review tries to address this issue.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| |
Collapse
|
30
|
von Meyenn L, Bertschi NL, Schlapbach C. Targeting T Cell Metabolism in Inflammatory Skin Disease. Front Immunol 2019; 10:2285. [PMID: 31608068 PMCID: PMC6769046 DOI: 10.3389/fimmu.2019.02285] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/10/2019] [Indexed: 12/31/2022] Open
Abstract
A properly functioning T cell compartment is crucial to protect the host from infections, tumors, and environmental substances. In recent years, it has become increasingly clear that the processes underlying proper T cell activation, proliferation, and differentiation require well-tuned and dynamic changes in T cell metabolism. Thus, proper metabolic reprogramming in T cells is crucial to ensure proper immunity in the context of infection and anti-tumor immunity. Conversely, aberrant regulation of T cell metabolism can impair T cell function and thereby contribute to T cell-mediated disease. In this review, the relevance of recent insights into T cell metabolism for prototypical T cell-mediated skin diseases will be discussed and their therapeutic potential will be outlined. First, the major modules of T cell metabolism are summarized. Then, the importance of T cell metabolism for T cell-mediated skin diseases such as psoriasis and allergic contact dermatitis is discussed, based on the current state of our understanding thereof. Finally, novel therapeutic opportunities for inflammatory skin disease that might emerge from investigations in T cell metabolism are outlined.
Collapse
Affiliation(s)
| | | | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
31
|
Liu R, Nikolajczyk BS. Tissue Immune Cells Fuel Obesity-Associated Inflammation in Adipose Tissue and Beyond. Front Immunol 2019; 10:1587. [PMID: 31379820 PMCID: PMC6653202 DOI: 10.3389/fimmu.2019.01587] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity-associated inflammation stems from a combination of cell-intrinsic changes of individual immune cell subsets and the dynamic crosstalk amongst a broad array of immune cells. Although much of the focus of immune cell contributions to metabolic disease has focused on adipose tissue-associated cells, these potent sources of inflammation inhabit other metabolic regulatory tissues, including liver and gut, and recirculate to promote systemic inflammation and thus obesity comorbidities. Tissue-associated immune cells, especially T cell subpopulations, have become a hotspot of inquiry based on their contributions to obesity, type 2 diabetes, non-alcoholic fatty liver diseases and certain types of cancers. The cell-cell interactions that take place under the stress of obesity are mediated by intracellular contact and cytokine production, and constitute a complicated network that drives the phenotypic alterations of immune cells and perpetuates a feed-forward loop of metabolic decline. Herein we discuss immune cell functions in various tissues and obesity-associated cancers from the viewpoint of inflammation. We also emphasize recent advances in the understanding of crosstalk amongst immune cell subsets under obese conditions, and suggest future directions for focused investigations with clinical relevance.
Collapse
Affiliation(s)
- Rui Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Barbara S. Nikolajczyk
- Department of Pharmacology and Nutritional Sciences, Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
32
|
Oliveira BM, Rasteiro AM, Correia A, Pinto A, Meireles P, Ferreira PG, Vilanova M, Teixeira L. T cells in mesenteric and subcutaneous adipose tissue of Holstein-Friesian cows. Sci Rep 2019; 9:3413. [PMID: 30833655 PMCID: PMC6399275 DOI: 10.1038/s41598-019-39938-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/05/2019] [Indexed: 12/11/2022] Open
Abstract
The importance of immune cells present in the adipose tissue to metabolic homeostasis has been increasingly recognized. Nevertheless, in bovines few studies have so far addressed the immune cell populations resident in this tissue. Here we developed an eight-colour flow cytometry panel to address T cell populations present in bovine adipose tissue. Our results showed that γδ T cells, CD4+ and CD8+ CD3+ non-γδ T cells, as well as NK cells, are present in the mesenteric and subcutaneous adipose tissue of Holstein-Friesian cows. The frequency of both γδ T cells and CD8+ non-γδ T cells was found higher in mesenteric than in subcutaneous adipose tissue. The majority of T cells in adipose tissue presented a CD45RO+CD62L- phenotype, characteristic of effector memory cells, and the frequency of these cellular populations was higher than in the blood. The ratio of CD4+ T cells over CD8+ T cells was similar between subcutaneous and mesenteric adipose tissue but different from the one found in blood. Overall, our results highlight particular phenotypic characteristics of bovine adipose tissue T cell populations.
Collapse
Affiliation(s)
- Bárbara M Oliveira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB -Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Ana M Rasteiro
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB -Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Alexandra Correia
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Ana Pinto
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB -Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Pedro Meireles
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,SVAExpleite, Rua D. Sancho I, 3202, 4760-485, Vila Nova de Famalicão, Portugal
| | - Paula G Ferreira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB -Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Manuel Vilanova
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Luzia Teixeira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal. .,UMIB -Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
33
|
Reiche ME, den Toom M, Willemsen L, van Os B, Gijbels MJJ, Gerdes N, Aarts SABM, Lutgens E. Deficiency of T cell CD40L has minor beneficial effects on obesity-induced metabolic dysfunction. BMJ Open Diabetes Res Care 2019; 7:e000829. [PMID: 31908798 PMCID: PMC6936585 DOI: 10.1136/bmjdrc-2019-000829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/30/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Obesity-associated metabolic dysfunction increases the risk of multiple diseases such as type 2 diabetes and cardiovascular disease. The importance of the co-stimulatory CD40-CD40L dyad in diet-induced obesity (DIO), with opposing phenotypes arising when either the receptor (aggravating) or the ligand (protective) is deleted, has been described previously. The functions of CD40 and CD40L are cell type dependent. As co-stimulation via T cell-mediated CD40L is essential for driving inflammation, we here investigate the role of T cell CD40L in DIO. RESEARCH DESIGN AND METHODS CD4CreCD40Lfl/fl mice on a C57BL/6 background were generated and subjected to DIO by administration of 15 weeks of high fat diet (HFD). RESULTS HFD-fed CD4CreCD40Lfl/fl mice had similar weight gain, adipocyte sizes, plasma cholesterol and triglyceride levels as their wild-type (WT) counterparts. Insulin and glucose tolerance were comparable, although CD4CreCD40Lfl/fl mice did have a decreased plasma insulin concentration, suggesting a minor improvement of insulin resistance. Furthermore, although the degree of hepatosteatosis was similar in both genotypes, the gene expression of fatty acid synthase 1 and ATP-citrate lyase had decreased, whereas expression of peroxisome proliferator-activated receptor-α had increased in livers of CD4CreCD40Lfl/fl mice, suggesting decreased hepatic lipid uptake in absence of T cell CD40L.Moreover, CD4CreCD40Lfl/fl mice displayed significantly lower numbers of effector memory CD4+ T cells and regulatory T cells in blood and lymphoid organs compared with WT. However, immune cell composition and inflammatory status of the adipose tissue was similar in CD4CreCD40Lfl/fl and WT mice. CONCLUSIONS T cell CD40L deficiency results in a minor improvement of insulin sensitivity and hepatic steatosis in DIO, despite the strong decrease in effector T cells and regulatory T cells in blood and lymphoid organs. Our data indicate that other CD40L-expressing cell types are more relevant in the pathogenesis of obesity-associated metabolic dysfunction.
Collapse
Affiliation(s)
- Myrthe E Reiche
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Myrthe den Toom
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Lisa Willemsen
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Bram van Os
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Marion J J Gijbels
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
- Pathology, CARIM, Maastricht, The Netherlands
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Suzanne A B M Aarts
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Esther Lutgens
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Munich, Germany
| |
Collapse
|