1
|
Reguero M, Reglero G, Quintela JC, Ramos-Ruiz R, Ramírez de Molina A, Gómez de Cedrón M. Silymarin-Enriched Extract from Milk Thistle Activates Thermogenesis in a Preclinical Model of High-Fat-Diet-Induced Obesity to Relieve Systemic Meta-Inflammation. Nutrients 2024; 16:4166. [PMID: 39683558 DOI: 10.3390/nu16234166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Obesity and aging are associated with the progressive loss of brown adipose tissue (BAT), an increase in visceral white adipose tissue (vWAT), and a reduction in subcutaneous white adipose tissue (sWAT). The progressive expansion of visceral obesity promotes a low grade of systemic chronic inflammation (meta-inflammation), contributing to the onset of comorbidities such as type 2 diabetes mellitus (T2DM), metabolic syndrome, and even cancer. Thus, preserving the thermogenic activity of adipose tissue and improving the metabolic flexibility of sWAT could be an effective strategy to prevent the development of metabolic chronic diseases and promote healthy aging. Precision nutrition has emerged as a complementary approach to control the metabolic alterations associated with unhealthy obesity and aging. In a previous work, we described that a silymarin-enriched extract from milk thistle (Mthistle) increased markers of browning and thermogenesis in vitro in human differentiated adipocytes (SGBS). OBJECTIVES/METHODS Therefore, this study aims to evaluate the potential of Mthistle to activate thermogenesis in a preclinical model of high-fat diet (HFD)-induced obesity (DIO). RESULTS Our results demonstrate that Mthistle increases systemic energy expenditure (EE), preserves body temperature after cold exposure, improves insulin resistance, and reduces inflammatory markers in WAT. CONCLUSIONS Based on these results, silymarin-enriched extract from Mthistle may be proposed as a nutraceutical for the management of metabolic chronic diseases and/or accelerated aging.
Collapse
Affiliation(s)
- Marina Reguero
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
- NATAC BIOTECH, Electronica 7, E28923 Madrid, Spain
| | - Guillermo Reglero
- Production and Characterization of Novel Foods Department, Institute of Food Science Research CIAL, CEI UAM + CSIC, E28049 Madrid, Spain
| | | | - Ricardo Ramos-Ruiz
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| | - Ana Ramírez de Molina
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| | - Marta Gómez de Cedrón
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
- Cell Metabolism Unit, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| |
Collapse
|
2
|
Zhu S, Liu B, Fu G, Yang L, Wei D, Zhang L, Zhang Q, Gao Y, Sun D, Wei W. PKC-θ is an important driver of fluoride-induced immune imbalance of regulatory T cells/effector T cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 934:173081. [PMID: 38754514 DOI: 10.1016/j.scitotenv.2024.173081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/28/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
Fluoride is unnecessary in the human body. Long-term fluoride exposure may lead to immune system abnormalities. However, the mechanism remains unclear. This study aim to explore the mechanism of fluoride interference in the immune system and also identify the key indicators of fluoride-induced immune damage. Questionnaires were used to collect basic information. Multiple linear analyses and other statistical methods were used in order to process the data. Flow cytometry was used to detect relevant immunomarkers and analyze immune damage. Simultaneously, Wistar rats and cell models exposed to fluoride were established to detect the effects of fluoride on immune homeostasis. The results showed that sex, residence time, smoking, and Corona Virus Disease 2019 (COVID-19) infection may indirectly influence fluoride-induced immune damage. In residents of fluoride-exposed areas, there was a significant decrease in CD3+ T lymphocytes and CD4+ and CD8+ cells and a downward trend in the CD4+/CD8+ cell ratio. CD4+CD8+/CD4+, regulatory T cells (Tregs), and Tregs/effector T cells (Teffs) ratios showed opposite changes. Fluoride inhibits T cell activation by inhibiting the expression and phosphorylation of Protein Kinase C-θ (PKC-θ), hinders the internalization of T cell receptors, and affects NF-kB and c-Jun protein expression, leading to homeostatic Treg/Teff imbalance in vivo and in vitro experiments. This study represents the first evidence suggesting that PKC-θ may be the key to immune imbalance in the body under fluoride exposure. It is possible that Tregs/Teffs cell ratio provide a reference point for the diagnosis and treatment of fluoride-induced immune damage.
Collapse
Affiliation(s)
- Siqi Zhu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Bingshu Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Guiyu Fu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Liu Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Dan Wei
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Liwei Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health Harbin Medical University, Harbin 150081, China
| | - Qiong Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health Harbin Medical University, Harbin 150081, China.
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health Harbin Medical University, Harbin 150081, China.
| | - Wei Wei
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
3
|
Valentine Y, Nikolajczyk BS. T cells in obesity-associated inflammation: The devil is in the details. Immunol Rev 2024; 324:25-41. [PMID: 38767210 DOI: 10.1111/imr.13354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Obesity presents a significant health challenge, affecting 41% of adults and 19.7% of children in the United States. One of the associated health challenges of obesity is chronic low-grade inflammation. In both mice and humans, T cells in circulation and in the adipose tissue play a pivotal role in obesity-associated inflammation. Changes in the numbers and frequency of specific CD4+ Th subsets and their contribution to inflammation through cytokine production indicate declining metabolic health, that is, insulin resistance and T2D. While some Th subset alterations are consistent between mice and humans with obesity, some changes mainly characterize male mice, whereas female mice often resist obesity and inflammation. However, protection from obesity and inflammation is not observed in human females, who can develop obesity-related T-cell inflammation akin to males. The decline in female sex hormones after menopause is also implicated in promoting obesity and inflammation. Age is a second underappreciated factor for defining and regulating obesity-associated inflammation toward translating basic science findings to the clinic. Weight loss in mice and humans, in parallel with these other factors, does not resolve obesity-associated inflammation. Instead, inflammation persists amid modest changes in CD4+ T cell frequencies, highlighting the need for further research into resolving changes in T-cell function after weight loss. How lingering inflammation after weight loss affecting the common struggle to maintain lower weight is unknown. Semaglutide, a newly popular pharmaceutical used for treating T2D and reversing obesity, holds promise for alleviating obesity-associated health complications, yet its impact on T-cell-mediated inflammation remains unexplored. Further work in this area could significantly contribute to the scientific understanding of the impacts of weight loss and sex/hormones in obesity and obesity-associated metabolic decline.
Collapse
Affiliation(s)
- Yolander Valentine
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Barbara S Nikolajczyk
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
4
|
Oliveira BM, Sidónio B, Correia A, Pinto A, Azevedo MM, Sampaio P, Ferreira PG, Vilanova M, Teixeira L. Cytokine production by bovine adipose tissue stromal vascular fraction cells upon Neospora caninum stimulation. Sci Rep 2024; 14:8444. [PMID: 38600105 PMCID: PMC11006870 DOI: 10.1038/s41598-024-58885-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
In bovines few studies addressed the contribution of adipose tissue to the host immune response to infection. Here we evaluated the in vitro response of bovine adipose tissue stromal vascular fraction (SVF) cells to the protozoan parasite Neospora caninum, using live and freeze-killed tachyzoites. Live N. caninum induced the production of IL-6, IL-1β and IL-10 by SVF cells isolated from subcutaneous adipose tissue (SAT), while in mesenteric adipose tissue (MAT) SVF cell cultures only IL-1β and IL-10 production was increased, showing slight distinct responses between adipose tissue depots. Whereas a clear IL-8 increase was detected in peripheral blood leucocytes (PBL) culture supernatants in response to live N. caninum, no such increase was observed in SAT or MAT SVF cell cultures. Nevertheless, in response to LPS, increased IL-8 levels were detected in all cell cultures. IL-10 levels were always increased in response to stimulation (live, freeze-killed N. caninum and LPS). Overall, our results show that bovine adipose tissue SVF cells produce cytokines in response to N. caninum and can therefore be putative contributors to the host immune response against this parasite.
Collapse
Affiliation(s)
- Bárbara M Oliveira
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Beatriz Sidónio
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Alexandra Correia
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
| | - Ana Pinto
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal
| | - Maria M Azevedo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Paula Sampaio
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Paula G Ferreira
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal
| | - Manuel Vilanova
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
| | - Luzia Teixeira
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal.
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal.
| |
Collapse
|
5
|
Soedono S, Sharlene S, Vo DHN, Averia M, Rosalie EE, Lee YK, Cho KW. Obese visceral adipose dendritic cells downregulate regulatory T cell development through IL-33. Front Immunol 2024; 15:1335651. [PMID: 38566998 PMCID: PMC10985834 DOI: 10.3389/fimmu.2024.1335651] [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: 11/09/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
Regulatory T cells (Tregs) residing in visceral adipose tissue (VAT) play a pivotal role in regulating tissue inflammation and metabolic dysfunction associated with obesity. However, the specific phenotypic and functional characteristics of Tregs in obese VAT, as well as the regulatory mechanisms shaping them, remain elusive. This study demonstrates that obesity selectively reduces Tregs in VAT, characterized by restrained proliferation, heightened PD-1 expression, and diminished ST2 expression. Additionally, obese VAT displays distinctive maturation of dendritic cells (DCs), marked by elevated expressions of MHC-II, CD86, and PD-L1, which are inversely correlated with VAT Tregs. In an in vitro co-culture experiment, only obese VAT DCs, not macrophages or DCs from subcutaneous adipose tissue (SAT) and spleen, result in decreased Treg differentiation and proliferation. Furthermore, Tregs differentiated by obese VAT DCs exhibit distinct characteristics resembling those of Tregs in obese VAT, such as reduced ST2 and IL-10 expression. Mechanistically, obesity lowers IL-33 production in VAT DCs, contributing to the diminished Treg differentiation. These findings collectively underscore the critical role of VAT DCs in modulating Treg generation and shaping Treg phenotype and function during obesity, potentially contributing to the regulation of VAT Treg populations.
Collapse
Affiliation(s)
- Shindy Soedono
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Sharlene Sharlene
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Dan Hoang Nguyet Vo
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Maria Averia
- Magister of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Eufrasia Elaine Rosalie
- Faculty of Biotechnology, Department of Food Technology, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Kae Won Cho
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| |
Collapse
|
6
|
Apaza CJ, Días M, García Tejedor A, Boscá L, Laparra Llopis JM. Contribution of Nucleotide-Binding Oligomerization Domain-like (NOD) Receptors to the Immune and Metabolic Health. Biomedicines 2024; 12:341. [PMID: 38397943 PMCID: PMC10886542 DOI: 10.3390/biomedicines12020341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Nucleotide-binding oligomerization domain-like (NOD) receptors rely on the interface between immunity and metabolism. Dietary factors constitute critical players in the activation of innate immunity and modulation of the gut microbiota. The latter have been involved in worsening or improving the control and promotion of diseases such as obesity, type 2 diabetes, metabolic syndrome, diseases known as non-communicable metabolic diseases (NCDs), and the risk of developing cancer. Intracellular NODs play key coordinated actions with innate immune 'Toll-like' receptors leading to a diverse array of gene expressions that initiate inflammatory and immune responses. There has been an improvement in the understanding of the molecular and genetic implications of these receptors in, among others, such aspects as resting energy expenditure, insulin resistance, and cell proliferation. Genetic factors and polymorphisms of the receptors are determinants of the risk and severity of NCDs and cancer, and it is conceivable that dietary factors may have significant differential consequences depending on them. Host factors are difficult to influence, while environmental factors are predominant and approachable with a preventive and/or therapeutic intention in obesity, T2D, and cancer. However, beyond the recognition of the activation of NODs by peptidoglycan as its prototypical agonist, the underlying molecular response(s) and its consequences on these diseases remain ill-defined. Metabolic (re)programming is a hallmark of NCDs and cancer in which nutritional strategies might play a key role in preventing the unprecedented expansion of these diseases. A better understanding of the participation and effects of immunonutritional dietary ingredients can boost integrative knowledge fostering interdisciplinary science between nutritional precision and personalized medicine against cancer. This review summarizes the current evidence concerning the relationship(s) and consequences of NODs on immune and metabolic health.
Collapse
Affiliation(s)
- César Jeri Apaza
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra Cantoblanco, 8, 28049 Madrid, Spain;
| | - Marisol Días
- Center of Biological Enginneering (CEB), Iberian Nantotechnology Laboratory (INL), University of Minho, 4715-330 Braga, Portugal;
| | - Aurora García Tejedor
- Bioactivity and Nutritional Immunology Group (BIOINUT), Faculty of Health Sciences, Universidad Internacional de Valencia (VIU), Pintor Sorolla 21, 46002 Valencia, Spain;
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols-Morreale (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029 Madrid, Spain
| | - José Moisés Laparra Llopis
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra Cantoblanco, 8, 28049 Madrid, Spain;
| |
Collapse
|
7
|
Liu Y, Hu G, Jia Y, Qin L, Xu L, Chang Y, Li B, Li H. Wnt10b knockdown regulates the relative balance of adipose tissue-resident T cells and inhibits white fat deposition. Mol Biol Rep 2024; 51:272. [PMID: 38302806 DOI: 10.1007/s11033-024-09249-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/12/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Wnt10b is one of critical Wnt family members that being involved in networks controlling stemness, pluripotency and cell fate decisions. However, its role in adipose-resident T lymphocytes and further in fat metabolism yet remains largely unknown. METHODS AND RESULTS In the present study, we demonstrated a distinctive effect for Wnt10b on the relative balance of T lymphocytes in adipose tissue by using a Wnt10b knockdown mouse model. Wnt10b knockdown led to a reduction of adipose-resident CD4+ T cells and an elevation of Foxp3+/CD4+ Treg cells. Wnt10b-knockdown mice fed with standard diet showed less white fat deposition owing to the suppressed adipogenic process. Moreover, under high fat diet conditions, Wnt10b knockdown resulted in an alleviated obesity symptoms, as well as an improvement of glucose homeostasis and hepatic steatosis. CONCLUSIONS Collectively, we reveal an unexpected and novel function for Wnt10b in mediating the frequency of adipose-resident T cell subsets, that when knockdown skewing toward a Treg-dominated phenotype and further improving fat metabolism.
Collapse
Affiliation(s)
- Yan Liu
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Geng Hu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Yanxin Jia
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Lining Qin
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Longfei Xu
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Yaxin Chang
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Bin Li
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Haifang Li
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China.
| |
Collapse
|
8
|
Gao F, Litchfield B, Wu H. Adipose tissue lymphocytes and obesity. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:5. [PMID: 38455510 PMCID: PMC10919906 DOI: 10.20517/jca.2023.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Obesity is associated with chronic inflammation in adipose tissue (AT), mainly evidenced by infiltration and phenotypic changes of various types of immune cells. Macrophages are the major innate immune cells and represent the predominant immune cell population within AT. Lymphocytes, including T cells and B cells, are adaptive immune cells and constitute another important immune cell population in AT. In obesity, CD8+ effector memory T cells, CD4+ Th1 cells, and B2 cells are increased in AT and promote AT inflammation, while regulatory T cells and Th2 cells, which usually function as immune regulatory or type 2 inflammatory cells, are reduced in AT. Immune cells may regulate the metabolism of adipocytes and other cells through various mechanisms, contributing to the development of metabolic diseases, including insulin resistance and type 2 diabetes. Efforts targeting immune cells and inflammation to prevent and treat obesity-linked metabolic disease have been explored, but have not yielded significant success in clinical studies. This review provides a concise overview of the changes in lymphocyte populations within AT and their potential role in AT inflammation and the regulation of metabolic functions in the context of obesity.
Collapse
Affiliation(s)
- Feng Gao
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
9
|
Śnieżewska A, Anisiewicz A, Gdesz-Birula K, Wietrzyk J, Filip-Psurska B. Age-Dependent Effect of Calcitriol on Mouse Regulatory T and B Lymphocytes. Nutrients 2023; 16:49. [PMID: 38201878 PMCID: PMC10780377 DOI: 10.3390/nu16010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
The hormonally active vitamin D3 metabolite, calcitriol, functions as an important modulator of the immune system. We assumed that calcitriol exerts different effects on immune cells and cytokine production, depending on the age of the animal; therefore, we analyzed its effects on regulatory T lymphocytes and regulatory B lymphocytes in healthy young and old female C57Bl/6/Foxp3GFP mice. In the lymph nodes of young mice, calcitriol decreased the percentage of Tregs, including tTregs and pTregs, and the expression of GITR, CD103, and CD101; however, calcitriol increased the level of IL-35 in adipose tissue. In the case of aged mice, calcitriol decreased the percentages of tTregs and CD19+ cells in lymph nodes and the level of osteopontin in the plasma. Additionally, increases in the levels of IgG and the lowest levels of IFN-γ, IL-10, and IL-35 were observed in the adipose tissue of aged mice. This study showed that calcitriol treatment had different effects, mainly on Treg phenotypes and cytokine secretion, in young and old female mice; it seemed that calcitriol enhanced the immunosuppressive properties of the lymphatic organs and adipose tissue of healthy young mice but not of healthy aged mice, where the opposite effects were observed.
Collapse
Affiliation(s)
| | | | | | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.Ś.); (A.A.); (K.G.-B.)
| | - Beata Filip-Psurska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.Ś.); (A.A.); (K.G.-B.)
| |
Collapse
|
10
|
Mukherjee S, Skrede S, Haugstøyl M, López M, Fernø J. Peripheral and central macrophages in obesity. Front Endocrinol (Lausanne) 2023; 14:1232171. [PMID: 37720534 PMCID: PMC10501731 DOI: 10.3389/fendo.2023.1232171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/28/2023] [Indexed: 09/19/2023] Open
Abstract
Obesity is associated with chronic, low-grade inflammation. Excessive nutrient intake causes adipose tissue expansion, which may in turn cause cellular stress that triggers infiltration of pro-inflammatory immune cells from the circulation as well as activation of cells that are residing in the adipose tissue. In particular, the adipose tissue macrophages (ATMs) are important in the pathogenesis of obesity. A pro-inflammatory activation is also found in other organs which are important for energy metabolism, such as the liver, muscle and the pancreas, which may stimulate the development of obesity-related co-morbidities, including insulin resistance, type 2 diabetes (T2D), cardiovascular disease (CVD) and non-alcoholic fatty liver disease (NAFLD). Interestingly, it is now clear that obesity-induced pro-inflammatory signaling also occurs in the central nervous system (CNS), and that pro-inflammatory activation of immune cells in the brain may be involved in appetite dysregulation and metabolic disturbances in obesity. More recently, it has become evident that microglia, the resident macrophages of the CNS that drive neuroinflammation, may also be activated in obesity and can be relevant for regulation of hypothalamic feeding circuits. In this review, we focus on the action of peripheral and central macrophages and their potential roles in metabolic disease, and how macrophages interact with other immune cells to promote inflammation during obesity.
Collapse
Affiliation(s)
- Sayani Mukherjee
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Silje Skrede
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Martha Haugstøyl
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Johan Fernø
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
11
|
Ni H, Chen Y. Differentiation, regulation and function of regulatory T cells in non-lymphoid tissues and tumors. Int Immunopharmacol 2023; 121:110429. [PMID: 37327512 DOI: 10.1016/j.intimp.2023.110429] [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: 03/03/2023] [Revised: 05/22/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023]
Abstract
Regulatory T cells (Tregs) play a substantial role in inhibiting excessive immune response. A large number of studies have focused on the tissue homeostasis maintenance and remodeling characteristics of Tregs in non-lymphoid tissues, such as the skin, colon, lung, brain, muscle, and adipose tissues. Herein, we overview the kinetics of Treg migration to non-lymphoid tissues and adaptation to the specific tissue microenvironment through the development of tissue-specific chemokine receptors, transcription factors, and phenotypes. Additionally, tumor-infiltrating Tregs (Ti-Tregs) play an important role in tumor generation and immunotherapy resistance. The phenotypes of Ti-Tregs are related to the histological location of the tumor and there is a large overlap between the transcripts of Ti-Tregs and those of tissue-specific Tregs. We recapitulate the molecular underpinnings of tissue-specific Tregs, which might shed new light on Treg-based therapeutic targets and biomarkers for inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Hongbo Ni
- The First Clinical Medicine Faculty, China Medical University, Shenyang 110001, China
| | - Yinghan Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
12
|
Camacho-Muñoz D, Niven J, Kucuk S, Cucchi D, Certo M, Jones SW, Fischer DP, Mauro C, Nicolaou A. Omega-3 polyunsaturated fatty acids reverse the impact of western diets on regulatory T cell responses through averting ceramide-mediated pathways. Biochem Pharmacol 2022; 204:115211. [PMID: 35985403 DOI: 10.1016/j.bcp.2022.115211] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/02/2022]
Abstract
Western diet (WD), high in sugar and fat, promotes obesity and associated chronic low-grade pro-inflammatory environment, leading to impaired immune function, reprogramming of innate and adaptive immune cells, and development of chronic degenerative diseases, including cardiovascular disease. Increased concentrations of circulating and tissue ceramides contribute to inflammation and cellular dysfunction common in immune metabolic and cardiometabolic disease. Therefore, ceramide-lowering interventions have been considered as strategies to improve adipose tissue health. Here, we report the ability of omega-3 polyunsaturated fatty acids (n-3PUFA) to attenuate inflammatory phenotypes promoted by WD, through ceramide-dependent pathways. Using an animal model, we show that enrichment of WD diet with n-3PUFA, reduced the expression of ceramide synthase 2 (CerS2), and lowered the concentration of long-chain ceramides (C23-C26) in plasma and adipose tissues. N-3PUFA also increased prevalence of the anti-inflammatory CD4+Foxp3+ and CD4+Foxp3+CD25+ Treg subtypes in lymphoid organs. The CerS inhibitor FTY720 mirrored the effect of n-3PUFA. Treatment of animal and human T cells with ceramide C24 in vitro, reduced CD4+Foxp3+ Treg polarisation and IL-10 production, and increased IL-17, while it decreased Erk and Akt phosphorylation downstream of T cell antigen receptors (TCR). These findings suggest that molecular mechanisms mediating the adverse effect of ceramides on regulatory T lymphocytes, progress through reduced TCR signalling. Our findings suggest that nutritional enrichment of WD with fish oil n-3PUFA can partially mitigate its detrimental effects, potentially improving the low-grade inflammation associated with immune metabolic disease. Compared to pharmacological interventions, n-3PUFA offer a simpler approach that can be accommodated as lifestyle choice.
Collapse
Affiliation(s)
- Dolores Camacho-Muñoz
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Jennifer Niven
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK
| | - Salih Kucuk
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK
| | - Danilo Cucchi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Michelangelo Certo
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK
| | - Simon W Jones
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK
| | - Deborah P Fischer
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK; William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Anna Nicolaou
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, UK.
| |
Collapse
|
13
|
The Mitigatory Effect of Shen-Qi Compound on the Diabetic Thoracic Aortic Complications through Inhibiting the Inflammatory Microenvironment by miR-223-3p/RBP-J/IRF8 Axis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6686931. [PMID: 36212957 PMCID: PMC9534610 DOI: 10.1155/2022/6686931] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/18/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022]
Abstract
Background Disruption of the vascular immunological inflammatory microenvironment is linked to metabolic memory impairment. Even though it has been proven that the Shen-Qi compound (SQC) can efficiently halt metabolic memory and preserve vascular endothelial cells, extensive studies need to be done to investigate if it can also change the vascular immune-inflammatory microenvironment by regulating the immune system. This will help figure out the role of stopping metabolic memory. Methods After 4 weeks on a high-fat diet (HFD), GK rats were used to create a model for diabetic thoracic aortic problems. The effect and mechanisms of SQC on diabetic thoracic aortic complications were assessed by hematoxylin-eosin (H&E) staining, enzyme-linked immunosorbent assay (ELISA), biochemical analysis, terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling (TUNEL), reverse transcription, real-time polymerase chain reaction (RT-qPCR), immunofluorescence (IF), western blot, and luciferase reporter assays. Results SQC treatment ameliorates the HFD-induced pathological symptoms as well as the HFD-induced increased concentrations of fasting blood glucose (FBG), fasting insulin (FINS), total cholesterol (TC), triglycerides (TGs), and low-density lipoprotein cholesterol (LDL-C) and decreased concentrations of high-density lipoprotein cholesterol (HDL-C). Besides, SQC counteracted the HFD-induced average fluorescence intensity of vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1), as well as the concentrations of endothelin-1 (ET-1) and monocyte chemoattractant protein-1 (MCP-1), while rescuing the HFD-induced concentrations of nitric oxide (NO) and nitric oxide synthetase (NOS). Also, SQC decreases apoptosis and oxidative stress in rats with diabetic thoracic aortic complications. In addition, SQC facilitated the polarization of macrophages, stimulated the activation of dendritic cells, and regulated the inflammatory milieu in rats with diabetic thoracic aortic complications. Furthermore, SQC also modulated the miR-223-3p/RBP-J/IRF8 axis in the macrophages of rats with diabetic thoracic aortic complications. Conclusion SQC ameliorated diabetic thoracic aortic complications through the regulation of apoptosis, oxidative stress, and inflammatory microenvironment mediating by the miR-223-3p/RBP-J/IRF8 axis.
Collapse
|
14
|
Horckmans M, Diaz Villamil E, Bianchini M, De Roeck L, Communi D. Central role of PD-L1 in cardioprotection resulting from P2Y4 nucleotide receptor loss. Front Immunol 2022; 13:1006934. [PMID: 36248854 PMCID: PMC9555169 DOI: 10.3389/fimmu.2022.1006934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
A better understanding of the immune function of pericardial adipose tissue is essential to adapt treatments after myocardial infarction. We showed previously that inactivation of mouse P2Y4 nucleotide receptor induces adiponectin overexpression and protection against myocardial infarction. We investigated here the inflammatory state of pericardial adipose tissue in ischemic P2Y4-deficient mice. We demonstrated that P2Y4-deficient mice displayed adipocyte beiging with increased PD-L1 expression and a higher number of regulatory leukocytes in their pericardial adipose tissue after left anterior descending artery ligation, compared to wild type mice. Effectively, a higher level of anti-inflammatory M2c macrophages and regulatory T cells was observed in pericardial adipose tissue of P2Y4 KO mice and correlated with reduced post-ischemic expansion of fat-associated lymphoid clusters. Interestingly, the anti-inflammatory effects observed in P2Y4 KO mice, were no more observed in P2Y4/adiponectin double KO ischemic mice. Finally, the reduction of T cell infiltration and cardiac fibrosis observed in P2Y4-deficient heart was lost after injection of anti-PD-L1 blocking antibody in ischemic mice. The present study defines P2Y4 as a regulator of PD-L1 and adiponectin, and as a potential target for anti-inflammatory therapies to improve myocardial infarction outcome. The combined effect of P2Y4 loss on adipocyte beiging and regulatory leukocyte increase highlights this nucleotide receptor as an important player in post-ischemic cardiac response.
Collapse
Affiliation(s)
- Michael Horckmans
- Institute of Interdisciplinary Research, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Free University of Brussels, Brussels, Belgium
| | - Esteban Diaz Villamil
- Institute of Interdisciplinary Research, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Free University of Brussels, Brussels, Belgium
| | - Mariaelvy Bianchini
- Institute for Cardio-vascular Prevention, Ludwig-Maximilians-Universität (LMU), Munich, German
| | - Lucas De Roeck
- Institute of Interdisciplinary Research, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Free University of Brussels, Brussels, Belgium
| | - Didier Communi
- Institute of Interdisciplinary Research, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Free University of Brussels, Brussels, Belgium
- *Correspondence: Didier Communi,
| |
Collapse
|
15
|
Bradley D, Smith AJ, Blaszczak A, Shantaram D, Bergin SM, Jalilvand A, Wright V, Wyne KL, Dewal RS, Baer LA, Wright KR, Stanford KI, Needleman B, Brethauer S, Noria S, Renton D, Joseph JJ, Lovett-Racke A, Liu J, Hsueh WA. Interferon gamma mediates the reduction of adipose tissue regulatory T cells in human obesity. Nat Commun 2022; 13:5606. [PMID: 36153324 PMCID: PMC9509397 DOI: 10.1038/s41467-022-33067-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
Decreased adipose tissue regulatory T cells contribute to insulin resistance in obese mice, however, little is known about the mechanisms regulating adipose tissue regulatory T cells numbers in humans. Here we obtain adipose tissue from obese and lean volunteers. Regulatory T cell abundance is lower in obese vs. lean visceral and subcutaneous adipose tissue and associates with reduced insulin sensitivity and altered adipocyte metabolic gene expression. Regulatory T cells numbers decline following high-fat diet induction in lean volunteers. We see alteration in major histocompatibility complex II pathway in adipocytes from obese patients and after high fat ingestion, which increases T helper 1 cell numbers and decreases regulatory T cell differentiation. We also observe increased expression of inhibitory co-receptors including programmed cell death protein 1 and OX40 in visceral adipose tissue regulatory T cells from patients with obesity. In human obesity, these global effects of interferon gamma to reduce regulatory T cells and diminish their function appear to instigate adipose inflammation and suppress adipocyte metabolism, leading to insulin resistance.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
- Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Alan J Smith
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Alecia Blaszczak
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Dharti Shantaram
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Stephen M Bergin
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Anahita Jalilvand
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Valerie Wright
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kathleen L Wyne
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Revati S Dewal
- Diabetes and Metabolism Research Center, Division of Physiology and Cell Biology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Lisa A Baer
- Diabetes and Metabolism Research Center, Division of Physiology and Cell Biology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Katherine R Wright
- Diabetes and Metabolism Research Center, Division of Physiology and Cell Biology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kristin I Stanford
- Diabetes and Metabolism Research Center, Division of Physiology and Cell Biology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Bradley Needleman
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Stacy Brethauer
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Sabrena Noria
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - David Renton
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Joshua J Joseph
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Amy Lovett-Racke
- Department of Microbial Immunity and Infection, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Joey Liu
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Willa A Hsueh
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
16
|
de Wit S, Glen C, de Boer RA, Lang NN. Mechanisms shared between cancer, heart failure, and targeted anti-cancer therapies. Cardiovasc Res 2022; 118:3451-3466. [PMID: 36004495 PMCID: PMC9897696 DOI: 10.1093/cvr/cvac132] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/12/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023] Open
Abstract
Heart failure (HF) and cancer are the leading causes of death worldwide and accumulating evidence demonstrates that HF and cancer affect one another in a bidirectional way. Patients with HF are at increased risk for developing cancer, and HF is associated with accelerated tumour growth. The presence of malignancy may induce systemic metabolic, inflammatory, and microbial alterations resulting in impaired cardiac function. In addition to pathophysiologic mechanisms that are shared between cancer and HF, overlaps also exist between pathways required for normal cardiac physiology and for tumour growth. Therefore, these overlaps may also explain the increased risk for cardiotoxicity and HF as a result of targeted anti-cancer therapies. This review provides an overview of mechanisms involved in the bidirectional connection between HF and cancer, specifically focusing upon current 'hot-topics' in these shared mechanisms. It subsequently describes targeted anti-cancer therapies with cardiotoxic potential as a result of overlap between their anti-cancer targets and pathways required for normal cardiac function.
Collapse
Affiliation(s)
- Sanne de Wit
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, PO Box 30.001, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Claire Glen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, PO Box 30.001, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | | |
Collapse
|
17
|
Abstract
Adipose tissue is a complex dynamic organ with whole-body immunometabolic influence. Much of the work into understanding the role of immune cells in adipose tissue has been in the context of obesity. These investigations have also uncovered a range of typical (immune) and non-typical functions exerted by adipose tissue leukocytes. Here we provide an overview of the adipose tissue immune system, including its role as an immune reservoir in the whole-body response to infection and as a site of parasitic and viral infections. We also describe the functional roles of specialized immunological structures found within adipose tissue. However, our main focus is on the recently discovered 'non-immune' functions of adipose tissue immune cells, which include the regulation of adipocyte homeostasis, as well as responses to changing nutrient status and body temperature. In doing so, we outline the therapeutic potential of the adipose tissue immune system in health and disease.
Collapse
|
18
|
Zhang Q, Wang S, Ji S. Trifolirhizin regulates the balance of Th17/Treg cells and inflammation in the ulcerative colitis mice through inhibiting the TXNIP-mediated activation of NLRP3 inflammasome. Clin Exp Pharmacol Physiol 2022; 49:787-796. [PMID: 35575951 DOI: 10.1111/1440-1681.13654] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 11/27/2022]
Abstract
Ulcerative colitis (UC) is a chronic and recurrent autoimmune disease, characterized by recurrence and remission of mucosal inflammation. Although the understanding of the pathogenesis of UC has been improved, effective therapeutic drugs are required for treating patients with UC. In current work, the mouse model of colitis was established. Trifolirhizin was demonstrated to improve symptom in dextran sulfate sodium (DSS)-induced colitis mice. The body weight of mice was elevated, while the disease activity index (DAI) was reduced. Moreover, Trifolirhizin was involved in inhibition of inflammation and regulation of Th17/Treg cell balance in DSS-induced colitis mice. Further, the activation NLRP3 inflammasome was suppressed by Trifolirhizin in DSS-induced colitis mice. Trifolirhizin was also identified to regulate AMPK-TXNIP pathway. The Trifolirhizin-mediated anti-inflammatory effect was inhibited by suppressing AMPK in DSS-induced UC mice. In summary, the research suggested that administration of Trifolirhizin significantly improved the symptoms and the pathological damage in DSS-induced UC mice. Trifolirhizin regulated the balance of Th17/Treg cells and inflammation in the UC mice through inhibiting the TXNIP-mediated activation of NLRP3 inflammasome. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Digestive Internal Medicine, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu, China
| | - Shufang Wang
- Department of Digestive Internal Medicine, Liayunngang Second People's Hospital, Lianyungang, Jiangsu, China
| | - Shanyun Ji
- Department of Digestive Internal Medicine, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu, China
| |
Collapse
|
19
|
Chen H, Sun L, Feng L, Yin Y, Zhang W. Role of Innate lymphoid Cells in Obesity and Insulin Resistance. Front Endocrinol (Lausanne) 2022; 13:855197. [PMID: 35574038 PMCID: PMC9091334 DOI: 10.3389/fendo.2022.855197] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity, a growing chronic metabolic disease, greatly increases the risk of metabolic syndrome which includes type 2 diabetes, fatty liver and cardiovascular diseases. Obesity-associated metabolic diseases significantly contribute to mortality and reduce life expectancy. Recently, innate lymphoid cells (ILCs) have emerged as crucial regulators of metabolic homeostasis and tissue inflammation. This review focuses on the roles of ILCs in different metabolic tissues, including adipose tissue, liver, pancreas, and intestine. We briefly outline the relationship between obesity, inflammation, and insulin resistance. We then discuss how ILCs in distinct metabolic organs may function to maintain metabolic homeostasis and contribute to obesity and its associated metabolic diseases. The potential of ILCs as the therapeutic target for obesity and insulin resistance is also addressed.
Collapse
Affiliation(s)
- Hong Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Lu Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
- *Correspondence: Weizhen Zhang, ; Yue Yin,
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, United States
- *Correspondence: Weizhen Zhang, ; Yue Yin,
| |
Collapse
|
20
|
Dahlquist KJ, Camell CD. Aging Leukocytes and the Inflammatory Microenvironment of the Adipose Tissue. Diabetes 2022; 71:23-30. [PMID: 34995348 PMCID: PMC8763870 DOI: 10.2337/dbi21-0013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/18/2021] [Indexed: 01/03/2023]
Abstract
Age-related immunosenescence, defined as an increase in inflammaging and the decline of the immune system, leads to tissue dysfunction and increased risk for metabolic disease. The elderly population is expanding, leading to a heightened need for therapeutics to improve health span. With age, many alterations of the immune system are observed, including shifts in the tissue-resident immune cells, increased expression of inflammatory factors, and the accumulation of senescent cells, all of which are responsible for a chronic inflammatory loop. Adipose tissue and the immune cell activation within are of particular interest for their well-known roles in metabolic disease. Recent literature reveals that adipose tissue is an organ in which signs of initial aging occur, including immune cell activation. Aged adipose tissue reveals changes in many innate and adaptive immune cell subsets, revealing a complex interaction that contributes to inflammation, increased senescence, impaired catecholamine-induced lipolysis, and impaired insulin sensitivity. Here, we will describe current knowledge surrounding age-related changes in immune cells while relating those findings to recent discoveries regarding immune cells in aged adipose tissue.
Collapse
Affiliation(s)
| | - Christina D. Camell
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN
| |
Collapse
|
21
|
Tomás-Cortázar J, Bossi L, Quinn C, Reynolds CJ, Butler DK, Corcoran N, Murchú MÓ, McMahon E, Singh M, Rongkard P, Anguita J, Blanco A, Dunachie SJ, Altmann D, Boyton RJ, Arnold J, Giltaire S, McClean S. BpOmpW Antigen Stimulates the Necessary Protective T-Cell Responses Against Melioidosis. Front Immunol 2021; 12:767359. [PMID: 34966388 PMCID: PMC8710444 DOI: 10.3389/fimmu.2021.767359] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/19/2021] [Indexed: 11/24/2022] Open
Abstract
Melioidosis is a potentially fatal bacterial disease caused by Burkholderia pseudomallei and is estimated to cause 89,000 deaths per year in endemic areas of Southeast Asia and Northern Australia. People with diabetes mellitus are most at risk of melioidosis, with a 12-fold increased susceptibility for severe disease. Interferon gamma (IFN-γ) responses from CD4 and CD8 T cells, but also from natural killer (NK) and natural killer T (NKT) cells, are necessary to eliminate the pathogen. We previously reported that immunization with B. pseudomallei OmpW (BpOmpW antigen) protected mice from lethal B. pseudomallei challenge for up to 81 days. Elucidating the immune correlates of protection of the protective BpOmpW vaccine is an essential step prior to clinical trials. Thus, we immunized either non-insulin-resistant C57BL/6J mice or an insulin-resistant C57BL/6J mouse model of type 2 diabetes (T2D) with a single dose of BpOmpW. BpOmpW induced strong antibody responses, stimulated effector CD4+ and CD8+ T cells and CD4+ CD25+ Foxp3+ regulatory T cells, and produced higher IFN-γ responses in CD4+, CD8+, NK, and NKT cells in non-insulin-resistant mice. The T-cell responses of insulin-resistant mice to BpOmpW were comparable to those of non-insulin-resistant mice. In addition, as a precursor to its evaluation in human studies, humanized HLA-DR and HLA-DQ (human leukocyte antigen DR and DQ isotypes, respectively) transgenic mice elicited IFN-γ recall responses in an enzyme-linked immune absorbent spot (ELISpot)-based study. Moreover, human donor peripheral blood mononuclear cells (PBMCs) exposed to BpOmpW for 7 days showed T-cell proliferation. Finally, plasma from melioidosis survivors with diabetes recognized our BpOmpW vaccine antigen. Overall, the range of approaches used strongly indicated that BpOmpW elicits the necessary immune responses to combat melioidosis and bring this vaccine closer to clinical trials.
Collapse
MESH Headings
- Animals
- Antigens, Bacterial/immunology
- Bacterial Outer Membrane Proteins/immunology
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/immunology
- Burkholderia pseudomallei/immunology
- Burkholderia pseudomallei/metabolism
- Burkholderia pseudomallei/physiology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/microbiology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/microbiology
- Cells, Cultured
- Diabetes Mellitus, Type 2/immunology
- Humans
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/microbiology
- Male
- Melioidosis/immunology
- Melioidosis/microbiology
- Melioidosis/prevention & control
- Mice, Inbred C57BL
- Mice, Transgenic
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/microbiology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/microbiology
- Mice
Collapse
Affiliation(s)
- Julen Tomás-Cortázar
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Lorenzo Bossi
- Immunxperts SA, a Nexelis Company, Gosselies, Belgium
| | - Conor Quinn
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Catherine J. Reynolds
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Lung Division, Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - David K. Butler
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Lung Division, Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Niamh Corcoran
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Maitiú Ó Murchú
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Eve McMahon
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Mahavir Singh
- LIONEX Diagnostics and Therapeutics GmbH, Brunswick, Germany
| | - Patpong Rongkard
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
- Oxford Centre for Global Health Research, University of Oxford, Oxford, United Kingdom
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Lab, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Alfonso Blanco
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Susanna J. Dunachie
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
- Oxford Centre for Global Health Research, University of Oxford, Oxford, United Kingdom
| | - Daniel Altmann
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Lung Division, Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Rosemary J. Boyton
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Lung Division, Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Johan Arnold
- Immunxperts SA, a Nexelis Company, Gosselies, Belgium
| | | | - Siobhán McClean
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
22
|
Gao Z, Xu X, Li Y, Sun K, Yang M, Zhang Q, Wang S, Lin Y, Lou L, Wu A, Liu W, Nie B. Mechanistic Insight into PPARγ and Tregs in Atherosclerotic Immune Inflammation. Front Pharmacol 2021; 12:750078. [PMID: 34658891 PMCID: PMC8511522 DOI: 10.3389/fphar.2021.750078] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/17/2021] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis (AS) is the main pathological cause of acute cardiovascular and cerebrovascular diseases, such as acute myocardial infarction and cerebral apoplexy. As an immune-mediated inflammatory disease, the pathogenesis of AS involves endothelial cell dysfunction, lipid accumulation, foam cell formation, vascular smooth muscle cell (VSMC) migration, and inflammatory factor infiltration. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) plays an important role in lipid metabolism, inflammation, and apoptosis by antagonizing the Wnt/β-catenin pathway and regulating cholesterol efflux and inflammatory factors. Importantly, PPARγ-dependant fatty acid uptake is critical for metabolic programming. Activated PPARγ can exert an anti-atherosclerotic effect by inhibiting the expression of various inflammatory factors, improving endothelial cell function, and restraining the proliferation and migration of VSMCs. Regulatory T cells (Tregs) are the only subset of T lymphocytes that have a completely negative regulatory effect on the autoimmune response. They play a critical role in suppressing excessive immune responses and inflammatory reactions and widely affect AS-associated foam cell formation, plaque rupture, and other processes. Recent studies have shown that PPARγ activation promotes the recruitment of Tregs to reduce inflammation, thereby exerting its anti-atherosclerotic effect. In this review, we provide an overview of the anti-AS roles of PPARγ and Tregs by discussing their pathological mechanisms from the perspective of AS and immune-mediated inflammation, with a focus on basic research and clinical trials of their efficacies alone or in combination in inhibiting atherosclerotic inflammation. Additionally, we explore new ideas for AS treatment and plaque stabilization and establish a foundation for the development of natural PPARγ agonists with Treg recruitment capability.
Collapse
Affiliation(s)
- Zhao Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China.,Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Institute of Nephrology, Guangdong Medical University, Zhanjiang, China
| | - Xinrui Xu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Yang Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kehan Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Manfang Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingyue Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Shuqi Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yiyi Lin
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Lixia Lou
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Aiming Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China.,Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Institute of Nephrology, Guangdong Medical University, Zhanjiang, China
| | - Bo Nie
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to BeijingUniversity of Chinese Medicine, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
23
|
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
|
24
|
Wan M, Han J, Ding L, Hu F, Gao P. Novel Immune Subsets and Related Cytokines: Emerging Players in the Progression of Liver Fibrosis. Front Med (Lausanne) 2021; 8:604894. [PMID: 33869241 PMCID: PMC8047058 DOI: 10.3389/fmed.2021.604894] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a pathological process caused by persistent chronic injury of the liver. Kupffer cells, natural killer (NK) cells, NKT cells, and dendritic cells (DCs), which are in close contact with T and B cells, serve to bridge innate and adaptive immunity in the liver. Meanwhile, an imbalanced inflammatory response constitutes a challenge in liver disease. The dichotomous roles of novel immune cells, including T helper 17 (Th17), regulatory T cells (Tregs), mucosa-associated invariant T cells (MAIT), and innate lymphoid cells (ILCs) in liver fibrosis have gradually been revealed. These cells not only induce damage during liver fibrosis but also promote tissue repair. Hence, immune cells have unique, and often opposing, roles during the various stages of fibrosis. Due to this heterogeneity, the treatment, or reversal of fibrosis through the target of immune cells have attracted much attention. Moreover, activation of hepatic stellate cells (HSCs) constitutes the core of fibrosis. This activation is regulated by various immune mediators, including Th17, Th22, and Th9, MAIT, ILCs, and γδ T cells, as well as their related cytokines. Thus, liver fibrosis results from the complex interaction of these immune mediators, thereby complicating the ability to elucidate the mechanisms of action elicited by each cell type. Future developments in biotechnology will certainly aid in this feat to inform the design of novel therapeutic targets. Therefore, the aim of this review was to summarize the role of specific immune cells in liver fibrosis, as well as biomarkers and treatment methods related to these cells.
Collapse
Affiliation(s)
- Minjie Wan
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China.,Central Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jiawen Han
- Central Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Lili Ding
- Central Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China.,Intensive Care Unit, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Feng Hu
- Department of Hepatology and Gastroenterology, The Second Part of First Hospital, Jilin University, Changchun, China
| | - Pujun Gao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
25
|
Wittwer J, Bradley D. Clusterin and Its Role in Insulin Resistance and the Cardiometabolic Syndrome. Front Immunol 2021; 12:612496. [PMID: 33717095 PMCID: PMC7946829 DOI: 10.3389/fimmu.2021.612496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
The cardiometabolic syndrome involves a clustering of metabolic and cardiovascular factors which increase the risk of patients developing both Type 2 Diabetes Mellitus and cardio/cerebrovascular disease. Although the mechanistic underpinnings of this link remain uncertain, key factors include insulin resistance, excess visceral adiposity, atherogenic dyslipidemia, and endothelial dysfunction. Of these, a state of resistance to insulin action in overweight/obese patients appears to be central to the pathophysiologic process. Given the increasing prevalence of obesity-related Type 2 Diabetes, coupled with the fact that cardiovascular disease is the number one cause of mortality in this patient population, a more thorough understanding of the cardiometabolic syndrome and potential options to mitigate its risk is imperative. Inherent in the pathogenesis of insulin resistance is an underlying state of chronic inflammation, at least partly in response to excess adiposity. Within obese adipose tissue, an immunomodulatory shift occurs, involving a preponderance of pro-inflammatory immune cells and cytokines/adipokines, along with antigen presentation by adipocytes. Therefore, various adipokines differentially expressed by obese adipocytes may have a significant effect on cardiometabolism. Clusterin is a molecular chaperone that is widely produced by many tissues throughout the body, but is also preferentially overexpressed by obese compared lean adipocytes and relates strongly to multiple components of the cardiometabolic syndrome. Herein, we summarize the known and potential roles of circulating and adipocyte-specific clusterin in cardiometabolism and discuss potential further investigations to determine if clusterin is a viable target to attenuate both metabolic and cardiovascular disease.
Collapse
Affiliation(s)
- Jennifer Wittwer
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| | - David Bradley
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
26
|
Adipose Tissue T Regulatory Cells: Implications for Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:125-139. [PMID: 33523447 DOI: 10.1007/978-981-15-6407-9_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity dramatically increases the risk of numerous conditions, including type 2 diabetes mellitus and other components of the metabolic syndrome. Pro-inflammatory changes that occur in adipose tissue are critical to the pathogenesis of these obesity-induced complications. Adipose tissue is one of the body's largest endocrine organs, and the cells that comprise the adipose tissue immunoenvironment secrete multiple factors (including adipokines and cytokines) that impact systemic metabolism. In particular, immunosuppressive regulatory T cells (Tregs) decline in obesity, partly in response to its complex interaction with adipocytes, and this decline contributes to disruption of the typical homeostasis observed in lean adipose tissue. Although the regulation of Treg differentiation, function, and enrichment is incompletely understood, factors including various cell-surface co-stimulatory molecules, certain lipid species, and cytokines such as PPARγ, adiponectin, and leptin are important mediators. It is also clear that there may be depot-specific differences in Tregs, rendering adipose tissue Tregs distinct from lymphoid or circulating Tregs, with implications on maintenance and functionality. While most of these findings are derived from studies in murine models, comparatively little is known about the human adipose tissue Treg signature, which requires further investigation.
Collapse
|
27
|
Dimachkie MD, Bechtel MD, Robertson HL, Michel C, Lee EK, Sullivan DK, Chalise P, Thrasher JB, Parker WP, Godwin AK, Pathak HB, DiGiovanni J, Shivappa N, Hébert JR, Hamilton-Reeves JM. Exploration of biomarkers from a pilot weight management study for men undergoing radical prostatectomy. Urol Oncol 2021; 39:495.e7-495.e15. [PMID: 33563536 DOI: 10.1016/j.urolonc.2021.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/03/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Several biologic mechanisms, including inflammation and immune changes, have been proposed to explain the role of obesity in prostate cancer (CaP) progression. Compared to men of a healthy weight, overweight and obese men are more likely to have CaP recurrence post-prostatectomy. Obesity is related to inflammation and immune dysregulation; thus, weight loss may be an avenue to reduce inflammation and reverse these immune processes. OBJECTIVES This study explores the reversibility of the biological mechanisms through intentional weight loss using a comprehensive weight management program in men undergoing prostatectomy. Outcomes include blood and tissue biomarkers, microtumor environment gene expression, inflammation markers and Dietary Inflammatory Index (DII) scores. METHODS Twenty overweight men undergoing prostatectomy participated in this study. Fifteen men chose the intervention and 5 men chose the nonintervention group. The intervention consisted of a comprehensive weight loss program prior to prostatectomy and a weight maintenance program following surgery. Prostate tissue samples were obtained from diagnostic biopsies before the intervention and prostatectomy samples after weight loss. Blood samples and diet records were collected at baseline, pre-surgery after weight loss and at study end after weight maintenance. Immunohistochemistry and NanoString analysis were used to analyze the tissue samples. Flow cytometry was used to assess circulating immune markers. Inflammation markers were measured using Luminex panels. RESULTS The intervention group lost >5% body weight prior to surgery. DII scores improved during the weight loss intervention from baseline to pre-surgery (P = 0.002); and between group differences were significant (P = 0.02). DII scores were not associated with IL-6 nor hsCRP. In the intervention, CXCL12, CXCR7, and CXCR4 (C-X-C motif chemokine ligand/receptor) and Ki67 expression decreased in the prostate tissue from biopsy to surgery (P = 0.06), yet plasma CXCL12 increased during the same timeframe (P = 0.009). The downregulation of several genes (FDR<0.001) was observed in the intervention compared to the non-intervention. Changes in immune cells were not significant in either group. CONCLUSION This feasibility study demonstrates that in overweight men with localized CaP, weight loss alters blood, and tissue biomarkers, as well as tumor gene expression. More research is needed to determine the biological and clinical significance of these findings.
Collapse
Affiliation(s)
| | - Misty D Bechtel
- Department of Urology, University of Kansas Medical Center, Kansas City, KS
| | - Hilary L Robertson
- Department of Urology, University of Kansas Medical Center, Kansas City, KS
| | - Carrie Michel
- Department of Urology, University of Kansas Medical Center, Kansas City, KS
| | - Eugene K Lee
- Department of Urology, University of Kansas Medical Center, Kansas City, KS
| | - Debra K Sullivan
- Department of Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, KS
| | - Prabhakar Chalise
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS
| | | | - William P Parker
- Department of Urology, University of Kansas Medical Center, Kansas City, KS
| | - Andrew K Godwin
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Harsh B Pathak
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX
| | - Nitin Shivappa
- Cancer Prevention and Control Program, University of South Carolina, Columbia, SC 29208; Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC; Connecting Health Innovations LLC, Columbia, SC
| | - James R Hébert
- Cancer Prevention and Control Program, University of South Carolina, Columbia, SC 29208; Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC; Connecting Health Innovations LLC, Columbia, SC
| | - Jill M Hamilton-Reeves
- Department of Urology, University of Kansas Medical Center, Kansas City, KS; Department of Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, KS.
| |
Collapse
|
28
|
Wara AK, Wang S, Wu C, Fang F, Haemmig S, Weber BN, Aydogan CO, Tesmenitsky Y, Aliakbarian H, Hawse JR, Subramaniam M, Zhao L, Sage PT, Tavakkoli A, Garza A, Lynch L, Banks AS, Feinberg MW. KLF10 Deficiency in CD4 + T Cells Triggers Obesity, Insulin Resistance, and Fatty Liver. Cell Rep 2020; 33:108550. [PMID: 33378664 PMCID: PMC7816773 DOI: 10.1016/j.celrep.2020.108550] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 11/15/2019] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
CD4+ T cells regulate inflammation and metabolism in obesity. An imbalance of CD4+ T regulatory cells (Tregs) is critical in the development of insulin resistance and diabetes. Although cytokine control of this process is well understood, transcriptional regulation is not. KLF10, a member of the Kruppel-like transcription factor family, is an emerging regulator of immune cell function. We generated CD4+-T-cell-specific KLF10 knockout (TKO) mice and identified a predisposition to obesity, insulin resistance, and fatty liver due to defects of CD4+ Treg mobilization to liver and adipose tissue depots and decreased transforming growth factor β3 (TGF-β3) release in vitro and in vivo. Adoptive transfer of wild-type CD4+ Tregs fully rescued obesity, insulin resistance, and fatty liver. Mechanistically, TKO Tregs exhibit reduced mitochondrial respiration and glycolysis, phosphatidylinositol 3-kinase (PI3K)-Akt-mTOR signaling, and consequently impaired chemotactic properties. Collectively, our study identifies CD4+ T cell KLF10 as an essential regulator of obesity and insulin resistance by altering Treg metabolism and mobilization.
Collapse
Affiliation(s)
- Akm Khyrul Wara
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shijia Wang
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Wu
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang Fang
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Stefan Haemmig
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Brittany N Weber
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ceren O Aydogan
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Cerrahpasa District, Kocamustafapasa Street, Number 34/E, Fatih, Istanbul, Turkey
| | - Yevgenia Tesmenitsky
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hassan Aliakbarian
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Lei Zhao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peter T Sage
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ali Tavakkoli
- Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Cerrahpasa District, Kocamustafapasa Street, Number 34/E, Fatih, Istanbul, Turkey
| | - Amanda Garza
- Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lydia Lynch
- Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alexander S Banks
- Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Mark W Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
29
|
Ramos-Ramírez P, Malmhäll C, Johansson K, Adner M, Lötvall J, Bossios A. Lung Regulatory T Cells Express Adiponectin Receptor 1: Modulation by Obesity and Airway Allergic Inflammation. Int J Mol Sci 2020; 21:ijms21238990. [PMID: 33256137 PMCID: PMC7730828 DOI: 10.3390/ijms21238990] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/26/2022] Open
Abstract
Regulatory T cells (Tregs) decrease in the adipose tissue upon weight gain, contributing to persistent low-grade inflammation in obesity. We previously showed that adipose tissue Tregs express the adiponectin receptor 1 (AdipoR1); however, the expression in lung Tregs is still unknown. Here, we aimed to determine whether Helios+ and Helios- Treg subsets expressed AdipoR1 in the lungs of obese mice and whether different obesity grades affected the expression upon allergic lung inflammation. For diet-induced obesity (DIO), mice were fed a high-fat diet (HFD) for up to 15 weeks (overweight), 21 weeks (obesity), and 26 weeks (morbid obesity). Overweight and morbidly obese mice were sensitized and challenged with ovalbumin (OVA) to induce allergic lung inflammation. The AdipoR1 expression was reduced significantly in the lung Helios+ and Helios- Tregs of obese mice compared with lean mice. Airway allergic inflammation showed reduced AdipoR1 expression in lung Foxp3+ Tregs. Obesity significantly exacerbated the eosinophilic airway inflammation and reduced the number of Helios+ Tregs in lung and adipose tissue in the obesity-associated asthma model. Upon further weight gain, AdipoR1-expressing Tregs in the lungs of allergic mice were increased, whereas AdipoR1-expressing Tregs in adipose tissue were reduced. These data suggest that obesity-associated adipose tissue inflammation may exacerbate allergic inflammation by downregulating the AdipoR1+ Tregs in the lungs.
Collapse
Affiliation(s)
- Patricia Ramos-Ramírez
- Krefting Research Centre, Department for Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; (P.R.-R.); (C.M.); (K.J.); (J.L.)
| | - Carina Malmhäll
- Krefting Research Centre, Department for Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; (P.R.-R.); (C.M.); (K.J.); (J.L.)
| | - Kristina Johansson
- Krefting Research Centre, Department for Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; (P.R.-R.); (C.M.); (K.J.); (J.L.)
| | - Mikael Adner
- Institute of Environmental Medicine, Karolinska Institutet, SE-171 65 Stockholm, Sweden;
| | - Jan Lötvall
- Krefting Research Centre, Department for Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; (P.R.-R.); (C.M.); (K.J.); (J.L.)
| | - Apostolos Bossios
- Krefting Research Centre, Department for Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; (P.R.-R.); (C.M.); (K.J.); (J.L.)
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Huddinge, SE-141 86 Stockholm, Sweden
- Department of Medicine, Huddinge, Karolinska Institutet, SE-141 86 Stockholm, Sweden
- Correspondence: ; Tel.: +46-8-58586734
| |
Collapse
|
30
|
Aging and Immunometabolic Adaptations to Thermogenesis. Ageing Res Rev 2020; 63:101143. [PMID: 32810648 DOI: 10.1016/j.arr.2020.101143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/20/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022]
Abstract
Brown and subcutaneous adipose tissues play a key role in non-shivering thermogenesis both in mice and human, and their activation by adrenergic stimuli promotes energy expenditure, reduces adiposity, and protects against age-related metabolic diseases such as type 2 diabetes (T2D). Low-grade inflammation and insulin resistance characterize T2D. Even though the decline of thermogenic adipose tissues is well-established during ageing, the mechanisms by which this event affects immune system and contributes to the development of T2D is still poorly defined. It is emerging that activation of thermogenic adipose tissues promotes type 2 immunity skewing, limiting type 1 inflammation. Of note, metabolic substrates sustaining type 1 inflammation (e.g. glucose and succinate) are also used by activated adipocytes to promote thermogenesis. Keeping in mind this aspect, a nutrient competition between adipocytes and adipose tissue immune cell infiltrates could be envisaged. Herein, we reviewed the metabolic rewiring of adipocytes during thermogenesis in order to give important insight into the anti-inflammatory role of thermogenic adipose tissues and delineate how their decline during ageing may favor the setting of low-grade inflammatory states that predispose to type 2 diabetes in elderly. A brief description about the contribution of adipokines secreted by thermogenic adipocytes in modulation of immune cell activation is also provided. Finally, we have outlined experimental flow chart procedures and provided technical advices to investigate the physiological processes leading to thermogenic adipose tissue impairment that are behind the immunometabolic decline during aging.
Collapse
|
31
|
The omentum harbors unique conditions in the peritoneal cavity to promote healing and regeneration for diaphragm muscle repair in mdx mice. Cell Tissue Res 2020; 382:447-455. [PMID: 32661578 DOI: 10.1007/s00441-020-03238-1] [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: 01/09/2020] [Accepted: 06/05/2020] [Indexed: 10/23/2022]
Abstract
Although the primary cause of Duchenne muscular dystrophy (DMD) is a genetic mutation, the inflammatory response contributes directly to severity and exacerbation of the diaphragm muscle pathology. The omentum is a lymphoid organ with unique structural and immune functions serving as a sanctuary of hematopoietic and mesenchymal progenitors that coordinate immune responses in the peritoneal cavity. Upon activation, these progenitors expand and the organ produces large amounts of growth factors orchestrating tissue regeneration. The omentum of mdx mouse, a DMD murine model, is rich in milky spots and produces growth factors that promote diaphragm muscle regeneration. The present review summarizes the current knowledge of the omentum as an important immunologic structure and highlights its contribution to resolution of dystrophic muscle injury by providing an adequate environment for muscle regeneration, thus being a potential site for therapeutic interventions in DMD.
Collapse
|
32
|
Putilin DA, Evchenko SY, Fedoniuk LY, Tokarskyy OS, Kamyshny OM, Migenko LM, Andreychyn SM, Hanberher II, Bezruk TO. The Influence of Metformin to the Transcriptional Activity of the mTOR and FOX3 Genes in Parapancreatic Adipose Tissue of Streptozotocin-Induced Diabetic Rats. J Med Life 2020; 13:50-55. [PMID: 32341701 PMCID: PMC7175427 DOI: 10.25122/jml-2020-0029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The mammalian target of rapamycin is not only a central regulator of lipid metabolism that controls the processes of adipogenesis and lipolysis but also a regulator of the immunometabolism of immune cells that infiltrate adipose tissue. In turn, the level of progression of diabetes is significantly influenced by the Treg subpopulation, the complexity and heterogeneity of which is confirmed by the detection of numerous tissue-specific Tregs, including the so-called VAT Tregs (visceral adipose tissue CD4+Foxp3+ regulatory T cells). Therefore, the purpose of the study was to determine the mRNA expression levels of mTOR, Foxp3, IL1β, and IL17A genes in rat parapancreatic adipose tissue with experimental streptozotocin-induced diabetes mellitus, with or without metformin administration. The experiments were performed on male Wistar rats with induced diabetes as a result of streptozotocin administration. Molecular genetic studies were performed using real-time reverse transcription-polymerase chain reaction. The development of diabetes caused transcriptional activation of the mammalian target of rapamycin protein kinase gene, as well as increased mRNA expression of the pro-inflammatory cytokines IL1β and IL17A, but did not affect Foxp3 mRNA expression. The intervention with metformin in diabetic rats inhibited the mammalian target of rapamycin mRNA expression and caused an increase in the transcriptional activity of the Foxp3 gene in parapancreatic adipose tissue.
Collapse
Affiliation(s)
| | - Sergey Yuryevich Evchenko
- Department of Microbiology, Virology and Immunology, Zaporizhzhia State Medical University, Zaporizhzhia, Ukraine
| | | | | | - Oleksandr Mikhailovich Kamyshny
- Department of Microbiology, Virology and Immunology, Molecular Genetics Laboratory, Zaporizhzhia State Medical University, Zaporizhzhia, Ukraine
| | | | - Serhiy Mikhailovich Andreychyn
- Department of Propedeutics of Internal Medicine and Phthisiology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Ihorivna Hanberher
- Department of Propedeutics of Internal Medicine and Phthisiology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Tetyana Oleksandrivna Bezruk
- Department of Internal Medicine and Infectious Diseases, Bukovinian State Medical University, Chernivtsi, Ukraine
| |
Collapse
|
33
|
Arroyo Hornero R, Hamad I, Côrte-Real B, Kleinewietfeld M. The Impact of Dietary Components on Regulatory T Cells and Disease. Front Immunol 2020; 11:253. [PMID: 32153577 PMCID: PMC7047770 DOI: 10.3389/fimmu.2020.00253] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
The rise in the prevalence of autoimmune diseases in developed societies has been associated with a change in lifestyle patterns. Among other factors, increased consumption of certain dietary components, such as table salt and fatty acids and excessive caloric intake has been associated with defective immunological tolerance. Dietary nutrients have shown to modulate the immune response by a direct effect on the function of immune cells or, indirectly, by acting on the microbiome of the gastrointestinal tract. FOXP3+ regulatory T cells (Tregs) suppress immune responses and are critical for maintaining peripheral tolerance and immune homeostasis, modulating chronic tissue inflammation and autoimmune disease. It is now well-recognized that Tregs show certain degree of plasticity and can gain effector functions to adapt their regulatory function to different physiological situations during an immune response. However, plasticity of Tregs might also result in conversion into effector T cells that may contribute to autoimmune pathogenesis. Yet, which environmental cues regulate Treg plasticity and function is currently poorly understood, but it is of significant importance for therapeutic purposes. Here we review the current understanding on the effect of certain dietary nutrients that characterize Western diets in Treg metabolism, stability, and function. Moreover, we will discuss the role of Tregs linking diet and autoimmunity and the potential of dietary-based interventions to modulate Treg function in disease.
Collapse
Affiliation(s)
- Rebeca Arroyo Hornero
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Ibrahim Hamad
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Beatriz Côrte-Real
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| |
Collapse
|
34
|
Kim HR, Choi EJ, Kie JH, Lee JH, Seoh JY. Deficiency of glutathione peroxidase-1 and catalase attenuated diet-induced obesity and associated metabolic disorders. Acta Diabetol 2020; 57:151-161. [PMID: 31372751 DOI: 10.1007/s00592-019-01388-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/24/2022]
Abstract
AIMS Oxidative stress has been considered to contribute to the development of obesity-related metabolic disorders including insulin resistance. To the contrary, deficiency of an anti-oxidizing enzyme, glutathione peroxidase (GPx)-1, was reported to enhance insulin signaling, suggesting that oxidative stress may inhibit the development of type 2 diabetes. However, the beneficial effects of the absence of GPx-1 in metabolic homeostasis, including body weight control, have not yet been clearly manifested. To clarify the relationship between oxidative stress and obesity-related metabolic disorders, we investigated another mouse deficient with both GPx-1 and catalase (Cat). METHODS C57BL/6J wild-type and GPx-1-/- × Cat-/- mice were fed with a high-fat diet (60% fat) or a normal chow diet for 16 weeks and were investigated for metabolic and histological studies. RESULTS Body weight gain was significantly reduced, and glucose metabolism as well as hepatic steatosis was obviously improved in the GPx-1-/- × Cat-/- mice. The serum levels of insulin and total cholesterol were also significantly lowered. For the underlying mechanism, inflammation was attenuated and expression of markers for fat browning was enhanced in the visceral white adipose tissues. CONCLUSIONS Oxidative stress due to deficiency of GPx-1 and Cat may improve obesity-related metabolic disorders through attenuation of inflammation and fat browning.
Collapse
Affiliation(s)
- Hyung-Ran Kim
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Magokdongro 2-25, Gangseo-Gu, Seoul, 07804, Republic of Korea
| | - Eun-Jeong Choi
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Magokdongro 2-25, Gangseo-Gu, Seoul, 07804, Republic of Korea
| | - Jeong-Hae Kie
- Department of Pathology, National Health Insurance Cooperation Ilsan Hospital, Koyang, Republic of Korea
| | - Joo-Ho Lee
- Department of Surgery, Ewha Womans University Graduate School of Medicine, Gangseo-Gu, Seoul, Republic of Korea.
- Mediplant Research Institute of Bioscience, Mapo-Gu, Seoul, Republic of Korea.
| | - Ju-Young Seoh
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Magokdongro 2-25, Gangseo-Gu, Seoul, 07804, Republic of Korea.
- Mediplant Research Institute of Bioscience, Mapo-Gu, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Griffin DM, Bitner BR, Criss Ii Z, Marcano D, Berlin JM, Kent TA, Tour JM, Samson SL, Pautler RG. Use of a bioengineered antioxidant in mouse models of metabolic syndrome. Expert Opin Investig Drugs 2020; 29:209-219. [PMID: 31937152 DOI: 10.1080/13543784.2020.1716216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Background: Oxidative stress has been implicated in metabolic syndrome (MetS); however, antioxidants such as vitamin E have had limited success in the clinic. This prompts the question of what effects amore potent antioxidant might produce. A prime candidate is the recently developed bioengineered antioxidant, poly(ethylene glycol)-functionalizedhydrophilic carbon clusters (PEG-HCCs), which are capable of neutralizing the reactive oxygen species (ROS) superoxide anion and hydroxyl radical at106/molecule of PEG-HCC. In this project, we tested the potential of PEG-HCCs as a possible therapeutic for MetS.Results: PEG-HCC treatment lessened lipid peroxidation, aspartate aminotransferase levels, non-fastingblood glucose levels, and JNK phosphorylation inob/ob mice. PEG-HCC-treated WT mice had an increased response to insulin by insulin tolerance tests and adecrease in blood glucose by glucose tolerance tests. These effects were not observed in HFD-fed mice, regardless of treatment. PEG-HCCs were observed in the interstitial space of liver, spleen, skeletal muscle, and adipose tissue. No significant difference was shown in gluconeogenesis or inflammatory gene expression between treatment and dietary groups.Expert Opinion: PEG-HCCs improved some parameters of disease possibly due to a resulting increase in peripheral insulin sensitivity. However, additional studies are needed to elucidate how PEG-HCCsare producing these effects.
Collapse
Affiliation(s)
- Deric M Griffin
- Interdepartmental Program in Translation Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Brittany R Bitner
- Interdepartmental Program in Translation Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Zachary Criss Ii
- Interdepartmental Program in Translation Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Daniela Marcano
- Department of Chemistry, Rice University, Houston, TX, USA.,Smalley-Curl Institute for and Nanocarbon Center, Rice University, Houston, TX, USA
| | - Jacob M Berlin
- Department of Chemistry, Rice University, Houston, TX, USA.,Smalley-Curl Institute for and Nanocarbon Center, Rice University, Houston, TX, USA.,Molecular Medicine, City of Hope, Duarte, CA, USA
| | - Thomas A Kent
- Interdepartmental Program in Translation Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Neurology, Baylor College of Medicine, Houston, TX, USA.,Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - James M Tour
- Department of Chemistry, Rice University, Houston, TX, USA.,Smalley-Curl Institute for and Nanocarbon Center, Rice University, Houston, TX, USA
| | - Susan L Samson
- Department of Chemistry, Rice University, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Robia G Pautler
- Interdepartmental Program in Translation Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
36
|
Calcaterra V, Croce S, Vinci F, De Silvestri A, Cordaro E, Regalbuto C, Zuccotti GV, Mameli C, Albertini R, Avanzini MA. Th17 and Treg Balance in Children With Obesity and Metabolically Altered Status. Front Pediatr 2020; 8:591012. [PMID: 33330284 PMCID: PMC7710792 DOI: 10.3389/fped.2020.591012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/09/2020] [Indexed: 01/14/2023] Open
Abstract
Background: Chronic low-grade inflammation and activation of the immune system are hallmark pathogenic mechanisms involved in metabolic dysfunction and are related to obesity. In particular, the involvement of regulatory and pro-inflammatory lymphocyte subpopulations has been reported in adults. We evaluated the Th17/Treg lymphocyte balance in obese and normal weight children, in relation with their metabolic status. Methods: We enrolled 50 pediatric patients. According to metabolic status, subjects were classified into: metabolically healthy (MH) and metabolically unhealthy (MU) groups. MU phenotype was defined as the presence of at least one of the following risk factors: blood pressure >90th percentile, glycemia>100 mg/dl, HDL cholesterol <40 mg/dl, triglycerides>100 mg/dl (<10 years) or >130 mg/dl (>10 years), impaired insulin sensitivity with HOMA-IR>97.5th percentile. Patient Treg and Th17 profiles were also evaluated. Results: Based on the presence of metabolic and/or cardiovascular pathological parameters, we classified 15 MU (30%) and 35 MH (70%) children; all MU children were obese. Analyzing the correlations between lymphocyte subpopulations and metabolic data, we noted a correlation between Th17 percentage and systolic hypertension (p = 0.01, r = -0.37); Treg/Th17 ratio and HOMA-IR (p = 0.02, r = 0.32) and systolic hypertension (p = 0.05, r = 0.30). Conclusion: Children with obesity have a high risk of developing metabolic and cardiovascular complications. The Th17/Treg lymphocyte balance appears to be involved in glycemic homeostasis and blood pressure control. Careful and early monitoring of the immune system would facilitate new early preventive strategies in pediatric metabolic diseases.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy.,Department of Pediatrics, Children's Hospital "V. Buzzi", Milan, Italy
| | - Stefania Croce
- Cell Factory, Pediatric Hematology Oncology Unit, Immunology and Transplantation Laboratory, Department of Maternal and Children's Health, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo, Pavia, Italy
| | - Federica Vinci
- Pediatric Unit, Department of the Mother and Child Health, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Annalisa De Silvestri
- Biometry & Clinical Epidemiology, Scientific Direction, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Erika Cordaro
- Cell Factory, Pediatric Hematology Oncology Unit, Immunology and Transplantation Laboratory, Department of Maternal and Children's Health, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo, Pavia, Italy
| | - Corrado Regalbuto
- Pediatric Unit, Department of the Mother and Child Health, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, Children's Hospital "V. Buzzi", Milan, Italy.,Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Chiara Mameli
- Department of Pediatrics, Children's Hospital "V. Buzzi", Milan, Italy.,Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Riccardo Albertini
- Laboratory of Clinical Chemistry, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Maria Antonietta Avanzini
- Cell Factory, Pediatric Hematology Oncology Unit, Immunology and Transplantation Laboratory, Department of Maternal and Children's Health, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo, Pavia, Italy
| |
Collapse
|
37
|
Bourgeois C, Gorwood J, Barrail-Tran A, Lagathu C, Capeau J, Desjardins D, Le Grand R, Damouche A, Béréziat V, Lambotte O. Specific Biological Features of Adipose Tissue, and Their Impact on HIV Persistence. Front Microbiol 2019; 10:2837. [PMID: 31921023 PMCID: PMC6927940 DOI: 10.3389/fmicb.2019.02837] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Although white AT can contribute to anti-infectious immune responses, it can also be targeted and perturbed by pathogens. The AT's immune involvement is primarily due to strong pro-inflammatory responses (with both local and paracrine effects), and the large number of fat-resident macrophages. Adipocytes also exert direct antimicrobial responses. In recent years, it has been found that memory T cells accumulate in AT, where they provide efficient secondary responses against viral pathogens. These observations have prompted researchers to re-evaluate the links between obesity and susceptibility to infections. In contrast, AT serves as a reservoir for several persistence pathogens, such as human adenovirus Ad-36, Trypanosoma gondii, Mycobacterium tuberculosis, influenza A virus, and cytomegalovirus (CMV). The presence and persistence of bacterial DNA in AT has led to the concept of a tissue-specific microbiota. The unexpected coexistence of immune cells and pathogens within the specific AT environment is intriguing, and its impact on anti-infectious immune responses requires further evaluation. AT has been recently identified as a site of HIV persistence. In the context of HIV infection, AT is targeted by both the virus and the antiretroviral drugs. AT's intrinsic metabolic features, large overall mass, and wide distribution make it a major tissue reservoir, and one that may contribute to the pathophysiology of chronic HIV infections. Here, we review the immune, metabolic, viral, and pharmacological aspects that contribute to HIV persistence in AT. We also evaluate the respective impacts of both intrinsic and HIV-induced factors on AT's involvement as a viral reservoir. Lastly, we examine the potential consequences of HIV persistence on the metabolic and immune activities of AT.
Collapse
Affiliation(s)
- Christine Bourgeois
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Jennifer Gorwood
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Aurélie Barrail-Tran
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| | - Claire Lagathu
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Jacqueline Capeau
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Delphine Desjardins
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Roger Le Grand
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Abderaouf Damouche
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Véronique Béréziat
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Olivier Lambotte
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| |
Collapse
|
38
|
Deng G, Song X, Fujimoto S, Piccirillo CA, Nagai Y, Greene MI. Foxp3 Post-translational Modifications and Treg Suppressive Activity. Front Immunol 2019; 10:2486. [PMID: 31681337 PMCID: PMC6813729 DOI: 10.3389/fimmu.2019.02486] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (Tregs) are engaged in maintaining immune homeostasis and preventing autoimmunity. Treg cells include thymic Treg cells and peripheral Treg cells, both of which can suppress the immune response via multiple distinct mechanisms. The differentiation, proliferation, suppressive function and survival of Treg cells are affected by distinct energy metabolic programs. Tissue-resident Treg cells hold unique features in comparison with the lymphoid organ Treg cells. Foxp3 transcription factor is a lineage master regulator for Treg cell development and suppressive activity. Accumulating evidence indicates that the activity of Foxp3 protein is modulated by various post-translational modifications (PTMs), including phosphorylation, O-GlcNAcylation, acetylation, ubiquitylation and methylation. These modifications affect multiple aspects of Foxp3 function. In this review, we define features of Treg cells and roles of Foxp3 in Treg biology, and summarize current research in PTMs of Foxp3 protein involved in modulating Treg function. This review also attempts to define Foxp3 dimer modifications relevant to mediating Foxp3 activity and Treg suppression. Understanding Foxp3 protein features and modulation mechanisms may help in the design of rational therapies for immune diseases and cancer.
Collapse
Affiliation(s)
- Guoping Deng
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | - Xiaomin Song
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | | | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada.,Centre of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Yasuhiro Nagai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mark I Greene
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
39
|
López S, García-Serrano S, Gutierrez-Repiso C, Rodríguez-Pacheco F, Ho-Plagaro A, Santiago-Fernandez C, Alba G, Cejudo-Guillen M, Rodríguez-Cañete A, Valdes S, Garrido-Sanchez L, Pozo D, García-Fuentes E. Tissue-Specific Phenotype and Activation of iNKT Cells in Morbidly Obese Subjects: Interaction with Adipocytes and Effect of Bariatric Surgery. Obes Surg 2019; 28:2774-2782. [PMID: 29619756 DOI: 10.1007/s11695-018-3215-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The immune response of visceral adipose tissue (VAT) in obesity, in particular the role of invariant natural killer T (iNKT) cells, has not yet been fully elucidated. OBJECTIVE To characterize iNKT cells and its activation status in VAT and peripheral blood mononuclear cells (PBMC) in morbidly obese subjects (MO), and to analyze their association with metabolic parameters. SUBJECTS AND METHODS Twenty non-obese and 20 MO subjects underwent Roux-en-Y gastric bypass (RYGB) and were studied before and 6 months after RYGB. VAT and PBMC were obtained. RESULTS A decrease in VAT iNKT cells from MO was found, however, not in PBMC. Visceral adipocytes from MO presented increased CD1d expression (p = 0.032). MO presented an increase in early activated CD69+ iNKT cells in PBMC before RYGB (p < 0.001), but not after RYGB nor in VAT, and an increase in later activated CD25+ iNKT in VAT (p = 0.046), without differences in PBMC. The co-expression of early and later markers (CD69+CD25+) in iNKT cells was increased in MO in VAT (p = 0.050) and PBMC (p = 0.006), decreasing after RYGB (p = 0.050). CD69+ iNKT and CD69+CD25+ iNKT cells in PBMC after RYGB correlated negatively with glucose, insulin, and insulin resistance levels. CONCLUSIONS There is a tissue-specific phenotype and activation of iNKT cells in VAT in morbid obesity, which could be involved in VAT immunometabolism dysregulation. Also, the increase in CD1d expression could be to offset the lack of VAT iNKT cells.
Collapse
Affiliation(s)
- Soledad López
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Seville, Spain. .,CABIMER-Andalusian Center for Molecular Biology and Regenerative Medicine (CSIC-University of Seville-UPO-Junta de Andalucia), Seville, Spain. .,Dpto. Bioquímica Médica, Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain.
| | - Sara García-Serrano
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Malaga, Spain.,CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Malaga, Spain
| | - Carolina Gutierrez-Repiso
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Malaga, Spain
| | - Francisca Rodríguez-Pacheco
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Malaga, Spain
| | - Ailec Ho-Plagaro
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Malaga, Spain
| | - Concepción Santiago-Fernandez
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Malaga, Spain
| | - Gonzalo Alba
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Seville, Spain
| | - Marta Cejudo-Guillen
- CABIMER-Andalusian Center for Molecular Biology and Regenerative Medicine (CSIC-University of Seville-UPO-Junta de Andalucia), Seville, Spain
| | - Alberto Rodríguez-Cañete
- Unidad de Gestión Clínica de Cirugía General, Digestiva y Trasplantes, Hospital Regional Universitario, Malaga, Spain
| | - Sergio Valdes
- CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Malaga, Spain.,Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Malaga, Spain
| | - Lourdes Garrido-Sanchez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Malaga, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Malaga, Spain
| | - David Pozo
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Seville, Spain.,CABIMER-Andalusian Center for Molecular Biology and Regenerative Medicine (CSIC-University of Seville-UPO-Junta de Andalucia), Seville, Spain
| | - Eduardo García-Fuentes
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Malaga, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Malaga, Spain. .,Laboratorio de Investigación, Hospital Civil, Plaza del Hospital Civil s/n, 29009, Malaga, Spain.
| |
Collapse
|
40
|
Kane H, Lynch L. Innate Immune Control of Adipose Tissue Homeostasis. Trends Immunol 2019; 40:857-872. [DOI: 10.1016/j.it.2019.07.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/19/2022]
|
41
|
Sasaki T, Moro K, Kubota T, Kubota N, Kato T, Ohno H, Nakae S, Saito H, Koyasu S. Innate Lymphoid Cells in the Induction of Obesity. Cell Rep 2019; 28:202-217.e7. [DOI: 10.1016/j.celrep.2019.06.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 04/16/2019] [Accepted: 06/04/2019] [Indexed: 12/21/2022] Open
|
42
|
Tang CL, Yu XH, Li Y, Zhang RH, Xie J, Liu ZM. Schistosoma japonicum Soluble Egg Antigen Protects Against Type 2 Diabetes in Lepr db/db Mice by Enhancing Regulatory T Cells and Th2 Cytokines. Front Immunol 2019; 10:1471. [PMID: 31297120 PMCID: PMC6607994 DOI: 10.3389/fimmu.2019.01471] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 06/13/2019] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes is a metabolic disorder characterized by persistently elevated glucose levels. There is no effective treatment strategy for this condition, and it poses a massive economic burden globally. Schistosoma soluble egg antigen (SEA)-induced immunomodulatory mechanisms have been reported in the treatment of autoimmune disease. This study aimed to determine the ability of Schistosoma japonicum SEA to protect against type 2 diabetes in Leprdb/db mice and understand the associated mechanisms. The mice were divided into four groups: C57BL/6 (the normal group), SEA (C57BL/6 mice treated with SEA), Leprdb/db, and SEA and Leprdb/db co-treatment groups. The mice in the SEA and co-treatment groups were injected with 50 μg of SEA (twice a week for 6 weeks), and the same volume of PBS was used as control. Blood glucose, insulin, and HOMA-IR levels were measured in all mice, which were sacrificed 6 weeks after the last SEA administration. Flow cytometry was used to detect the percentages of regulatory T cells in splenocytes. ELISA was used to detect the levels of IFN-γ, IL-2, IL-4, and IL-5 in cell culture supernatants. Compared with the mice in the Leprdb/db group, the mice in the SEA + Leprdb/db group exhibited significantly reduced insulin resistance, as evidenced by the enhancement of wound healing. The frequency of spleen regulatory T cells increased significantly after SEA administration; meanwhile, the secretion of IL-4 and IL-5 in spleen cells was elevated. These results indicate that SEA can reduce insulin resistance and provide new targets for the treatment of type 2 diabetes. The potential mechanisms might be associated with increases in regulatory T cells and Th2 cytokines in Leprdb/db mice, which warrants further investigation.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Xiao-Hong Yu
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Yan Li
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Rong-Hui Zhang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jun Xie
- Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Zhi-Ming Liu
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to examine the evidence describing adipose tissue as a reservoir for HIV-1 and how this often expansive anatomic compartment contributes to HIV persistence. RECENT FINDINGS Memory CD4 T cells and macrophages, the major host cells for HIV, accumulate in adipose tissue during HIV/SIV infection of humans and rhesus macaques. Whereas HIV and SIV proviral DNA is detectable in CD4 T cells of multiple fat depots in virtually all infected humans and monkeys examined, viral RNA is less frequently detected, and infected macrophages may be less prevalent in adipose tissue. However, based on viral outgrowth assays, adipose-resident CD4 T cells are latently infected with virus that is replication-competent and infectious. Additionally, adipocytes interact with CD4 T cells and macrophages to promote immune cell activation and inflammation which may be supportive for HIV persistence. Antiviral effector cells, such as CD8 T cells and NK/NKT cells, are abundant in adipose tissue during HIV/SIV infection and typically exceed CD4 T cells, whereas B cells are largely absent from adipose tissue of humans and monkeys. Additionally, CD8 T cells in adipose tissue of HIV patients are activated and have a late differentiated phenotype, with unique TCR clonotypes of less diversity relative to blood CD8 T cells. With respect to the distribution of antiretroviral drugs in adipose tissue, data is limited, but there may be class-specific penetration of fat depots. The trafficking of infected immune cells within adipose tissues is a common event during HIV/SIV infection of humans and monkeys, but the virus may be mostly transcriptionally dormant. Viral replication may occur less in adipose tissue compared to other major reservoirs, such as lymphoid tissue, but replication competence and infectiousness of adipose latent virus are comparable to other tissues. Due to the ubiquitous nature of adipose tissue, inflammatory interactions among adipocytes and CD4 T cells and macrophages, and selective distribution of antiretroviral drugs, the sequestration of infected immune cells within fat depots likely represents a major challenge for cure efforts.
Collapse
Affiliation(s)
- Jacob Couturier
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 2.112, Houston, TX, 77030, USA
| | - Dorothy E Lewis
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 2.112, Houston, TX, 77030, USA.
| |
Collapse
|
44
|
Cellular Metabolic Regulation in the Differentiation and Function of Regulatory T Cells. Cells 2019; 8:cells8020188. [PMID: 30795546 PMCID: PMC6407031 DOI: 10.3390/cells8020188] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 12/29/2022] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining immune tolerance and preventing autoimmune and inflammatory diseases. The activity and function of Tregs are in large part determined by various intracellular metabolic processes. Recent findings have focused on how intracellular metabolism can shape the development, trafficking, and function of Tregs. In this review, we summarize and discuss current research that reveals how distinct metabolic pathways modulate Tregs differentiation, phenotype stabilization, and function. These advances highlight numerous opportunities to alter Tregs frequency and function in physiopathologic conditions via metabolic manipulation and have important translational implications.
Collapse
|
45
|
Raud B, McGuire PJ, Jones RG, Sparwasser T, Berod L. Fatty acid metabolism in CD8 + T cell memory: Challenging current concepts. Immunol Rev 2019; 283:213-231. [PMID: 29664569 DOI: 10.1111/imr.12655] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD8+ T cells are key members of the adaptive immune response against infections and cancer. As we discuss in this review, these cells can present diverse metabolic requirements, which have been intensely studied during the past few years. Our current understanding suggests that aerobic glycolysis is a hallmark of activated CD8+ T cells, while naive and memory (Tmem ) cells often rely on oxidative phosphorylation, and thus mitochondrial metabolism is a crucial determinant of CD8+ Tmem cell development. Moreover, it has been proposed that CD8+ Tmem cells have a specific requirement for the oxidation of long-chain fatty acids (LC-FAO), a process modulated in lymphocytes by the enzyme CPT1A. However, this notion relies heavily on the metabolic analysis of in vitro cultures and on chemical inhibition of CPT1A. Therefore, we introduce more recent studies using genetic models to demonstrate that CPT1A-mediated LC-FAO is dispensable for the development of CD8+ T cell memory and protective immunity, and question the use of chemical inhibitors to target this enzyme. We discuss insights obtained from those and other studies analyzing the metabolic characteristics of CD8+ Tmem cells, and emphasize how T cells exhibit flexibility in their choice of metabolic fuel.
Collapse
Affiliation(s)
- Brenda Raud
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Peter J McGuire
- Metabolism, Infection, and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Russell G Jones
- Department of Physiology, Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Luciana Berod
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| |
Collapse
|
46
|
Laparra A, Tricot S, Le Van M, Damouche A, Gorwood J, Vaslin B, Favier B, Benoist S, Ho Tsong Fang R, Bosquet N, Le Grand R, Chapon C, Lambotte O, Bourgeois C. The Frequencies of Immunosuppressive Cells in Adipose Tissue Differ in Human, Non-human Primate, and Mouse Models. Front Immunol 2019; 10:117. [PMID: 30804937 PMCID: PMC6371887 DOI: 10.3389/fimmu.2019.00117] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 01/15/2019] [Indexed: 12/20/2022] Open
Abstract
Although the metabolic properties of white adipose tissue have been extensively characterized, the tissue's immune properties are now attracting renewed interest. Early experiments in a mouse model suggested that white adipose tissue contains a high density of regulatory T cells (Tregs), and so it was assumed that all adipose tissue has an immunosuppressive profile—even though the investigation was limited to visceral body fat in relatively old male mice. This observation was also corroborated by high frequencies of other cell subsets with immunoregulatory properties, such as anti-inflammatory M2 macrophages, and regulatory B cells. Many studies have since evidenced the persistence of pathogens (trypanosomes, Mycobacterium tuberculosis, HIV, etc.) in adipose tissue. However, a recent report identified adipose tissue as a reservoir of memory T cells capable of protecting animals upon rechallenge. The immune potential of lean adipose tissue thus remains to be further investigated. Here, we compared the relative proportions of immune cells (and Tregs in particular) in lean adipose tissue collected from humans, a non-human primate (the cynomolgus macaque), and three mouse models. We demonstrated that the proportion of Foxp3+ Tregs in visceral adipose tissue was low in all models other than the C57Bl/6 mouse. These low values were not linked to correspondingly low proportions of effector cells because T lymphocytes (a main target of Treg suppression) were more frequent in cynomolgus macaques than in C57Bl/6 mice and (to a lesser extent) humans. In contrast, the proportions of macrophages and B cells were lower in cynomolgus macaques than in C57Bl/6 mice. We also observed a higher proportion of CD34+CD45- cells (which predominantly correspond to mesenchymal stem cells) in C57Bl/6 mouse and cynomolgus macaques than in humans and both for subcutaneous and visceral adipose tissues. Lastly, a microscopy analysis confirmed predominant proportion of adipocytes within adipose tissue, and highlighted a marked difference in adipocyte size among the three species studied. In conclusion, our study of lean, middle-aged, male individuals showed that the immune compartment of adipose tissue differed markedly in humans vs. mice, and suggesting the presence of a more inflammatory steady-state profile in humans than mice.
Collapse
Affiliation(s)
- Ariane Laparra
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Sabine Tricot
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Mélanie Le Van
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Abderaouf Damouche
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Jennifer Gorwood
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Bruno Vaslin
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Benoit Favier
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Stéphane Benoist
- Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Service de Chirurgie Digestive et Oncologique, Le Kremlin-Bicêtre, France
| | - Raphael Ho Tsong Fang
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Nathalie Bosquet
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Roger Le Grand
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Catherine Chapon
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Olivier Lambotte
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,Université Paris Sud, Le Kremlin Bicêtre, France.,Assistance Publique-Hôpitaux de Paris, Service de Médecine Interne et Immunologie Clinique, Groupe Hospitalier Universitaire Paris Sud, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Christine Bourgeois
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| |
Collapse
|
47
|
Wu D, Han JM, Yu X, Lam AJ, Hoeppli RE, Pesenacker AM, Huang Q, Chen V, Speake C, Yorke E, Nguyen N, Sampath S, Harris D, Levings MK. Characterization of regulatory T cells in obese omental adipose tissue in humans. Eur J Immunol 2019; 49:336-347. [DOI: 10.1002/eji.201847570] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 11/09/2018] [Accepted: 12/17/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Dan Wu
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Jonathan M. Han
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Xin Yu
- Prevention of Organ Failure (PROOF) Centre of Excellence Vancouver BC Canada
| | - Avery J. Lam
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Romy E. Hoeppli
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Anne M. Pesenacker
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Qing Huang
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Virginia Chen
- Prevention of Organ Failure (PROOF) Centre of Excellence Vancouver BC Canada
| | - Cate Speake
- Diabetes Clinical Research ProgramBenaroya Research Institute Seattle WA USA
| | - Ekua Yorke
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- Richmond Metabolic and Bariatric Surgery ProgramRichmond Hospital Richmond BC Canada
| | - Nam Nguyen
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- Richmond Metabolic and Bariatric Surgery ProgramRichmond Hospital Richmond BC Canada
| | - Sharadh Sampath
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- Richmond Metabolic and Bariatric Surgery ProgramRichmond Hospital Richmond BC Canada
| | - David Harris
- Richmond Metabolic and Bariatric Surgery ProgramRichmond Hospital Richmond BC Canada
- Department of MedicineUniversity of British Columbia Vancouver BC Canada
| | - Megan K. Levings
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| |
Collapse
|
48
|
Wang Q, Wu H. T Cells in Adipose Tissue: Critical Players in Immunometabolism. Front Immunol 2018; 9:2509. [PMID: 30459770 PMCID: PMC6232870 DOI: 10.3389/fimmu.2018.02509] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/10/2018] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue performs immunoregulatory functions in addition to fat storage. Various T cells in different fat depots either help maintain metabolic homeostasis under healthy conditions or contribute to metabolic disorders in pathological conditions such as obesity, diabetes, cardiovascular diseases, or even cancer. These T cells play critical roles in immunometabolism, which refers to the intersection of immunity and metabolism. Numerous studies have examined the presence and changes of different T cell subsets, including helper T cells, regulatory T cells, cytotoxic T cells, and natural killer T cells, in adipose depots in health and diseases. In this review, we will discuss the adipose tissue niches that influence the patterns and functions of T cell subsets and in turn the impact of these T cells on cell- or body-based immunometabolism accounting for health and obesity.
Collapse
Affiliation(s)
- Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Huaizhu Wu
- Department of Medicine and Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
49
|
Lopreiato V, Hosseini A, Rosa F, Zhou Z, Alharthi A, Trevisi E, Loor JJ. Dietary energy level affects adipose depot mass but does not impair in vitro subcutaneous adipose tissue response to short-term insulin and tumor necrosis factor-α challenge in nonlactating, nonpregnant Holstein cows. J Dairy Sci 2018; 101:10206-10219. [PMID: 30146294 DOI: 10.3168/jds.2018-14389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 07/04/2018] [Indexed: 11/19/2022]
Abstract
We assessed effects of overfeeding energy to nonlactating and nonpregnant Holstein cows during a length of time similar to a typical dry period on body lipid storage and the abundance of genes related to insulin signaling, inflammation, and ubiquitination in subcutaneous adipose tissue (SAT) in vitro challenged with insulin and recombinant bovine tumor necrosis factor-α. Fourteen cows were randomly assigned to either a high-energy (OVE; net energy for lactation = 1.60 Mcal/kg of dry matter; n = 7) or control (CON; net energy for lactation = 1.30 Mcal/kg of dry matter; n = 7) diet for 6 wk. Immediately after slaughter, liver, kidneys, and mammary gland were separated and weighed. The adipose tissue mass in the omental, mesenteric, and perirenal depots was dissected and weighed. Subcutaneous adipose tissue was collected from the tail-head region and was used as follows: control, bovine insulin (INS) at 1 µmol/L, tumor necrosis factor-α at 5 ng/mL (TNF), and their combination. Despite a lack of difference in final body condition score, OVE cows had greater energy intake and were heavier than CON cows. Furthermore, overfeeding led to greater mass of mesenteric and perirenal adipose, liver, and mammary gland. Overall, SAT incubated with INS had an upregulation of insulin receptor (INSR), interleukin-10 (IL10), small ubiquitin-like modifier 3 (SUMO3), and ubiquitin conjugating enzyme E2I (UBC9), whereas TNF upregulated peroxisome proliferator-activated receptor gamma (PPARG), diacylglycerol O-acyltransferase 2 (DGAT2), interleukin-6 (IL6), nuclear factor kappa B subunit 1 (NFKB1), small ubiquitin-like modifier 2 (SUMO2), and UBC9. Regardless of in vitro treatment, feeding OVE upregulated PPARG, fatty acid synthase (FASN), and insulin induced gene 1 (INSIG1). Abundance of PPARG was greater in SAT of OVE cows cultured individually with INS and TNF. The interaction between diet and in vitro treatment revealed that sterol regulatory element binding transcription factor 1 (SREBF1) had greater abundance in SAT from the CON group in response to culture with INS, whereas SAT from OVE cows had greater SREBF1 abundance in response to culture with TNF. The mRNA abundance of IL6 and NFKB1 was greater in response to TNF treatment and overall in CON cows. Furthermore, SAT from these cows had greater IL10 abundance when cultured with INS and TNF. Overall, data highlighted that overfeeding energy increases adipose tissue mass in part by stimulating transcription of key genes associated with insulin signaling, adipogenesis, and lipogenesis. Because SAT thickness or mass was not measured, the lack of effect of overfeeding on body condition score limits its use to predict overall body lipid storage. An overt inflammatory response in SAT after a 6-wk period of over-consumption of energy could not be discerned.
Collapse
Affiliation(s)
- V Lopreiato
- Interdepartmental Services Centre of Veterinary for Human and Animal Health, Department of Health Science, Magna Græcia University, Catanzaro, 88100, Italy
| | - A Hosseini
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - F Rosa
- Dairy and Food Science Department, South Dakota State University, 1111 College Ave., 113H Alfred Dairy Science Hall, Brookings 57007
| | - Z Zhou
- Department of Animal and Veterinary Sciences, Clemson University, Clemson, SC 29634
| | - A Alharthi
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - E Trevisi
- Department of Animal Sciences, Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, Via Emilia Parmense, 84, 29122 Piacenza, Italy
| | - J J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
50
|
Modulation of the Gastrointestinal Microbiome with Nondigestible Fermentable Carbohydrates To Improve Human Health. Microbiol Spectr 2018; 5. [PMID: 28936943 DOI: 10.1128/microbiolspec.bad-0019-2017] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is a clear association between the gastrointestinal (GI) microbiome and the development of chronic noncommunicable diseases, providing a rationale for the development of strategies that target the GI microbiota to improve human health. In this article, we discuss the potential of supplementing the human diet with nondigestible fermentable carbohydrates (NDFCs) to modulate the composition, structure, diversity, and metabolic potential of the GI microbiome in an attempt to prevent or treat human disease. The current concepts by which NDFCs can be administered to humans, including prebiotics, fermentable dietary fibers, and microbiota-accessible carbohydrates, as well as the mechanisms by which these carbohydrates exert their health benefits, are discussed. Epidemiological research presents compelling evidence for the health effects of NDFCs, with clinical studies providing further support for some of these benefits. However, rigorously designed human intervention studies with well-established clinical markers and microbial endpoints are still essential to establish (i) the clinical efficiency of specific NDFCs, (ii) the causal role of the GI microbiota in these effects, (iii) the underlying mechanisms involved, and (iv) the degree by which inter-individual differences between GI microbiomes influence these effects. Such studies would provide the mechanistic understanding needed for a systematic application of NDFCs to improve human health via GI microbiota modulation while also allowing the personalization of these dietary strategies.
Collapse
|