101
|
Lieber AD, Beier UH, Xiao H, Wilkins BJ, Jiao J, Li XS, Schugar RC, Strauch CM, Wang Z, Brown JM, Hazen SL, Bokulich NA, Ruggles KV, Akimova T, Hancock WW, Blaser MJ. Loss of HDAC6 alters gut microbiota and worsens obesity. FASEB J 2019; 33:1098-1109. [PMID: 30102568 PMCID: PMC6355060 DOI: 10.1096/fj.201701586r] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 07/23/2018] [Indexed: 01/09/2023]
Abstract
Alterations in gut microbiota are known to affect intestinal inflammation and obesity. Antibiotic treatment can affect weight gain by elimination of histone deacetylase (HDAC) inhibitor-producing microbes, which are anti-inflammatory by augmenting regulatory T (Treg) cells. We asked whether mice that lack HDAC6 and have potent suppressive Treg cells are protected from microbiota-induced accelerated weight gain. We crossed wild-type and HDAC6-deficient mice and subjected the offspring to perinatal penicillin, inducing weight gain via microbiota disturbance. We observed that male HDAC6-deficient mice were not protected and developed profoundly accelerated weight gain. The antibiotic-exposed HDAC6-deficient mice showed a mixed immune phenotype with increased CD4+ and CD8+ T-cell activation yet maintained the enhanced Treg cell-suppressive function phenotype characteristic of HDAC6-deficient mice. 16S rRNA sequencing of mouse fecal samples reveals that their microbiota diverged with time, with HDAC6 deletion altering microbiome composition. On a high-fat diet, HDAC6-deficient mice were depleted in representatives of the S24-7 family and Lactobacillus but enriched with Bacteroides and Parabacteroides; these changes are associated with obesity. Our findings further our understanding of the influence of HDACs on microbiome composition and are important for the development of HDAC6 inhibitors in the treatment of human diseases.-Lieber, A. D., Beier, U. H., Xiao, H., Wilkins, B. J., Jiao, J., Li, X. S., Schugar, R. C., Strauch, C. M., Wang, Z., Brown, J. M., Hazen, S. L., Bokulich, N. A., Ruggles, K. V., Akimova, T., Hancock, W. W., Blaser, M. J. Loss of HDAC6 alters gut microbiota and worsens obesity.
Collapse
Affiliation(s)
- Arnon D. Lieber
- Department of Medicine New York University School of Medicine (NYUSM), New York, New York, USA
- Department of Microbiology, New York University School of Medicine (NYUSM), New York, New York, USA
| | - Ulf H. Beier
- Division of Nephrology, Department of Pediatrics University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Haiyan Xiao
- Division of Nephrology, Department of Pediatrics University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benjamin J. Wilkins
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jing Jiao
- Division of Nephrology, Department of Pediatrics University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xinmin S. Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Rebecca C. Schugar
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Christopher M. Strauch
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Zeneng Wang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - J. Mark Brown
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stanley L. Hazen
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicholas A. Bokulich
- Department of Medicine New York University School of Medicine (NYUSM), New York, New York, USA
- Department of Microbiology, New York University School of Medicine (NYUSM), New York, New York, USA
| | - Kelly V. Ruggles
- Applied Bioinformatics Laboratories, New York University School of Medicine (NYUSM), New York, New York, USA
- Division of Translational Medicine, Department of Medicine, New York University School of Medicine (NYUSM), New York, New York, USA
| | - Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Biesecker Center for Pediatric Liver Disease, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Biesecker Center for Pediatric Liver Disease, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Martin J. Blaser
- Department of Medicine New York University School of Medicine (NYUSM), New York, New York, USA
- Department of Microbiology, New York University School of Medicine (NYUSM), New York, New York, USA
- New York Harbor Department of Veterans Affairs Medical Center, New York, New York, USA
| |
Collapse
|
102
|
Nonalcoholic Fatty Liver Disease: Basic Pathogenetic Mechanisms in the Progression From NAFLD to NASH. Transplantation 2019; 103:e1-e13. [DOI: 10.1097/tp.0000000000002480] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
103
|
Patel S, Mandaliya D, Prajapati B, Kumar S, Seshadri S. Cefdinir Microsphere Modulated Microflora and Liver Immunological Response to Diet Induced Diabetes in Mice. Endocr Metab Immune Disord Drug Targets 2018; 19:349-357. [PMID: 30582487 DOI: 10.2174/1871530319666181224122115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Gut microbiota is currently targeted for various diseases especially metabolic disorders such as diabetes. Our strategy is to alter gut microflora via specific antibiotic to reduce load of inflammation in the liver that increases as a result of high carbohydrate diet. Th1, Th17 and Treg are important immune cell types which decide the type of inflammatory response. Liver is tolerogenic in nature with low Th17/Treg ratio. In diabetics, this ratio decreases even more, and can cause liver trauma. METHOD The present study tries to find relationship between gut flora and immune cells such as Th1/Th17/Treg and their role in liver metabolism using diet induced diabetic mice model. RESULT Upon alteration of flora using Cefdinir in different forms, one could help lower the level of Treg cells thus increasing the ratio. Gut flora is strongly associated with the immunity in the liver. Targeted alteration of gut flora helps us to restore insulin sensitivity. CONCLUSION Colon targeted Cefdinir gives more promising results, opens colonic bacteria as target for improving gut, liver inflammation and insulin sensitivity.
Collapse
Affiliation(s)
- Sweta Patel
- Institute of Science, Nirma University, S G Highway, Ahmedabad, India
| | - Dipeeka Mandaliya
- Institute of Science, Nirma University, S G Highway, Ahmedabad, India
| | - Bhumika Prajapati
- Institute of Science, Nirma University, S G Highway, Ahmedabad, India.,Indian Institute of Public Health, Gandhinagar, India
| | - Sunny Kumar
- Institute of Science, Nirma University, S G Highway, Ahmedabad, India
| | - Sriram Seshadri
- Institute of Science, Nirma University, S G Highway, Ahmedabad, India
| |
Collapse
|
104
|
Kalathookunnel Antony A, Lian Z, Wu H. T Cells in Adipose Tissue in Aging. Front Immunol 2018; 9:2945. [PMID: 30619305 PMCID: PMC6299975 DOI: 10.3389/fimmu.2018.02945] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Similar to obesity, aging is associated with visceral adiposity and insulin resistance. Inflammation in adipose tissue, mainly evidenced by increased accumulation and proinflammatory polarization of T cells and macrophages, has been well-documented in obesity and may contribute to the associated metabolic dysfunctions including insulin resistance. Studies show that increased inflammation, including inflammation in adipose tissue, also occurs in aging, so-called "inflamm-aging." Aging-associated inflammation in adipose tissue has some similarities but also differences compared to obesity-related inflammation. In particular, conventional T cells are elevated in adipose tissue in both obesity and aging and have been implicated in metabolic functions in obesity. However, the changes and also possibly functions of regulatory T cells (Treg) in adipose tissue are different in aging and obesity. In this review, we will summarize recent advances in research on the changes of these immune cells in adipose tissue with aging and obesity and discuss their possible contributions to metabolism and the potential of these immune cells as novel therapeutic targets for prevention and treatment of metabolic diseases associated with aging or obesity.
Collapse
Affiliation(s)
| | - Zeqin Lian
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
105
|
Ilan Y. Immune rebalancing by oral immunotherapy: A novel method for getting the immune system back on track. J Leukoc Biol 2018; 105:463-472. [PMID: 30476347 DOI: 10.1002/jlb.5ru0718-276rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/14/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
Immune modulating treatments are often associated with immune suppression or an opposing anti-inflammatory paradigm. As such, there is a risk of exposing patients to infections and malignancies. Contrarily, eliciting only mild immune modulation can be insufficient for alleviating immune-mediated damage. Oral immunotherapy is a novel approach that uses the inherent ability of the gut immune system to generate signals that specifically suppress inflammation at affected sites, without inducing generalized immune suppression. Oral immunotherapy is being developed as a method to rebalance systemic immunity and restore balance, getting it back on track, rather than pushing the immune response too much or too little in opposing directions. Here, I review recent preclinical and clinical data examining the technique and describe its primary advantages.
Collapse
Affiliation(s)
- Yaron Ilan
- Gastroenterology and Liver Units, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
106
|
Zhou T, Hu Z, Yang S, Sun L, Yu Z, Wang G. Role of Adaptive and Innate Immunity in Type 2 Diabetes Mellitus. J Diabetes Res 2018; 2018:7457269. [PMID: 30533447 PMCID: PMC6250017 DOI: 10.1155/2018/7457269] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 09/10/2018] [Indexed: 12/14/2022] Open
Abstract
After the recognition of the essential role of the immune system in the progression of type 2 diabetes mellitus, more studies are focused on the effects produced by the abnormal differentiation of components of the immune system. In patients suffering from obesity or T2DM, there were alterations in proliferation of T cells and macrophages, and impairment in function of NK cells and B cells, which represented abnormal innate and adaptive immunity. The abnormality of either innate immunity, adaptive immunity, or both was involved and interacted with each other during the progression of T2DM. Although previous studies have revealed the functional involvement of T cells in T2DM, and the regulation of metabolism by the innate or adaptive immune system during the pathogenesis of T2DM, there has been a lack of literature reviewing the relevant role of adaptive and innate immunity in the progression of T2DM. Here, we will review their relevant roles, aiming to provide new thought for the development of immunotherapy in T2DM.
Collapse
Affiliation(s)
- Tong Zhou
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zheng Hu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Shuo Yang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhenxiang Yu
- Department of Respiration, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
107
|
Cani PD, Jordan BF. Gut microbiota-mediated inflammation in obesity: a link with gastrointestinal cancer. Nat Rev Gastroenterol Hepatol 2018; 15:671-682. [PMID: 29844585 DOI: 10.1038/s41575-018-0025-6] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Overweight and obesity are associated with increased risk of developing metabolic disorders such as diabetes and cardiovascular diseases. However, besides these metabolic diseases, excess body weight is also associated with different cancers, including gastrointestinal cancers, such as liver, pancreatic and colon cancers. Inflammation is a common feature of both obesity and cancer; however, the origin of this inflammation has been largely debated. Over the past decade, growing evidence has shown that the composition of the gut microbiota and its activity might be associated not only with the onset of inflammation but also with metabolic disorders and cancer. Here, we review the links between the gut microbiota, gut barrier function and the onset of low-grade inflammation in the development of gastrointestinal cancer. We also describe the mechanisms by which specific microorganism-associated molecular patterns crosstalk with the immune system and how the metabolic activity of bacteria induces specific signalling pathways beyond the gut that eventually trigger carcinogenesis.
Collapse
Affiliation(s)
- Patrice D Cani
- Université catholique de Louvain, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Metabolism and Nutrition Research Group, Brussels, Belgium.
| | - Benedicte F Jordan
- Université catholique de Louvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Brussels, Belgium
| |
Collapse
|
108
|
Zhao Y, Lin L, Li J, Xiao Z, Chen B, Wan L, Li M, Wu X, Hin Cho C, Shen J. CD4+ T cells in obesity and obesity-associated diseases. Cell Immunol 2018; 332:1-6. [DOI: 10.1016/j.cellimm.2018.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/29/2018] [Accepted: 08/20/2018] [Indexed: 02/08/2023]
|
109
|
Zeng Q, Sun X, Xiao L, Xie Z, Bettini M, Deng T. A Unique Population: Adipose-Resident Regulatory T Cells. Front Immunol 2018; 9:2075. [PMID: 30323806 PMCID: PMC6172295 DOI: 10.3389/fimmu.2018.02075] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 08/21/2018] [Indexed: 01/15/2023] Open
Abstract
Regulatory T (Treg) cell is well known for its anti-inflammatory function in a variety of tissues in health and disease. Accordingly, Treg cells that reside in adipose tissue exhibit specific phenotypes. Their numbers are regulated by age, gender and environmental factors, such as diet and cold stimulation. Adipose-resident Treg cells have been suggested to be critical regulators of immune and metabolic microenvironment in adipose tissue, as well as involved in pathogenesis of obesity-related metabolic disorders. This review surveys existing information on adipose-resident Treg cells. We first describe the origin, phenotype and function of adipose-resident Treg cells. We then describe the major regulators of adipose-resident Treg cells, and discuss how the adipose-resident Treg cells are regulated in lean and obese conditions, especially in humans. Finally, we highlight their therapeutic potential in obesity-related disorders.
Collapse
Affiliation(s)
- Qin Zeng
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiao Sun
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha, China
| | - Liuling Xiao
- Center for Bioenergetics, Weill Cornell Medical College, Houston Methodist Research Institute, Houston, TX, United States
| | - Zhiguo Xie
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Maria Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, McNair Medical Institute, Texas Children's Hospital, Houston, TX, United States
| | - Tuo Deng
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha, China.,Center for Bioenergetics, Weill Cornell Medical College, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
110
|
Th17 and Treg lymphocytes in obesity and Type 2 diabetic patients. Clin Immunol 2018; 197:77-85. [PMID: 30218707 DOI: 10.1016/j.clim.2018.09.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/09/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022]
Abstract
Assumption that the pathogenesis of obesity-associated type 2 diabetes (T2DM) encompasses inflammation and autoimmune aspects is increasingly recognized. In the state of obesity and T2DM, the imbalance of T helper 17 (Th17) cells and regulatory T (Treg) cells are observed. These alterations reflect a loss of T cell homeostasis, which may contribute to tissue and systemic inflammation and immunity in T2DM. In this review we will discuss the accumulating data supporting the concept that Th17/Treg mediated immune responses are present in obesity-related T2DM pathogenesis, and provide evidences that restoration of Th17/Treg imbalance may be a possible therapeutic avenue for the prevention and treatment of T2DM and its complications.
Collapse
|
111
|
Zhou H, Liu F. Regulation, Communication, and Functional Roles of Adipose Tissue-Resident CD4 + T Cells in the Control of Metabolic Homeostasis. Front Immunol 2018; 9:1961. [PMID: 30233575 PMCID: PMC6134258 DOI: 10.3389/fimmu.2018.01961] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/09/2018] [Indexed: 01/21/2023] Open
Abstract
Evidence accumulated over the past few years has documented a critical role for adipose tissue (AT)-resident immune cells in the regulation of local and systemic metabolic homeostasis. In the lean state, visceral adipose tissue (VAT) is predominated by anti-inflammatory T-helper 2 (Th2) and regulatory T (Treg) cell subsets. As obesity progresses, the population of Th2 and Treg cells decreases while that of the T-helper 1 (Th1) and T-helper 17 (Th17) cells increases, leading to augmented inflammation and insulin resistance. Notably, recent studies also suggest a potential role of CD4+ T cells in the control of thermogenesis and energy homeostasis. In this review, we have summarized recent advances in understanding the characteristics and functional roles of AT CD4+ T cell subsets during obesity and energy expenditure. We have also discussed new findings on the crosstalk between CD4+ T cells and local antigen-presenting cells (APCs) including adipocytes, macrophages, and dendritic cells (DCs) to regulate AT function and metabolic homeostasis. Finally, we have highlighted the therapeutic potential of targeting CD4+ T cells as an effective strategy for the treatment of obesity and its associated metabolic diseases.
Collapse
Affiliation(s)
- Haiyan Zhou
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center of Central South University, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Liu
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center of Central South University, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
112
|
Gonzalez LL, Garrie K, Turner MD. Type 2 diabetes - An autoinflammatory disease driven by metabolic stress. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3805-3823. [PMID: 30251697 DOI: 10.1016/j.bbadis.2018.08.034] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/27/2018] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes has traditionally been viewed as a metabolic disorder characterised by chronic high glucose levels, insulin resistance, and declining insulin secretion from the pancreas. Modern lifestyle, with abundant nutrient supply and reduced physical activity, has resulted in dramatic increases in the rates of obesity-associated disease conditions, including diabetes. The associated excess of nutrients induces a state of systemic low-grade chronic inflammation that results from production and secretion of inflammatory mediators from the expanded pool of activated adipocytes. Here, we review the mechanisms by which obesity induces adipose tissue dysregulation, detailing the roles of adipose tissue secreted factors and their action upon other cells and tissues central to glucose homeostasis and type 2 diabetes. Furthermore, given the emerging importance of adipokines, cytokines and chemokines in disease progression, we suggest that type 2 diabetes should now be viewed as an autoinflammatory disease, albeit one that is driven by metabolic dysregulation.
Collapse
Affiliation(s)
- Laura L Gonzalez
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| | - Karin Garrie
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| | - Mark D Turner
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom.
| |
Collapse
|
113
|
Loss of ovarian function in association with a high-fat diet promotes insulin resistance and disturbs adipose tissue immune homeostasis. J Nutr Biochem 2018; 57:93-102. [DOI: 10.1016/j.jnutbio.2018.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 12/15/2022]
|
114
|
Woo HJ, Reifman J. Genetic interaction effects reveal lipid-metabolic and inflammatory pathways underlying common metabolic disease risks. BMC Med Genomics 2018; 11:54. [PMID: 29925367 PMCID: PMC6011398 DOI: 10.1186/s12920-018-0373-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Common metabolic diseases, including type 2 diabetes, coronary artery disease, and hypertension, arise from disruptions of the body's metabolic homeostasis, with relatively strong contributions from genetic risk factors and substantial comorbidity with obesity. Although genome-wide association studies have revealed many genomic loci robustly associated with these diseases, biological interpretation of such association is challenging because of the difficulty in mapping single-nucleotide polymorphisms (SNPs) onto the underlying causal genes and pathways. Furthermore, common diseases are typically highly polygenic, and conventional single variant-based association testing does not adequately capture potentially important large-scale interaction effects between multiple genetic factors. METHODS We analyzed moderately sized case-control data sets for type 2 diabetes, coronary artery disease, and hypertension to characterize the genetic risk factors arising from non-additive, collective interaction effects, using a recently developed algorithm (discrete discriminant analysis). We tested associations of genes and pathways with the disease status while including the cumulative sum of interaction effects between all variants contained in each group. RESULTS In contrast to non-interacting SNP mapping, which produced few genome-wide significant loci, our analysis revealed extensive arrays of pathways, many of which are involved in the pathogenesis of these metabolic diseases but have not been directly identified in genetic association studies. They comprised cell stress and apoptotic pathways for insulin-producing β-cells in type 2 diabetes, processes covering different atherosclerotic stages in coronary artery disease, and elements of both type 2 diabetes and coronary artery disease risk factors (cell cycle, apoptosis, and hemostasis) associated with hypertension. CONCLUSIONS Our results support the view that non-additive interaction effects significantly enhance the level of common metabolic disease associations and modify their genetic architectures and that many of the expected genetic factors behind metabolic disease risks reside in smaller genotyping samples in the form of interacting groups of SNPs.
Collapse
Affiliation(s)
- Hyung Jun Woo
- Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| | - Jaques Reifman
- Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA.
| |
Collapse
|
115
|
Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The role of infiltrating immune cells in dysfunctional adipose tissue. Cardiovasc Res 2018; 113:1009-1023. [PMID: 28838042 PMCID: PMC5852626 DOI: 10.1093/cvr/cvx108] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue (AT) dysfunction, characterized by loss of its homeostatic functions, is a hallmark of non-communicable diseases. It is characterized by chronic low-grade inflammation and is observed in obesity, metabolic disorders such as insulin resistance and diabetes. While classically it has been identified by increased cytokine or chemokine expression, such as increased MCP-1, RANTES, IL-6, interferon (IFN) gamma or TNFα, mechanistically, immune cell infiltration is a prominent feature of the dysfunctional AT. These immune cells include M1 and M2 macrophages, effector and memory T cells, IL-10 producing FoxP3+ T regulatory cells, natural killer and NKT cells and granulocytes. Immune composition varies, depending on the stage and the type of pathology. Infiltrating immune cells not only produce cytokines but also metalloproteinases, reactive oxygen species, and chemokines that participate in tissue remodelling, cell signalling, and regulation of immunity. The presence of inflammatory cells in AT affects adjacent tissues and organs. In blood vessels, perivascular AT inflammation leads to vascular remodelling, superoxide production, endothelial dysfunction with loss of nitric oxide (NO) bioavailability, contributing to vascular disease, atherosclerosis, and plaque instability. Dysfunctional AT also releases adipokines such as leptin, resistin, and visfatin that promote metabolic dysfunction, alter systemic homeostasis, sympathetic outflow, glucose handling, and insulin sensitivity. Anti-inflammatory and protective adiponectin is reduced. AT may also serve as an important reservoir and possible site of activation in autoimmune-mediated and inflammatory diseases. Thus, reciprocal regulation between immune cell infiltration and AT dysfunction is a promising future therapeutic target.
Collapse
Affiliation(s)
- Tomasz J Guzik
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Dominik S Skiba
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Rhian M Touyz
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - David G Harrison
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Department of Clinical Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
116
|
Shi HJ, Li YF, Ji WJ, Lin ZC, Cai W, Chen T, Yuan B, Niu XL, Li HY, Shu W, Li YM, Yuan F, Zhou X, Zhang Z. Evaluation of Visceral Adipose Tissue Oxygenation by Blood Oxygen Level-Dependent MRI in Zucker Diabetic Fatty Rats. Obesity (Silver Spring) 2018; 26:1017-1025. [PMID: 29732719 DOI: 10.1002/oby.22191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 03/15/2018] [Accepted: 03/17/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE This study aimed to investigate the feasibility of blood oxygen level-dependent magnetic resonance imaging (BOLD-MRI) to evaluate visceral adipose tissue (VAT) oxygenation in Zucker diabetic fatty (ZDF) rats and its associations with systemic metaflammation. METHODS Five-week-old ZDF rats and Zucker lean (ZL) rats were fed a high-fat diet (HFD) for 18 weeks. A baseline BOLD-MRI scan of perirenal adipose tissue was performed after 8 weeks of HFD feeding, and then the rats were randomized to receive pioglitazone or a vehicle for the following 10 weeks. At sacrifice, BOLD-MRI scan, Hypoxyprobe-1 injection, and circulating T helper 17 (Th17), regulatory T (Treg) cells, and monocyte subtype flow cytometry analysis were performed. RESULTS HFD feeding led to a significant increase in VAT BOLD-MRI R2* signals (20.14 ± 0.23 per second vs. 21.53 ± 0.20 per second; P = 0.012), an indicator for decreased oxygenation. R2* signal was significantly correlated with VAT pimonidazole adduct-positive area, insulin resistance, Th17 and Treg cells, CD43 + and CD43+ + monocyte subtypes, and VAT macrophage infiltration. Pioglitazone treatment improved the insulin resistance and was associated with a delayed progression of VAT oxygenation. CONCLUSIONS This work demonstrated the feasibility of BOLD-MRI for detecting the VAT oxygenation status in ZDF rats, and the BOLD-MRI signals were associated with insulin resistance and systemic metaflammation in ZDF rats during the development of obesity.
Collapse
Affiliation(s)
- Hong-Jian Shi
- Department of Radiology, Pingjin Hospital, Logistics University of PAPF, Tianjin, China
| | - Yan-Feng Li
- Department of Radiology, Pingjin Hospital, Logistics University of PAPF, Tianjin, China
| | - Wen-Jie Ji
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Zhi-Chun Lin
- Department of Radiology, Pingjin Hospital, Logistics University of PAPF, Tianjin, China
| | - Wei Cai
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Tao Chen
- Department of Radiology, Pingjin Hospital, Logistics University of PAPF, Tianjin, China
| | - Bin Yuan
- Department of Radiology, Pingjin Hospital, Logistics University of PAPF, Tianjin, China
| | - Xiu-Long Niu
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Han-Ying Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Wen Shu
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Yu-Ming Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Fei Yuan
- Department of Radiology, Pingjin Hospital, Logistics University of PAPF, Tianjin, China
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Zhuoli Zhang
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
117
|
Ou X, Guan J, Chen JS, Ying JC, Liu XP, Tian PK, Liu JK, Nie LP, Zhao Y, Yu GY. LAP +CD4 + T cells are elevated among the peripheral blood mononuclear cells and tumor tissue of patients with hepatocellular carcinoma. Exp Ther Med 2018; 16:788-796. [PMID: 30116333 PMCID: PMC6090257 DOI: 10.3892/etm.2018.6229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 04/26/2018] [Indexed: 12/13/2022] Open
Abstract
The purpose of the present study was to investigate the role of latency-associated peptide (LAP)+CD4+T cells in hepatocellular carcinoma (HCC) immunity. Flow cytometric analysis was performed to detect the proportion of LAP+CD4+ T cells among the peripheral blood mononuclear cells (PBMCs) of 30 HBV-infected HCC patients at the pre-operative and post-operative stages, as well as 30 hepatitis B virus (HBV)-infected volunteers as a control group. Furthermore, tumor tissues and peri-tumor tissues from 28 patients with HCC, as well as hepatic tissues from 28 HBV-infected patients with benign lesions were subjected to immunohistochemical analysis with double staining for LAP and CD4, and the average number of the LAP+CD4+T cells in each visual field was quantified. The results indicated that the proportion of LAP+CD4+ T cells in the PBMCs of patients with HCC was significantly higher than that in the control group (1.84±0.85 vs. 0.73±0.39%, P=0.019), while it was significantly reduced after the operation (1.07±0.35, P=0.021), but still slightly, if not significantly, higher compared with that in the control group (P=0.342). Furthermore, the number of LAP+CD4+ T cells per high-magnification microscopic field (magnification, ×400) in the HCC tissues was 11.25±3.00, which was significantly higher than that in the peri-cancer tissues (5.75±1.00) and that in the HBV-infected hepatic tissues around benign lesions (2.61±0.83). In peri-cancer tissues, LAP+CD4+ T cells were also significantly more abundant than in control tissues. Furthermore, in the HCC tissues, LAP+CD4+ T cells were present as clusters in the tumor stroma and closely associated with CD4+ T lymphocytes. By contrast, in the peri-cancer liver tissues and HBV-infected hepatic tissues around benign lesions, LAP+CD4+ T cells were sparsely distributed. LAP+CD4+ T cells have marked inhibitory effects, and in the peripheral blood and tumor tissues of patients with HCC, they have an important role in the suppression of anti-tumor immunity and in the immune evasion of tumor cells.
Collapse
Affiliation(s)
- Xi Ou
- Department of Hepatobiliary and Laparoscopic Surgery, Shenzhen Hospital, Peking University, Shenzhen, Guangdong 518036, P.R. China
| | - Jing Guan
- Department of Obstetrics and Gynecology, Xiamen University Affiliated Zhongshan Hospital, Xiamen, Fujian 361004, P.R. China
| | - Jing-Sen Chen
- Department of Hepatobiliary and Laparoscopic Surgery, Shenzhen Hospital, Peking University, Shenzhen, Guangdong 518036, P.R. China
| | - Jie-Cao Ying
- Department of General Surgery, Jinhua People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| | - Xiao-Ping Liu
- Department of Hepatobiliary and Laparoscopic Surgery, Shenzhen Hospital, Peking University, Shenzhen, Guangdong 518036, P.R. China
| | - Pei-Kai Tian
- Department of Hepatobiliary and Laparoscopic Surgery, Shenzhen Hospital, Peking University, Shenzhen, Guangdong 518036, P.R. China
| | - Ji-Kui Liu
- Department of Hepatobiliary and Laparoscopic Surgery, Shenzhen Hospital, Peking University, Shenzhen, Guangdong 518036, P.R. China
| | - Li-Ping Nie
- Department of Clinical Laboratory, Shenzhen Hospital, Peking University, Shenzhen, Guangdong 518036, P.R. China
| | - Yang Zhao
- Department of Pathology, Shenzhen Hospital, Peking University, Shenzhen, Guangdong 518036, P.R. China
| | - Guang-Yin Yu
- Department of Pathology, Shenzhen Hospital, Peking University, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
118
|
Drori A, Rotnemer-Golinkin D, Zolotarov L, Ilan Y. Oral Administration of CardioAid and Lunasin Alleviates Liver Damage in a High-Fat Diet Nonalcoholic Steatohepatitis Model. Digestion 2018; 96:110-118. [PMID: 28796993 DOI: 10.1159/000479734] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 07/20/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND Several of the drugs in development for treatment of nonalcoholic steatohepatitis (NASH) target liver fibrosis or have side effects that prohibit their long-term use in patients with mild to moderate disease. Lunasin is a soy-derived peptide with anti-inflammatory properties. ADM's CardioAid™ is a plant sterol extract that exerts cholesterol- and triacylglycerol-lowering effects. AIM To determine the immunomodulatory effects of CardioAid and lunasin in a high-fat diet (HFD) animal model of NASH. METHODS C57BL/6 mice on an HFD were orally administered CardioAid or lunasin for 25 weeks. The effects on the immune system, liver function, insulin resistance and lipid profile were studied. RESULTS Treatment with CardioAid and lunasin was associated with a significant decrease in the CD4/CD8 ratio and an increase in CD4+CD25+ lymphocytes. A decrease in interleukin 1-alpha serum levels and an increase in transforming growth factor beta serum levels were noted. These were associated with alleviation of liver damage as indicated by a significant decrease in liver enzymes and improvement in the histological nonalcoholic fatty liver disease activity score (NAS). Decreases in both serum triglyceride and serum glucose levels were observed in treated mice. A decrease in total body fat measured by EchoMRI was also observed in treated mice. CONCLUSIONS CardioAid and lunasin exerted hepatoprotective and glucose-protective effects in an HFD NASH model. These data and the high-safety profiles of CardioAid and Lunasin support their use in patients in the early stages of NASH to prevent deterioration due to the disease.
Collapse
Affiliation(s)
- Ariel Drori
- Gastroenterology and Liver Units, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | |
Collapse
|
119
|
Yang M, Su L, Tao Q, Zhang C, Wu Y, Liu J. Depletion of Regulatory T Cells in Visceral Adipose Tissues Contributes to Insulin Resistance in Hashimoto's Thyroiditis. Front Physiol 2018. [PMID: 29541033 PMCID: PMC5835527 DOI: 10.3389/fphys.2018.00136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Hashimoto's Thyroiditis (HT) is a common organ-specific autoimmune disorder associated with a high incidence, and insulin resistance is highly related to autoimmune. Here, we examined the insulin sensitivity in HT patients and found decreased insulin sensitivity occurred in HT patients. To explore the relationship between impaired insulin sensitivity and immune status, we established HT model mice which showed similar pathological features and immune features to HT patients. In HT model mice, reinfusion of regulatory T cells (Tregs) from peripheral blood of normal mice could improve insulin sensitivity and decrease the inflammation. Anti-CD25 antibodies blocked beneficial effects from reinfusion of Tregs, but delayed administration of anti-CD25 antibodies could not abolished the effect from Tregs. Delayed administration of anti-CD25 antibodies abolished exogenous Tregs in peripheral blood, but there were increased exogenous Tregs located to visceral adipose tissues (VATs) which modulated the expression of cytokines in VATs. These findings suggest that insulin resistance exists in HT patients and it associates with the decreased Tregs and increased inflammation in the VATs.
Collapse
Affiliation(s)
- Min Yang
- Department of Endocrinology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Li Su
- Testing and Analysis Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Qin Tao
- Department of Endocrinology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Chenxi Zhang
- Testing and Analysis Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yueyue Wu
- Department of Endocrinology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jun Liu
- Department of Endocrinology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
120
|
Ursini F, Russo E, Ruscitti P, Giacomelli R, De Sarro G. The effect of non-TNF-targeted biologics and small molecules on insulin resistance in inflammatory arthritis. Autoimmun Rev 2018; 17:399-404. [PMID: 29452240 DOI: 10.1016/j.autrev.2017.11.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 11/25/2017] [Indexed: 12/14/2022]
Abstract
Inflammatory arthritides are chronic diseases characterised by an increase in cardiovascular risk, largely attributable to the synergy between high-grade systemic inflammation and an elevated prevalence of traditional cardiovascular risk factors. Amongst the latter, insulin resistance and type 2 diabetes (T2D) play a key position. Previous studies demonstrated a potential insulin-sensitizing effect of anti-TNF biologic medications. For converse, less is known about the role of newer biologics or small molecules. For this reason, we performed a systematic review of the literature in order to identify the available data on the effect on insulin resistance of non-TNF targeting biologics and small molecules approved for the treatment of inflammatory arthritides. The search strategy initially retrieved 486 records of which only 10 articles were selected for inclusion in the final review. According to the available evidence, some of the newest molecules, in particular tocilizumab and abatacept, may have a role in improving insulin sensitivity; for converse, anakinra-mediated effect on glucose metabolism may exploit different facets of T2D pathophysiology, such as the preservation of beta-cell function. However, the data available on this issue are largely inconsistent and future, adequately designed studies are still needed to clarify the differential impact of novel therapeutics on individual pathophysiological features of T2D and other emerging cardiovascular risk factors.
Collapse
Affiliation(s)
- Francesco Ursini
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy; Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
| | - Emilio Russo
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Piero Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
| | - Roberto Giacomelli
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
| | | |
Collapse
|
121
|
Winger RC, Zamvil SS. Your nose knows how to target brain inflammation. Brain 2018; 139:1866-9. [PMID: 27343218 PMCID: PMC4939693 DOI: 10.1093/brain/aww121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Ryan C Winger
- Department of Neurology and Program in Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Scott S Zamvil
- Department of Neurology and Program in Immunology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
122
|
Tang CL, Liu ZM, Gao YR, Xiong F. Schistosoma Infection and Schistosoma-Derived Products Modulate the Immune Responses Associated with Protection against Type 2 Diabetes. Front Immunol 2018; 8:1990. [PMID: 29387059 PMCID: PMC5776330 DOI: 10.3389/fimmu.2017.01990] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/21/2017] [Indexed: 12/27/2022] Open
Abstract
Studies on parasite-induced immunoregulatory mechanisms could contribute to the development of new therapies for inflammatory diseases such as type 2 diabetes (T2D), which is a chronic inflammatory disease characterized by persistent elevated glucose levels due to insulin resistance. The association between previous Schistosoma infection and T2D has been confirmed—Schistosoma infection and Schistosoma-derived products modulate the immune system, including innate and acquired immune responses, contributing to T2D disease control. Schistosoma infections and Schistosoma-derived molecules affect the immune cell composition in adipose tissue, dampening inflammation and improving glucose tolerance. This protective role includes the polarization of immune cells to alternatively activated macrophages, dendritic cells, eosinophils, and group 2 innate lymphoid cells. Furthermore, Schistosoma infection and Schistosoma products are effective for the treatment of T2D, as they increase the number of type 2 helper T cells (Th2) and regulatory T cells (Tregs) and decrease type 1 helper T cells (Th1) and type 17 helper T cells (Th17) cells. Thus, our aim was to comprehensively review the mechanism through which Schistosoma infection and Schistosoma products modulate the immune response against T2D.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Department of Science and Education, Wuchang Hospital, Wuhan, China
| | - Zhi-Ming Liu
- Department of Science and Education, Wuchang Hospital, Wuhan, China
| | - Yan Ru Gao
- Medical Department, City College, Wuhan University of Science and Technology, Wuhan, China
| | - Fei Xiong
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
123
|
Tschöpe C, Van Linthout S, Kherad B. Heart Failure with Preserved Ejection Fraction and Future Pharmacological Strategies: a Glance in the Crystal Ball. Curr Cardiol Rep 2017; 19:70. [PMID: 28656481 DOI: 10.1007/s11886-017-0874-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW The current definition of heart failure is mainly based on an inappropriate measure of cardiac function, i.e., left ventricular ejection fraction (LVEF). The initial sole entity, heart failure with reduced ejection fraction (HFrEF, LVEF <40%), was complemented by the addition of heart failure with preserved ejection fraction (HFpEF, LVEF ≥50%) and most recently, heart failure with mid-range ejection fraction (HFmrEF, LVEF 40-49%). Initially, HFpEF was believed to be a purely left ventricular diastolic dysfunction. Pathophysiological concepts of HFpEF have changed considerably during the last years. In addition to intrinsic cardiac mechanisms, the heart failure pathogenesis is increasingly considered as driven by non-cardiac systemic processes including metabolic disorders, ischemic conditions, and pro-inflammatory/pro-fibrotic or immunological alterations. Presentation and pathophysiology of HFpEF is heterogeneous, and its management remains a challenge since evidence of therapeutic benefits is scarce. Up to now, there are no therapies improving survival in patients with HFpEF. RECENT FINDINGS Several results from clinical and preclinical interventions targeting non-cardiac mechanisms or non-pharmacological interventions including new anti-diabetic or anti-inflammatory drugs, mitochondrial-targeted anti-oxidants, anti-fibrotic strategies, microRNases incl. antagomirs, cell therapeutic options, and high-density lipoprotein-raising strategies are promising and under further investigation. This review addresses mechanisms and available data of current best clinical practice and novel approaches towards HFpEF.
Collapse
Affiliation(s)
- Carsten Tschöpe
- Department of Cardiology, Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany. .,Berliner Zentrum für Regenerative Therapien (BCRT), Campus Virchow Klinikum (CVK), Berlin, Germany. .,Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Berlin, Germany. .,Campus Virchow Clinic, Department of Cardiology, Charité - Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany.
| | - Sophie Van Linthout
- Berliner Zentrum für Regenerative Therapien (BCRT), Campus Virchow Klinikum (CVK), Berlin, Germany.,Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Berlin, Germany.,Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Föhrerstrasse 15, 13353, Berlin, Germany
| | - Behrouz Kherad
- Department of Cardiology, Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany.,Campus Virchow Clinic, Department of Cardiology, Charité - Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany.,Privatpraxis Dr. Kherad, Große Hamburger Strasse 5-11, 10115, Berlin, Germany
| |
Collapse
|
124
|
Integrated Immunomodulatory Mechanisms through which Long-Chain n-3 Polyunsaturated Fatty Acids Attenuate Obese Adipose Tissue Dysfunction. Nutrients 2017; 9:nu9121289. [PMID: 29186929 PMCID: PMC5748740 DOI: 10.3390/nu9121289] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is a global health concern with rising prevalence that increases the risk of developing other chronic diseases. A causal link connecting overnutrition, the development of obesity and obesity-associated co-morbidities is visceral adipose tissue (AT) dysfunction, characterized by changes in the cellularity of various immune cell populations, altered production of inflammatory adipokines that sustain a chronic state of low-grade inflammation and, ultimately, dysregulated AT metabolic function. Therefore, dietary intervention strategies aimed to halt the progression of obese AT dysfunction through any of the aforementioned processes represent an important active area of research. In this connection, fish oil-derived dietary long-chain n-3 polyunsaturated fatty acids (PUFA) in the form of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been demonstrated to attenuate obese AT dysfunction through multiple mechanisms, ultimately affecting AT immune cellularity and function, adipokine production, and metabolic signaling pathways, all of which will be discussed herein.
Collapse
|
125
|
Morel S, Kwak B, Rohner-Jeanrenaud F, Steffens S, Molica F. Adipokines at the crossroad between obesity and cardiovascular disease. Thromb Haemost 2017; 113:553-66. [DOI: 10.1160/th14-06-0513] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/18/2014] [Indexed: 12/31/2022]
Abstract
SummaryObesity, and especially excessive visceral adipose tissue accumulation, is considered as a low-grade inflammatory state that is responsible for adipocyte dysfunction and associated metabolic disorders. Adipose tissue displays endocrine functions by releasing pro- or antiinflammatory bioactive molecules named adipokines. An altered expression of these molecules, provoked by obesity or adipocyte dysregulation, contributes to major metabolic diseases such as insulin resistance and type 2 diabetes mellitus that are important risk factors for cardiovascular disease. However, obesity is also characterised by the expansion of perivascular adipose tissue that acts locally via diffusion of adipokines into the vascular wall. Local inflammation within blood vessels induced by adipokines contributes to the onset of endothelial dysfunction, atherosclerosis and thrombosis, but also to vascular remodelling and hypertension. A fast expansion of obesity is expected in the near future, which will rapidly increase the incidence of these cardiovascular diseases. The focus of this review is to summarise the link between metabolic and cardiovascular disease and discuss current treatment approaches, limitations and future perspectives for more targeted therapies.
Collapse
|
126
|
Zhan J, Huang L, Ma H, Chen H, Yang Y, Tan S, Song W, Zhao W, Dai X. Reduced inflammatory responses of follicular helper T cell promote the development of regulatory B cells after Roux-en-Y gastric bypass. Clin Exp Pharmacol Physiol 2017; 44:556-565. [PMID: 28222218 DOI: 10.1111/1440-1681.12740] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 02/06/2023]
Abstract
Bariatric surgery is currently the most effective strategy in treating severe obesity and its comorbidities, such as type 2 diabetes (T2D). However, the mechanism through which bariatric surgery mediates its benefits is not completely understood. Since obesity and T2D represent yet another inflammatory disease, and follicular helper T (Tfh) cells play important roles in inflammatory disorders, we investigated whether the Tfh activity was altered after Roux-en-Y gastric bypass (RYGB), one of the most common bariatric surgery procedures. We found that the Tfh cells after RYGB were not significantly changed in number, but presented altered cytokine secretion profile, including lower interferon (IFN)-γ, interleukin (IL)-2, IL-4, and IL-17 secretion. Tfh cells after RYGB also downregulated inducible co-stimulator and programmed death-1. Interestingly, after Tfh cell-naive B cell coculture, Tfh cells after RYGB secreted more IL-10 than autologous Tfh cells before RYGB. The frequencies of IL-10-expressing and transforming growth factor (TGF)-β-expressing regulatory B cells after Tfh cell-naive B cell coculture were directly correlated with the frequency of IL-10-expressing Tfh cells. Depletion of IL-10 in the coculture, however, resulted in fewer regulatory B cells. Finally, patients with greater increase in IL-10-expressing Tfh cells presented further reductions in body mass index, glycaemia, and body fat percentage. Together, these data demonstrated that the Tfh cells after RYGB presented lower inflammatory status and secreted higher IL-10, through which these Tfh cells promoted the development of regulatory B cells. Higher IL-10-expressing Tfh cell level also predicted better patient response to RYGB.
Collapse
Affiliation(s)
- Junfang Zhan
- Health Management Center, Guangzhou First People's Hospital, Guangzhou Medical College, Guangzhou, China
| | - Liyu Huang
- Surgical Center for Obesity and Diabetes, Jinshazhou Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiyong Ma
- Surgical Center for Obesity and Diabetes, Jinshazhou Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huan Chen
- Surgical Center for Obesity and Diabetes, Jinshazhou Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuan Yang
- Surgical Center for Obesity and Diabetes, Jinshazhou Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sheng Tan
- Surgical Center for Obesity and Diabetes, Jinshazhou Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wendy Song
- DICAT Biomedical Computation Centre, Vancouver, BC, Canada
| | - Weiguo Zhao
- Surgical Center for Obesity and Diabetes, Jinshazhou Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojiang Dai
- Surgical Center for Obesity and Diabetes, Jinshazhou Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
127
|
Harrison CA, Taren D. How poverty affects diet to shape the microbiota and chronic disease. Nat Rev Immunol 2017; 18:279-287. [PMID: 29109542 DOI: 10.1038/nri.2017.121] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Here, we discuss the link between nutrition, non-communicable chronic diseases and socio-economic standing, with a special focus on the microbiota. We provide a theoretical framework and several lines of evidence from both animal and human studies that support the idea that income inequality is an underlying factor for the maladaptive changes seen in the microbiota in certain populations. We propose that this contributes to the health disparities that are seen between lower-income and higher-income populations in high-income countries.
Collapse
Affiliation(s)
- Christy A Harrison
- Departments of Immunobiology and Pediatrics, University of Arizona, Tucson, USA
| | - Douglas Taren
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, USA
| |
Collapse
|
128
|
Borovcanin MM, Jovanovic I, Radosavljevic G, Pantic J, Minic Janicijevic S, Arsenijevic N, Lukic ML. Interleukin-6 in Schizophrenia-Is There a Therapeutic Relevance? Front Psychiatry 2017; 8:221. [PMID: 29163240 PMCID: PMC5681495 DOI: 10.3389/fpsyt.2017.00221] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022] Open
Abstract
Renewing interest in immune aspects of schizophrenia and new findings about the brain-fat axis encourage us to discuss the possible role of interleukin-6 (IL-6) in schizophrenia. Previously, it was suggested that a primary alteration of the innate immune system may be relevant in schizophrenia. Functional dichotomy of IL-6 suggests that this chemical messenger may be responsible for regulating the balance between pro- and anti-inflammatory responses, with tissue-specific properties at the periphery and in the central nervous system. Specific phase of this chronic and deteriorating disorder must be considered, which can involve IL-6 in acute or possible chronic inflammation and/or autoimmunity. We give an overview of IL-6 role in the onset and progression of this disorder, also considering cognitive impairment and metabolic changes in patients with schizophrenia. Data suggest that decreased serum level of IL-6 following antipsychotic therapy could be predisposing factor for the development of obesity and obesity-related metabolic disorders in schizophrenia. As we reviewed, the IL-6 plays significant role in disease genesis and progression, so the use of specific inhibitors may not only be beneficial for exacerbation and alleviation of positive symptoms, but may attenuate cognitive impairment in patients with schizophrenia.
Collapse
Affiliation(s)
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Gordana Radosavljevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jelena Pantic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag L. Lukic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
129
|
Khoury T, Rotnemer-Golinkin D, Shabat Y, Zolotarovya L, Ilan Y. Oral Co-administration of Soy-derived Extracts with Alcohol or with Sugar-sweetened Beverages Exerts Liver and Sugar Protective Effects. J Clin Transl Hepatol 2017; 5:208-215. [PMID: 28936402 PMCID: PMC5606967 DOI: 10.14218/jcth.2017.00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/14/2017] [Accepted: 04/21/2017] [Indexed: 12/04/2022] Open
Abstract
Background and Aims: Both alcoholic drinks and high sugar-containing soft drinks cause major health problems worldwide. Oral administration of OS and M1 soy-derived extracts has been shown to alleviate liver injury in animal models. The aim of the present study was to determine the liver- and sugar-protective effect of OS and M1 soy-derived extracts when added to alcohol and sugar-enriched drinks. Methods: Mice were treated with alcohol or high sugar-containing drinks, with and without administration of a combination of OS and M1 soy extracts. Mice were observed for the effects on liver injury, glucose metabolism, and the immune system. Results: Co-administration of the soy extracts OS and M1 significantly alleviated the liver injury induced by acute alcohol, as evidenced by decreased liver enzymes. These beneficial effects were associated with promotion of subsets of regulatory T lymphocytes and with a trend towards a pro-inflammatory to an anti-inflammatory cytokine shift. Co-administration of OS M1 soy extracts with sugar-sweetened beverages significantly alleviated the increases in serum sugar levels. Conclusions: OS and M1 extracts exert a synergistic hepato- and glucose-protective effect in models of alcohol-induced liver damage and soft drinks-associated increases in serum glucose. These extracts may provide a solution to the two pressing health problems.
Collapse
Affiliation(s)
- Tawfik Khoury
- *Correspondence to: Tawfik Khoury, Gastroenterology and Liver Units, Department of Medicine, Hebrew University-Hadassah Medical Center, PO Box 1200, Jerusalem IL 91120, Israel. Tel: +972-509870611, E-mails: ,
| | | | | | | | | |
Collapse
|
130
|
Becker M, Levings MK, Daniel C. Adipose-tissue regulatory T cells: Critical players in adipose-immune crosstalk. Eur J Immunol 2017; 47:1867-1874. [PMID: 28849586 DOI: 10.1002/eji.201646739] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/13/2017] [Accepted: 08/24/2017] [Indexed: 12/19/2022]
Abstract
Obesity and type-2 diabetes (T2D) are associated with metabolic defects and inflammatory processes in fat depots. FoxP3+ regulatory T cells (Tregs) control immune tolerance, and have an important role in controlling tissue-specific inflammation. In this mini-review we will discuss current insights into how cross-talk between T cells and adipose tissue shapes the inflammatory environment in obesity-associated metabolic diseases, focusing on the role of CD4+ T cells and Tregs. We will also highlight potential opportunities for how the immunoregulatory properties of Tregs could be harnessed to control inflammation in obesity and T2D and emphasize the critical need for more research on humans to establish mechanisms that are conserved in both mice and humans.
Collapse
Affiliation(s)
- Maike Becker
- Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Munich, Germany.,Deutsches Zentrum für Diabetesforschung (DZD), Munich, Germany
| | - Megan K Levings
- Department of Surgery University of British Columbia and BC Children's Hospital Research Institute, Vancouver, B.C. Canada
| | - Carolin Daniel
- Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Munich, Germany.,Deutsches Zentrum für Diabetesforschung (DZD), Munich, Germany
| |
Collapse
|
131
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an overview of shared dysregulation of the hypothalamic-pituitary-adrenal (HPA) and brain-gut-microbiome (BGM) axes associated with depression and type 2 diabetes (T2D). Clinical implications and future research are also discussed. RECENT FINDINGS Both depression and T2D are associated with dysregulation of the HPA and BGM axes. These pathways regulate immune function, glucose metabolism, and sleep, which are altered in both illnesses. Dysregulation of homeostatic brain-body pathways may be positively influenced through different therapeutic actions, including psychotherapy, healthy eating, physical activity, sleep promotion, and certain anti-inflammatory or antidepressant medications. While the causal nature of the relationship between depression and T2D remains unclear, these conditions share dysregulation of homeostatic brain-body pathways that are central to mental and physical health. Better understanding of this dysregulation may provide opportunities for interventions that could benefit both conditions. Future research should examine the additive burden of depression and T2D on HPA and BGM dysregulation and better differentiate depression from emotional distress.
Collapse
Affiliation(s)
- Claire J Hoogendoorn
- Ferkauf Graduate School of Psychology, Yeshiva University, 1165 Morris Park Avenue, Rousso Building, Bronx, NY, 10461, USA.
| | - Juan F Roy
- Ferkauf Graduate School of Psychology, Yeshiva University, 1165 Morris Park Avenue, Rousso Building, Bronx, NY, 10461, USA
| | - Jeffrey S Gonzalez
- Ferkauf Graduate School of Psychology, Yeshiva University, 1165 Morris Park Avenue, Rousso Building, Bronx, NY, 10461, USA
- Department of Medicine (Endocrinology), Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
132
|
Ogura M, Deng S, Preston-Hurlburt P, Ogura H, Shailubhai K, Kuhn C, Weiner HL, Herold KC. Oral treatment with foralumab, a fully human anti-CD3 monoclonal antibody, prevents skin xenograft rejection in humanized mice. Clin Immunol 2017; 183:240-246. [PMID: 28739191 DOI: 10.1016/j.clim.2017.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/14/2017] [Accepted: 07/14/2017] [Indexed: 01/12/2023]
Abstract
Oral administration of biologics may be a feasible approach for immune therapy that improves drug safety and potentiates mechanisms of tolerance at mucosal barriers. We tested the ability of a fully human non-FcR binding anti-CD3 mAb, foralumab, to prevent skin xenograft rejection in mice with human immune systems. At an intragastric dose of 15μg, the drug could transit through the small bowel. Serum absorption and binding of lymphoid cells was seen and proliferative responses of splenic CD8+ T cells to mitogen were reduced. Five consecutive daily doses, then weekly dosing led to indefinite graft acceptance without depletion of peripheral T cells. Proliferative and cytokine responses to activation of splenocytes with PHA were reduced. The serum levels of IL-10 but not TNF were increased 6days after application of the skin graft. Oral treatment with anti-CD3 mAb may represent a feasible approach for immune modulation.
Collapse
Affiliation(s)
- Mineko Ogura
- Department of Immunobiology, Yale University, New Haven, CT, United States
| | - Songyan Deng
- Department of Immunobiology, Yale University, New Haven, CT, United States
| | | | - Hideki Ogura
- Department of Immunobiology, Yale University, New Haven, CT, United States
| | - Kunwar Shailubhai
- Tiziana Life Sciences, R&D Center, 3805 Old Easton Road, Doylestown, PA 18902, United States
| | - Chantal Kuhn
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT, United States; Department of Internal Medicine, Yale University, New Haven, CT, United States.
| |
Collapse
|
133
|
Deng T, Liu J, Deng Y, Minze L, Xiao X, Wright V, Yu R, Li XC, Blaszczak A, Bergin S, DiSilvestro D, Judd R, Bradley D, Caligiuri M, Lyon CJ, Hsueh WA. Adipocyte adaptive immunity mediates diet-induced adipose inflammation and insulin resistance by decreasing adipose Treg cells. Nat Commun 2017. [PMCID: PMC5510177 DOI: 10.1038/ncomms15725] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
134
|
de Candia P, De Rosa V, Gigantino V, Botti G, Ceriello A, Matarese G. Immunometabolism of human autoimmune diseases: from metabolites to extracellular vesicles. FEBS Lett 2017. [PMID: 28649760 DOI: 10.1002/1873-3468.12733] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunometabolism focuses on the mechanisms regulating the impact of metabolism on lymphocyte activity and autoimmunity outbreak. The adipose tissue is long known to release adipokines, either pro- or anti-inflammatory factors bridging nutrition and immune function. More recently, adipocytes were discovered to also release extracellular vesicles (EVs) containing a plethora of biological molecules, including metabolites and microRNAs, which can regulate cell function/metabolism in distant tissues, suggesting that immune regulatory function by the adipose tissue may be far more complex than originally thought. Moreover, EVs were also identified as important mediators of immune cell-to-cell communication, adding a further microenvironmental mechanism of plasticity to fine-tune specific lymphocyte responses. This Review will first focus on the known mechanisms by which metabolism impacts immune function, presenting a systemic (nutrition and long-ranged adipokines) and a cellular point of view (metabolic pathway derangement in autoimmunity). It will then discuss the new discoveries concerning how EVs may act as nanometric vehicles integrating immune/metabolic responses at the level of the extracellular environment and affecting pathological processes.
Collapse
Affiliation(s)
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy
| | | | - Gerardo Botti
- IRCCS Istituto Nazionale Tumori, Fondazione G. Pascale, Naples, Italy
| | | | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli 'Federico II', Naples, Italy
| |
Collapse
|
135
|
Cao H, Tuo L, Tuo Y, Xia Z, Fu R, Liu Y, Quan Y, Liu J, Yu Z, Xiang M. Immune and Metabolic Regulation Mechanism of Dangguiliuhuang Decoction against Insulin Resistance and Hepatic Steatosis. Front Pharmacol 2017; 8:445. [PMID: 28736524 PMCID: PMC5500616 DOI: 10.3389/fphar.2017.00445] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/21/2017] [Indexed: 12/14/2022] Open
Abstract
Dangguiliuhuang decoction (DGLHD) is a traditional Chinese medicine (TCM) formula, which mainly consists of angelica, radix rehmanniae, radix rehmanniae praeparata, scutellaria baicalensis, coptis chinensis, astragalus membranaceus, and golden cypress, and used for the treatment of diabetes and some autoimmune diseases. In this study, we explored the potential mechanism of DGLHD against insulin resistance and fatty liver in vivo and in vitro. Our data revealed that DGLHD normalized glucose and insulin level, increased the expression of adiponectin, diminished fat accumulation and lipogenesis, and promoted glucose uptake. Metabolomic analysis also demonstrated that DGLHD decreased isoleucine, adenosine, and cholesterol, increased glutamine levels in liver and visceral adipose tissue (VAT) of ob/ob mice. Importantly, DGLHD promoted the shift of pro-inflammatory to anti-inflammatory cytokines, suppressed T lymphocytes proliferation, and enhanced regulatory T cells (Tregs) differentiation. DGLHD also inhibited dendritic cells (DCs) maturation, attenuated DCs-stimulated T cells proliferation and secretion of IL-12p70 cytokine from DCs, and promoted the interaction of DCs with Tregs. Further studies indicated that the changed PI3K/Akt signaling pathway and elevated PPAR-γ expression were not only observed with the ameliorated glucose and lipid metabolism in adipocytes and hepatocytes, but also exhibited in DCs and T cells by DGLHD. Collectively, our results suggest that DGLHD exerts anti-insulin resistant and antisteatotic effects by improving abnormal immune and metabolic homeostasis. And DGLHD may be a novel approach to the treatment of obesity-related insulin resistance and hepatic steatosis.
Collapse
Affiliation(s)
- Hui Cao
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Lingling Tuo
- Department of Traditional Chinese Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Yali Tuo
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Ziyun Xia
- Department of Pharmacy, China Pharmaceutical UniversityNanjing, China
| | - Rong Fu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Yang Liu
- Synergy Innovation Center of Biological Peptide Antidiabetics of Hubei Province, School of Life Science, Wuchang University of TechnologyWuhan, China
| | - Yihong Quan
- Department of Traditional Chinese Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Jue Liu
- Department of Traditional Chinese Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Zhihong Yu
- Department of Traditional Chinese Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| |
Collapse
|
136
|
Inflammation, metaflammation and immunometabolic disorders. Nature 2017; 542:177-185. [PMID: 28179656 DOI: 10.1038/nature21363] [Citation(s) in RCA: 1384] [Impact Index Per Article: 197.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/05/2017] [Indexed: 12/11/2022]
Abstract
Proper regulation and management of energy, substrate diversity and quantity, as well as macromolecular synthesis and breakdown processes, are fundamental to cellular and organismal survival and are paramount to health. Cellular and multicellular organization are defended by the immune response, a robust and critical system through which self is distinguished from non-self, pathogenic signals are recognized and eliminated, and tissue homeostasis is safeguarded. Many layers of evolutionarily conserved interactions occur between immune response and metabolism. Proper maintenance of this delicate balance is crucial for health and has important implications for many pathological states such as obesity, diabetes, and other chronic non-communicable diseases.
Collapse
|
137
|
Almon E, Khoury T, Drori A, Gingis-Velitski S, Alon S, Chertkoff R, Mushkat M, Shaaltiel Y, Ilan Y. An oral administration of a recombinant anti-TNF fusion protein is biologically active in the gut promoting regulatory T cells: Results of a phase I clinical trial using a novel oral anti-TNF alpha-based therapy. J Immunol Methods 2017; 446:21-29. [PMID: 28392436 DOI: 10.1016/j.jim.2017.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/30/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND An orally administered BY-2 plant cell-expressed recombinant anti-TNF fusion protein (PRX-106) consists of the soluble form of the human TNF receptor (TNFR) fused to the Fc component of a human IgG1 domain. Aim This study aim at determining the safety and the immune modulatory effect of an oral administration of PRX-106 in humans. METHODS Three different doses (2, 8 or 16mg/day) of PRX-106 were orally administered for five consecutive days in 14 healthy volunteered participants. Subjects were followed for safety parameters and for an effect on T lymphocytes subsets and cytokine levels. RESULTS An oral administration of PRX-106 was safe and well tolerated. The PK study showed that PRX106 is not absorbed. No effect on white blood cells and lymphocytes counts were noted. A dose dependent effect was noted on systemic lymphocytes. The oral administration of all three dosages was associated with an increase in CD4+CD25+ and CD8+CD25+ subset of suppressor lymphocytes. A marked increase in CD4+CD25+FoxP3 regulatory T cells was noted in the 8mg treated group. In addition, NKT regulatory cells, CD3+CD69+ and CD4+CD62 lymphocyte subsets increased with treatment. No changes in serum TNF alpha were observed. CONCLUSION An oral administration of the non-absorbable recombinant anti-TNF fusion protein, PRX-106, is safe, not associated with immune suppression, while inducing a favorable anti-inflammatory immune modulation. The PRX-106 may provide a safe orally administered effective anti-TNF alpha-based immune therapy for inflammatory bowel diseases and non-alcoholic steatohepatitis, as well as other autoimmune, TNF-mediated diseases.
Collapse
Affiliation(s)
| | - Tawfik Khoury
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel Drori
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | - Mordechai Mushkat
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | - Yaron Ilan
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
138
|
Rezende RM, Weiner HL. History and mechanisms of oral tolerance. Semin Immunol 2017; 30:3-11. [DOI: 10.1016/j.smim.2017.07.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/13/2017] [Indexed: 12/26/2022]
|
139
|
Zhu F, Wang A, Li Y, Liang R, Li D, Li B. Adipose Tissue-Resident Regulatory T Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1011:153-162. [DOI: 10.1007/978-94-024-1170-6_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
140
|
Xia C, Rao X, Zhong J. Role of T Lymphocytes in Type 2 Diabetes and Diabetes-Associated Inflammation. J Diabetes Res 2017; 2017:6494795. [PMID: 28251163 PMCID: PMC5307004 DOI: 10.1155/2017/6494795] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 12/30/2016] [Accepted: 01/12/2017] [Indexed: 12/20/2022] Open
Abstract
Although a critical role of adaptive immune system has been confirmed in driving local and systemic inflammation in type 2 diabetes and promoting insulin resistance, the underlying mechanism is not completely understood. Inflammatory regulation has been focused on innate immunity especially macrophage for a long time, while increasing evidence suggests T cells are crucial for the development of metabolic inflammation and insulin resistance since 2009. There was growing evidence supporting the critical implication of T cells in the pathogenesis of type 2 diabetes. We will discuss the available effect of T cells subsets in adaptive immune system associated with the procession of T2DM, which may unveil several potential strategies that could provide successful therapies in the future.
Collapse
Affiliation(s)
- Chang Xia
- College of Health Science & Nursing, Wuhan Polytechnic University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Xiaoquan Rao
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Jixin Zhong
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, USA
- *Jixin Zhong:
| |
Collapse
|
141
|
King BC, Blom AM. Non-traditional roles of complement in type 2 diabetes: Metabolism, insulin secretion and homeostasis. Mol Immunol 2016; 84:34-42. [PMID: 28012560 DOI: 10.1016/j.molimm.2016.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/01/2016] [Accepted: 12/06/2016] [Indexed: 12/20/2022]
Abstract
Type 2 Diabetes (T2D) is a disease of increasing importance and represents a growing burden on global healthcare and human health. In T2D, loss of effectiveness of insulin signaling in peripheral tissues cannot be compensated for by adequate insulin secretion, leading to hyperglycemia and resultant complications. In recent years, inflammation has been identified as a central component of T2D, both in inducing peripheral insulin resistance as well as in the pancreatic islet, where it contributes to loss of insulin secretion and death of insulin-secreting beta cells. In this review we will focus on non-traditional roles of complement proteins which have been identified in T2D-associated inflammation, beta cell secretory function, and in maintaining homeostasis of the pancreatic islet. Improved understanding of both traditional and novel roles of complement proteins in T2D may lead to new therapeutic approaches for this global disease.
Collapse
Affiliation(s)
- Ben C King
- Lund University, Department of Translation Medicine, Division of Medical Protein Chemistry, Skåne University Hospital, Malmö, Sweden.
| | - Anna M Blom
- Lund University, Department of Translation Medicine, Division of Medical Protein Chemistry, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
142
|
Abstract
PURPOSE OF REVIEW Purpose of review: It is becoming increasingly clear that some obese individuals do not develop dyslipidemia and instead remain healthy, while some normal weight individuals become dyslipidemic and unhealthy. RECENT FINDINGS The present review examines the similarities and differences between healthy and unhealthy individuals with and without obesity and discusses putative underlying mechanisms of dyslipidemia. The presence of dyslipidemia and compromised metabolic health in both lean and obese individuals suggests that the obese phenotype per se does not represent a main independent risk factor for the development of dyslipidemia and that dyslipidemia, rather than obesity, may be the driver of metabolic diseases. Notably, adipose tissue dysfunction and ectopic lipid deposition, in particular in the liver, seems a common trait of unhealthy individuals.
Collapse
Affiliation(s)
- David H Ipsen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark
| | - Pernille Tveden-Nyborg
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark
| | - Jens Lykkesfeldt
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark.
| |
Collapse
|
143
|
Ilan Y. Review article: novel methods for the treatment of non-alcoholic steatohepatitis - targeting the gut immune system to decrease the systemic inflammatory response without immune suppression. Aliment Pharmacol Ther 2016; 44:1168-1182. [PMID: 27778363 PMCID: PMC5216447 DOI: 10.1111/apt.13833] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 07/28/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND The systemic immune system plays a role in inflammation and fibrogenesis associated with non-alcoholic steatohepatitis (NASH) and has become a potential target for drug development. In particular, the gut immune system has been suggested as a means for generating signals that can target the systemic immune system. AIM To describe seven novel methods being developed for the treatment of NASH that target the gut immune system for alleviation of the systemic inflammatory response, including oral administration of fatty-liver-derived proteins, anti-CD3 antibodies, tumour necrosis factor fusion protein, anti-lipopolysaccharide antibodies, glucosylceramide, delayed-release mercaptopurine, and soy-derived extracts. METHODS A search for these methods for oral immunotherapy for NASH was conducted. RESULTS Oral administration of these compounds provides an opportunity for immune modulation without immune suppression, with the advantage of being independent of a single molecular/inflammatory pathway. These modes of oral immune therapy demonstrate superior safety profiles, such that the patient is not exposed to general immune suppression. Moreover, these approaches target the whole spectrum of the disease and may serve as adjuvants to other therapies, such that they provide a platform for treatment of concomitant disorders in patients with NASH, including diabetes and hyperlipidaemia. Most of the compounds reviewed are currently in phase II trials, and it is anticipated that the acquisition of more clinical data in the next few years will enable the use of this new class of drugs for the treatment of NASH. CONCLUSION Oral immunotherapy may provide a novel platform for the treatment of NASH.
Collapse
Affiliation(s)
- Y. Ilan
- Gastroenterology and Liver UnitsDepartment of MedicineHadassah Hebrew University Medical CenterJerusalemIsrael
| |
Collapse
|
144
|
|
145
|
Sutti S, Bruzzì S, Albano E. The role of immune mechanisms in alcoholic and nonalcoholic steatohepatitis: a 2015 update. Expert Rev Gastroenterol Hepatol 2016; 10:243-53. [PMID: 26634783 DOI: 10.1586/17474124.2016.1111758] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
So far, innate immune mechanisms have been recognized as the main responsible for the evolution of both alcoholic steatohepatitis (ASH) and nonalcoholic steatohepatitis (NASH). However, increasing evidence points toward the possible role of adaptive immune responses, as an additional factor in promoting hepatic inflammation in steatohepatitis. In this article, we discuss recent data involving circulating antibodies and lymphocyte-mediated responses in sustaining the progression of ASH and NASH to fibrosis, as well as the possible mechanisms implicated in favoring the onset of adaptive immunity in the setting of steatohepatitis.
Collapse
Affiliation(s)
- Salvatore Sutti
- a Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases , University "Amedeo Avogadro" of East Piedmont , Novara , Italy
| | - Stefania Bruzzì
- a Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases , University "Amedeo Avogadro" of East Piedmont , Novara , Italy
| | - Emanuele Albano
- a Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases , University "Amedeo Avogadro" of East Piedmont , Novara , Italy
| |
Collapse
|
146
|
Ilan Y, Gingis-Velitski S, Ben Ya'aco A, Shabbat Y, Zolotarov L, Almon E, Shaaltiel Y. A plant cell-expressed recombinant anti-TNF fusion protein is biologically active in the gut and alleviates immune-mediated hepatitis and colitis. Immunobiology 2016; 222:544-551. [PMID: 27832933 DOI: 10.1016/j.imbio.2016.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 01/02/2023]
Abstract
The orally administered BY-2 plant cell-expressed recombinant anti-TNF fusion protein (PRX-106) (n=6) consists of the soluble form of the human TNF receptor (TNFR) fused to the Fc component of a human antibody IgG1 domain. AIM To evaluate the immune modulatory effect of the oral administration of plant cells expressing PRX-106. METHODS Mice treated with Concanavalin A (ConA) to induce immune hepatitis was orally treated with cells expressing PRX-106 containing 0.5 or 5μg PRX 106. In the colitis model, TNBS-colitis was induced in mice followed by the oral administration of plant cells expressing PRX-106. The immune modulatory effect was determined through follow-up to assess the clinical effect, histology, and serum cytokine levels and by FACS analysis for lymphocyte subsets. RESULTS The oral administration of BY-2 cells expressing PRX-106 alleviated immune-mediated liver injury. Serum AST and ALT levels decreased and were comparable to those of mice that had received high-dose steroids. The beneficial effect was also observed as a marked decrease in hepatic necrosis. In the colitis model, the oral administration of BY-2 plant cells expressing PRX-106 alleviated weight loss associated with immune-mediated colitis and improved bowel histology. A reduction in I-IkB-alpha phosphorylation in treated mice was also observed. These effects were associated with a significant alteration in the distribution of CD4+CD25+FOXP3+ cells. CONCLUSIONS Plant cells expressing recombinant anti-TNF fusion protein show biological activity when orally administered, exerting an immune modulatory effect through the alleviation of immune-mediated hepatitis and immune-mediated colitis.
Collapse
Affiliation(s)
- Yaron Ilan
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| | | | - Ami Ben Ya'aco
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yehudit Shabbat
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Lidya Zolotarov
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | |
Collapse
|
147
|
Ilan Y, Ben Ya'acov A, Shabbat Y, Gingis-Velitski S, Almon E, Shaaltiel Y. Oral administration of a non-absorbable plant cell-expressed recombinant anti-TNF fusion protein induces immunomodulatory effects and alleviates nonalcoholic steatohepatitis. World J Gastroenterol 2016; 22:8760-8769. [PMID: 27818591 PMCID: PMC5075550 DOI: 10.3748/wjg.v22.i39.8760] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/21/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the immunomodulatory effect of oral administration of PRX-106 in the high-fat diet model. METHODS For 22 wk, C57BL/6 HFD-fed mice received daily oral treatments with BY-2 cells expressing recombinant anti-tumor necrosis factor alpha fusion protein (PRX-106). Mice were followed for serum liver enzyme and triglyceride levels, liver histology and intrahepatic and systemic FACS. RESULTS The orally administered non-absorbable PRX-106 was biologically active. Altered distribution of CD4+CD25+FoxP3+ between the liver and spleen and an increase in the intrasplenic-to-intrahepatic CD4+CD25+FoxP3+ ratio and a decrease in the intrasplenic-to-intrahepatic CD8+CD25+FoxP3+ ratio were observed. An increase in intrahepatic NKT cells and a decrease in the intrasplenic-to-intrahepatic NKT ratio were noted. Assessment of the CD4-to-CD8 ratios showed sequestration of CD8+ lymphocytes in the liver. These effects were associated with a decrease in serum triglyceride levels, decrease in the aspartate aminotransferase levels, serum glucose levels, and HOMA-IR score. A decrease in hepatic triglycerides content was observed in the high dose-treated mice. CONCLUSION Orally administered PRX-106 shows biological activity and exerts an immunomodulatory effect, alleviating liver damage. The data suggest that PRX-106 may provide an oral immunotherapy for nonalcoholic steatohepatitis.
Collapse
|
148
|
Abstract
The immune system has evolved to defend the organism against an almost infinite number of pathogens in a locally confined and antigen-specific manner while at the same time preserving tolerance to harmless antigens and self. Regulatory T (Treg) cells essentially contribute to an immunoregulatory network preventing excessive immune responses and immunopathology. There is emerging evidence that Treg cells not only operate in secondary lymphoid tissue but also regulate immune responses directly at the site of inflammation. Hence, the classification of Treg cells might need to be further extended by Treg cell subsets that are functionally and phenotypically polarized by their residency. In this review, we discuss recent findings on these tissue-resident Treg cell subsets and how these cells may operate in a tissue- and context-dependent manner.
Collapse
|
149
|
Bradley D, Hsueh W. Type 2 Diabetes in the Elderly: Challenges in a Unique Patient Population. JOURNAL OF GERIATRIC MEDICINE AND GERONTOLOGY 2016; 2:14. [PMID: 31692858 PMCID: PMC6831098 DOI: 10.23937/2469-5858/1510014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the older patient population, rates of Type 2 Diabetes (T2D) and obesity are reaching epidemic proportions. In fact, older patients will soon constitute the majority of patients with T2D in most developed countries. The higher prevalence of T2D in older individuals is seen in both men and women and across racial and ethnic groups. However, certain ethnic groups are disproportionately affected and successful strategies must account for these fundamental differences. T2D in old age is associated with traditional diabetes-associated complications including micro- and macro vascular disease, but is also closely related to numerous other comorbidities including cognitive impairment, urinary incontinence, sarcopenia, and increased fall risk. An overall state of chronic inflammation and dysregulated immune system may underlie these increased risks; yet our understanding of immunometabolism during the aging process remains incomplete. In addition, optimal recognition and treatment of diabetes in the elderly is hampered by a lack of relevant, high-quality studies, as the majority of clinical trial data establishing risk profiles, glycemic targets, and therapeutic interventions for T2D are not applicable for large segments of the older patient population. Simply acknowledging this gap is inadequate. We need strong evidence-based data upon which to successfully identify diabetic patients and then intervene in ways that are targeted to specific individuals within a heterogeneous group of elderly patients with T2D.
Collapse
Affiliation(s)
- David Bradley
- Department of Internal Medicine, Wexner Medical Center, The Ohio State University, USA
| | - Willa Hsueh
- Department of Internal Medicine, Wexner Medical Center, The Ohio State University, USA
| |
Collapse
|
150
|
Kuhn C, Weiner HL. Therapeutic anti-CD3 monoclonal antibodies: from bench to bedside. Immunotherapy 2016; 8:889-906. [DOI: 10.2217/imt-2016-0049] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The induction of tolerance is a major goal of immunotherapy. Investigations over the last 20 years have shown that anti-CD3 monoclonal antibodies (mAbs) effectively treat autoimmune disease in animal models and have also shown promise in clinical trials. Tolerance induction by anti-CD3 mAbs is related to the induction of Tregs that control pathogenic autoimmune responses. Here, we review preclinical and clinical studies in which intravenous or mucosal administration of anti-CD3 mAbs has been employed and provide an outlook on future developments to enhance the efficacy of this promising therapeutic approach.
Collapse
Affiliation(s)
- Chantal Kuhn
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|