1
|
Farid A, Moussa P, Youssef M, Haytham M, Shamy A, Safwat G. Melatonin relieves diabetic complications and regenerates pancreatic beta cells by the reduction in NF-kB expression in streptozotocin induced diabetic rats. Saudi J Biol Sci 2022; 29:103313. [PMID: 35707823 PMCID: PMC9189213 DOI: 10.1016/j.sjbs.2022.103313] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 04/18/2022] [Accepted: 05/18/2022] [Indexed: 12/25/2022] Open
Abstract
Melatonin, a pleiotropic hormone, has many regulatory effects on the circadian and seasonal rhythms, sleep and body immune system. It is used in the treatment of blind circadian rhythm sleep disorders, delayed sleep phase and insomnia. It is a potent antioxidant, anti-inflammatory, free radical scavenger, helpful in fighting infectious disease and cancer treatment. Decreased level of circulating melatonin was associated with an increased blood glucose level, losing the anti-oxidant protection and anti-inflammatory responses. We aimed to evaluate the effect of melatonin administration, in streptozotocin (STZ) induced diabetic rats, on blood glucose level and pancreatic beta (β) cells. Diabetes mellitus was induced in Sprague dawley male rats by the intravenous (i.v) injection of 65 mg/kg of STZ. Diabetic rats received melatonin at a dose of 10 mg/kg daily for 8 weeks by oral routes. The results showed, after 8 weeks of melatonin administration, a reduction in: 1- fasting blood glucose (FBG) and fructosamine (FTA) levels, 2- kidney and liver function parameters, 3- levels of serum triglycerides, cholesterol and LDL-C, 4- malondialdehyde (MDA), 5- NF-κB expression in treated group, 6- pro-inflammatory cytokines (IL-1β and IL-12) and immunoglobulins (IgA, IgE and IgG). Furthermore, an elevation in insulin secretion was noticed in melatonin treated group that indicated β cells regeneration. Therefore, melatonin administration, in STZ induced diabetic rats; reduced hyperglycemia, hyperlipidemia and oxidative stress. Melatonin acted as an anti-inflammatory agent that reduced pro-inflammatory cytokines (IL-1β and IL-12) and oxidative stress biomarkers (MDA). Melatonin succeeded in protecting β cells under severe inflammatory situations, which was apparent by the regeneration of islets of Langerhans in treated diabetic rats. Moreover, these results can open a gate for diabetes management and treatment.
Collapse
Affiliation(s)
- Alyaa Farid
- Zoology Dep., Faculty of Science, Cairo University, Giza, Egypt
| | - Passant Moussa
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Manar Youssef
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Merna Haytham
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Ali Shamy
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Gehan Safwat
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| |
Collapse
|
2
|
Zeng Q, Song J, Wang D, Sun X, Xiao Y, Zhang H, Xiao Y, Zhou Z, Deng T. Identification of Sorafenib as a Treatment for Type 1 Diabetes. Front Immunol 2022; 13:740805. [PMID: 35242127 PMCID: PMC8886732 DOI: 10.3389/fimmu.2022.740805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Th1 cell activation is considered a key mediator of the pathogenesis of type 1 diabetes. Targeting IL-12-induced Th1 cell differentiation seems to be an effective way to block the development of type 1 diabetes. However, given the critical function of Th1 in the immune system, the potential side effects hinder the application of anti-Th1 therapy in the treatment of type 1 diabetes. To identify safe anti-Th1 treatment(s), we screened the FDA-approved tyrosine kinase inhibitor (TKI) drug library using an IL-12-induced Th1 differentiation cell model. We found that among the TKIs with little effect on T cell viability, sorafenib is the top contender for the inhibition of Th1 differentiation. Treatment of NOD mice with sorafenib significantly impeded the development of type 1 diabetes and ameliorated insulitis, which coincided with a specifically decreased accumulation of Th1 cell population in the pancreas but not in peripheral immune organs. Mechanistically, sorafenib indirectly inhibited janus kinase 2 (JAK2) activity and blocked IL-12-induced phosphorylations of JAK2 and signal transducer and activator of transcription 4 (STAT4). Since sorafenib is classified as an FDA-approved drug, it serves as a preliminary lead point for additional experimentation and may be a promising therapy for type 1 diabetes in humans.
Collapse
Affiliation(s)
- Qin Zeng
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jianfeng Song
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Dandan Wang
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoxiao Sun
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yalun Xiao
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haowei Zhang
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tuo Deng
- National Clinical Research Center for Metabolic Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology, Ministry of Education, and Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Immunology Center, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
3
|
Jhala G, Krishnamurthy B, Brodnicki TC, Ge T, Akazawa S, Selck C, Trivedi PM, Pappas EG, Mackin L, Principe N, Brémaud E, De George DJ, Boon L, Smyth I, Chee J, Kay TWH, Thomas HE. Interferons limit autoantigen-specific CD8 + T-cell expansion in the non-obese diabetic mouse. Cell Rep 2022; 39:110747. [PMID: 35476975 DOI: 10.1016/j.celrep.2022.110747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/24/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022] Open
Abstract
Interferon gamma (IFNγ) is a proinflammatory cytokine implicated in autoimmune diseases. However, deficiency or neutralization of IFNγ is ineffective in reducing disease. We characterize islet antigen-specific T cells in non-obese diabetic (NOD) mice lacking all three IFN receptor genes. Diabetes is minimally affected, but at 125 days of age, antigen-specific CD8+ T cells, quantified using major histocompatibility complex class I tetramers, are present in 10-fold greater numbers in Ifngr-mutant NOD mice. T cells from Ifngr-mutant mice have increased proliferative responses to interleukin-2 (IL-2). They also have reduced phosphorylated STAT1 and its target gene, suppressor of cytokine signaling 1 (SOCS-1). IFNγ controls the expansion of antigen-specific CD8+ T cells by mechanisms which include increased SOCS-1 expression that regulates IL-2 signaling. The expanded CD8+ T cells are likely to contribute to normal diabetes progression despite reduced inflammation in Ifngr-mutant mice.
Collapse
Affiliation(s)
- Gaurang Jhala
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia
| | - Balasubramanian Krishnamurthy
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia; Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Thomas C Brodnicki
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia; Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia; Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Tingting Ge
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia; Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Satoru Akazawa
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia
| | - Claudia Selck
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia
| | - Prerak M Trivedi
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia
| | - Evan G Pappas
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia
| | - Leanne Mackin
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia
| | - Nicola Principe
- National Centre of Asbestos-Related Diseases, Institute of Respiratory Health, School of Biomedical Science, University of Western Australia, Nedlands, WA 6009, Australia
| | - Erwan Brémaud
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia
| | - David J De George
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia; Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Louis Boon
- Polpharma Biologics, 3584 CM Utrecht, the Netherlands
| | - Ian Smyth
- Australian Phenomics Network, Monash Genome Modification Platform, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Jonathan Chee
- National Centre of Asbestos-Related Diseases, Institute of Respiratory Health, School of Biomedical Science, University of Western Australia, Nedlands, WA 6009, Australia
| | - Thomas W H Kay
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia; Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia.
| | - Helen E Thomas
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC 3065, Australia; Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
4
|
Lee JH, Kim DY, Pantha R, Lee EH, Bae JH, Han E, Song DK, Kwon TK, Im SS. Identification of Pre-Diabetic Biomarkers in the Progression of Diabetes Mellitus. Biomedicines 2021; 10:72. [PMID: 35052752 PMCID: PMC8773205 DOI: 10.3390/biomedicines10010072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/25/2021] [Accepted: 12/29/2021] [Indexed: 01/11/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a major global health issue. The development of T2DM is gradual and preceded by the pre-diabetes mellitus (pre-DM) stage, which often remains undiagnosed. This study aimed to identify novel pre-DM biomarkers in a high-fat diet (HFD)-induced pre-DM mouse model. Male C57BL/6J mice were fed either a chow diet or HFD for 12 weeks. Serum and liver samples were isolated in a time-dependent manner. Semi-quantitative assessment of secretory cytokines was performed by cytokine array analysis, and 13 cytokines were selected for further analysis based on the changes in expression levels in the pre-DM and T2DM stages. HFD-fed mice gained body weight and exhibited high serum lipid, liver enzyme, glucose, and insulin levels during the progression of pre-DM to T2DM. The mRNA expression of inflammatory and lipogenic genes was elevated in HFD-fed mice The mRNA expression of Fc receptor, IgG, low affinity Iib, lectin, galactose binding, soluble 1, vascular cell adhesion molecule 1, insulin-like growth factor binding protein 5, and growth arrest specific 6 was elevated in the pre-DM, which was confirmed by measuring protein levels. Our study identified novel pre-DM biomarkers that may help to delay or prevent the progression of T2DM.
Collapse
Affiliation(s)
- Jae-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea; (J.-H.L.); (D.-Y.K.); (R.P.); (E.-H.L.); (J.-H.B.); (D.-K.S.)
| | - Do-Young Kim
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea; (J.-H.L.); (D.-Y.K.); (R.P.); (E.-H.L.); (J.-H.B.); (D.-K.S.)
| | - Rubee Pantha
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea; (J.-H.L.); (D.-Y.K.); (R.P.); (E.-H.L.); (J.-H.B.); (D.-K.S.)
| | - Eun-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea; (J.-H.L.); (D.-Y.K.); (R.P.); (E.-H.L.); (J.-H.B.); (D.-K.S.)
| | - Jae-Hoon Bae
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea; (J.-H.L.); (D.-Y.K.); (R.P.); (E.-H.L.); (J.-H.B.); (D.-K.S.)
| | - Eugene Han
- Department of Internal Medicine, Division of Endocrinology, Keimyung University School of Medicine, Daegu 42601, Korea;
| | - Dae-Kyu Song
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea; (J.-H.L.); (D.-Y.K.); (R.P.); (E.-H.L.); (J.-H.B.); (D.-K.S.)
| | - Taeg Kyu Kwon
- Department of Immunology, Keimyung University School of Medicine, Daegu 42601, Korea;
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea; (J.-H.L.); (D.-Y.K.); (R.P.); (E.-H.L.); (J.-H.B.); (D.-K.S.)
| |
Collapse
|
5
|
Abdelmajed SS, El-Dessouky MA, SalahElDin DS, Hassan NAM, Zaki ME, Ismail S. Assessing the association of rs7574865 STAT4 gene variant and type 1 diabetes mellitus among Egyptian patients. J Genet Eng Biotechnol 2021; 19:112. [PMID: 34342790 PMCID: PMC8333174 DOI: 10.1186/s43141-021-00214-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022]
Abstract
Background Variants in the signal transducer and activator of transcription 4 (STAT4) gene have an important role in the incident of multiple autoimmune diseases including type 1 diabetes mellitus (T1D). It is a genetically related auto-immune disorder that resulted from T cell-mediated destruction of pancreatic cells that are in control for the production of insulin in the blood. The current study aimed to clarify the role of STAT4 (rs7574865) variant allelic and genotypic variations in the susceptibility to type 1 diabetes among Egyptians by using the real-time PCR. Results A total of 100 patients and 100 controls were genotyped for rs7574865, and the biochemical and anthropometric parameters were measured to show that type 1 diabetic patients had significantly higher levels of HbA1c and triglycerides compared to non-diabetic individuals (P < 0.05). And genetically, the T allele and GT genotype have a significant correlation with diabetes type 1. Conclusion It was confirmed by this study that the rs7574865 T allele and GT genotype have a significant correlation with diabetes type 1 incidence among Egyptian patients.
Collapse
Affiliation(s)
| | - Mohamed A El-Dessouky
- Chemistry Department (Biochemistry Division), Faculty of Science, Cairo University, Giza, Egypt
| | - Doaa S SalahElDin
- Chemistry Department (Biochemistry Division), Faculty of Science, Cairo University, Giza, Egypt
| | - Naglaa Abu-Mandil Hassan
- Biological Anthropology Department, Medical Research Division, National Research Centre, Giza, Egypt
| | - Moushira Erfan Zaki
- Biological Anthropology Department, Medical Research Division, National Research Centre, Giza, Egypt
| | - Somaia Ismail
- Medical Molecular Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Giza, Egypt
| |
Collapse
|
6
|
Deng C, Peng N, Tang Y, Yu N, Wang C, Cai X, Zhang L, Hu D, Ciccia F, Lu L. Roles of IL-25 in Type 2 Inflammation and Autoimmune Pathogenesis. Front Immunol 2021; 12:691559. [PMID: 34122457 PMCID: PMC8194343 DOI: 10.3389/fimmu.2021.691559] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/13/2021] [Indexed: 12/19/2022] Open
Abstract
Interleukin-17E (IL-25) is a member of the IL-17 cytokine family that includes IL-17A to IL-17F. IL-17 family cytokines play a key role in host defense responses and inflammatory diseases. Compared with other IL-17 cytokine family members, IL-25 has relatively low sequence similarity to IL-17A and exhibits a distinct function from other IL-17 cytokines. IL-25 binds to its receptor composed of IL-17 receptor A (IL-17RA) and IL-17 receptor B (IL-17RB) for signal transduction. IL-25 has been implicated as a type 2 cytokine and can induce the production of IL-4, IL-5 and IL-13, which in turn inhibits the differentiation of T helper (Th) 17. In addition to its anti-inflammatory properties, IL-25 also exhibits a pro-inflammatory effect in the pathogenesis of Th17-dominated diseases. Here, we review recent advances in the roles of IL-25 in the pathogenesis of inflammation and autoimmune diseases.
Collapse
Affiliation(s)
- Chong Deng
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China
| | - Na Peng
- Department of Rheumatology and Nephrology, The Second People's Hospital, China Three Gorges University, Yichang, China
| | - Yuan Tang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China
| | - Na Yu
- Department of Rheumatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Cuicui Wang
- Department of Rheumatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaoyan Cai
- Department of Rheumatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lijun Zhang
- Department of Rheumatology, Shenzhen Hospital, The University of Hong Kong, Shenzhen, China
| | - Dajun Hu
- Department of Rheumatology and Nephrology, The Second People's Hospital, China Three Gorges University, Yichang, China
| | - Francesco Ciccia
- Dipartimento di Medicina di Precisione, Section of Rheumatology, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
7
|
Preisser TM, da Cunha VP, Santana MP, Pereira VB, Cara DC, Souza BM, Miyoshi A. Recombinant Lactococcus lactis Carrying IL-4 and IL-10 Coding Vectors Protects against Type 1 Diabetes in NOD Mice and Attenuates Insulitis in the STZ-Induced Model. J Diabetes Res 2021; 2021:6697319. [PMID: 33604389 PMCID: PMC7872750 DOI: 10.1155/2021/6697319] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 01/01/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that culminates in beta cell destruction in the pancreas and, subsequently, deficiency in insulin production. Cytokines play a crucial role in the development of diabetes, orchestrating the recruitment and action of immune cells, to not only destroy insulin-producing cells but also preserve them. Therefore, the aim of this study was to investigate the effect of orally administered Lactococcus lactis MG1363 FnBPA+ strains carrying plasmids encoding IL-4 and IL-10 in the streptozotocin- (STZ-) induced diabetes model and in nonobese diabetic (NOD) mice. The STZ-induced mice that were treated with combined bacterial strains carrying plasmids encoding IL-4 and IL-10 showed lower incidence of diabetes and more preserved pancreatic islets than the mice that received the individual bacterial strains. Combined administration of L. lactis MG1363 FnBPA+ (pValac::dts::IL-4) and L. lactis MG1363 FnBPA+ (pValac::IL-10) resulted in protection against diabetes in NOD mice. It was shown that the combined treatment with recombinant bacterial by oral route prevented hyperglycemia and reduced the pancreatic islets-destruction in NOD mice. In addition, increased levels of IL-4 and IL-10 in serum and pancreatic tissue revealed a systemic effect of the treatment and also favored an anti-inflammatory microenvironment. Reduced concentrations of IL-12 in pancreas were essential to the regulation of inflammation, resulting in no incidence of diabetes in treated NOD mice. Normal levels of intestinal sIgA after long-term treatment with the L. lactis strains carrying plasmids encoding IL-4 and IL-10 indicate the development of oral tolerance and corroborate the use of this potent tool of mucosal delivery. For the first time, L. lactis MG1363 FnBPA+ strains carrying eukaryotic expression vectors encoding IL-4 and IL-10 are tested in STZ-induced and NOD mouse models. Therefore, our study demonstrates this innovative strategy provides immunomodulatory potential for further investigations in T1D and other autoimmune diseases.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- Colon/immunology
- Colon/metabolism
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/prevention & control
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/prevention & control
- Female
- Genetic Therapy
- Genetic Vectors
- Immunoglobulin A, Secretory/metabolism
- Insulin/blood
- Interleukin-10/biosynthesis
- Interleukin-10/blood
- Interleukin-10/genetics
- Interleukin-4/biosynthesis
- Interleukin-4/blood
- Interleukin-4/genetics
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Lactococcus lactis/genetics
- Lactococcus lactis/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice
Collapse
Affiliation(s)
- Tatiane M. Preisser
- Laboratory of Genetic Technology, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais ZIP/Post Code: 31270-901, Brazil
| | - Vanessa P. da Cunha
- Laboratory of Genetic Technology, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais ZIP/Post Code: 31270-901, Brazil
| | - Mariana P. Santana
- Laboratory of Genetic Technology, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais ZIP/Post Code: 31270-901, Brazil
| | - Vanessa B. Pereira
- Laboratory of Genetic Technology, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais ZIP/Post Code: 31270-901, Brazil
| | - Denise C. Cara
- Center for Gastrointestinal Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais ZIP/Post Code: 31270-901, Brazil
| | - Bianca M. Souza
- Laboratory of Genetic Technology, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais ZIP/Post Code: 31270-901, Brazil
| | - Anderson Miyoshi
- Laboratory of Genetic Technology, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais ZIP/Post Code: 31270-901, Brazil
| |
Collapse
|
8
|
Qin K, Jian D, Xue Y, Cheng Y, Zhang P, Wei Y, Zhang J, Xiong H, Zhang Y, Yuan X. DDX41 regulates the expression and alternative splicing of genes involved in tumorigenesis and immune response. Oncol Rep 2021; 45:1213-1225. [PMID: 33650667 PMCID: PMC7859996 DOI: 10.3892/or.2021.7951] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022] Open
Abstract
DEAD‑box helicase 41 (DDX41) is an RNA helicase and accumulating evidence has suggested that DDX41 is involved in pre‑mRNA splicing during tumor development. However, the role of DDX41 in tumorigenesis remains unclear. In order to determine the function of DDX41, the human DDX41 gene was cloned and overexpressed in HeLa cells. The present study demonstrated that DDX41 overexpression inhibited proliferation and promoted apoptosis in HeLa cells. RNA‑sequencing analysis of the transcriptomes in overexpressed and normal control samples. DDX41 regulated 959 differentially expressed genes compared with control cells. Expression levels of certain oncogenes were also regulated by DDX41. DDX41 selectively regulated the alternative splicing of genes in cancer‑associated pathways including the EGFR and FGFR signaling pathways. DDX41 selectively upregulated the expression levels of five antigen processing and presentation genes (HSPA1A, HSPA1B, HSPA6, HLA‑DMB and HLA‑G) and downregulated other immune‑response genes in HeLa cells. Additionally, DDX41‑regulated oncogenes and antigen processing and presentation genes were associated with patient survival rates. Moreover, DDX41 expression was associated with immune infiltration in cervical and endocervical squamous cancer. The present findings showed that DDX41 regulated the cancer cell transcriptome at both the transcriptional and alternative splicing levels. The DDX41 regulatory network predicted the biological function of DDX41 in suppressing tumor cell growth and regulating cancer immunity, which may be important for developing anticancer therapeutics.
Collapse
Affiliation(s)
- Kai Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Danni Jian
- Department of Otolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yaqiang Xue
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, Wuhan, Hubei 430075, P.R. China
| | - Yi Cheng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yaxun Wei
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, Wuhan, Hubei 430075, P.R. China
| | - Jing Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yi Zhang
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, Wuhan, Hubei 430075, P.R. China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
9
|
Ullrich KAM, Schulze LL, Paap EM, Müller TM, Neurath MF, Zundler S. Immunology of IL-12: An update on functional activities and implications for disease. EXCLI JOURNAL 2020; 19:1563-1589. [PMID: 33408595 PMCID: PMC7783470 DOI: 10.17179/excli2020-3104] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022]
Abstract
As its first identified member, Interleukin-12 (IL-12) named a whole family of cytokines. In response to pathogens, the heterodimeric protein, consisting of the two subunits p35 and p40, is secreted by phagocytic cells. Binding of IL-12 to the IL-12 receptor (IL-12R) on T and natural killer (NK) cells leads to signaling via signal transducer and activator of transcription 4 (STAT4) and subsequent interferon gamma (IFN-γ) production and secretion. Signaling downstream of IFN-γ includes activation of T-box transcription factor TBX21 (Tbet) and induces pro-inflammatory functions of T helper 1 (TH1) cells, thereby linking innate and adaptive immune responses. Initial views on the role of IL-12 and clinical efforts to translate them into therapeutic approaches had to be re-interpreted following the discovery of other members of the IL-12 family, such as IL-23, sharing a subunit with IL-12. However, the importance of IL-12 with regard to immune processes in the context of infection and (auto-) inflammation is still beyond doubt. In this review, we will provide an update on functional activities of IL-12 and their implications for disease. We will begin with a summary on structure and function of the cytokine itself as well as its receptor and outline the signal transduction and the transcriptional regulation of IL-12 secretion. In the second part of the review, we will depict the involvement of IL-12 in immune-mediated diseases and relevant experimental disease models, while also providing an outlook on potential translational approaches.
Collapse
Affiliation(s)
- Karen A.-M. Ullrich
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Lisa Lou Schulze
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Eva-Maria Paap
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Tanja M. Müller
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Markus F. Neurath
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Sebastian Zundler
- Department of Medicine and Deutsches Zentrum Immuntherapie, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| |
Collapse
|
10
|
He X, Li W, Xie Y, Zhao Y. Long-term inhibition of dipeptidyl-peptidase 4 reduces islet infiltration and downregulates IL-1β and IL-12 in NOD mice. Int Immunopharmacol 2020; 88:106945. [PMID: 33182020 PMCID: PMC7510641 DOI: 10.1016/j.intimp.2020.106945] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/11/2020] [Accepted: 08/25/2020] [Indexed: 11/17/2022]
Abstract
DPP-4 inhibition reduced CD4+T cells infiltration and ameliorated insulitis. DPP-4 inhibition downregulated serum IL-1β and IL-12. LPS increased CD11b+ cells to infiltrate into islets.
Dipeptidyl-peptidase 4 (DPP-4) inhibitor (sitagliptin) is a novel anti-hyperglycemia drug in the treatment of type 2 diabetes. However, its potential in type 1 diabetes is still unclear. Recent studies show that increased infection, especially respiratory tract infection, is significantly associated with DPP-4 inhibitors. In this study, we aimed to explore the effects of long-term inhibition of DPP- 4 on innate immunity in type 1 diabetes. Forty mice were randomly divided into 4 groups (n = 10 in each group): control group, lipopolysaccharide (LPS) group, sitagliptin group and sitagliptin + LPS group. The concentrations of IL-1β, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, TNF-α and IFN-γ were measured with Mesco Scale Discovery multiplexed-assay kit. Immunohistochemistry staining of pancreases was performed and insulitis scores for each islet were determined. The results showed that DPP-4 inhibition has no effect on incident rate of diabetes and metabolic parameters in NOD mice. Long-term inhibition of DPP-4 reduced CD4+T cells to infiltrate into islets and ameliorated insulitis in NOD mice. DPP-4 inhibition downregulated serum interleukin IL-1β and IL-12 in NOD mice. However, it had no significant effect on LPS-induced IL-1β, IL-6, IL-10, IL-12, tumor necrosis factor (TNF)-α and interferon (IFN)-γ in NOD mice. In conclusion, Long-term inhibition of DPP-4 exists anti-inflammatory effect in type 1 diabetes probably by reducing CD4+T cells to infiltrate into islets and downregulating L-1β and IL-12 in serum.
Collapse
Affiliation(s)
- Xinran He
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, The East Chang-Gang Road, Guangzhou, China
| | - Wangen Li
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, The East Chang-Gang Road, Guangzhou, China
| | - Yunliang Xie
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, The East Chang-Gang Road, Guangzhou, China
| | - Yunjuan Zhao
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, The East Chang-Gang Road, Guangzhou, China.
| |
Collapse
|
11
|
Yang C, Mai H, Peng J, Zhou B, Hou J, Jiang D. STAT4: an immunoregulator contributing to diverse human diseases. Int J Biol Sci 2020; 16:1575-1585. [PMID: 32226303 PMCID: PMC7097918 DOI: 10.7150/ijbs.41852] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Signal transducer and activator of transcription 4 (STAT4) is a member of the STAT family and localizes to the cytoplasm. STAT4 is phosphorylated after a variety of cytokines bind to the membrane, and then dimerized STAT4 translocates to the nucleus to regulate gene expression. We reviewed the essential role played by STAT4 in a wide variety of cells and the pathogenesis of diverse human diseases, especially many kinds of autoimmune and inflammatory diseases, via activation by different cytokines through the Janus kinase (JAK)-STAT signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | - Deke Jiang
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, GuangZhou, China
| |
Collapse
|
12
|
Stefan-van Staden RI, Popa-Tudor I, Ionescu-Tirgoviste C, Stoica RA. Molecular Recognition and Determination of Interleukins 1β, 6, 12, and 17 in Whole Blood from Diabetic Patients. ANAL LETT 2020. [DOI: 10.1080/00032719.2020.1728293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Raluca-Ioana Stefan-van Staden
- Laboratory of Electrochemistry and PATLAB, National Institute of Research for Electrochemistry and Condensed Matter, Bucharest-6, Romania
- Faculty of Applied Chemistry and Material Science, Politehnica University of Bucharest, Bucharest, Romania
| | - Ioana Popa-Tudor
- Laboratory of Electrochemistry and PATLAB, National Institute of Research for Electrochemistry and Condensed Matter, Bucharest-6, Romania
- Faculty of Applied Chemistry and Material Science, Politehnica University of Bucharest, Bucharest, Romania
| | | | - Roxana-Adriana Stoica
- Faculty of General Medicine, “Carol Davila” University of Medicine and Pharmacy Bucharest, Romania
| |
Collapse
|
13
|
STAT4 sequence variant and elevated gene expression are associated with type 1 diabetes in Polish children. Cent Eur J Immunol 2020; 45:22-28. [PMID: 32425676 PMCID: PMC7226553 DOI: 10.5114/ceji.2019.92492] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/23/2019] [Indexed: 12/23/2022] Open
Abstract
Introduction Type 1 diabetes (T1D) is caused by the autoimmune destruction of pancreatic β cells, resulting from coincident genetic predisposition and some environmental triggers. Signal transducer and activator of transcription 4 (STAT4) gene encodes a transcription factor, which promotes Th1 cell differentiation, interferon γ production, and development of Th17 cells. Polymorphisms of STAT4 are associated with several autoimmune conditions, while studies in T1D provided inconsistent results. This analysis was designed to investigate the association of STAT4 rs7574865 with T1D in Polish children and to assess STAT4 expression in newly diagnosed subjects. Material and methods Rs7574865 was genotyped in 656 T1D children and 782 healthy individuals. STAT4 mRNA expression was analyzed in peripheral blood mononuclear cells (PBMCs) from 29 children with T1D and 27 age-matched controls. β-cell and thyroid-specific serum autoantibodies were assessed with radioimmunoassays. Results The distribution of rs7574865 genotypes and alleles demonstrated significant difference (p = 0.002, p < 0.001, respectively) between patients vs. controls. Carriers of the minor T allele presented earlier T1D onset (p = 0.017). No differences were found in γ-cell autoantibody in genotype-stratified patients (p > 0.050), while anti-thyroid antibodies were more frequent in carriers of the minor allele(p = 0.039 for anti-thyroperoxidase, p = 0.007 for anti-thyroglobulin antibodies, respectively). STAT4 was overexpressed in PBMCs from T1D patients (p = 0.008), especially subjects with two/three circulating β-cell antibodies (p < 0.001). Conclusions The study confirms an association of STAT4 rs7574865 with T1D in Polish patients, and provides an evidence for its relationship with an earlier disease onset and concomitant thyroid autoimmunity. STAT4 expression appears elevated in T1D, especially with more severe reaction against β-cell antigens.
Collapse
|
14
|
Zirnheld AL, Villard M, Harrison AM, Kosiewicz MM, Alard P. β-Catenin stabilization in NOD dendritic cells increases IL-12 production and subsequent induction of IFN-γ-producing T cells. J Leukoc Biol 2019; 106:1349-1358. [PMID: 31568613 DOI: 10.1002/jlb.3a0919-244r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/08/2019] [Accepted: 09/13/2019] [Indexed: 11/08/2022] Open
Abstract
Dendritic cells (DC) from diabetes-prone NOD mice and patients with type 1 diabetes (T1D) produce excess IL-12 that drives development of β-cell-destroying IFN-γ-producing T cells. The molecular mechanisms that control IL-12 production in T1D are unclear. In this study, we report that β-catenin, a multifunctional protein involved in inflammation, is dramatically increased in DC from NOD mice. We further investigated the mechanisms leading to accumulation of β-catenin in NOD DC and its role in the inflammatory pathogenic responses associated with T1D. Hyperphosphorylation of β-catenin at a stabilizing residue, serine 552, mediated by activation of Akt, appears to lead to β-catenin accumulation in NOD DC. Elevated β-catenin in DC correlated with IL-12 production and induction of IFN-γ-producing CD4 cells. On the one hand, knockdown/inhibition of β-catenin significantly reduced NOD DC production of IL-12 and their ability to induce IFN-γ-producing CD4 cells. On the other hand, overexpression of β-catenin in control DC resulted in increased IL-12 production and induction of IFN-γ-production in T cells. Additionally, we found that β-catenin inhibitors decreased NF-κB activation in NOD DC and IFN-γ production by NOD T cells in vivo. These data strongly suggest that accumulation of β-catenin in DC from NOD mice drives IL-12 production, and consequently, development of pathogenic IFN-γ-producing T cells. Targeting the defect responsible for β-catenin accumulation and subsequent overproduction of pro-inflammatory cytokines by NOD DC could be an effective therapeutic strategy for the prevention and/or treatment of T1D.
Collapse
Affiliation(s)
- Arin L Zirnheld
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Marine Villard
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA.,Hospices Civils, Lyon, France
| | - Alisha M Harrison
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA.,Department of Biomedical Sciences, Midwestern University, Glendale, Arizona, USA
| | - Michele M Kosiewicz
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Pascale Alard
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
15
|
Yakoub AM, Schülke S. A Model for Apoptotic-Cell-Mediated Adaptive Immune Evasion via CD80-CTLA-4 Signaling. Front Pharmacol 2019; 10:562. [PMID: 31214024 PMCID: PMC6554677 DOI: 10.3389/fphar.2019.00562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 05/06/2019] [Indexed: 12/22/2022] Open
Abstract
Apoptotic cells carry a plethora of self-antigens but they suppress eliciting of innate and adaptive immune responses to them. How apoptotic cells evade and subvert adaptive immune responses has been elusive. Here, we propose a novel model to understand how apoptotic cells regulate T cell activation in different contexts, leading mostly to tolerogenic responses, mainly via taking control of the CD80-CTLA-4 coinhibitory signal delivered to T cells. This model may facilitate understanding of the molecular mechanisms of autoimmune diseases associated with dysregulation of apoptosis or apoptotic cell clearance, and it highlights potential therapeutic targets or strategies for treatment of multiple immunological disorders.
Collapse
Affiliation(s)
- Abraam M Yakoub
- Department of Molecular and Cellular Physiology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Stefan Schülke
- Vice President's Research Group: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
16
|
Arshad Z, Rezapour-Firouzi S, Ebrahimifar M, Mosavi Jarrahi A, Mohammadian M. Association of Delta-6-Desaturase Expression with Aggressiveness of Cancer, Diabetes Mellitus, and Multiple Sclerosis: A Narrative Review. Asian Pac J Cancer Prev 2019; 20:1005-1018. [PMID: 31030467 PMCID: PMC6948902 DOI: 10.31557/apjcp.2019.20.4.1005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/12/2019] [Indexed: 01/01/2023] Open
Abstract
Background: The phosphatidylinositol 3-kinase/ protein kinase B /mammalian target of rapamycin (PI3K/Akt/ mTOR) signaling regulates multiple cellular processes and organizes cell proliferation, survival, and differentiation with the available nutrients, in particular, fatty acids. Polyunsaturated fatty acids (PUFAs) are cytotoxic to cancer cells and play a critical role in the treatment of multiple sclerosis (MS) and diabetes mellitus (DM). PUFAs are produced in the body by desaturases and elongases from dietary essential fatty acids (EFAs), primarily involving delta-6-desaturase (D6D). D6D is a rate-limiting enzyme for maintaining many aspects of lipid homeostasis and normal health. D6D is important to recognize the mechanisms that regulate the expression of this enzyme in humans. A lower level of D6D was seen in breast tumors compared to normal tissues. Interestingly, the elevated serum level of D6D was seen in MS and DM, which explains the critical role of D6D in inflammatory diseases. Methods: We searched databases of PubMed, Web of Science (WOS), Google Scholar, Scopus and related studies by predefined eligibility criteria. We assessed their quality and extracted data. Results: Regarding the mTOR signaling pathway, there is remarkable contributions of many inflammatory diseases to attention to common metabolic pathways are depicted. Of course, we need to have the insights into each disorder and their pathological process. The first step in balancing the intake of EFAs is to prevent the disruption of metabolism and expression of the D6D enzyme. Conclusions: The ω6 and ω3 pathways are two major pathways in the biosynthesis of PUFAs. In both of these, D6D is a vital bifunctional enzyme desaturating linoleic acid or alpha-linolenic acid. Therefore, if ω6 and ω3 EFAs are given together in a ratio of 2: 1, the D6D expression will be down-regulated and normalized.
Collapse
Affiliation(s)
- Zhila Arshad
- Department of Pathology of Anatomy, School of medicine, Baku University of Medical Sciences, Baku, Azerbaijan,
| | | | - Meysam Ebrahimifar
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia,
| | - Alireza Mosavi Jarrahi
- Department of Toxicology, Faculty of Pharmacy, Islamic Azad University, Shahreza Branch, Shahreza,
| | - Mahshid Mohammadian
- Department of Social Medicine, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Anchi P, Khurana A, Swain D, Samanthula G, Godugu C. Dramatic improvement in pharmacokinetic and pharmacodynamic effects of sustain release curcumin microparticles demonstrated in experimental type 1 diabetes model. Eur J Pharm Sci 2019; 130:200-214. [DOI: 10.1016/j.ejps.2019.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 10/17/2018] [Accepted: 02/03/2019] [Indexed: 12/19/2022]
|
18
|
Zhang J, Qiu H, Huang J, Ding S, Huang B, Wu Q, Jiang Q. Establishment of a diabetic myocardial hypertrophy model in Mus musculus castaneus mouse. Int J Exp Pathol 2018; 99:295-303. [PMID: 30614094 PMCID: PMC6384502 DOI: 10.1111/iep.12296] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/31/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
The aim of this study was to establish a robust model of diabetic myocardial hypertrophy in Mus musculus castaneus mice. Mice were fed a high-fat diet for four weeks and then given streptozotocin (STZ, 40 mg kg-1 d-1 for 5 days, intraperitoneally) and fasting blood glucose (FBG) levels were tested after seven days. Mice with FBG levels above 11.1 mmol/L were considered diabetic. Diabetic mice continued to have access to the high-fat diet until cardiac hypertrophy developed. FBG and body weight (BW) were measured weekly. Myocardial hypertrophy was confirmed by left ventricle (LV) hypertrophy index (LVHI), LV/BW, LV histopathological observation and atrial natriuretic factor (ANF) mRNA expression. Serum insulin and plasma haemoglobin A1c (HbA1c) levels, total cholesterol (TCH) and triglyceride (TG) were measured, and then an insulin resistance index (HOMA.IR) was calculated. The level of FBG in the model group remained above 11.1 mmol/L, and the BW showed significant weight loss, compared with the control group (P < 0.01). The high levels of HbA1c, HOME.IR, TCH and TG, and the low level of insulin suggested that glucose metabolism was not balanced with insulin resistance; meanwhile, higher TCH and TG showed that dyslipidaemia had also developed. After the diabetic mice were kept on the high-energy diet for another four weeks, histopathological observation showed myocardial injuries, much more surface area and collagen fibres, higher LVHI and LV/BW, and elevated expression of ANF mRNA (P < 0.01), suggesting that myocardial hypertrophy had appeared in Mus musculus castaneus mice under the current experimental conditions. Thus a robust model of diabetic myocardial hypertrophy was established four weeks after confirmation of diabetes, which was induced by feeding a high-fat diet for four weeks combined with a repeated low-dose STZ exposure, in Mus musculus castaneus mice.
Collapse
Affiliation(s)
- Jie Zhang
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| | - Hongmei Qiu
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| | - Jiajun Huang
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| | - Shumei Ding
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| | - Bo Huang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityGuizhouChina
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of EducationZunyi Medical UniversityGuizhouChina
| | - Qingsong Jiang
- Department of PharmacologyChongqing Key Laboratory of Biochemistry and Molecular PharmacologyChongqing Medical UniversityChongqingChina
| |
Collapse
|
19
|
Loo TT, Gao Y, Lazarevic V. Transcriptional regulation of CD4 + T H cells that mediate tissue inflammation. J Leukoc Biol 2018; 104:1069-1085. [PMID: 30145844 DOI: 10.1002/jlb.1ri0418-152rr] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/15/2022] Open
Abstract
Acquired and genetic immunodeficiencies have revealed an indispensable role for CD4+ T cells in the induction of protective host immune responses against a myriad of microbial pathogens. Influenced by the cytokines present in the microenvironment, activated CD4+ T cells may differentiate into several highly-specialized helper subsets defined by the production of distinct signature cytokines tailored to combat diverse classes of pathogens. The process of specification and differentiation is controlled by networks of core, master, and accessory transcription factors, which ensure that CD4+ T helper (TH ) cell responses mounted against an invading microbe are of the correct specificity and type. However, aberrant activation or inactivation of transcription factors can result in sustained and elevated expression of immune-related genes, leading to chronic activation of CD4+ TH cells and organ-specific autoimmunity. In this review, we provide an overview of the molecular basis of CD4+ TH cell differentiation and examine how combinatorial expression of transcription factors, which promotes genetic plasticity of CD4+ TH cells, can contribute to immunological dysfunction of CD4+ TH responses. We also discuss recent studies which highlight the potential of exploiting the genetic plasticity of CD4+ TH cells in the treatment of autoimmune and other immune-mediated disorders.
Collapse
Affiliation(s)
- Tiffany T Loo
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yuanyuan Gao
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Vanja Lazarevic
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
20
|
Yakoub AM, Schulz R, Seiffert M, Sadek M. Autoantigen-Harboring Apoptotic Cells Hijack the Coinhibitory Pathway of T Cell Activation. Sci Rep 2018; 8:10533. [PMID: 30002409 PMCID: PMC6043626 DOI: 10.1038/s41598-018-28901-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 06/28/2018] [Indexed: 01/11/2023] Open
Abstract
Apoptosis is an important physiological process in development and disease. Apoptotic cells (ACs) are a major source of self-antigens, but ACs usually evade immune responses. The mechanism by which ACs repress T cell adaptive immune responses is poorly understood. T cell activation is finely regulated by a balance of costimulatory signaling (mediated by the costimulatory receptor CD28 on T cells) and coinhibitory signaling (mediated by the coinhibitory ligands CD80 and PD-L1 and -2 on Antigen-Presenting Cells). Here, we found that ACs specifically upregulated the coinhibitory ligand CD80 on macrophages. Conversely, ACs did not exhibit a robust regulation of the other coinhibitory ligands on macrophages or the costimulatory receptor CD28 on T cells. We show that the robust positive regulation of CD80 by ACs requires phagocytosis of ACs by macrophages. We also demonstrate that CD80 modulation by dead cells is a specific effect of ACs, but not necrotic cells (which stimulate immune responses). These results indicate that ACs modulate the coinhibitory pathway of T cell activation via CD80, and suggest a role for CD80 in suppressing T cell responses by ACs. Understanding a mechanism of regulating adaptive immune responses to ACs, which harbor an abundance of self-antigens, may advance our understanding of mechanisms of regulating autoimmunity and facilitate future therapy development for autoimmune disorders.
Collapse
Affiliation(s)
- Abraam M Yakoub
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford University, Stanford, CA, 94305, USA.
| | - Ralph Schulz
- Division of Molecular Genetics, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Martina Seiffert
- Division of Molecular Genetics, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Heidelberg, Germany
| | - Mark Sadek
- Department of Pharmaceutical Biotechnology, University of Illinois College of Pharmacy, Chicago, IL, 60612, USA.,Department of Research and Development, Akorn Pharmaceuticals, Vernon Hills, IL, 60061, USA
| |
Collapse
|
21
|
Semeraro ML, Glenn LM, Morris MA. The Four-Way Stop Sign: Viruses, 12-Lipoxygenase, Islets, and Natural Killer Cells in Type 1 Diabetes Progression. Front Endocrinol (Lausanne) 2017; 8:246. [PMID: 28993759 PMCID: PMC5622285 DOI: 10.3389/fendo.2017.00246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/08/2017] [Indexed: 12/29/2022] Open
Abstract
Natural killer (NK) cells represent an important effector arm against viral infection, and mounting evidence suggests that viral infection plays a role in the development of type 1 diabetes (T1D) in at least a portion of patients. NK cells recognize their target cells through a delicate balance of inhibitory and stimulatory receptors on their surface. If unbalanced, NK cells have great potential to wreak havoc in the pancreas due to the beta cell expression of the as-yet-defined NKp46 ligand through interactions with the activating NKp46 receptor found on the surface of most NK cells. Blocking interactions between NKp46 and its ligand protects mice from STZ-induced diabetes, but differential expression non-diabetic and diabetic donor samples have not been tested. Additional studies have shown that peripheral blood NK cells from human T1D patients have altered phenotypes that reduce the lytic and functional ability of the NK cells. Investigations of humanT1D pancreas tissues have indicated that the presence of NK cells may be beneficial despite their infrequent detection. In non-obese diabetic (NOD) mice, we have noted that NK cells express high levels of the proinflammatory mediator 12/15-lipoxygenase (12/15-LO), and decreased levels of stimulatory receptors. Conversely, NK cells of 12/15-LO deficient NOD mice, which are protected from diabetes development, express significantly higher levels of stimulatory receptors. Furthermore, the human NK92 cell line expresses the ALOX12 protein [human 12-lipoxygenase (12-LO), related to mouse 12/15-LO] via Western blotting. Human 12-LO is upregulated in the pancreas of both T1D and T2D human donors with insulin-containing islets, showing a link between 12-LO expression and diabetes progression. Therefore, our hypothesis is that NK cells in those susceptible to developing T1D are unable to function properly during viral infections of pancreatic beta cells due to increased 12-LO expression and activation, which contributes to increased interferon-gamma production and an imbalance in activating and inhibitory NK cell receptors, and may contribute to downstream autoimmune T cell responses. The work presented here outlines evidence from our lab, as well as published literature, supporting our hypothesis, including novel data.
Collapse
Affiliation(s)
- Michele L. Semeraro
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Lindsey M. Glenn
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Margaret A. Morris
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA, United States
| |
Collapse
|
22
|
Kuhn C, Besançon A, Lemoine S, You S, Marquet C, Candon S, Chatenoud L. Regulatory mechanisms of immune tolerance in type 1 diabetes and their failures. J Autoimmun 2016; 71:69-77. [DOI: 10.1016/j.jaut.2016.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/07/2016] [Indexed: 12/11/2022]
|
23
|
Kabat AM, Pott J, Maloy KJ. The Mucosal Immune System and Its Regulation by Autophagy. Front Immunol 2016; 7:240. [PMID: 27446072 PMCID: PMC4916208 DOI: 10.3389/fimmu.2016.00240] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 06/07/2016] [Indexed: 12/20/2022] Open
Abstract
The gastrointestinal tract presents a unique challenge to the mucosal immune system, which has to constantly monitor the vast surface for the presence of pathogens, while at the same time maintaining tolerance to beneficial or innocuous antigens. In the intestinal mucosa, specialized innate and adaptive immune components participate in directing appropriate immune responses toward these diverse challenges. Recent studies provide compelling evidence that the process of autophagy influences several aspects of mucosal immune responses. Initially described as a “self-eating” survival pathway that enables nutrient recycling during starvation, autophagy has now been connected to multiple cellular responses, including several aspects of immunity. Initial links between autophagy and host immunity came from the observations that autophagy can target intracellular bacteria for degradation. However, subsequent studies indicated that autophagy plays a much broader role in immune responses, as it can impact antigen processing, thymic selection, lymphocyte homeostasis, and the regulation of immunoglobulin and cytokine secretion. In this review, we provide a comprehensive overview of mucosal immune cells and discuss how autophagy influences many aspects of their physiology and function. We focus on cell type-specific roles of autophagy in the gut, with a particular emphasis on the effects of autophagy on the intestinal T cell compartment. We also provide a perspective on how manipulation of autophagy may potentially be used to treat mucosal inflammatory disorders.
Collapse
Affiliation(s)
- Agnieszka M Kabat
- Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| | - Johanna Pott
- Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| |
Collapse
|
24
|
Weaver JR, Nadler JL, Taylor-Fishwick DA. Interleukin-12 (IL-12)/STAT4 Axis Is an Important Element for β-Cell Dysfunction Induced by Inflammatory Cytokines. PLoS One 2015; 10:e0142735. [PMID: 26555476 PMCID: PMC4640700 DOI: 10.1371/journal.pone.0142735] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/26/2015] [Indexed: 12/14/2022] Open
Abstract
Pathology driving β-cell loss in diabetes is poorly defined. Chronic subclinical inflammation is associated with β-cell dysfunction. Acute in vitro exposure of islets and β-cells to an inflammatory cytokine cocktail (IL-1β/TNF-α/IFN-γ) results in loss of cell function and viability. The contribution of each cytokine alone or in combination has been evaluated in homogeneous mouse β-cell lines and primary mouse islets. Cytokine cooperation is required for β-cell apoptosis with the most potent combinations including IL-1β. Single cytokine exposure did not induce β-cell apoptosis. Expression of endogenous interleukin-12 in β-cells correlated with inflammatory cytokine combinations that induced β-cell apoptosis. Uncoupling of the IL-12 axis by a block of IL-12 production, inhibition of IL-12 receptor/ligand interaction or disruption of IL-12 receptor signaling conferred protection to β-cells from apoptosis induced by inflammatory cytokine stimulation. Signaling through STAT4 is indicated since disruption of IL-12 concomitantly reduced inflammatory cytokine stimulation of endogenous IFN-γ expression. Primary mouse islets isolated from mice deficient in STAT4 show resistance to inflammatory-cytokine-induced cell death when compared to islets isolated from wild type mice. Collectively, the data identify IL-12 as an important mediator of inflammation induced β-cell apoptosis. Modulation of IL-12/STAT4 signaling may be a valuable therapeutic strategy to preserve islet/β-cell viability in established diabetes.
Collapse
Affiliation(s)
- Jessica R. Weaver
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Jerry L. Nadler
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - David A. Taylor-Fishwick
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
- * E-mail:
| |
Collapse
|
25
|
Flores RR, Kim E, Zhou L, Yang C, Zhao J, Gambotto A, Robbins PD. IL-Y, a synthetic member of the IL-12 cytokine family, suppresses the development of type 1 diabetes in NOD mice. Eur J Immunol 2015; 45:3114-25. [PMID: 26260044 DOI: 10.1002/eji.201445403] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 07/03/2015] [Accepted: 08/05/2015] [Indexed: 12/31/2022]
Abstract
The IL-12 family of heterodimeric cytokines, consisting of IL-12, IL-23, IL-27, and IL-35, has important roles in regulating the immune response. IL-12 family members are comprised of a heterodimer consisting of α and β chains: IL-12 (p40 and p35), IL-23 (p40 and p19), IL-27 (Ebi3 and p28), and IL-35 (Ebi3 and p35). Given the combinatorial nature of the IL-12 family, we generated adenoviral vectors expressing two putative IL-12 family members not yet found naturally, termed IL-X (Ebi3 and p19) and IL-Y (p40 and p28), as single-chain molecules. Single chain IL-Y (scIL-Y), but not scIL-X, was able to stimulate significantly a unique cytokine/chemokine expression profile as well as activate STAT3 in mice, in part, through a pathway involving IL-27Rα in splenocytes. Adenoviral-mediated, intratumoral delivery of scIL-Y increased tumor growth in contrast to the anti-tumor effects of scIL-12 and scIL-23. Similarly, treatment of prediabetic NOD mice by intravenous injection of Ad.scIL-Y prevented the onset of hyperglycemia. Analysis of cells from Ad.scIL-Y-treated NOD mice demonstrated that scIL-Y reduced expression of inflammatory mediators such as IFN-γ. Our data demonstrate that a novel, synthetic member of the IL-12 family, termed IL-Y, confers unique immunosuppressive effects in two different disease models and thus could have therapeutic applications.
Collapse
Affiliation(s)
- Rafael R Flores
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, USA
| | - Eun Kim
- Departments of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Liqiao Zhou
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, USA
| | - Chenjie Yang
- Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jing Zhao
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, USA
| | - Andrea Gambotto
- Departments of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paul D Robbins
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, USA
- Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
26
|
Zundler S, Neurath MF. Interleukin-12: Functional activities and implications for disease. Cytokine Growth Factor Rev 2015; 26:559-68. [PMID: 26182974 DOI: 10.1016/j.cytogfr.2015.07.003] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/01/2015] [Indexed: 02/01/2023]
Abstract
Interleukin-12 (IL-12) was the first member of the IL-12 family of cytokines to be identified and has therefore become its eponym. It is a heterodimeric protein of two subunits (p35, p40) secreted by phagocytic cells in response to pathogens and mainly acts through STAT4 to induce IFN-γ production in T and NK cells. IFN-γ in turn mediates proinflammatory functions and activates T-bet. As IL-12 engages in TH1 development, it is believed to represent an important link between innate and adaptive immunity. Following its identification and the finding of its association to TH1 commitment, great hopes were placed in IL-12 to become a target for therapeutic applications in multiple settings of autoimmunity and cancer. Though, the discovery of the related members of the IL-12 family and several rather disappointing attempts to translate experimental results into clinical practice, have relativized these hopes. Nevertheless, IL-12 remains a cytokine of outstanding importance with lots of unresolved questions. In this review, we will first briefly depict the biochemistry of the cytokine, its receptor and the related signal transduction, before summarizing the regulation of IL-12 production and its biological functions. We will then describe the current knowledge about the implication of IL-12 in different murine disease models as well as in the corresponding human conditions and comment on possible consequences for future clinical applications.
Collapse
Affiliation(s)
- Sebastian Zundler
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Medical Clinic 1, Ulmenweg 18, 91054 Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Medical Clinic 1, Ulmenweg 18, 91054 Erlangen, Germany.
| |
Collapse
|
27
|
Pane JA, Coulson BS. Lessons from the mouse: potential contribution of bystander lymphocyte activation by viruses to human type 1 diabetes. Diabetologia 2015; 58:1149-59. [PMID: 25794781 DOI: 10.1007/s00125-015-3562-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/04/2015] [Indexed: 02/07/2023]
Abstract
Viruses are considered to be potential key modulators of type 1 diabetes mellitus, with several possible mechanisms proposed for their modes of action. Here we discuss the evidence for virus involvement, including pancreatic infection and the induction of T cell-mediated molecular mimicry. A particular focus of this review is the further possibility that virus infection triggers bystander activation of pre-existing autoreactive lymphocytes. In this scenario, the virus triggers dendritic cell maturation and proinflammatory cytokine secretion by engaging pattern recognition receptors. These proinflammatory cytokines provoke bystander autoreactive lymphocyte activation in the presence of cognate autoantigen, which leads to enhanced beta cell destruction. Importantly, this mechanism does not necessarily involve pancreatic virus infection, and its virally non-specific nature suggests that it might represent a means commonly employed by multiple viruses. The ability of viruses specifically associated with type 1 diabetes, including group B coxsackievirus, rotavirus and influenza A virus, to induce these responses is also examined. The elucidation of a mechanism shared amongst several viruses for accelerating progression to type 1 diabetes would facilitate the identification of important targets for disease intervention.
Collapse
Affiliation(s)
- Jessica A Pane
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, VIC, 3010, Australia
| | | |
Collapse
|
28
|
Kozlovskaya V, Xue B, Lei W, Padgett LE, Tse HM, Kharlampieva E. Hydrogen-bonded multilayers of tannic acid as mediators of T-cell immunity. Adv Healthc Mater 2015; 4:686-94. [PMID: 25491369 DOI: 10.1002/adhm.201400657] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 11/17/2014] [Indexed: 01/07/2023]
Abstract
Type 1 diabetes is an autoimmune-mediated disease resulting in the destruction of insulin-secreting pancreatic β-cells. Transplantation of insulin-producing islets is a viable treatment to restore euglycemia in Type 1 diabetics; however, the clinical application remains limited due to the use of toxic immunosuppressive therapies to prevent immune-mediated rejection. A nanothin polymer material with dual antioxidant and immunosuppressive properties capable of modulating both innate and adaptive immune responses crucial for transplantation outcome is presented. Through the use of hollow microparticles (capsules) composed of hydrogen-bonded multilayers of natural polyphenol (tannic acid) with poly(N-vinylpyrrolidone) (TA/PVPON) and with poly(N-vinylcaprolactam) (TA/PVCL), proinflammatory reactive oxygen and nitrogen species are efficiently dissipated and the production of interferon (IFN)-γ and tumor necrosis factor (TNF)-α proinflammatory cytokines is attenuated by cognate antigen-stimulated autoreactive CD4+ T cells. These results provide evidence that TA-containing capsules are efficacious in immunomodulation and may provide physical transplant protection and prevent diabetogenic autoreactive T-cell responses. Future studies will determine if xeno- and allotransplantation with (TA/PVPON)- or (TA/PVCL)-coated pancreatic islets will decrease the risk of graft rejection due to attenuation of oxidative stress and IFN-γ, and restore euglycemia in Type 1 diabetics.
Collapse
Affiliation(s)
- Veronika Kozlovskaya
- Department of Chemistry; University of Alabama at Birmingham; Birmingham AL 35294 USA
| | - Bing Xue
- Department of Chemistry; University of Alabama at Birmingham; Birmingham AL 35294 USA
| | - Weiqi Lei
- Department of Microbiology; University of Alabama at Birmingham; Birmingham AL 35294 USA
| | - Lindsey E. Padgett
- Department of Microbiology; University of Alabama at Birmingham; Birmingham AL 35294 USA
| | - Hubert M. Tse
- Department of Microbiology; University of Alabama at Birmingham; Birmingham AL 35294 USA
| | - Eugenia Kharlampieva
- Department of Chemistry; University of Alabama at Birmingham; Birmingham AL 35294 USA
- Center for Nanoscale Materials and Biointegration; University of Alabama at Birmingham; Birmingham AL 35294 USA
| |
Collapse
|
29
|
|
30
|
Alkan M, Machavoine F, Rignault R, Dam J, Dy M, Thieblemont N. Histidine Decarboxylase Deficiency Prevents Autoimmune Diabetes in NOD Mice. J Diabetes Res 2015; 2015:965056. [PMID: 26090474 PMCID: PMC4452174 DOI: 10.1155/2015/965056] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/13/2015] [Accepted: 04/15/2015] [Indexed: 12/31/2022] Open
Abstract
Recent evidence has highlighted the role of histamine in inflammation. Since this monoamine has also been strongly implicated in the pathogenesis of type-1 diabetes, we assessed its effect in the nonobese diabetic (NOD) mouse model. To this end, we used mice (inactivated) knocked out for the gene encoding histidine decarboxylase, the unique histamine-forming enzyme, backcrossed on a NOD genetic background. We found that the lack of endogenous histamine in NOD HDC(-/-) mice decreased the incidence of diabetes in relation to their wild-type counterpart. Whereas the proportion of regulatory T and myeloid-derived suppressive cells was similar in both strains, histamine deficiency was associated with increased levels of immature macrophages, as compared with wild-type NOD mice. Concerning the cytokine pattern, we found a decrease in circulating IL-12 and IFN-γ in HDC(-/-) mice, while IL-6 or leptin remained unchanged, suggesting that histamine primarily modulates the inflammatory environment. Paradoxically, exogenous histamine given to NOD HDC(-/-) mice provided also protection against T1D. Our study supports the notion that histamine is involved in the pathogenesis of diabetes, thus providing additional evidence for its role in the regulation of the immune response.
Collapse
Affiliation(s)
- Manal Alkan
- Université Paris Descartes, 75014 Paris, France
- CNRS UMR 8147, Hôpital Necker, 75015 Paris, France
- CNRS UMR 8104, Cochin Institute, 75014 Paris, France
- INSERM U1016, Cochin Institute, 75014 Paris, France
- Center of Excellence, LABEX Inflamex, 75014 Paris, France
| | - François Machavoine
- Université Paris Descartes, 75014 Paris, France
- CNRS UMR 8147, Hôpital Necker, 75015 Paris, France
| | - Rachel Rignault
- Université Paris Descartes, 75014 Paris, France
- CNRS UMR 8147, Hôpital Necker, 75015 Paris, France
| | - Julie Dam
- Université Paris Descartes, 75014 Paris, France
- CNRS UMR 8104, Cochin Institute, 75014 Paris, France
- INSERM U1016, Cochin Institute, 75014 Paris, France
| | - Michel Dy
- Université Paris Descartes, 75014 Paris, France
- CNRS UMR 8147, Hôpital Necker, 75015 Paris, France
| | - Nathalie Thieblemont
- Université Paris Descartes, 75014 Paris, France
- CNRS UMR 8147, Hôpital Necker, 75015 Paris, France
- CNRS UMR 8104, Cochin Institute, 75014 Paris, France
- INSERM U1016, Cochin Institute, 75014 Paris, France
- Center of Excellence, LABEX Inflamex, 75014 Paris, France
- *Nathalie Thieblemont:
| |
Collapse
|
31
|
Ikeda T, Hirata S, Takamatsu K, Haruta M, Tsukamoto H, Ito T, Uchino M, Ando Y, Nagafuchi S, Nishimura Y, Senju S. Suppression of Th1-mediated autoimmunity by embryonic stem cell-derived dendritic cells. PLoS One 2014; 9:e115198. [PMID: 25522369 PMCID: PMC4270741 DOI: 10.1371/journal.pone.0115198] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 11/19/2014] [Indexed: 11/18/2022] Open
Abstract
We herein demonstrate the immune-regulatory effect of embryonic stem cell-derived dendritic cells (ES-DCs) using two models of autoimmune disease, namely non-obese diabetic (NOD) mice and experimental autoimmune encephalomyelitis (EAE). Treatment of pre-diabetic NOD mice with ES-DCs exerted almost complete suppression of diabetes development during the observation period for more than 40 weeks. The prevention of diabetes by ES-DCs was accompanied with significant reduction of insulitis and decreased number of Th1 and Th17 cells in the spleen. Development of EAE was also inhibited by the treatment with ES-DCs, and the therapeutic effect was obtained even if ES-DCs were administrated after the onset of clinical symptoms. Treatment of EAE-induced mice with ES-DCs reduced the infiltration of inflammatory cells into the spinal cord and suppressed the T cell response to the myelin antigen. Importantly, the ES-DC treatment did not affect T cell response to an exogenous antigen. As the mechanisms underlying the reduction of the number of infiltrating Th1 cells, we observed the inhibition of differentiation and proliferation of Th1 cells by ES-DCs. Furthermore, the expression of VLA-4α on Th1 cells was significantly inhibited by ES-DCs. Considering the recent advances in human induced pluripotent stem cell-related technologies, these results suggest a clinical application for pluripotent stem cell-derived dendritic cells as a therapy for T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Tokunori Ikeda
- Department of Immunogenetics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Japan Science and Technology Agency, CREST, Kawaguchi, Japan
- * E-mail:
| | - Shinya Hirata
- Department of Hematology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Koutaro Takamatsu
- Department of Immunogenetics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Japan Science and Technology Agency, CREST, Kawaguchi, Japan
- Department of Neurology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Miwa Haruta
- Department of Immunogenetics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Japan Science and Technology Agency, CREST, Kawaguchi, Japan
| | - Hirotake Tsukamoto
- Department of Immunogenetics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Takaaki Ito
- Department of Pathology and Experimental Medicine, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | | | - Yukio Ando
- Department of Neurology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Seiho Nagafuchi
- Department of Medical Science and Technology, Kyushu University, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Japan Science and Technology Agency, CREST, Kawaguchi, Japan
| |
Collapse
|
32
|
Varzaneh FN, Keller B, Unger S, Aghamohammadi A, Warnatz K, Rezaei N. Cytokines in common variable immunodeficiency as signs of immune dysregulation and potential therapeutic targets - a review of the current knowledge. J Clin Immunol 2014; 34:524-43. [PMID: 24827633 DOI: 10.1007/s10875-014-0053-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/05/2014] [Indexed: 01/19/2023]
Abstract
Common variable immunodeficiency (CVID) is characterized by low levels of circulating immunoglobulins and compromised specific antibody response leading to frequent infections. Cytokines play an important role in the orchestration of the antibody response. Several previous studies have attempted to identify distinct cytokines responsible for the inflammatory changes and different manifestations of CVID, but there are conflicting results regarding the cytokine profiles in CVID patients. In light of this, an extensive review regarding the level of various cytokines and their potential therapeutic role in CVID patients was performed. This review delineates the contribution of interleukin (IL)-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-10, IL-12, IL-21, interferons, tumor necrosis factor (TNF)-α, IL-17, APRIL (a proliferation inducing ligand) and BAFF (B cell activating factor) in CVID disease and outline their potential therapeutic implications in these patients.
Collapse
Affiliation(s)
- Farnaz Najmi Varzaneh
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
33
|
Kota DJ, Wiggins LL, Yoon N, Lee RH. TSG-6 produced by hMSCs delays the onset of autoimmune diabetes by suppressing Th1 development and enhancing tolerogenicity. Diabetes 2013; 62:2048-58. [PMID: 23349496 PMCID: PMC3661629 DOI: 10.2337/db12-0931] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Genetic and immunological screening for type 1 diabetes has led to the possibility of preventing disease in susceptible individuals. Here, we show that human mesenchymal stem/stromal cells (hMSCs) and tumor necrosis factor-α-stimulated gene 6 (TSG-6), a protein produced by hMSCs in response to signals from injured tissues, delayed the onset of spontaneous autoimmune diabetes in NOD mice by inhibiting insulitis and augmenting regulatory T cells (Tregs) within the pancreas. Importantly, hMSCs with a knockdown of tsg-6 were ineffective at delaying insulitis and the onset of diabetes in mice. TSG-6 inhibited the activation of both T cells and antigen-presenting cells (APCs) in a CD44-dependent manner. Moreover, multiple treatments of TSG-6 rendered APCs more tolerogenic, capable of enhancing Treg generation and delaying diabetes in an adoptive transfer model. Therefore, these results could provide the basis for a novel therapy for the prevention of type 1 diabetes.
Collapse
|
34
|
Poudel B, Ki HH, Lee YM, Kim DK. Induction of IL-12 production by the activation of discoidin domain receptor 2 via NF-κB and JNK pathway. Biochem Biophys Res Commun 2013; 434:584-8. [DOI: 10.1016/j.bbrc.2013.03.118] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 03/21/2013] [Indexed: 01/21/2023]
|
35
|
Nepom GT, Ehlers M, Mandrup-Poulsen T. Anti-cytokine therapies in T1D: Concepts and strategies. Clin Immunol 2013; 149:279-85. [PMID: 23510726 DOI: 10.1016/j.clim.2013.02.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 02/05/2013] [Indexed: 12/24/2022]
Abstract
Therapeutic targeting of proinflammatory cytokines is clinically beneficial in several autoimmune disorders. Several of these cytokines are directly implicated in the pathogenesis of type 1 diabetes, suggesting opportunities for design of clinical trials in type 1 diabetes that incorporate selective cytokine blockade as a component of preventative or interventional immunotherapy. The rationale and status of inhibitory therapy directed against IL-1, TNF, IL-12, IL-23, and IL-6 are discussed, towards a goal of using cytokine inhibition as a therapeutic platform to establish an in vivo milieu suitable for modulating the immune response in T1D.
Collapse
Affiliation(s)
- Gerald T Nepom
- Benaroya Research Institute, 1201 Ninth Av, Seattle, WA 98101 USA.
| | | | | |
Collapse
|
36
|
Tsuda M, Zhang W, Yang GX, Tsuneyama K, Ando Y, Kawata K, Park O, Leung PS, Coppel RL, Ansari AA, Ridgway WM, Gao B, Lian ZX, Flavell R, He XS, Gershwin ME. Deletion of interleukin (IL)-12p35 induces liver fibrosis in dominant-negative TGFβ receptor type II mice. Hepatology 2013; 57:806-16. [PMID: 22576253 PMCID: PMC3424295 DOI: 10.1002/hep.25829] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 05/01/2012] [Indexed: 12/20/2022]
Abstract
Mice with a dominant-negative transforming growth factor β receptor restricted to T cells (dnTGFβRII mice) develop an inflammatory biliary ductular disease that strongly resembles human primary biliary cirrhosis (PBC). Furthermore, deletion of the gene encoding interleukin (IL)-12p40 resulted in a strain (IL-12p40(-/-) dnTGFβRII) with dramatically reduced autoimmune cholangitis. To further investigate the role of the IL-12 cytokine family in dnTGFβRII autoimmune biliary disease, we deleted the gene encoding the IL-12p35 subunit from dnTGFβRII mice, resulting in an IL-12p35(-/-) dnTGFβRII strain which is deficient in two members of the IL-12 family, IL-12 and IL-35. In contrast to IL-12p40(-/-) mice, the IL-12p35(-/-) mice developed liver inflammation and bile duct damage with similar severity but delayed onset as the parental dnTGFβRII mice. The p35(-/-) mice also demonstrated a distinct cytokine profile characterized by a shift from a T-helper 1 (Th1) to a Th17 response. Strikingly, liver fibrosis was frequently observed in IL-12p35(-/-) mice. In conclusion, IL-12p35(-/-) dnTGFβRII mice, histologically and immunologically, reflect key features of PBC, providing a useful generic model to understand the immunopathology of human PBC.
Collapse
Affiliation(s)
- Masanobu Tsuda
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616,Department of Emergency and Critical Care Medicine, Kansai Medical University, Osaka 570-8506, Japan
| | - Weici Zhang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616
| | - Guo-Xiang Yang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616
| | - Koichi Tsuneyama
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616,Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama 930-0194, Japan
| | - Yugo Ando
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616
| | - Kazuhito Kawata
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616
| | - Ogyi Park
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Patrick S.C. Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616
| | - Ross L. Coppel
- Department of Microbiology, Monash University, Victoria, 3168, Australia
| | - Aftab A. Ansari
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322
| | - William M. Ridgway
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Zhe-Xiong Lian
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616,Institute of Immunology and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Richard Flavell
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Xiao-Song He
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616
| |
Collapse
|
37
|
Taylor-Fishwick DA, Weaver JR, Grzesik W, Chakrabarti S, Green-Mitchell S, Imai Y, Kuhn N, Nadler JL. Production and function of IL-12 in islets and beta cells. Diabetologia 2013; 56:126-35. [PMID: 23052055 PMCID: PMC3651896 DOI: 10.1007/s00125-012-2732-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 08/09/2012] [Indexed: 10/27/2022]
Abstract
AIMS/HYPOTHESIS IL-12 is an important cytokine in early inflammatory responses and is implicated in the immune-mediated pathogenesis of pancreatic islets in diabetes. However, little is known about the direct effects of IL-12 on islets and beta cells. METHODS In this study, beta cell function, gene expression and protein production were assessed in primary human donor islets and murine beta cell lines in response to stimulation with IL-12 or a pro-inflammatory cytokine cocktail (TNF-α, IL-1β and IFN-γ). RESULTS The pro-inflammatory cytokine cocktail induced islet dysfunction and potently increased the expression and production of IL-12 ligand and IL-12 receptor in human islets. In human islets, the receptor for IL-12 co-localised to the cell surface of insulin-producing cells. Both IL-12 ligand and IL-12 receptor are expressed in the homogeneous beta cell line INS-1. IL-12 induced changes in gene expression, including a dose-dependent upregulation of IFNγ (also known as IFNG), in INS-1 cells. A neutralising antibody to IL-12 directly inhibited IFNγ gene expression in human donor islets induced by either IL-12 or pro-inflammatory cytokine stimulation. Functionally, IL-12 impaired glucose-stimulated insulin secretion (GSIS) in INS-1 cells and human donor islets. A neutralising antibody to IL-12 reversed the beta cell dysfunction (uncoupling of GSIS or induction of caspase-3 activity) induced by pro-inflammatory cytokines. CONCLUSIONS/INTERPRETATION These data identify beta cells as a local source of IL-12 ligand and suggest a direct role of IL-12 in mediating beta cell pathology.
Collapse
Affiliation(s)
- D A Taylor-Fishwick
- Strelitz Diabetes Center, Department of Internal Medicine, Eastern Virginia Medical School, 700 W. Olney Road, Norfolk, VA 23501, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Kozlovskaya V, Zavgorodnya O, Chen Y, Ellis K, Tse HM, Cui W, Thompson JA, Kharlampieva E. Ultrathin polymeric coatings based on hydrogen-bonded polyphenol for protection of pancreatic islet cells. ADVANCED FUNCTIONAL MATERIALS 2012; 22:3389-3398. [PMID: 23538331 PMCID: PMC3607452 DOI: 10.1002/adfm.201200138] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Though transplantation of pancreatic islet cells has emerged as a promising treatment for Type 1 diabetes its clinical application remains limited due to a number of limitations including both pathogenic innate and adaptive immune responses. We report here on a novel type of multifunctional cytoprotective material applied to coat living pancreatic islets. The coating utilizes hydrogen-bonded interactions of a natural polyphenol (tannic acid) with poly(N-vinylpyrrolidone) deposited on the islet surface via non-ionic layer-by-layer assembly. We demonstrate that the coating is conformal over the surface of mammalian islets including those derived from rat, non-human primate (NHP), and human. In contrast to unmodified controls, the coated islets maintain their viability and β-cell functionality for at least 96 hours in vitro. We also determine that the coating demonstrates immunomodulatory cytoprotective properties suppressing pro-inflammatory cytokine synthesis in stimulated bone marrow-derived macrophages and diabetogenic BDC-2.5 T cells. The coating material combines high chemical stability under physiologically relevant conditions with capability of suppressing cytokine synthesis, crucial parameters for prolonged islet integrity, viability, and function in vivo. Our study offers new opportunities in the area of advanced multifunctional materials to be used for a cell-based transplantation therapy.
Collapse
Affiliation(s)
- Veronika Kozlovskaya
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294 (USA)
| | - Oleksandra Zavgorodnya
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294 (USA)
| | - Yi Chen
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294 (USA)
| | - Kristin Ellis
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294 (USA)
| | - Hubert M. Tse
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294 (USA)
| | - Wanxing Cui
- Department of Surgery, Division of Transplantation, the University of Alabama at Birmingham, Birmingham, AL 35294 (USA)
| | - J. Anthony Thompson
- Department of Surgery, Division of Transplantation, the University of Alabama at Birmingham, Birmingham, AL 35294 (USA)
| | - Eugenia Kharlampieva
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294 (USA)
| |
Collapse
|
39
|
van Wanrooij RLJ, Zwiers A, Kraal G, Bouma G. Genetic variations in interleukin-12 related genes in immune-mediated diseases. J Autoimmun 2012; 39:359-68. [PMID: 22819329 DOI: 10.1016/j.jaut.2012.06.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 06/13/2012] [Accepted: 06/24/2012] [Indexed: 12/20/2022]
Abstract
The interleukin-12 (IL-12) family comprises a group of heterodimeric cytokines and their respective receptors that play key roles in immune responses. A growing number of autoimmune diseases has been found to be associated with genetic variation in these genes. Based on their respective associations with the IL-12 genes, autoimmune diseases appear to cluster in two groups that either show strong associations with the Th1/Th17 pathway (as indicated by genetic association with IL12B and IL23R) or the Th1/IL-35 pathway as the consequence of their association with polymorphisms in the IL12A gene region. The genetic associations are described in relation to what is known of the functionality of these genes in the various diseases. Comparing association data for gene families in different diseases may lead to better insight in the function of the genes in the onset and course of the disease.
Collapse
Affiliation(s)
- R L J van Wanrooij
- Department of Gastroenterology and Hepatology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
40
|
Anti-IL-7 receptor-α reverses established type 1 diabetes in nonobese diabetic mice by modulating effector T-cell function. Proc Natl Acad Sci U S A 2012; 109:12674-9. [PMID: 22733769 DOI: 10.1073/pnas.1203795109] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Genetic variation in the IL-7 receptor-α (IL-7R) gene is associated with susceptibility to human type 1 diabetes (T1D). Here we investigate the therapeutic efficacy and mechanism of IL-7Rα antibody in a mouse model of T1D. IL-7Rα antibody induces durable, complete remission in newly onset diabetic mice after only two to three injections. IL-7 increases, whereas IL-7Rα antibody therapy reduces, the IFN-γ-producing CD4(+) (T(H)1) and IFN-γ-producing CD8(+) T cells. Conversely, IL-7 decreases and IL-7Rα antibody enhances the inhibitory receptor Programmed Death 1 (PD-1) expression in the effector T cells. Programmed Death 1 blockade reversed the immune tolerance mediated by the IL-7Rα antibody therapy. Furthermore, IL-7Rα antibody therapy increases the frequency of regulatory T cells without affecting their suppressor activity. The durable efficacy and the multipronged tolerogenic mechanisms of IL-7Rα antibody therapy suggest a unique disease-modifying approach to T1D.
Collapse
|
41
|
Yaghini N, Mahmoodi M, Hassanshahi G, Asadikaram G, Arababadi MK, Rezaeian M, Sajjadi SMA, Kennedy D. Genetic variation of IL-12B (+1188 region) is associated with its decreased circulating levels and susceptibility to Type 2 diabetes. Biomark Med 2012; 6:89-95. [PMID: 22296201 DOI: 10.2217/bmm.11.84] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus is one of the most common types of endocrine disease and the immune system plays a predominant role in its pathogenesis. AIMS The present study aimed to examine known gene polymorphisms within IL-12B (+1188) region and its circulating serum levels in Type 2 diabetic patients from the southeastern region of Iran and compare them with unrelated controls. MATERIALS & METHODS In this clinical study, peripheral blood was collected from 114 Type 2 diabetic patients and 100 healthy controls. Serum levels of IL-12B were measured by ELISA. Genomic DNA was extracted from peripheral blood samples and polymorphisms at the +1188 position of the IL-12B gene were assessed using PCR restriction fragment-length polymorphism. RESULTS Our findings demonstrated that the AA genotype and the A allele of IL-12B were increased significantly in Type 2 diabetic patients when compared with controls. Our results also showed that the circulating levels of IL-12B were significantly decreased in Type 2 diabetic patients when compared with controls. CONCLUSION According to the findings of the current study, we concluded that IL-12B and its +1188 polymorphism may play a prominent role in the pathogenesis of Type 2 diabetes. Further replicative investigations using a larger sample size are essential to identify additional IL-12B genetic variants associated with a risk of Type 2 diabetes.
Collapse
Affiliation(s)
- Narges Yaghini
- Department of Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Iran
| | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
IFN-γ induced by IL-12 administration prevents diabetes by inhibiting pathogenic IL-17 production in NOD mice. J Autoimmun 2012; 38:20-8. [DOI: 10.1016/j.jaut.2011.11.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 11/08/2011] [Accepted: 11/28/2011] [Indexed: 11/17/2022]
|
44
|
Tersey SA, Carter JD, Rosenberg L, Taylor-Fishwick DA, Mirmira RG, Nadler JL. Amelioration of type 1 diabetes following treatment of non-obese diabetic mice with INGAP and lisofylline. ACTA ACUST UNITED AC 2012; 2:251-257. [PMID: 26473085 DOI: 10.4236/jdm.2012.22040] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Type 1 diabetes mellitus results from the autoimmune and inflammatory destruction of insulin-producing islet β cells, rendering individuals devoid of insulin production. Recent studies suggest that combination therapies consisting of anti-inflammatory agents and islet growth-promoting factors have the potential to cause sustained recovery of β cell mass, leading to amelioration or reversal of type 1 diabetes in mouse models. In this study, we hypothesized that the combination of the anti-inflammatory agent lisofylline (LSF) with an active peptide fragment of islet neogenesis associated protein (INGAP peptide) would lead to remission of type 1 diabetes in the non-obese diabetic (NOD) mouse. We treated groups of spontaneously diabetic NOD mice with combinations of LSF, INGAP peptide, or control saline parenterally for up to 6 weeks. Our results demonstrate that the mice receiving combined treatment with LSF and INGAP peptide exhibited partial remission of diabetes with increased plasma insulin levels. Histologic assessment of pancreata in mice receiving combined therapy revealed the presence of islet insulin staining, increased β cell replication, and evidence of Pdx1-positivity in ductal cells. By contrast, diabetic animals showed severe insulitis with no detectible insulin or Pdx1 staining. We conclude that the novel combination treatment with LSF and INGAP peptide has the potential to ameliorate hyperglycemia in the setting of established type 1 diabetes via the recovery of endogenous β cells and warrant further studies.
Collapse
Affiliation(s)
- Sarah A Tersey
- Department of Pediatrics, Indiana University, Indianapolis, USA ; Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, USA
| | - Jeffery D Carter
- Department of Medicine, University of Virginia, Charlottesville, USA
| | | | - David A Taylor-Fishwick
- Department of Medicine and the Strelitz Diabetes Center, Eastern Virginia Medicial School, Norfolk, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics, Indiana University, Indianapolis, USA ; Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, USA ; Department of Medicine, Indiana University, Indianapolis, USA ; Department of Cellular and Integrative Physiology, Indiana University, Indianapolis, USA
| | - Jerry L Nadler
- Department of Medicine and the Strelitz Diabetes Center, Eastern Virginia Medicial School, Norfolk, USA
| |
Collapse
|
45
|
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an inflammatory demyelinating disease of the central nervous system that is induced in laboratory animals by the generation of an immune response against myelin epitopes. It has been used as a prototype of Th1- and/or Th17-driven, organ-specific autoimmunity and as a model for the human disease, multiple sclerosis. In this chapter we describe two classic protocols for EAE induction (active immunization and adoptive transfer of Th1- or Th17-polarized cells) in Subheadings 3.1 and 3.2, respectively. Subheading 3.3 describes methods for rating clinical disease in symptomatic animals. Subheading 3.4 includes instructions for the isolation of mononuclear cells from the inflamed spinal cords of mice with EAE. Subheading 3.5 describes a method for performing the enzyme-linked immunospot assay.
Collapse
Affiliation(s)
- Praveen Rao
- Department of Neurology, University of Michigan Multiple Sclerosis Center and Holtom-Garrett Program in Neuroimmunology, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
46
|
Wang YL, Chou FC, Chen SJ, Lin SH, Chang DM, Sytwu HK. Targeting pre-ligand assembly domain of TNFR1 ameliorates autoimmune diseases - an unrevealed role in downregulation of Th17 cells. J Autoimmun 2011; 37:160-170. [PMID: 21689905 DOI: 10.1016/j.jaut.2011.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 05/06/2011] [Accepted: 05/14/2011] [Indexed: 01/09/2023]
Abstract
The pre-ligand assembly domain (PLAD) of tumor necrosis factor receptors mediates specific ligand-independent receptor assembly and subsequent signaling. However, the physiological role of PLAD in the regulation of TNFR-mediated immune responses in autoimmunity is still unclear. By using the recombinant PLAD.Fc protein to block TNFR1 assembly, we demonstrated that PLAD.Fc treatment significantly reduced the TNFR1-driving proinflammatory cytokines and protected NOD mice from diabetes. Strikingly, Th17 differentiation was significantly inhibited in PLAD.Fc-treated NOD and TNFR1-deficient mice, indicating a TNFR1-dependent Th17 development. PLAD.Fc-modulated effects on DCs, in terms of the downregulation of Th17-inducing cytokines, IL-6 and TGF-β, explained the potential mechanism for Th17 suppression. Finally, we provided an additional result that PLAD.Fc administration diminished the infiltration of Th17 cells in the central nervous system and ameliorated the experimental autoimmune encephalomyelitis in mice. Collectively, these data demonstrated that targeting PLAD of TNFR1 provides protection from autoimmune diseases through the downregulation of Th17 and suggested a therapeutic potential of PLAD-modulation in TNF-involved inflammatory diseases.
Collapse
MESH Headings
- Animals
- Autoimmunity/drug effects
- Autoimmunity/genetics
- Cell Differentiation
- Cell Movement/drug effects
- Cell Movement/immunology
- Central Nervous System/drug effects
- Central Nervous System/immunology
- Central Nervous System/pathology
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Down-Regulation
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Female
- Humans
- Interleukin-6/biosynthesis
- Interleukin-6/immunology
- Jurkat Cells
- Mice
- Mice, Inbred NOD
- Molecular Targeted Therapy
- Plasmids
- Protein Structure, Tertiary
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/immunology
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- Recombinant Fusion Proteins/pharmacology
- Signal Transduction/genetics
- Signal Transduction/immunology
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Transfection
- Transforming Growth Factor beta/biosynthesis
- Transforming Growth Factor beta/immunology
Collapse
Affiliation(s)
- Yen-Ling Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Neihu, Taipei 114, Taiwan
| | | | | | | | | | | |
Collapse
|
47
|
Singh B, Nikoopour E, Huszarik K, Elliott JF, Jevnikar AM. Immunomodulation and regeneration of islet Beta cells by cytokines in autoimmune type 1 diabetes. J Interferon Cytokine Res 2011; 31:711-9. [PMID: 21851268 DOI: 10.1089/jir.2011.0025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Juvenile or type 1 diabetes (T1D) involves autoimmune-mediated destruction of insulin-producing β cells in the islets of Langerhans in the pancreas. Lack of insulin prevents the absorption and metabolism of glucose throughout the body by interfering with cell signaling. Cytokines have been shown to play a key role in β cell destruction and regulation of autoimmunity in T1D. The multiple roles of cytokines in T1D pathogenesis, regulation, and regeneration of β cells presents both promise and challenge for their use in immunotherapy. We found that mycobacterial adjuvants induce various regulatory T cells in the non-obese diabetic (NOD) mouse model of T1D. Cytokines produced by these cells not only regulate innate and adaptive immunity but also prevent the development of diabetes and partially restored normoglycemia in diabetic NOD mice. We discovered that adjuvant immunotherapy upregulated Regenerating (Reg) genes in the islets and induced interleukin 22 (IL-22)-producing Th17 cells. IL-22 is known to upregulate Reg gene expression in islets and could potentially induce regeneration of β cells and prevent their apoptosis. Therefore, cytokines both induce and regulate T1D and have the potential to regenerate and preserve insulin-producing β cells in the islets.
Collapse
Affiliation(s)
- Bhagirath Singh
- Centre for Human Immunology, Department of Microbiology and Immunology and Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | |
Collapse
|
48
|
Ganesh BB, Bhattacharya P, Gopisetty A, Prabhakar BS. Role of cytokines in the pathogenesis and suppression of thyroid autoimmunity. J Interferon Cytokine Res 2011; 31:721-31. [PMID: 21823922 DOI: 10.1089/jir.2011.0049] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Autoimmune thyroid diseases (AITD) are one of the most common organ-specific autoimmune disorders, of which Hashimoto's thyroiditis (HT) and Graves' disease (GD) are 2 of the most common clinical expressions. HT is characterized by hypothyroidism that results from the destruction of the thyroid by thyroglobulin-specific T cell-mediated autoimmune response. In contrast, GD is characterized by hyperthyroidism due to excessive production of thyroid hormone induced by thyrotropin receptor-specific stimulatory autoantibodies. Cytokines play a crucial role in modulating immune responses that affect the balance between maintenance of self-tolerance and initiation of autoimmunity. However, the role of cytokines is often confusing and is neither independent nor exclusive of other immune mediators. A regulatory cytokine may either favor induction of tolerance against thyroid autoimmune disease or favor activation and/or exacerbation of autoimmune responses. These apparently contradictory functions of a given cytokine are primarily influenced by the nature of co-signaling delivered by other cytokines. Consequently, a thorough understanding of the role of a particular cytokine in the context of a specific immune response is essential for the development of appropriate strategies to modulate cytokine responses to maintain or restore health. This review provides a summary of recent research pertaining to the role of cytokines in the pathogenesis of AITD with a particular emphasis on the therapeutic applications of cytokine modulation.
Collapse
Affiliation(s)
- Balaji B Ganesh
- Department of Microbiology and Immunology, Research Resources Center, College of Medicine, University of Illinois at Chicago, Illinois, USA
| | | | | | | |
Collapse
|
49
|
Ganesh BB, Bhattacharya P, Gopisetty A, Sheng J, Vasu C, Prabhakar BS. IL-1β promotes TGF-β1 and IL-2 dependent Foxp3 expression in regulatory T cells. PLoS One 2011; 6:e21949. [PMID: 21779356 PMCID: PMC3136935 DOI: 10.1371/journal.pone.0021949] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 06/15/2011] [Indexed: 12/23/2022] Open
Abstract
Earlier, we have shown that GM-CSF-exposed CD8α− DCs that express low levels of pro-inflammatory cytokines IL-12 and IL-1β can induce Foxp3+ Tregs leading to suppression of autoimmunity. Here, we examined the differential effects of IL-12 and IL-1β on Foxp3 expression in T cells when activated in the presence and absence of DCs. Exogenous IL-12 abolished, but IL-1β enhanced, the ability of GM-CSF-exposed tolerogenic DCs to promote Foxp3 expression. Pre-exposure of DCs to IL-1β and IL-12 had only a modest effect on Foxp3− expressing T cells; however, T cells activated in the absence of DCs but in the presence of IL-1β or IL-12 showed highly significant increase and decrease in Foxp3+ T cell frequencies respectively suggesting direct effects of these cytokines on T cells and a role for IL-1β in promoting Foxp3 expression. Importantly, purified CD4+CD25+ cells showed a significantly higher ability to maintain Foxp3 expression when activated in the presence of IL-1β. Further analyses showed that the ability of IL-1β to maintain Foxp3 expression in CD25+ T cells was dependent on TGF-β1 and IL-2 expression in Foxp3+Tregs and CD25− effectors T cells respectively. Exposure of CD4+CD25+ T cells to IL-1β enhanced their ability to suppress effector T cell response in vitro and ongoing experimental autoimmune thyroidits in vivo. These results show that IL-1β can help enhance/maintain Tregs, which may play an important role in maintaining peripheral tolerance during inflammation to prevent and/or suppress autoimmunity.
Collapse
Affiliation(s)
- Balaji B. Ganesh
- Departments of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Palash Bhattacharya
- Departments of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Anupama Gopisetty
- Departments of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jianrong Sheng
- Departments of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Chenthamarakshan Vasu
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Bellur S. Prabhakar
- Departments of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
50
|
Daniel C, Weigmann B, Bronson R, von Boehmer H. Prevention of type 1 diabetes in mice by tolerogenic vaccination with a strong agonist insulin mimetope. ACTA ACUST UNITED AC 2011; 208:1501-10. [PMID: 21690251 PMCID: PMC3135372 DOI: 10.1084/jem.20110574] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Subimmunogenic vaccination with an agonist mimetope of insulin converts naive T cells into regulatory T cells and prevents type 1 diabetes in NOD mice. Type 1 diabetes (T1D) results from the destruction of insulin-secreting pancreatic β cells by autoreactive T cells. Insulin is an essential target of the autoimmune attack. Insulin epitopes recognized by diabetogenic T cell clones bind poorly to the class II I-Ag7 molecules of nonobese diabetic (NOD) mice, which results in weak agonistic activity of the peptide MHC complex. Here, we describe a strongly agonistic insulin mimetope that effectively converts naive T cells into Foxp3+ regulatory T cells in vivo, thereby completely preventing T1D in NOD mice. In contrast, natural insulin epitopes are ineffective. Subimmunogenic vaccination with strongly agonistic insulin mimetopes might represent a novel strategy to prevent T1D in humans at risk for the disease.
Collapse
Affiliation(s)
- Carolin Daniel
- Laboratory of Lymphocyte Biology, Department of Cancer Immunology and AIDS, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | |
Collapse
|