1
|
Banerjee A, Singh J. Remodeling adipose tissue inflammasome for type 2 diabetes mellitus treatment: Current perspective and translational strategies. Bioeng Transl Med 2020; 5:e10150. [PMID: 32440558 PMCID: PMC7237149 DOI: 10.1002/btm2.10150] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/07/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022] Open
Abstract
Obesity-associated type 2 diabetes mellitus (T2DM) is characterized by low-grade chronic systemic inflammation that arises primarily from the white adipose tissue. The interplay between various adipose tissue-derived chemokines drives insulin resistance in T2DM and has therefore become a subject of rigorous investigation. The adipocytokines strongly associated with glucose homeostasis include tumor necrosis factor-α, various interleukins, monocyte chemoattractant protein-1, adiponectin, and leptin, among others. Remodeling the adipose tissue inflammasome in obesity-associated T2DM is likely to treat the underlying cause of the disease and bring significant therapeutic benefit. Various strategies have been adopted or are being investigated to modulate the serum/tissue levels of pro- and anti-inflammatory adipocytokines to improve glucose homeostasis in T2DM. These include use of small molecule agonists/inhibitors, mimetics, antibodies, gene therapy, and other novel formulations. Here, we discuss adipocytokines that are strongly associated with insulin activity and therapies that are under investigation for modulation of their levels in the treatment of T2DM.
Collapse
Affiliation(s)
- Amrita Banerjee
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth Dakota
| | - Jagdish Singh
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth Dakota
| |
Collapse
|
2
|
Lu J, Liu J, Li L, Lan Y, Liang Y. Cytokines in type 1 diabetes: mechanisms of action and immunotherapeutic targets. Clin Transl Immunology 2020; 9:e1122. [PMID: 32185024 PMCID: PMC7074462 DOI: 10.1002/cti2.1122] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/31/2020] [Accepted: 03/01/2020] [Indexed: 12/17/2022] Open
Abstract
Cytokines play crucial roles in orchestrating complex multicellular interactions between pancreatic β cells and immune cells in the development of type 1 diabetes (T1D) and are thus potential immunotherapeutic targets for this disorder. Cytokines that can induce regulatory functions-for example, IL-10, TGF-β and IL-33-are thought to restore immune tolerance and prevent β-cell damage. By contrast, cytokines such as IL-6, IL-17, IL-21 and TNF, which promote the differentiation and function of diabetogenic immune cells, are thought to lead to T1D onset and progression. However, targeting these dysregulated cytokine networks does not always result in consistent effects because anti-inflammatory or proinflammatory functions of cytokines, responsible for β-cell destruction, are context dependent. In this review, we summarise the current knowledge on the involvement of well-known cytokines in both the initiation and destruction phases of T1D and discuss advances in recently discovered roles of cytokines. Additionally, we emphasise the complexity and implications of cytokine modulation therapy and discuss the ways in which this strategy has been translated into clinical trials.
Collapse
Affiliation(s)
- Jingli Lu
- Department of Pharmacy The First Affiliated Hospital of Zhengzhou University Zhengzhou China.,Henan Key Laboratory of Precision Clinical Pharmacy Zhengzhou University Zhengzhou China
| | - Jiyun Liu
- Department of Pharmacy The First Affiliated Hospital of Zhengzhou University Zhengzhou China.,Henan Key Laboratory of Precision Clinical Pharmacy Zhengzhou University Zhengzhou China
| | - Lulu Li
- Department of Pharmacy Wuhan No.1 Hospital Wuhan China
| | - Yan Lan
- Department of Pharmacy Huangshi Center Hospital Huangshi China
| | - Yan Liang
- Department of Pharmacy The First Affiliated Hospital of Zhengzhou University Zhengzhou China.,Henan Key Laboratory of Precision Clinical Pharmacy Zhengzhou University Zhengzhou China
| |
Collapse
|
3
|
Colli ML, Szymczak F, Eizirik DL. Molecular Footprints of the Immune Assault on Pancreatic Beta Cells in Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:568446. [PMID: 33042023 PMCID: PMC7522353 DOI: 10.3389/fendo.2020.568446] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/17/2020] [Indexed: 12/25/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic disease caused by the selective destruction of the insulin-producing pancreatic beta cells by infiltrating immune cells. We presently evaluated the transcriptomic signature observed in beta cells in early T1D and compared it with the signatures observed following in vitro exposure of human islets to inflammatory or metabolic stresses, with the aim of identifying "footprints" of the immune assault in the target beta cells. We detected similarities between the beta cell signatures induced by cytokines present at different moments of the disease, i.e., interferon-α (early disease) and interleukin-1β plus interferon-γ (later stages) and the beta cells from T1D patients, identifying biological process and signaling pathways activated during early and late stages of the disease. Among the first responses triggered on beta cells was an enrichment in antiviral responses, pattern recognition receptors activation, protein modification and MHC class I antigen presentation. During putative later stages of insulitis the processes were dominated by T-cell recruitment and activation and attempts of beta cells to defend themselves through the activation of anti-inflammatory pathways (i.e., IL10, IL4/13) and immune check-point proteins (i.e., PDL1 and HLA-E). Finally, we mined the beta cell signature in islets from T1D patients using the Connectivity Map, a large database of chemical compounds/drugs, and identified interesting candidates to potentially revert the effects of insulitis on beta cells.
Collapse
Affiliation(s)
- Maikel L. Colli
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
- *Correspondence: Maikel L. Colli
| | - Florian Szymczak
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Welbio, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Indiana Biosciences Research Institute, Indianapolis, IN, United States
| |
Collapse
|
4
|
Završnik M, Letonja J, Makuc J, Šeruga M, Cilenšek I, Petrovič D. Interleukin-4 (IL4) -590C/T (rs2243250) gene polymorphism is not associated with diabetic nephropathy (DN) in Caucasians with type 2 diabetes mellitus (T2DM). Bosn J Basic Med Sci 2018. [PMID: 29514038 DOI: 10.17305/bjbms.2018.2688] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Diabetic nephropathy (DN) is a microvascular complication that affects up to 40% of diabetic patients and can lead to end-stage kidney disease. Inflammatory cytokines such as interleukin 1 (IL-1), IL-6, IL-18 and tumor necrosis factor-α (TNFα) have been linked to the development and progression of DN. The aim of our study was to examine the relationship between interleukin-4 (IL4) -590C/T (rs2243250) gene polymorphism and DN in patients with type 2 diabetes mellitus (T2DM). This study is a continuation of our previous research on the association between angiotensinogen (AGT) gene polymorphisms and DN in patients with T2DM. We included 651 unrelated Slovenian (Caucasian) patients who had had T2DM for at least 10 years. The participants were classified into a group of T2DM patients with DN (276 cases) and a group without DN (375 controls). IL4 rs2243250 polymorphism was analyzed using a TaqMan SNP genotyping assay and StepOne Real-Time PCR System. The frequencies of rs2243250 TT, CT and CC (wild type) genotypes were 3.2%, 29.4% and 67.4%, respectively in patients with DN, and 2.7%, 34.4% and 62.9%, respectively in controls. Our logistic regression analysis adjusted for gender, age, diabetes duration, and glycated hemoglobin showed no association between rs2243250 and the risk for DN (OR 1.06; CI 0.37-3.05; p = 0.9). IL4 rs2243250 is not associated with DN in our subset of Slovenian patients with T2DM.
Collapse
Affiliation(s)
- Matej Završnik
- Department for Diabetes and Metabolic Diseases, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia.
| | | | | | | | | | | |
Collapse
|
5
|
Wang H, Hogquist KA. How Lipid-Specific T Cells Become Effectors: The Differentiation of iNKT Subsets. Front Immunol 2018; 9:1450. [PMID: 29997620 PMCID: PMC6028555 DOI: 10.3389/fimmu.2018.01450] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/12/2018] [Indexed: 12/24/2022] Open
Abstract
In contrast to peptide-recognizing T cells, invariant natural killer T (iNKT) cells express a semi-invariant T cell receptor that specifically recognizes self- or foreign-lipids presented by CD1d molecules. There are three major functionally distinct effector states for iNKT cells. Owning to these innate-like effector states, iNKT cells have been implicated in early protective immunity against pathogens. Yet, growing evidence suggests that iNKT cells play a role in tissue homeostasis as well. In this review, we discuss current knowledge about the underlying mechanisms that regulate the effector states of iNKT subsets, with a highlight on the roles of a variety of transcription factors and describe how each subset influences different facets of thymus homeostasis.
Collapse
Affiliation(s)
- Haiguang Wang
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Kristin A Hogquist
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
6
|
Van Kaer L, Wu L. Therapeutic Potential of Invariant Natural Killer T Cells in Autoimmunity. Front Immunol 2018; 9:519. [PMID: 29593743 PMCID: PMC5859017 DOI: 10.3389/fimmu.2018.00519] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 02/28/2018] [Indexed: 11/13/2022] Open
Abstract
Tolerance against self-antigens is regulated by a variety of cell types with immunoregulatory properties, such as CD1d-restricted invariant natural killer T (iNKT) cells. In many experimental models of autoimmunity, iNKT cells promote self-tolerance and protect against autoimmunity. These findings are supported by studies with patients suffering from autoimmune diseases. Based on these studies, the therapeutic potential of iNKT cells in autoimmunity has been explored. Many of these studies have been performed with the potent iNKT cell agonist KRN7000 or its structural variants. These findings have generated promising results in several autoimmune diseases, although mechanisms by which iNKT cells modulate autoimmunity remain incompletely understood. Here, we will review these preclinical studies and discuss the prospects for translating their findings to patients suffering from autoimmune diseases.
Collapse
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
7
|
Pinto LC, Cerqueira-Lima AT, Suzarth SDS, de Souza R, Tosta BR, da Silva HB, Pires ADO, Queiroz GDA, Teixeira TO, Dourado KMC, Costa VO, Baqueiro V, Oliveira DP, Brandão HR, de Souza CO, Druzian JI, Medeiros KCDP, Dantas Alves CDA, Lopes MV, Figueiredo CAV. Anonna muricata L. (soursop) seed oil improves type 1 diabetes parameters in vivo and in vitro. PHARMANUTRITION 2018. [DOI: 10.1016/j.phanu.2017.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
8
|
Gianchecchi E, Delfino DV, Fierabracci A. NK cells in autoimmune diseases: Linking innate and adaptive immune responses. Autoimmun Rev 2018; 17:142-154. [DOI: 10.1016/j.autrev.2017.11.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
9
|
Wang-Fischer Y, Garyantes T. Improving the Reliability and Utility of Streptozotocin-Induced Rat Diabetic Model. J Diabetes Res 2018; 2018:8054073. [PMID: 30345315 PMCID: PMC6174751 DOI: 10.1155/2018/8054073] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/07/2018] [Accepted: 08/13/2018] [Indexed: 12/24/2022] Open
Abstract
The Streptozotocin- (STZ-) induced diabetic model is widely used; however, unexplained acute toxicity has given the model an unreliable reputation. To improve the reliability and utility of this model, we characterize the age dependence of STZ toxicity and introduce novel endpoints to assess diabetic complications and reveal possible mechanisms for diabetic development. Diabetes was induced by STZ injection into male, 6 to 23 weeks old, Sprague-Dawley rats. Their metabolic (glucose, lipids, and hormones), inflammatory (cytokines), histologic and behavioral endpoints were observed for 1.2 years. Analgesic compounds were assessed for efficacy treating neuropathy. Acute mortality, within a week of STZ injection (50-65 mg/kg i.v.), was inversely correlated to animal age. Only 3% of rats, age 6-11 weeks, died in the week following STZ injection, whereas 83% of rats 12 to 17 weeks old and 91% of rats 18 weeks or older died in the same week. Partial model recovery (normalized insulin, glucose and food/water intake) was observed starting at week 36; however, pain scores, kidney enlargement, and cataract formation continued to show progression consistent with the diabetic state. Unique noninvasive observational measurements, such as haircoat quality and diarrhea scores, served as useful endpoints for this model. The increased plasma cytokines (such as TNF-α, IL-4, and IL-6) and inflammatory cell infiltration into the pancreatic islets are strong evidence of inflammation in the STZ-induced diabetic model. Pancreatic tissue staining revealed total islet area reduction and confirmed STZ-specific pancreatic toxicity; however, the β-cell density per area in pancreatic islets and insulin levels statistically increased over time in the diabetic rats, suggesting a mechanism for partial recovery of diabetic symptoms. Voltage-gated sodium channel (NaV1.7 specific, peripherally restricted) blocker, CC4148, inhibited neuropathy without side effects as compared to a nonspecific sodium channel inhibitor, Mexiletine, or GABA analog, Pregabalin, which inhibited neuropathy with side effects.
Collapse
Affiliation(s)
- Yanlin Wang-Fischer
- Department of Therapeutics, Chromocell Corporation, 685 U.S. Highway One, North Brunswick, NJ 08902, USA
| | - Tina Garyantes
- Department of Therapeutics, Chromocell Corporation, 685 U.S. Highway One, North Brunswick, NJ 08902, USA
| |
Collapse
|
10
|
Bézie S, Usal C, Guillonneau C. In Vitro and In Vivo Assessment of T, B and Myeloid Cells Suppressive Activity and Humoral Responses from Transplant Recipients. J Vis Exp 2017. [PMID: 28829428 DOI: 10.3791/55510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The main concern in transplantation is to achieve specific tolerance through induction of regulatory cells. The understanding of tolerance mechanisms requires reliable models. Here, we describe models of tolerance to cardiac allograft in rat, induced by blockade of costimulation signals or by upregulation of immunoregulatory molecules through gene transfer. Each of these models allowed in vivo generation of regulatory cells such as regulatory T cells (Tregs), regulatory B cells (Bregs) or regulatory myeloid cells (RegMCs). In this manuscript, we describe two complementary protocols that have been used to identify and define in vitro and in vivo regulatory cell activity to determine their responsibility in tolerance induction and maintenance. First, an in vitro suppressive assay allowed rapid identification of cells with suppressive capacity on effector immune responses in a dose dependent manner, and can be used for further analysis such as cytokine measurement or cytotoxicity. Second, the adoptive transfer of cells from a tolerant treated recipient to a newly irradiated grafted recipient, highlighted the tolerogenic properties of these cells in controlling graft directed immune responses and/or converting new regulatory cells (termed infectious tolerance). These methods are not restricted to cells with known phenotypic markers and can be extended to any cell population. Furthermore, donor directed allospecificity of regulatory cells (an important goal in the field) can be assessed by using third party donor cells or graft either in vitro or in vivo. Finally, to determine the specific tolerogenic capacity of these regulatory cells, we provide protocols to assess the humoral anti-donor antibody responses and the capacity of the recipient to develop humoral responses against new or former known antigens. The models of tolerance described can be used to further characterize regulatory cells, to identify new biomarkers, and immunoregulatory molecules, and are adaptable to other transplantation models or autoimmune diseases in rodent or human.
Collapse
Affiliation(s)
- Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes
| | - Claire Usal
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes;
| |
Collapse
|
11
|
Gourdy P, Bourgeois EA, Levescot A, Pham L, Riant E, Ahui ML, Damotte D, Gombert JM, Bayard F, Ohlsson C, Arnal JF, Herbelin A. Estrogen Therapy Delays Autoimmune Diabetes and Promotes the Protective Efficiency of Natural Killer T-Cell Activation in Female Nonobese Diabetic Mice. Endocrinology 2016; 157:258-67. [PMID: 26485613 DOI: 10.1210/en.2015-1313] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Therapeutic strategies focused on restoring immune tolerance remain the main avenue to prevent type 1 diabetes (T1D). Because estrogens potentiate FoxP3+ regulatory T cells (Treg) and invariant natural killer T (iNKT) cells, two regulatory lymphocyte populations that are functionally deficient in nonobese diabetic (NOD) mice, we investigated whether estradiol (E2) therapy influences the course of T1D in this model. To this end, female NOD mice were sc implanted with E2- or placebo-delivering pellets to explore the course of spontaneous and cyclophosphamide-induced diabetes. Treg-depleted and iNKT-cell-deficient (Jα18(-/-)) NOD mice were used to assess the respective involvement of these lymphocyte populations in E2 effects. Early E2 administration (from 4 wk of age) was found to preserve NOD mice from both spontaneous and cyclophosphamide-induced diabetes, and a complete protection was also observed throughout treatment when E2 treatment was initiated after the onset of insulitis (from 12 wk of age). This delayed E2 treatment remained fully effective in Treg-depleted mice but failed to entirely protect Jα18(-/-) mice. Accordingly, E2 administration was shown to restore the cytokine production of iNKT cells in response to in vivo challenge with the cognate ligand α-galactosylceramide. Finally, transient E2 administration potentiated the previously described protective action of α-galactosylceramide treatment in NOD females. This study provides original evidence that E2 therapy strongly protects NOD mice from T1D and reveals the estrogen/iNKT cell axis as a new effective target to counteract diabetes onset at the stage of insulitis. Estrogen-based therapy should thus be considered for T1D prevention.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/prevention & control
- Cytokines/blood
- Cytokines/metabolism
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/prevention & control
- Drug Implants
- Estradiol/administration & dosage
- Estradiol/therapeutic use
- Estrogen Replacement Therapy
- Estrogens/administration & dosage
- Estrogens/therapeutic use
- Female
- Galactosylceramides/agonists
- Galactosylceramides/pharmacology
- Galactosylceramides/therapeutic use
- Immune Tolerance/drug effects
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/drug effects
- Lymphocyte Depletion/adverse effects
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Mutant Strains
- Ovariectomy/adverse effects
- Prediabetic State/drug therapy
- Prediabetic State/immunology
- Prediabetic State/metabolism
- Prediabetic State/prevention & control
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Pierre Gourdy
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Elvire A Bourgeois
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Anaïs Levescot
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Linh Pham
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Elodie Riant
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Marie-Louise Ahui
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Diane Damotte
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Jean-Marc Gombert
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Francis Bayard
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Claes Ohlsson
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Jean-François Arnal
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - André Herbelin
- INSERM Unité 1048 (P.G., E.R., F.B., J.-F.A.), Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; Toulouse University (P.G., J.-F.A.), 31059 Toulouse, France; Department of Diabetology (P.G.), Toulouse University Hospital, 31403 Toulouse, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 8147 (E.A.B., L.P., M.-L.A.), Necker Hospital, 75015 Paris, France; Paris Descartes University, Necker Hospital (E.A.B., L.P., M.-L.A., A.H.), 75014 Paris, France; INSERM Unité 1082 (A.L., A.H.), 86022 Poitiers, France; Paris-Sud-11 University (A.L.), 91405 Orsay, France; Department of Anatomy and Cytology (A.L., D.D.), Hôtel Dieu, 49033 Paris, France; Laboratory of Immunology (J.-M.G.), Poitiers, and Poitiers University (J.-M.G., A.H.), 86000 Poitiers, France; Centre Hospitalo-Universitaire de Poitiers (J.-M.G., A.H.), 86021 Poitiers, France; and Centre for Bone and Arthritis Research (C.O.), University of Gothenburg, S-405 30 Gothenburg, Sweden
| |
Collapse
|
12
|
Pathogen-expanded CD11b+ invariant NKT cells feedback inhibit T cell proliferation via membrane-bound TGF-β1. J Autoimmun 2015; 58:21-35. [DOI: 10.1016/j.jaut.2014.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/13/2014] [Accepted: 12/21/2014] [Indexed: 12/23/2022]
|
13
|
Karumuthil-Melethil S, Sofi MH, Gudi R, Johnson BM, Perez N, Vasu C. TLR2- and Dectin 1-associated innate immune response modulates T-cell response to pancreatic β-cell antigen and prevents type 1 diabetes. Diabetes 2015; 64:1341-57. [PMID: 25377877 PMCID: PMC4375080 DOI: 10.2337/db14-1145] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The progression of autoimmune diseases is dictated by deviations in the fine balance between proinflammatory versus regulatory responses, and pathogen recognition receptors (PRRs) play a key role in maintaining this balance. Previously, we have reported that ligation of Toll-like receptor 2 (TLR2) and Dectin 1 on antigen-presenting cells by zymosan results in a regulatory immune response that prevents type 1 diabetes (T1D). Here, we show that TLR2 and Dectin 1 engagement by zymosan promotes regulatory T-cell (Treg) responses against the pancreatic β-cell-specific antigen (Ag). Unlike the TLR4 ligand, bacterial lipopolysaccharide, which induced proinflammatory cytokines and pathogenic T cells, zymosan induced a mixture of pro- and anti-inflammatory factors and Tregs, both in vitro and in vivo. Ag-specific T cells that are activated using zymosan-exposed dendritic cells (DCs) expressed Foxp3 and produced large amounts of IL-10, TGF-β1, and IL-17. NOD mice that received β-cell-Ag-loaded, zymosan-exposed DCs showed delayed hyperglycemia. Injection of NOD mice at the prediabetic age and early hyperglycemic stage with β-cell-Ag, along with zymosan, results in a superior protection of the NOD mice from diabetes as compared with mice that received zymosan alone. This therapeutic effect was associated with increased frequencies of IL-10-, IL-17-, IL-4-, and Foxp3-positive T cells, especially in the pancreatic lymph nodes. These results show that zymosan can be used as an immune regulatory adjuvant for modulating the T-cell response to pancreatic β-cell-Ag and reversing early-stage hyperglycemia in T1D.
Collapse
Affiliation(s)
| | - M Hanief Sofi
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC
| | - Radhika Gudi
- Department of Surgery, College of Medicine, Medical University of South Carolina, Charleston, SC
| | - Benjamin M Johnson
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC
| | - Nicolas Perez
- Department of Surgery, University of Illinois at Chicago, Chicago, IL
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC Department of Surgery, College of Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
14
|
Beaudoin L, Diana J, Ghazarian L, Simoni Y, Boitard C, Lehuen A. Plasmacytoid dendritic cells license regulatory T cells, upon iNKT-cell stimulation, to prevent autoimmune diabetes. Eur J Immunol 2014; 44:1454-66. [PMID: 24481989 DOI: 10.1002/eji.201343910] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 12/17/2013] [Accepted: 01/27/2014] [Indexed: 12/26/2022]
Abstract
Invariant NKT (iNKT)-cell stimulation with exogenous specific ligands prevents the development of type 1 diabetes (T1D) in NOD mice. Studies based on anti-islet T-cell transfer showed that iNKT cells prevent the differentiation of these T cells into effector T cells in the pancreatic lymph nodes (PLNs). We hypothesize that this defective priming could be explained by the ability of iNKT cells to induce tolerogenic dendritic cells (DCs) in the PLNs. We evaluated the effect of iNKT-cell stimulation on T1D development by transferring naïve diabetogenic BDC2.5 T cells into proinsulin 2(-/-) NOD mice treated with a long-lasting α-galactosylceramide regimen. In this context, iNKT cells induce the conversion of BDC2.5 T cells into Foxp3(+) Treg cells in the PLNs accumulating in the pancreatic islets. Furthermore, tolerogenic plasmacytoid DCs (pDCs) characterized by low MHC class II molecule expression and TGF-β production are critical in the PLNs for the recruitment of Treg cells into the pancreatic islets by inducing CXCR3 expression. Accordingly, pDC depletion in α-galactosylceramide-treated proinsulin 2(-/-) NOD mice abrogates the protection against T1D. These findings reveal that upon repetitive iNKT-cell stimulation, pDCs are critical for the recruitment of Treg cells in the pancreatic islets and the prevention of T1D development.
Collapse
Affiliation(s)
- Lucie Beaudoin
- INSERM U1016, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France; Laboratoire d'Excellence INFLAMEX, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | | | | | | | | |
Collapse
|
15
|
Van Kaer L. Role of invariant natural killer T cells in immune regulation and as potential therapeutic targets in autoimmune disease. Expert Rev Clin Immunol 2014; 2:745-57. [DOI: 10.1586/1744666x.2.5.745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
16
|
Persistent STAT5 phosphorylation and epigenetic dysregulation of GM-CSF and PGS2/COX2 expression in Type 1 diabetic human monocytes. PLoS One 2013; 8:e76919. [PMID: 24204704 PMCID: PMC3799903 DOI: 10.1371/journal.pone.0076919] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 09/04/2013] [Indexed: 01/22/2023] Open
Abstract
STAT5 proteins are adaptor proteins for histone acetylation enzymes. Histone acetylation at promoter and enhancer chromosomal regions opens the chromatin and allows access of transcription enzymes to specific genes in rapid response cell signals, such as in inflammation. Histone acetylation-mediated gene regulation is involved in expression of 2 key inflammatory response genes: CSF2, encoding granulocyte-macrophage colony stimulating factor (GM-CSF), and PTGS2, encoding prostaglandin synthase 2/cyclooxygenase 2 (PGS2/COX2). Prolonged CSF2 expression, high GM-CSF production, and GM-CSF activation of PTGS2 gene expression all are seen in type 1 diabetes (T1D) monocytes. Persistent phosphorylation activation of monocyte STAT5 (STAT5Ptyr) is also found in individuals with or at-risk for T1D. To examine whether elevated T1D monocyte STAT5Ptyr may be associated with aberrant inflammatory gene expression in T1D, blood monocytes from non-autoimmune controls and T1D patients were analyzed by flow cytometry for STAT5Ptyr activation, and by chromatin immuno-precipitation (ChIP) analyses for STAT5Ptyr’s ability to bind at CSF2 and PTGS2 regulatory sites in association with histone acetylation. In unstimulated monocytes, STAT5Ptyr was elevated in 59.65% of T1D, but only 2.44% of control subjects (p<0.0001). Increased STAT5Ptyr correlated with T1D disease duration (p = 0.0030, r2 = 0.0784). Unstimulated (p = 0.140) and GM-CSF-stimulated (p = 0.0485) T1D monocytes, had greater STAT5Ptyr binding to epigenetic regulatory sites upstream of CSF2 than control monocytes. Increased STAT5Ptyr binding in T1D monocytes was concurrent with binding at these sites of STAT6Ptyr (p = 0.0283), CBP/P300 histone acetylase, acetylated histones H3, SMRT/NCoR histone deacetylase (p = 0.0040), and RNA Polymerase II (p = 0.0040). Our study indicates that in T1D monocytes, STAT5Ptyr activation is significantly higher and that STAT5Ptyr is found bound to CSF2 promoter and PTGS2 enhancer regions coincident with histone acetylation and RNA polymerase II. These findings suggest that the persistent activation of STAT5 by GM-CSF may be involved in altering the epigenetic regulation of these inflammatory response genes in T1D monocytes.
Collapse
|
17
|
Jervis P, Polzella P, Wojno J, Jukes JP, Ghadbane H, Garcia
Diaz YR, Besra GS, Cerundolo V, Cox LR. Design, synthesis, and functional activity of labeled CD1d glycolipid agonists. Bioconjug Chem 2013; 24:586-94. [PMID: 23458425 PMCID: PMC3630740 DOI: 10.1021/bc300556e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/21/2013] [Indexed: 02/01/2023]
Abstract
Invariant natural killer T cells (iNKT cells) are restricted by CD1d molecules and activated upon CD1d-mediated presentation of glycolipids to T cell receptors (TCRs) located on the surface of the cell. Because the cytokine response profile is governed by the structure of the glycolipid, we sought a method for labeling various glycolipids to study their in vivo behavior. The prototypical CD1d agonist, α-galactosyl ceramide (α-GalCer) 1, instigates a powerful immune response and the generation of a wide range of cytokines when it is presented to iNKT cell TCRs by CD1d molecules. Analysis of crystal structures of the TCR-α-GalCer-CD1d ternary complex identified the α-methylene unit in the fatty acid side chain, and more specifically the pro-S hydrogen at this position, as a site for incorporating a label. We postulated that modifying the glycolipid in this way would exert a minimal impact on the TCR-glycolipid-CD1d ternary complex, allowing the labeled molecule to function as a good mimic for the CD1d agonist under investigation. To test this hypothesis, the synthesis of a biotinylated version of the CD1d agonist threitol ceramide (ThrCer) was targeted. Both diastereoisomers, epimeric at the label tethering site, were prepared, and functional experiments confirmed the importance of substituting the pro-S, and not the pro-R, hydrogen with the label for optimal activity. Significantly, functional experiments revealed that biotinylated ThrCer (S)-10 displayed behavior comparable to that of ThrCer 5 itself and also confirmed that the biotin residue is available for streptavidin and antibiotin antibody recognition. A second CD1d agonist, namely α-GalCer C20:2 4, was modified in a similar way, this time with a fluorescent label. The labeled α-GalCer C20:2 analogue (11) again displayed functional behavior comparable to that of its unlabeled substrate, supporting the notion that the α-methylene unit in the fatty acid amide chain should be a suitable site for attaching a label to a range of CD1d agonists. The flexibility of the synthetic strategy, and late-stage incorporation of the label, opens up the possibility of using this labeling approach to study the in vivo behavior of a wide range of CD1d agonists.
Collapse
MESH Headings
- Animals
- Antigens, CD1d/chemistry
- Antigens, CD1d/drug effects
- Antigens, CD1d/immunology
- Cells, Cultured
- Cytokines/analysis
- Cytokines/biosynthesis
- Cytokines/immunology
- Drug Design
- Galactosylceramides/chemistry
- Galactosylceramides/immunology
- Galactosylceramides/pharmacology
- Humans
- Mice
- Mice, Inbred C57BL
- Models, Molecular
- Molecular Conformation
- Natural Killer T-Cells/chemistry
- Natural Killer T-Cells/drug effects
- Natural Killer T-Cells/immunology
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Peter
J. Jervis
- School of Chemistry, University
of Birmingham, Edgbaston, Birmingham B15
2TT, U.K
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15
2TT, U.K
| | - Paolo Polzella
- Medical Research
Council Human
Immunology Unit, Nuffield Department of Medicine, Weatherall Institute
of Molecular Medicine, University of Oxford, Oxford OX3 9DS, U.K
| | - Justyna Wojno
- School of Chemistry, University
of Birmingham, Edgbaston, Birmingham B15
2TT, U.K
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15
2TT, U.K
| | - John-Paul Jukes
- Medical Research
Council Human
Immunology Unit, Nuffield Department of Medicine, Weatherall Institute
of Molecular Medicine, University of Oxford, Oxford OX3 9DS, U.K
| | - Hemza Ghadbane
- Medical Research
Council Human
Immunology Unit, Nuffield Department of Medicine, Weatherall Institute
of Molecular Medicine, University of Oxford, Oxford OX3 9DS, U.K
| | - Yoel R. Garcia
Diaz
- School of Chemistry, University
of Birmingham, Edgbaston, Birmingham B15
2TT, U.K
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15
2TT, U.K
| | - Gurdyal S. Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15
2TT, U.K
| | - Vincenzo Cerundolo
- Medical Research
Council Human
Immunology Unit, Nuffield Department of Medicine, Weatherall Institute
of Molecular Medicine, University of Oxford, Oxford OX3 9DS, U.K
| | - Liam R. Cox
- School of Chemistry, University
of Birmingham, Edgbaston, Birmingham B15
2TT, U.K
| |
Collapse
|
18
|
Mishra PK, Patel N, Wu W, Bleich D, Gause WC. Prevention of type 1 diabetes through infection with an intestinal nematode parasite requires IL-10 in the absence of a Th2-type response. Mucosal Immunol 2013; 6:297-308. [PMID: 22806101 DOI: 10.1038/mi.2012.71] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Helminth infection can prevent type 1 diabetes (T1D); however, the regulatory mechanisms inhibiting disease remain largely undefined. In these studies, nonobese diabetic (NOD) IL-4(-/-) mice were infected with the strictly enteric nematode parasite, Heligmosomoides polygyrus. Short-term infection, 5-7 weeks of age, inhibited T1D onset, as late as 40 weeks of age. CD4(+) T-cell STAT6 phosphorylation was inhibited, while suppressed signal transducer and activator of transcription 1 phosphorylation was sustained, as were increases in FOXP3(-), CD4(+) T-cell interleukin (IL)-10 production. Blockade of IL-10 signaling in NOD-IL-4(-/-), but not in NOD, mice during this short interval abrogated protective effects resulting in pancreatic β-cell destruction and ultimately T1D. Transfer of CD4(+) T cells from H. polygyrus (Hp)-inoculated NOD IL-4(-/-) mice to NOD mice blocked the onset of T1D. These studies indicate that Hp infection induces non-T-regulatory cells to produce IL-10 independently of STAT6 signaling and that in this Th2-deficient environment IL-10 is essential for T1D inhibition.
Collapse
Affiliation(s)
- P K Mishra
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, USA
| | | | | | | | | |
Collapse
|
19
|
Zaccone P, Cooke A. Helminth mediated modulation of Type 1 diabetes (T1D). Int J Parasitol 2013; 43:311-8. [PMID: 23291464 DOI: 10.1016/j.ijpara.2012.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 12/05/2012] [Accepted: 12/07/2012] [Indexed: 01/11/2023]
Abstract
Type 1 diabetes is increasing dramatically in incidence in the developed world. While there may be several reasons for this, improved sanitation and public health measures have altered our interactions with certain infectious agents such as helminths. There is increasing interest in the use of helminths or their products to alleviate inflammatory or allergic conditions. Using rodent models of diabetes, it has been possible to explore the therapeutic potential of both live infections as well as helminth-derived products on the development of autoimmunity. This review provides an overview of the findings from animal models and additionally explores the potential for translation to the clinic.
Collapse
Affiliation(s)
- Paola Zaccone
- Department of Pathology, University of Cambridge, Tennis Court Rd, Cambridge CB2 1QP, UK
| | | |
Collapse
|
20
|
Pilones KA, Aryankalayil J, Demaria S. Invariant NKT cells as novel targets for immunotherapy in solid tumors. Clin Dev Immunol 2012; 2012:720803. [PMID: 23118781 PMCID: PMC3483734 DOI: 10.1155/2012/720803] [Citation(s) in RCA: 26] [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: 07/11/2012] [Revised: 09/02/2012] [Accepted: 09/02/2012] [Indexed: 12/15/2022]
Abstract
Natural killer T (NKT) cells are a small population of lymphocytes that possess characteristics of both innate and adaptive immune cells. They are uniquely poised to respond rapidly to infection and inflammation and produce cytokines that critically shape the ensuing adaptive cellular response. Therefore, they represent promising therapeutic targets. In cancer, NKT cells are attributed a role in immunosurveillance. NKT cells also act as potent activators of antitumor immunity when stimulated with a synthetic agonist in experimental models. However, in some settings, NKT cells seem to act as suppressors and regulators of antitumor immunity. Here we briefly review current data supporting these paradoxical roles of NKT cells and their regulation. Increased understanding of the signals that determine the function of NKT cells in cancer will be essential to improve current strategies for NKT-cell-based immunotherapeutic approaches.
Collapse
Affiliation(s)
- Karsten A. Pilones
- Department of Pathology, NYU School of Medicine, 550 First Avenue, MSB-521, New York, NY 10016, USA
| | - Joseph Aryankalayil
- Department of Pathology, NYU School of Medicine, 550 First Avenue, MSB-521, New York, NY 10016, USA
| | - Sandra Demaria
- Department of Pathology, NYU School of Medicine, 550 First Avenue, MSB-521, New York, NY 10016, USA
| |
Collapse
|
21
|
Novak J, Novakova L. Prevention and treatment of type 1 diabetes mellitus by the manipulation of invariant natural killer T cells. Clin Exp Med 2012; 13:229-37. [PMID: 22825586 DOI: 10.1007/s10238-012-0199-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 07/04/2012] [Indexed: 01/11/2023]
Abstract
Invariant natural killer T (iNKT) cells are CD1d-restricted T cells with regulatory functions. iNKT cells are numerically and functionally deficient in experimental models of type 1 diabetes mellitus (T1DM). Moreover, various experimental strategies correcting the defect of or stimulating iNKT cells prevent T1DM. Here, we review the data on the role of iNKT cells in the development of T1DM and discuss indications, obstacles and prospects of the use of iNKT cell manipulations in the prevention and treatment of human T1DM.
Collapse
Affiliation(s)
- Jan Novak
- 3rd Faculty of Medicine, Charles University in Prague, Ruska 87, 100 00, Prague 10, Czech Republic,
| | | |
Collapse
|
22
|
Zhou L, Li K, Shi YL, Hamzavi I, Gao TW, Henderson M, Huggins RH, Agbai O, Mahmoud B, Mi X, Lim HW, Mi QS. Systemic analyses of immunophenotypes of peripheral T cells in non-segmental vitiligo: implication of defective natural killer T cells. Pigment Cell Melanoma Res 2012; 25:602-11. [PMID: 22591262 DOI: 10.1111/j.1755-148x.2012.01019.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Although it is widely believed that non-segmental vitiligo (NSV) results from the autoimmune destruction of melanocytes, a clear understanding of defects in immune tolerance, which mediate this uncontrolled self-reactivity, is still lacking. In the present study, we systemically evaluated circulating regulatory T (Treg) cells, including CD4(+) CD25(+) FoxP3(+) Treg cells and invariant natural killer T (iNKT) cells, as well as naïve and memory CD4(+) and CD8(+) T cells and their cytokine production, in a cohort of 43 progressive NSV patients with race-, gender-, and age-matched healthy controls. We found that the general immunophenotypes of CD4(+) and CD8(+) T cells and the percentage of CD4(+) CD25(+) FoxP3(+) Tregs were comparable between NSV and healthy controls. However, percentages of peripheral iNKT cells were significantly decreased in NSV patients compared to that in healthy controls. Our data confirm the previous notion that the percentage of peripheral CD4(+) CD25(+) FoxP3(+) Tregs remains unaltered in NSV and suggests the involvement of defective iNKT cells in the pathogenesis of NSV.
Collapse
Affiliation(s)
- Li Zhou
- Henry Ford Immunology Program, Henry Ford Hospital, Detroit, MI, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Subramanian L, Blumenfeld H, Tohn R, Ly D, Aguilera C, Maricic I, Mansson JE, Buschard K, Kumar V, Delovitch TL. NKT cells stimulated by long fatty acyl chain sulfatides significantly reduce the incidence of type 1 diabetes in nonobese diabetic mice [corrected]. PLoS One 2012; 7:e37771. [PMID: 22649557 PMCID: PMC3359325 DOI: 10.1371/journal.pone.0037771] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 04/27/2012] [Indexed: 12/22/2022] Open
Abstract
Sulfatide-reactive type II NKT cells have been shown to regulate autoimmunity and anti-tumor immunity. Although, two major isoforms of sulfatide, C16:0 and C24:0, are enriched in the pancreas, their relative role in autoimmune diabetes is not known. Here, we report that sulfatide/CD1d-tetramer+ cells accumulate in the draining pancreatic lymph nodes, and that treatment of NOD mice with sulfatide or C24:0 was more efficient than C16:0 in stimulating the NKT cell-mediated transfer of a delay in onset from T1D into NOD.Scid recipients. Using NOD.CD1d−/− mice, we show that this delay of T1D is CD1d-dependent. Interestingly, the latter delay or protection from T1D is associated with the enhanced secretion of IL-10 rather than IFN-g by C24:0-treated CD4+ T cells and the deviation of the islet-reactive diabetogenic T cell response. Both C16:0 and C24:0 sulfatide isoforms are unable to activate and expand type I iNKT cells. Collectively, these data suggest that C24:0 stimulated type II NKT cells may regulate protection from T1D by activating DCs to secrete IL-10 and suppress the activation and expansion of type I iNKT cells and diabetogenic T cells. Our results raise the possibility that C24:0 may be used therapeutically to delay the onset and protect from T1D in humans.
Collapse
Affiliation(s)
- Lakshmimathy Subramanian
- Laboratory of Autoimmune Diabetes, Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Hartley Blumenfeld
- Laboratory of Autoimmune Diabetes, Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Robert Tohn
- Laboratory of Autoimmune Diabetes, Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Dalam Ly
- Laboratory of Autoimmune Diabetes, Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Harvard Medical School, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Carlos Aguilera
- Laboratory of Autoimmunity, Torrey Pines Institute for Molecular Studies, San Diego, California, United States of America
| | - Igor Maricic
- Laboratory of Autoimmunity, Torrey Pines Institute for Molecular Studies, San Diego, California, United States of America
| | - Jan-Eric Mansson
- Department of Neuroscience, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Vipin Kumar
- Laboratory of Autoimmunity, Torrey Pines Institute for Molecular Studies, San Diego, California, United States of America
- * E-mail: (TLD); (VK)
| | - Terry L. Delovitch
- Laboratory of Autoimmune Diabetes, Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- * E-mail: (TLD); (VK)
| |
Collapse
|
24
|
Heterogeneous Biological Network Visualization System: Case Study in Context of Medical Image Data. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 736:95-118. [DOI: 10.1007/978-1-4419-7210-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
25
|
Blumenfeld HJ, Tohn R, Haeryfar SMM, Liu Y, Savage PB, Delovitch TL. Structure-guided design of an invariant natural killer T cell agonist for optimum protection from type 1 diabetes in non-obese diabetic mice. Clin Exp Immunol 2011; 166:121-33. [PMID: 21910729 DOI: 10.1111/j.1365-2249.2011.04454.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Because invariant natural killer T (iNK T) cells link innate and adaptive immunity, the structure-dependent design of iNK T cell agonists may have therapeutic value as vaccines for many indications, including autoimmune disease. Previously, we showed that treatment of non-obese diabetic (NOD) mice with the iNK T cell activating prototypic glycolipid α-galactosylceramide (α-GalCer) protects them from type 1 diabetes (T1D). However, α-GalCer is a strong agonist that can hyperactivate iNK T cells, elicit several side effects and has shown only limited success in clinical trials. Here, we used a structure-guided design approach to identify an iNK T cell agonist that optimally protects from T1D with minimal side effects. Analyses of the kinetics and function of a panel of synthetic α-GalCer fatty acyl chain derivatives (C8:0-C16:0) were performed in NOD mice. C16:0 elicited the highest protection from insulitis and T1D, which was associated with a higher frequency and survival of iNK T cells and enhanced activity of tolerogenic dendritic cells (DCs) in draining pancreatic lymph nodes (PLN), inability to transactivate NK cells and a more rapid kinetics of induction and recovery of iNK T cells from anergy. We conclude that the length and structure of the acyl chain of α-GalCer regulates the level of protection against T1D in mice, and propose that the extent of this protection depends on the relative capacity of the acyl chain to accommodate an endogenous spacer lipid of appropriate length and structure. Thus, our findings with the α-GalCer C16:0 derivative suggest strongly that it be considered as a lead glycolipid candidate in clinical trials of T1D.
Collapse
Affiliation(s)
- H J Blumenfeld
- Laboratory of Autoimmune Diabetes, Robarts Research Institute Department of Microbiology and Immunology, Dental Science Building, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | |
Collapse
|
26
|
Van Kaer L, Parekh VV, Wu L. Invariant NK T cells: potential for immunotherapeutic targeting with glycolipid antigens. Immunotherapy 2011; 3:59-75. [PMID: 21174558 DOI: 10.2217/imt.10.85] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Invariant NK T (iNKT) cells are a subset of T lymphocytes that recognize glycolipid antigens bound with the antigen-presenting molecule CD1d. iNKT cells have potent immunoregulatory activities that can promote or suppress immune responses during different pathological conditions. These immunoregulatory properties can be harnessed for therapeutic purposes with cognate glycolipid antigens, such as the marine sponge-derived glycosphingolipid α-galactosylceramide. Preclinical studies have shown substantial promise for iNKT cell-based treatments of infections, cancer and autoimmune and inflammatory diseases. Translation of these preclinical studies to the clinic, while faced with some obstacles, has already had some initial success. In this article, we review the immunodulatory activities of iNKT cells and the potential for developing iNKT cell-based prophylactic and curative therapies of human disease.
Collapse
Affiliation(s)
- Luc Van Kaer
- Department of Microbiology & Immunology, Vanderbilt University School of Medicine, Medical Center North, Room A-5301, 1161 21st Avenue South, Nashville, TN 37232-32363, USA.
| | | | | |
Collapse
|
27
|
α-Galactosylceramide ameliorates autoimmune diabetes in non-obese diabetic mice through a suppressive effect mediated by CD8+ T cells. Immunol Lett 2011; 138:54-62. [PMID: 21392534 DOI: 10.1016/j.imlet.2011.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 01/14/2011] [Accepted: 03/01/2011] [Indexed: 11/22/2022]
Abstract
Type 1 diabetes is an autoimmune disorder resulting from lymphocyte-mediated destruction of insulin-producing β cells in pancreas. Natural killer T cells are regulatory immune components controlling autoreactivity and immune homeostasis. Although early studies supported that amelioration of autoimmune diabetes by natural killer T cells was associated with Th1/2 shift, other Th2-independent regulatory mechanisms were also suggested. Since natural killer T cells are critical for the generation of CD8(+) regulatory T cells controlling anterior chamber-associated immune deviation and CD8(+) regulatory T cells also participate in suppression of immune responses like experimental autoimmune encephalomyelitis, we investigate whether the similar suppressive effects are involved in α-galactosylceramide-induced immune tolerance in non-obese diabetic mice. We demonstrate that repeated exposure of α-galactosylceramide reveals a hyporesponsiveness of total or antigen-presenting cells-depleted splenocytes upon anti-CD3/28 antibodies stimulation. The dispensability of dendritic cells in the hyporesponsiveness is consistent with the comparable expression of costimulatory molecules on CD11c(+) subsets between α-galactosylceramide- and vehicle-treated mice. α-Galactosylceramide treatment not only affects the effector T cell subsets and their cytokine production but also increases the secretion of transforming growth factor-β by splenocytes, implying the suppressive regulation. The adoptive transfer experiments demonstrate the suppressive effect of T cells from α-galactosylceramide-treated non-obese diabetic mice when co-transferred with vehicle-treated littermates. Finally, it reveals that CD8(+) subset among antigen-presenting cells-depleted splenocytes tends to confer the suppression since the protective ability vanishes upon withdrawal of CD8(+) subset. These results suggest that repeated exposure of α-galactosylceramide ameliorates autoimmune diabetes in non-obese diabetic mice mediated by CD8(+) T cell-associated suppression.
Collapse
|
28
|
Tohn R, Blumenfeld H, Haeryfar SMM, Veerapen N, Besra GS, Porcelli SA, Delovitch TL. Stimulation of a shorter duration in the state of anergy by an invariant natural killer T cell agonist enhances its efficiency of protection from type 1 diabetes. Clin Exp Immunol 2011; 164:26-41. [PMID: 21361909 DOI: 10.1111/j.1365-2249.2011.04323.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We have reported previously that treatment of non-obese diabetic (NOD) mice with the invariant natural killer T (iNK T) cell agonist α-galactosylceramide C26:0 (α-GalCer) or its T helper type 2 (Th2)-biasing derivative α-GalCer C20:2 (C20:2) protects against type 1 diabetes (T1D), with C20:2 yielding greater protection. After an initial response to α-GalCer, iNK T cells become anergic upon restimulation. While such anergic iNK T cells can induce tolerogenic dendritic cells (DCs) that mediate protection from T1D, chronic administration of α-GalCer also results in long-lasting anergy accompanied by significantly reduced iNK T cell frequencies, which raises concerns about its long-term therapeutic use. In this study, our objective was to understand more clearly the roles of anergy and induction of tolerogenic DCs in iNK T cell-mediated protection from T1D and to circumvent potential complications associated with α-GalCer. We demonstrate that NOD iNK T cells activated during multi-dose (MD) treatment in vivo with C20:2 enter into and exit from anergy more rapidly than after activation by α-GalCer. Importantly, this shorter duration of iNK T cells in the anergic state promotes the more rapid induction of tolerogenic DCs and reduced iNK T cell death, and enables C20:2 stimulated iNK T cells to elicit enhanced protection from T1D. Our findings further that suggest C20:2 is a more effective therapeutic drug than α-GalCer for protection from T1D. Moreover, the characteristics of C20:2 provide a basis of selection of next-generation iNK T cell agonists for the prevention of T1D.
Collapse
Affiliation(s)
- R Tohn
- Laboratory of Autoimmune Diabetes, Robarts Research Institute Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
29
|
Jordan MA, Fletcher JM, Jose R, Chowdhury S, Gerlach N, Allison J, Baxter AG. Role of SLAM in NKT cell development revealed by transgenic complementation in NOD mice. THE JOURNAL OF IMMUNOLOGY 2011; 186:3953-65. [PMID: 21357537 DOI: 10.4049/jimmunol.1003305] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Allelic variation of SLAM expression on CD4(+)CD8(+) thymocytes has been proposed to play a major role in NKT cell development. In this article, this hypothesis is tested by the production of subcongenic mouse strains and Slamf1 transgenic lines. The long isoform of the C57BL/6 allele of Slamf1 was transgenically expressed on CD4(+)CD8(+) thymocytes under control of an hCD2 minigene. NOD.Nkrp1b.Tg(Slamf1)1 mice, which had a 2-fold increase in SLAM protein expression on CD4(+)CD8(+) thymocytes, had a 2-fold increase in numbers of thymic NKT cells. The additional thymic NKT cells in NOD.Nkrp1b.Tg(Slamf1)1 mice were relatively immature, with a similar subset distribution to those of congenic NOD.Nkrp1b.Nkt1 and NOD.Nkrp1b.Slamf1 mice, which also express increased levels of SLAM on CD4(+)CD8(+) thymocytes and produce larger numbers of NKT cells. Transgenic enhancement of SLAM expression also increased IL-4 and IL-17 production in response to TCR-mediated stimulation. Paradoxically, NOD.Nkrp1b.Tg(Slamf1)2 mice, which had a 7-fold increase in SLAM expression, showed no significant increase in NKT cells numbers; on the contrary, at high transgene copy number, SLAM expression levels correlated inversely with NKT cell numbers, consistent with a contribution to negative selection. These data confirm a role for SLAM in controlling NKT cell development and are consistent with a role in both positive and negative thymic selection of NKT cells.
Collapse
Affiliation(s)
- Margaret A Jordan
- Comparative Genomics Centre, James Cook University, Townsville, Queensland 4811, Australia
| | | | | | | | | | | | | |
Collapse
|
30
|
Novak J, Lehuen A. Mechanism of regulation of autoimmunity by iNKT cells. Cytokine 2010; 53:263-70. [PMID: 21185200 DOI: 10.1016/j.cyto.2010.11.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Revised: 10/06/2010] [Accepted: 11/04/2010] [Indexed: 02/07/2023]
Abstract
iNKT cells, CD1d dependent natural killer T cells are a unique population of T cells. The capacity of iNKT cells to produce regulatory cytokines first provided an indication of their regulatory potential. Later on, in experimental models as well as in patients afflicted with an auto-immune disease, such as Type 1 diabetes mellitus, multiple sclerosis, and systemic lupus erythematosus along with others, a deficit in iNKT cell number was observed, suggesting the role these cells may possibly have in the prevention of auto-immune diseases. More importantly, experimental strategies which focused on increasing the volume or stimulation of iNKT cells in laboratory animals, demonstrated an improved level of protection against the development of auto-immune diseases. This article reviews the mechanism of protection against autoimmunity by iNKT cells, discusses the obstacles against and indications for the potential use of iNKT cell manipulation in the treatment of human auto-immune diseases.
Collapse
Affiliation(s)
- Jan Novak
- 3rd Faculty of Medicine, Charles University in Prague, Centre of Research for Diabetes, Endocrinological Diseases and Clinical Nutrition, Czech Republic.
| | | |
Collapse
|
31
|
Li K, Seo KH, Gao T, Zheng Q, Qi RQ, Wang H, Weiland M, Dong Z, Mi QS, Zhou L. Invariant NKT cell development and function in microRNA-223 knockout mice. Int Immunopharmacol 2010; 11:561-8. [PMID: 21094288 DOI: 10.1016/j.intimp.2010.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 10/28/2010] [Accepted: 11/01/2010] [Indexed: 11/15/2022]
Abstract
Invariant natural killer T (iNKT) cells, potent regulators of diverse immune responses, have been implicated in a number of diseases. The detailed mechanisms that drive iNKT cell development and maturation are still not completely understood. MicroRNAs (miRNAs) are small noncoding RNAs that regulate vast networks of genes that share miRNA target sequences. Our previous studies indicate that Dicer-dependent miRNAs play important roles in iNKT cell development, maturation, and function, but the roles of specific single miRNAs in this context are still lacking. Accumulated studies indicated that the miRNA miR-223 is a myeloid-specific miRNA. Here we report that miR-223 is highly expressed in thymic immature and activated splenic iNKT cells. To identify the role of miR-223 in iNKT cell development and function, miRNA-223-deficient mice were used. We have found that miR-223 deletion does not significantly interrupt iNKT cell development in the thymus, and miR-223-deficient mice have a normal frequency and number of iNKT cells in the thymus and peripheral immune organs. Furthermore, cytokine production of iNKT cells activated in vivo and in vitro shows no significant differences between miR-223 deficient mice and wild-type control. Thus, our data suggest that miR-223 may not be required for iNKT cell development and function.
Collapse
Affiliation(s)
- Kai Li
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Caielli S, Conforti-Andreoni C, Di Pietro C, Usuelli V, Badami E, Malosio ML, Falcone M. On/off TLR signaling decides proinflammatory or tolerogenic dendritic cell maturation upon CD1d-mediated interaction with invariant NKT cells. THE JOURNAL OF IMMUNOLOGY 2010; 185:7317-29. [PMID: 21078913 DOI: 10.4049/jimmunol.1000400] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Invariant NKT (iNKT) cells play an effector/adjuvant function during antimicrobial and antitumoral immunity and a regulatory role to induce immune tolerance and prevent autoimmunity. iNKT cells that differentially modulate adaptive immunity do not bear a unique phenotype and/or specific cytokine secretion profile, thus opening questions on how a single T cell subset can exert opposite immunological tasks. In this study, we show that iNKT cells perform their dual roles through a single mechanism of action relying on the cognate interaction with myeloid dendritic cells (DCs) and leading to opposite effects depending on the presence of other maturation stimuli simultaneously acting on DCs. The contact of murine purified iNKT cells with immature autologous DCs directly triggers the tolerogenic maturation of DCs, rendering them able to induce regulatory T cell differentiation and prevent autoimmune diabetes in vivo. Conversely, the interaction of the same purified iNKT cells with DCs, in the presence of simultaneous TLR4 stimulation, significantly enhances proinflammatory DC maturation and IL-12 secretion. The different iNKT cell effects are mediated through distinct mechanisms and activation of different molecular pathways within the DC: CD1d signaling and activation of the ERK1/2 pathway for the tolerogenic action, and CD40-CD40L interaction and NF-κB activation for the adjuvant effect. Our data suggest that the DC decision to undergo proinflammatory or tolerogenic maturation results from the integration of different signals received at the time of iNKT cell contact and could have important therapeutic implications for exploiting iNKT cell adjuvant/regulatory properties in autoimmune diseases, infections, and cancer.
Collapse
Affiliation(s)
- Simone Caielli
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
33
|
Seo KH, Zhou L, Meng D, Xu J, Dong Z, Mi QS. Loss of microRNAs in thymus perturbs invariant NKT cell development and function. Cell Mol Immunol 2010; 7:447-53. [PMID: 20852654 DOI: 10.1038/cmi.2010.49] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
microRNAs (miRNAs) are small noncoding RNAs that mediate RNA interference to suppress protein expression at the translational level. Accumulated evidence indicates that miRNAs play critical roles in various biological processes and disease development, including autoimmune diseases. Invariant natural killer T (iNKT) cells are an unusual CD1d-restricted subset of thymus-derived T cells that are potent regulators of diverse immune responses. Our previous studies with the mouse model of bone marrow-specific Dicer deletion suggest the involvement of Dicer-dependent miRNAs in the development and function of iNKT cells. In the present study, to further dissect the functional levels of Dicer-dependent miRNAs in regulating iNKT cell development, we generated a mouse model with the Dicer deletion in the thymus. Our data indicate that lack of miRNAs following the deletion of Dicer in the thymus severely interrupted the development and maturation of iNKT cells in the thymus and significantly decreased the number of iNKT cells in the peripheral immune organs. miRNA-deficient peripheral iNKT cells display profound defects in activation and cytokine production upon α-galactosylceramide (α-GalCer) stimulation. Our results demonstrate a critical role of the miRNA-dependent pathway in the thymus in the regulation of iNKT cell development and function.
Collapse
Affiliation(s)
- Kook-Heon Seo
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, USA
| | | | | | | | | | | |
Collapse
|
34
|
Bai-Feng L, Yong-Feng L, Ying C. Silencing inducible nitric oxide synthase protects rat pancreatic islet. Diabetes Res Clin Pract 2010; 89:268-75. [PMID: 20541824 DOI: 10.1016/j.diabres.2010.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 05/13/2010] [Accepted: 05/17/2010] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the effect of inducible nitric oxide synthase (iNOS) RNA interference on cytokine-induced injury of pancreatic islet in rats. MATERIALS AND METHODS Islets from Wistar rats were cultured in vitro and then randomly divided into five groups: group A, islets were cultured exclusively; group B, islets were transfected with negative control siRNA; group C, islets were transfected with iNOS siRNA; group D, islets were transfected with iNOS siRNA and then treated with TNF-alpha+IL-1beta; group E, islets were treated with TNF-alpha+IL-1beta. The expression of iNOS, Bax and Fas was determined by RT-PCR and Western blot. The viability of islet was examined by AO/EB staining and function was examined by glucose-stimulated insulin secretion (GSIS) assay. RESULTS The expression of iNOS and the promoting apoptosis gene Bax and Fas were significantly up-regulated by the induction of IL-1beta and TNF-alpha. Thus they led to apoptosis increase and the insulin secretion index decrease (1.87+/-0.31 vs 3.83+/-1.40, P<0.01). Silencing iNOS by RNAi prevented the up-regulation of Bax and Fas induced by cytokine, thus reduced apoptosis of islets and recovered the insulin secretion index (3.43+/-0.24 vs 1.87+/-0.31, P<0.01). CONCLUSION The apoptosis from cytokines to islets mediated by iNOS could be suppressed by RNA interference, which favors the survival and function of islets.
Collapse
Affiliation(s)
- Li Bai-Feng
- Department of General Surgery, The First Hospital of China Medical University, North Nanjing Street No 155, Shenyang 110001, Liaoning Province, China.
| | | | | |
Collapse
|
35
|
Abstract
BACKGROUND Recombinant vaccinia virus (rVV) strains expressing the immunomodulatory cholera toxin B subunit (CTB) fused to the autoantigen glutamic acid decarboxylase (GAD) or the immunosuppressive cytokine interleukin-10 (IL-10) were independently able to generate only low levels of immune suppression of type 1 diabetes mellitus (T1DM). Here we suggest that a vaccinia virus (VV)-mediated combination of CTB::GAD fusion and IL-10 proteins promises a effective and durable immunotherapeutic strategy for T1DM. METHODS To explore this hypothesis, a CTB::GAD fusion gene was co-delivered with a gene encoding IL-10 by rVV infection (rVV-CTB::GAD + rVV-IL10) into 5-7-week-old non-obese diabetic (NOD) mice. The mice were assessed for vaccine protection against development of hyperglycemia from 12 to 64 weeks of age by assessment of pancreatic inflammation (insulitis) and splenocyte-secreted interferon-gamma and IL-10 cytokine levels. RESULTS By 36 weeks of age, from 54% to 80% of the mice in the negative control animal groups (either mock-infected or inoculated with unrelated plasmid or VV) had developed hyperglycemia. Similarly, no statistically significant improvement in protection against diabetes onset was achieved by inoculation with VV expressing CTB::GAD or IL-10 independently. Surprisingly, only 20% of mice co-inoculated with rVV-CTB::GAD + rVV-IL10 developed hyperglycemia by 28 weeks of age. Other treatment groups developed hyperglycemia by 32-36 weeks. After 36 weeks, diabetes incidence no longer increased in any groups until the end of experiment at 64 weeks of age. Histological analysis of pancreatic tissues of hyperglycemic mice revealed high levels of intra-islet insulitis. Analysis of insulitis at termination of the experiment showed that euglycemic mice co-inoculated with VV expressing CTB::GAD and IL-10 had more effectively reduced inflammation in comparison with the other groups. CONCLUSIONS A combinatorial vaccination strategy based on VV co-delivery of genes encoding the immunoenhanced autoantigen CTB::GAD and the anti-inflammatory cytokine IL-10 can maintain effective and durable euglycemia and immunological homeostasis in NOD mice with prediabetes.
Collapse
Affiliation(s)
- Béla Dénes
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, California
- Department of Immunology, Central Veterinary Institute, Budapest, Hungary
| | - István Fodor
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, California
| | - William H.R. Langridge
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, California
| |
Collapse
|
36
|
Caielli S, Sorini C, Falcone M. The dangerous liaison between iNKT cells and dendritic cells: does it prevent or promote autoimmune diseases? Autoimmunity 2010; 44:11-22. [PMID: 20672910 DOI: 10.3109/08916931003782130] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Invariant natural killer T (iNKT) cells represent an important regulatory T-cell subset that perceives signals of danger and/or cellular distress and modulate the adaptive immune response accordingly. In the presence of pathogens, iNKT cells acquire an adjuvant function that is fundamental to boost anti-microbial and anti-tumor immunity. At the same time, iNKT cells can play a negative regulatory function to maintain peripheral T-cell tolerance toward self-antigens and to prevent autoimmune disease. Both these effects of iNKT cells involve the modulation of the activity of dendritic cells (DCs) through cell-cell interaction. Indeed, iNKT cells can either boost Th1 immunity by enhancing maturation of pro-inflammatory DCs or promote immune tolerance through the maturation of tolerogenic DCs. This dual action of iNKT cells opens questions on the modalities by which a single-cell subset can exert opposite effects on DCs and may even put in question the overall immunosuppressive properties of iNKT cells. This review presents the large body of evidence that shows the ability of iNKT cells to negatively regulate autoimmunity and to prevent autoimmune diseases including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, and systemic lupus erythematosus. In addition, an update is provided on the mechanisms of iNKT-DCs interactions and how this can result in inflammatory or tolerogenic responses.
Collapse
Affiliation(s)
- Simone Caielli
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | | | | |
Collapse
|
37
|
Hegde S, Fox L, Wang X, Gumperz JE. Autoreactive natural killer T cells: promoting immune protection and immune tolerance through varied interactions with myeloid antigen-presenting cells. Immunology 2010; 130:471-83. [PMID: 20465577 DOI: 10.1111/j.1365-2567.2010.03293.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Natural killer T (NKT) cells are innate T lymphocytes that are restricted by CD1d antigen-presenting molecules and recognize lipids and glycolipids as antigens. NKT cells have attracted attention for their potent immunoregulatory effects. Like other types of regulatory lymphocytes, a high proportion of NKT cells appear to be autoreactive to self antigens. Thus, as myeloid antigen-presenting cells (APCs) such as monocytes, dendritic cells (DCs) and myeloid-derived suppressor cells (MDSCs) constitutively express CD1d, NKT cells are able to interact with these APCs not only during times of immune activation but also in immunologically quiescent periods. The interactions of NKT cells with myeloid APCs can have either pro-inflammatory or tolerizing outcomes, and a central question is how the ensuing response is determined. Here we bring together published results from a variety of model systems to highlight three critical factors that influence the outcome of the NKT-APC interaction: (i) the strength of the antigenic signal delivered to the NKT cell, as determined by antigen abundance and/or T-cell receptor (TCR) affinity; (ii) the presence or absence of cytokines that costimulate NKT cells [e.g. interleukin (IL)-12, IL-18 and interferon (IFN)-alpha]; (iii) APC intrinsic factors such as differentiation state (e.g. monocyte versus DC) and Toll-like receptor (TLR) stimulation. Together with recent findings that demonstrate new links between NKT cell activation and endogenous lipid metabolism, these results outline a picture in which the functions of NKT cells are closely attuned to the existing biological context. Thus, NKT cells may actively promote tolerance until a critical level of danger signals arises, at which point they switch to activating pro-inflammatory immune responses.
Collapse
Affiliation(s)
- Subramanya Hegde
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
38
|
La Torre D, Lernmark A. Immunology of beta-cell destruction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:537-83. [PMID: 20217514 DOI: 10.1007/978-90-481-3271-3_24] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pancreatic islet beta-cells are the target for an autoimmune process that eventually results in an inability to control blood glucose due to the lack of insulin. The different steps that eventually lead to the complete loss of the beta-cells are reviewed to include the very first step of a triggering event that initiates the development of beta-cell autoimmunity to the last step of appearance of islet-cell autoantibodies, which may mark that insulitis is about to form. The observations that the initial beta-cell destruction by virus or other environmental factors triggers islet autoimmunity not in the islets but in the draining pancreatic lymph nodes are reviewed along with possible basic mechanisms of loss of tolerance to islet autoantigens. Once islet autoimmunity is established the question is how beta-cells are progressively killed by autoreactive lymphocytes which eventually results in chronic insulitis. Many of these series of events have been dissected in spontaneously diabetic mice or rats, but controlled clinical trials have shown that rodent observations are not always translated into mechanisms in humans. Attempts are therefore needed to clarify the step 1 triggering mechanisms and the step to chronic autoimmune insulitis to develop evidence-based treatment approaches to prevent type 1 diabetes.
Collapse
Affiliation(s)
- Daria La Torre
- Lund University, CRC, Department of Clinical Sciences, University Hospital MAS, SE-205 02, Malmö, Sweden.
| | | |
Collapse
|
39
|
Ly D, Tohn R, Rubin B, Blumenfeld H, Besra GS, Veerapen N, Porcelli SA, Delovitch TL. An alpha-galactosylceramide C20:2 N-acyl variant enhances anti-inflammatory and regulatory T cell-independent responses that prevent type 1 diabetes. Clin Exp Immunol 2009; 160:185-98. [PMID: 20015094 DOI: 10.1111/j.1365-2249.2009.04074.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Protection from type 1 diabetes (T1D), a T helper type 1 (Th1)-mediated disease, is achievable in non-obese diabetic (NOD) mice by treatment with alpha-galactosylceramide (alpha-GalCer) glycolipids that stimulate CD1d-restricted invariant natural killer T (iNK T) cells. While we have reported previously that the C20:2 N-acyl variant of alpha-GalCer elicits a Th2-biased cytokine response and protects NOD mice from T1D more effectively than a form of alpha-GalCer that induces mixed Th1 and Th2 responses, it remained to determine whether this protection is accompanied by heightened anti-inflammatory responses. We show that treatment of NOD mice with C20:2 diminished the activation of 'inflammatory' interleukin (IL)-12 producing CD11c(high)CD8+ myeloid dendritic cells (mDCs) and augmented the function of 'tolerogenic' DCs more effectively than treatment with the prototypical iNKT cell activator KRN7000 (alpha-GalCer C26:0) that induces Th1- and Th2-type responses. These findings correlate with a reduced capacity of C20:2 to sustain the early transactivation of T, B and NK cells. They may also explain our observation that C20:2 activated iNK T cells depend less than KRN7000 activated iNK T cells upon regulation by regulatory T cells for cytokine secretion and protection from T1D. The enhanced anti-inflammatory properties of C20:2 relative to KRN7000 suggest that C20:2 should be evaluated further as a drug to induce iNK T cell-mediated protection from T1D in humans.
Collapse
Affiliation(s)
- D Ly
- Laboratory of Autoimmune Diabetes, Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Regulation of type 1 diabetes, tuberculosis, and asthma by parasites. J Mol Med (Berl) 2009; 88:27-38. [PMID: 19844667 DOI: 10.1007/s00109-009-0546-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 07/27/2009] [Accepted: 09/18/2009] [Indexed: 10/20/2022]
Abstract
Helminth infection is a worldwide health problem. In addition to directly causing disease, helminthic infection also affects the incidence and progression of other diseases by exerting immune modulatory effects. In animal models, infection with helminthic parasites can prevent autoimmune diseases and allergic inflammatory diseases, but worsens protective immunity to certain infectious pathogens. In this review, we summarize current findings regarding the effects of helminth infection on type 1 diabetes, tuberculosis, and asthma and discuss possible mechanisms through which helminthic parasites modulate host immunity. Investigating these mechanisms could lead to treatment strategies that specifically modulate the immune response as well as address fundamental questions in immunobiology.
Collapse
|
41
|
Fletcher MT, Baxter AG. Clinical application of NKT cell biology in type I (autoimmune) diabetes mellitus. Immunol Cell Biol 2009; 87:315-23. [PMID: 19223852 DOI: 10.1038/icb.2009.5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Type 1 natural killer T (NKT) cells are a population of CD1d-restricted, regulatory T cells that exhibit various NK cell characteristics and rapidly produce cytokines on stimulation with glycolipid antigen. In type I diabetes (TID), NKT cells are thought to have a tolerogenic function, evidenced by NKT cell numerical and functional deficiencies in the nonobese diabetic (NOD) mouse, which when corrected, can ameliorate disease. The mechanisms by which NKT cells can mediate their immunosuppressive effects in NOD mice are still poorly understood, which makes successful clinical translation of NKT- cell-based therapies challenging. However, new insights into the genetic control of NKT cell deficiencies have provided some understanding of the genes that may control NKT cell number and function, potentially offering a new avenue for assessing TID risk in humans. Here, we review the mechanisms by which NKT cells are thought to prevent TID, discuss the evidence for involvement of NKT cells in the regulation of human TID and examine the genetic control of NKT cell number and function. A greater understanding of these areas will increase the chances of successful clinical manipulation of NKT cells to prevent or treat TID.
Collapse
Affiliation(s)
- Marie T Fletcher
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | | |
Collapse
|
42
|
Ueno A, Wang J, Cheng L, Im JS, Shi Y, Porcelli SA, Yang Y. Enhanced Early Expansion and Maturation of Semi-Invariant NK T Cells Inhibited Autoimmune Pathogenesis in Congenic Nonobese Diabetic Mice. THE JOURNAL OF IMMUNOLOGY 2008; 181:6789-96. [DOI: 10.4049/jimmunol.181.10.6789] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
43
|
Dufour FD, Baxter AG, Silveira PA. Interactions between B-Lymphocytes and Type 1 NKT Cells in Autoimmune Diabetes. J Immunotoxicol 2008; 5:249-57. [DOI: 10.1080/15476910802131543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
44
|
Wang J, Cho S, Ueno A, Cheng L, Xu BY, Desrosiers MD, Shi Y, Yang Y. Ligand-Dependent Induction of Noninflammatory Dendritic Cells by Anergic Invariant NKT Cells Minimizes Autoimmune Inflammation. THE JOURNAL OF IMMUNOLOGY 2008; 181:2438-45. [DOI: 10.4049/jimmunol.181.4.2438] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
45
|
Hussain S, Delovitch TL. Intravenous Transfusion of BCR-Activated B Cells Protects NOD Mice from Type 1 Diabetes in an IL-10-Dependent Manner. THE JOURNAL OF IMMUNOLOGY 2007; 179:7225-32. [DOI: 10.4049/jimmunol.179.11.7225] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
46
|
Wu L, Van Kaer L. Role of NKT cells in the digestive system. II. NKT cells and diabetes. Am J Physiol Gastrointest Liver Physiol 2007; 293:G919-22. [PMID: 17600041 DOI: 10.1152/ajpgi.00242.2007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Natural killer T (NKT) cells are a subset of regulatory T lymphocytes that recognize glycolipid antigens presented by the major histocompatibility complex class I-related glycoprotein CD1d. NKT cells have been implicated in regulating the progression of Type 1 diabetes (T1D) in human patients and in an animal model for T1D. In addition, glycolipid agonists of NKT cells have been successful in preventing diabetes in mice, raising enthusiasm for the development of NKT cell-based therapies for T1D.
Collapse
Affiliation(s)
- Lan Wu
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Medical Center North, Nashville, TN 37232, USA.
| | | |
Collapse
|
47
|
Abstract
Recent studies indicate that invariant TCR+ CD1d-restricted natural killer T (iNKT) cells play an important role in regulating the development of asthma and allergy. iNKT cells can function to skew adaptive immunity toward Th2 responses, or can act directly as effector cells at mucosal surfaces in diseases such as ulcerative colitis and bronchial asthma. In mouse models of asthma, NKT cell-deficient strains fail to develop allergen-induced airway hyperreactivity (AHR), a cardinal feature of asthma, and NKT cells are found in the lungs of patients with chronic asthma, suggesting a critical role for NKT cells in the development of AHR. However, much work remains in characterizing iNKT cells and their function in asthma, and in understanding the relationship between the iNKT cells and conventional CD4+ T cells.
Collapse
Affiliation(s)
- E H Meyer
- Division of Immunology, Children's Hospital Boston, Harvard Medical School, One Blackfan Circle, Boston, MA 02115, USA
| | | | | |
Collapse
|
48
|
Novak J, Griseri T, Beaudoin L, Lehuen A. Regulation of type 1 diabetes by NKT cells. Int Rev Immunol 2007; 26:49-72. [PMID: 17454264 DOI: 10.1080/08830180601070229] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Type 1 diabetes is an autoimmune disease due to the destruction of insulin-producing pancreatic beta cells. Natural Killer T (NKT) cells are a T-cell subset that links the innate and adaptive immune systems. NKT cells play a key regulatory role in type 1 diabetes. The absence of NKT cells correlates with exacerbation of type 1 diabetes, whereas an increased frequency and/or activation of NKT cells prevents beta-cell autoimmunity. Various mechanisms are involved in the protective effect of NKT cells. The goal is now to translate knowledge gained from mouse models into human therapeutics.
Collapse
Affiliation(s)
- Jan Novak
- INSERM U561, Université René Descartes, Hôpital Cochin/Saint Vincent de Paul. Paris. France
| | | | | | | |
Collapse
|
49
|
Umetsu DT, Meyer EH, DeKruyff RH. Natural killer T cells regulate the development of asthma. Int Rev Immunol 2007; 26:121-40. [PMID: 17454267 DOI: 10.1080/08830180601070237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Dale T Umetsu
- Division of Immunology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
50
|
Cheng L, Ueno A, Cho S, Im JS, Golby S, Hou S, Porcelli SA, Yang Y. Efficient activation of Valpha14 invariant NKT cells by foreign lipid antigen is associated with concurrent dendritic cell-specific self recognition. THE JOURNAL OF IMMUNOLOGY 2007; 178:2755-62. [PMID: 17312118 DOI: 10.4049/jimmunol.178.5.2755] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A burst release of cytokines by Valpha14 invariant NKT (iNKT) cells upon their TCR engagement critically regulates innate and adaptive immune responses. However, it remains unclear in vivo why iNKT cells respond efficiently to microbial or intracellular lipid Ags that are at low levels or that possess suboptimal antigenicity. We found that dendritic cells (DCs) potentiated iNKT cells to respond to a minimal amount of ligand alpha-galactosylceramide (alphaGalCer) through CD1d-dependent autoreactive responses that require endosomal processing and CD1d trafficking. The ability of potentiation of NKT cells was DC specific and did not depend on costimulatory signals and IL-12 production by DCs. However, DCs that failed to synthesize a major endogenous lipid Ag isoglobotrihexosylceramide were unable to potentiate NKT cells for efficient activation. Further analysis showed that differences in the level and pattern of endogenous lipid Ag presentation differentiate DCs and B cells for effective potentiation and subsequent activation of iNKT cells in the presence of an exogenous Ag. Thus, CD1d-dependent potentiation by DCs may be crucial for iNKT cell-mediated immunity against infectious agents.
Collapse
MESH Headings
- Animals
- Antigen Presentation/immunology
- Antigens/immunology
- Antigens, CD1/immunology
- Antigens, CD1d
- B-Lymphocytes/immunology
- Dendritic Cells/immunology
- Endosomes/immunology
- Galactosylceramides/immunology
- Globosides/immunology
- Immunity, Cellular
- Interleukin-12/immunology
- Killer Cells, Natural/immunology
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, SCID
- Protein Transport/immunology
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Self Tolerance/immunology
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- Trihexosylceramides/immunology
Collapse
Affiliation(s)
- Lu Cheng
- Julia McFarlane Diabetes Research Centre, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|