Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Article in English | MEDLINE | ID: mdl-38421272

ABSTRACT

The immune system is regulated by a complex set of genetic, molecular, and cellular interactions. Rapid advances in the study of immunity and its network of interactions have been boosted by a spectrum of "omics" technologies that have generated huge amounts of data that have reached the status of big data (BD). With recent developments in artificial intelligence (AI), theoretical and clinical breakthroughs could emerge. Analyses of large data sets with AI tools will allow the formulation of new testable hypotheses open new research avenues and provide innovative strategies for regulating immunity and treating immunological diseases. This includes diagnosis and identification of rare diseases, prevention and treatment of autoimmune diseases, allergic disorders, infectious diseases, metabolomic disorders, cancer, and organ transplantation. However, ethical and regulatory challenges remain as to how these studies will be used to advance our understanding of basic immunology and how immunity might be regulated in health and disease. This will be particularly important for entities in which the complexity of interactions occurring at the same time and multiple cellular pathways have eluded conventional approaches to understanding and treatment. The analyses of BD by AI are likely to be complicated as both positive and negative outcomes of regulating immunity may have important ethical ramifications that need to be considered. We suggest there is an immediate need to develop guidelines as to how the analyses of immunological BD by AI tools should guide immune-based interventions to treat various diseases, prevent infections, and maintain health within an ethical framework.


Subject(s)
Autoimmune Diseases , Hypersensitivity , Humans , Artificial Intelligence , Big Data , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Cell Communication
2.
PLoS Pathog ; 16(5): e1008393, 2020 05.
Article in English | MEDLINE | ID: mdl-32433711

ABSTRACT

Infection with (SAg)-producing bacteria may precede or follow infection with or vaccination against influenza A viruses (IAVs). However, how SAgs alter the breadth of IAV-specific CD8+ T cell (TCD8) responses is unknown. Moreover, whether recall responses mediating heterosubtypic immunity to IAVs are manipulated by SAgs remains unexplored. We employed wild-type (WT) and mutant bacterial SAgs, SAg-sufficient/deficient Staphylococcus aureus strains, and WT, mouse-adapted and reassortant IAV strains in multiple in vivo settings to address the above questions. Contrary to the popular view that SAgs delete or anergize T cells, systemic administration of staphylococcal enterotoxin B (SEB) or Mycoplasma arthritidis mitogen before intraperitoneal IAV immunization enlarged the clonal size of 'select' IAV-specific TCD8 and reshuffled the hierarchical pattern of primary TCD8 responses. This was mechanistically linked to the TCR Vß makeup of the impacted clones rather than their immunodominance status. Importantly, SAg-expanded TCD8 retained their IFN-γ production and cognate cytolytic capacities. The enhancing effect of SEB on immunodominant TCD8 was also evident in primary responses to vaccination with heat-inactivated and live attenuated IAV strains administered intramuscularly and intranasally, respectively. Interestingly, in prime-boost immunization settings, the outcome of SEB administration depended strictly upon the time point at which this SAg was introduced. Accordingly, SEB injection before priming raised CD127highKLRG1low memory precursor frequencies and augmented the anamnestic responses of SEB-binding TCD8. By comparison, introducing SEB before boosting diminished recall responses to IAV-derived epitopes drastically and indiscriminately. This was accompanied by lower Ki67 and higher Fas, LAG-3 and PD-1 levels consistent with a pro-apoptotic and/or exhausted phenotype. Therefore, SAgs can have contrasting impacts on anti-IAV immunity depending on the naïve/memory status and the TCR composition of exposed TCD8. Finally, local administration of SEB or infection with SEB-producing S. aureus enhanced pulmonary TCD8 responses to IAV. Our findings have clear implications for superinfections and prophylactic vaccination.


Subject(s)
Immunologic Memory/immunology , Influenza A virus/immunology , Superantigens/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Female , Humans , Immunologic Memory/physiology , Influenza A virus/metabolism , Influenza, Human/immunology , Influenza, Human/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Staphylococcus aureus/immunology , Superantigens/physiology , Superinfection/immunology , Vaccination
3.
Cell Immunol ; 339: 29-32, 2019 05.
Article in English | MEDLINE | ID: mdl-30249342

ABSTRACT

Pathogenic lymphocytes aberrantly recognize and mount an immune response against self-antigens, leading to the destruction of healthy cells, tissues and organs. Recent studies have shown that both B and T lymphocytes contribute to the development, prevention and modulation of various autoimmune diseases. Regulatory T and B cell subsets appear to play a prominent role in the prevention of autoimmune diseases. The recent identification of novel regulatory Th17 cells, termed as Treg17 cells, has expanded the scope of regulatory T lymphocytes (Treg cells) in the prevention of autoimmune diseases. Similarly, novel regulatory B cell subsets, termed as Breg cells, acting on their own or by inducing Treg cells have extended the role of B lymphocytes in the prevention and regulation of autoimmune diseases. We suggest that Treg17 cells and Breg cells have an important immunoregulatory role in autoimmune diseases.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Humans
4.
Arch Immunol Ther Exp (Warsz) ; 66(1): 11-19, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28733878

ABSTRACT

Transforming growth factor (TGF)-ß has been implicated in regulation of the immune system, including autoimmunity. We have found that TGF-ß is readily produced by T cells following immunization with self-peptide epitopes that downregulate autoimmune responses in type 1 diabetes (T1D) prone nonobese diabetic (NOD) mice. These include multiple peptide epitopes derived from the islet ß-cell antigens GAD65 (GAD65 p202-221, GAD65 p217-236), GAD67 (GAD67 p210-229, GAD67 p225-244), IGRP (IGRP p123-145, IGRP p195-214) and insulin B-chain (Ins. B:9-23) that protected NOD mice from T1D. Immunization of NOD mice with the self-MHC class II I-Ag7 ß-chain-derived peptide, I-Aßg7 p54-76 also induced large amounts of TGF-ß and also protected these mice from diabetes development. These results indicate that peptides derived from disease related self-antigens and MHC class II molecules primarily induce TGF-ß producing regulatory Th3 and Tr1-like cells. TGF-ß produced by these cells could enhance the differentiation of induced regulatory iTreg and iTreg17 cells to prevent induction and progression of autoimmune diseases. We therefore suggest that peripheral immune tolerance could be induced and maintained by immunization with self-peptides that induce TGF-ß producing T cells.


Subject(s)
Diabetes Mellitus, Type 1/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/metabolism , Animals , Autoantigens/immunology , Autoimmunity , Humans , Immune Tolerance , Immunization , Mice , Mice, Inbred NOD , Peptides/immunology
5.
Nat Biotechnol ; 35(11): 1069-1076, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28967887

ABSTRACT

Technical variation in metagenomic analysis must be minimized to confidently assess the contributions of microbiota to human health. Here we tested 21 representative DNA extraction protocols on the same fecal samples and quantified differences in observed microbial community composition. We compared them with differences due to library preparation and sample storage, which we contrasted with observed biological variation within the same specimen or within an individual over time. We found that DNA extraction had the largest effect on the outcome of metagenomic analysis. To rank DNA extraction protocols, we considered resulting DNA quantity and quality, and we ascertained biases in estimates of community diversity and the ratio between Gram-positive and Gram-negative bacteria. We recommend a standardized DNA extraction method for human fecal samples, for which transferability across labs was established and which was further benchmarked using a mock community of known composition. Its adoption will improve comparability of human gut microbiome studies and facilitate meta-analyses.


Subject(s)
Chemical Fractionation/methods , DNA/chemistry , Feces/chemistry , Metagenomics , Bacteria/genetics , Computational Biology , Humans , Quality Control , Species Specificity
6.
J Immunol ; 198(7): 2805-2818, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28219889

ABSTRACT

Toxic shock syndrome (TSS) is caused by staphylococcal and streptococcal superantigens (SAgs) that provoke a swift hyperinflammatory response typified by a cytokine storm. The precipitous decline in the host's clinical status and the lack of targeted therapies for TSS emphasize the need to identify key players of the storm's initial wave. Using a humanized mouse model of TSS and human cells, we herein demonstrate that SAgs elicit in vitro and in vivo IL-17A responses within hours. SAg-triggered human IL-17A production was characterized by remarkably high mRNA stability for this cytokine. A distinct subpopulation of CD4+ effector memory T (TEM) cells that secrete IL-17A, but not IFN-γ, was responsible for early IL-17A production. We found mouse "TEM-17" cells to be enriched within the intestinal epithelium and among lamina propria lymphocytes. Furthermore, interfering with IL-17A receptor signaling in human PBMCs attenuated the expression of numerous inflammatory mediators implicated in the TSS-associated cytokine storm. IL-17A receptor blockade also abrogated the secondary effect of SAg-stimulated PBMCs on human dermal fibroblasts as judged by C/EBP δ expression. Finally, the early IL-17A response to SAgs was pathogenic because in vivo neutralization of IL-17A in humanized mice ameliorated hepatic and intestinal damage and reduced mortality. Together, our findings identify CD4+ TEM cells as a key effector of TSS and reveal a novel role for IL-17A in TSS immunopathogenesis. Our work thus elucidates a pathogenic, as opposed to protective, role for IL-17A during Gram-positive bacterial infections. Accordingly, the IL-17-IL-17R axis may provide an attractive target for the management of SAg-mediated illnesses.


Subject(s)
Interleukin-17/immunology , Shock, Septic/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Flow Cytometry , Gram-Positive Bacterial Infections/immunology , Humans , Immunologic Memory/immunology , Interleukin-17/biosynthesis , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Shock, Septic/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism
7.
J Infect Dis ; 215(5): 824-829, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28035011

ABSTRACT

During toxic shock syndrome (TSS), bacterial superantigens trigger a polyclonal T -cell response leading to a potentially catastrophic "cytokine storm". Whether innate-like invariant natural killer T (iNKT) cells, with remarkable immunomodulatory properties, participate in TSS is unclear. Using genetic and cell depletion approaches, we generated iNKT cell-deficient, superantigen-sensitive HLA-DR4-transgenic (DR4tg) mice, which were compared with their iNKT-sufficient counterparts for responsiveness to staphylococcal enterotoxin B (SEB). Both approaches indicate that iNKT cells are pathogenic in TSS. Importantly, treating DR4tg mice with a TH2-polarizing glycolipid agonist of iNKT cells reduced SEB-inflicted morbidity/mortality. Therefore, iNKT cells may constitute an attractive therapeutic target in superantigen-mediated illnesses.


Subject(s)
HLA-DR4 Antigen/genetics , Natural Killer T-Cells/immunology , Shock, Septic/immunology , Shock, Septic/prevention & control , Animals , Disease Models, Animal , Enterotoxins/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superantigens/blood , Superantigens/immunology
8.
10.
Cytokine ; 74(1): 35-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25467639

ABSTRACT

IL-22 as a cytokine is described with opposing pro-inflammatory and anti-inflammatory functions. Cell regeneration, tissue remodelling and balance between commensal bacteria in the gut and host immune system are considered as anti-inflammatory features of IL-22, whereas production of IL-22 from Th17 cells links this cytokine to pro-inflammatory pathways. Th17 cells and group 3 innate lymphoid cells (ILC3) are two major producers of IL-22 and both cell types express ROR-γt and Aryl hydrocarbon receptor (AhR) transcription factors. Typically, the immune system cells are the main producers of IL-22. However, targets of this cytokine are mostly non-hematopoietic cells such as hepatocytes, keratinocytes, and epithelial cells of lung and intestine. Association of IL-22 with other cytokines or transcription factors in different cell types might explain its contrasting role in health and disease. In this review we discuss the regulation of IL-22 production by AhR- and IL-23-driven pathways. A clear understanding of the biology of IL-22 will provide new opportunities for its application to improve human health involving many debilitating conditions.


Subject(s)
Autoimmunity , Epithelial Cells/physiology , Interleukin-23/metabolism , Interleukins/immunology , Interleukins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Differentiation , Cytokines/metabolism , Gene Expression Regulation , Humans , Interleukin-17/metabolism , Interleukins/biosynthesis , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Regeneration , Th17 Cells/immunology , Interleukin-22
11.
Inflamm Allergy Drug Targets ; 14(2): 84-93, 2015.
Article in English | MEDLINE | ID: mdl-26817477

ABSTRACT

Extensive analysis of the complexity and diversity of microbiota using metagenomics in the gut and other body sites has provided evidence that dysbiosis occurs in many disease states. With the application of next generation sequencing technology this research is starting to uncover the impact of microbiota on metabolic, physiological and immunological pathways and elucidate the cellular and molecular mechanisms involved. To highlight these advances we have focused on autoimmunity and gut and liver related diseases and discuss the opportunities and challenges of translating microbiome research towards its application in humans. Towards this goal we discuss the application of fecal microbiome transplantation (FMT) for the treatment of multiple chronic gut associated inflammatory diseases such as Clostridium difficile infection (CDI) and inflammatory bowel disease (IBD). The potential role of human migration across continents and cultures leading to alteration in their microbiome and its implication in health and disease is also discussed.


Subject(s)
Autoimmune Diseases/microbiology , Bacteria/growth & development , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Liver Diseases/microbiology , Animals , Anti-Bacterial Agents/therapeutic use , Autoimmune Diseases/epidemiology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Bacteria/drug effects , Bacteria/genetics , Bacteria/immunology , Fecal Microbiota Transplantation , Feces/microbiology , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/therapy , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/immunology , Human Migration , Humans , Liver Diseases/epidemiology , Liver Diseases/immunology , Liver Diseases/therapy , Protective Factors , Risk Factors
12.
Inflamm Allergy Drug Targets ; 13(2): 94-104, 2014.
Article in English | MEDLINE | ID: mdl-24678760

ABSTRACT

Adaptation of the whole microbial normal flora residing in a host to its natural habitat over an evolutionary peroid has resulted in peaceful coexistence with mutual benefits for both microbiota and host in steady state. This symbiotic relationship between host and microbiota has a significant impact on shaping the immune response in the host to achieve an immune tolerance to microbiota but retaining the ability to respond to invading pathogens. Perturbation of this balance by manipulation of microbial communities in the host can lead to immune dysregulation and susceptibility to diseases. By studying the host in the absence of microbiota or with alteration of microbiota the complexity of microbial impact on the immune system can be resolved. Conversely, the study of microbiota in the absence of immune system factors can show how the immune system contributes to preservation of the host-microbiota balance. The absence of molecules involved in innate or adaptive immunity in knockout models can perturb the balance between host and microbiota further adding to more immune dysregulation. A better understanding of Microbiome-immune system interaction provides a new opportunity to identify biomarkers and drug targets. This will allow the development of new therapeutic agents for modulating the immune system to improve health with little or no toxicity. The study of interplay between host and microbiota has a promising role in the design of therapeutic interventions for immunopathological diseases arising from imbalanced host and microbiota interactions.


Subject(s)
Health Status , Host-Pathogen Interactions , Immune System/immunology , Immune System/microbiology , Microbiota/immunology , Animals , Communicable Diseases/immunology , Communicable Diseases/microbiology , Communicable Diseases/therapy , Disease Susceptibility , Humans , Immune System/drug effects , Immune System/metabolism , Immune System Diseases/immunology , Immune System Diseases/microbiology , Immune System Diseases/therapy , Immunity, Cellular , Immunity, Humoral , Immunologic Factors/therapeutic use , Microbiota/drug effects , Probiotics/therapeutic use , Receptors, Immunologic/metabolism , Signal Transduction , Th17 Cells/immunology , Th17 Cells/microbiology
13.
PLoS One ; 9(2): e90439, 2014.
Article in English | MEDLINE | ID: mdl-24587363

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is a tryptophan-degrading enzyme known to suppress antitumor CD8(+) T cells (TCD8). The role of IDO in regulation of antiviral TCD8 responses is far less clear. In addition, whether IDO controls both immunodominant and subdominant TCD8 is not fully understood. This is an important question because the dominance status of tumor- and virus-specific TCD8 may determine their significance in protective immunity and in vaccine design. We evaluated the magnitude and breadth of cross-primed TCD8 responses to simian virus 40 (SV40) large T antigen as well as primary and recall TCD8 responses to influenza A virus (IAV) in the absence or presence of IDO. IDO(-/-) mice and wild-type mice treated with 1-methyl-D-tryptophan, a pharmacological inhibitor of IDO, exhibited augmented responses to immunodominant epitopes encoded by T antigen and IAV. IDO-mediated suppression of these responses was independent of CD4(+)CD25(+)FoxP3(+) regulatory T cells, which remained numerically and functionally intact in IDO(-/-) mice. Treatment with L-kynurenine failed to inhibit TCD8 responses, indicating that tryptophan metabolites are not responsible for the suppressive effect of IDO in our models. Immunodominant T antigen-specific TCD8 from IDO(-/-) mice showed increased Ki-67 expression, suggesting that they may have acquired a more vigorous proliferative capacity in vivo. In conclusion, IDO suppresses immunodominant TCD8 responses to tumor and viral antigens. Our work also demonstrates that systemic primary and recall TCD8 responses to IAV are controlled by IDO. Inhibition of IDO thus represents an attractive adjuvant strategy in boosting anticancer and antiviral TCD8 targeting highly immunogenic antigens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Tolerance/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Animals , Antigens, Polyomavirus Transforming/immunology , Antigens, Viral/immunology , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression , Immunity, Innate , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Influenza A virus/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Kynurenine/pharmacology , Lymphocyte Activation , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
14.
Eur J Immunol ; 44(4): 1170-80, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24443235

ABSTRACT

The chromogranin A (ChgA) 29-42 sequence is the antigenic epitope for the BDC2.5 CD4(+) T-cell receptor in NOD mice (H-2(g7) ). We have now characterized the binding register of the ChgA 29-42 peptide for the I-A(g7) molecule. Truncation of the peptide demonstrated that the KCVLEVISD sequence 34-42 is the binding register and extension of this sequence by flanking residues increased its binding affinity and antigenic capacity. We employed anti-ChgA peptide antibodies generated against different fragments of ChgA for immunostaining of pancreatic islet sections from NOD mice. A strong immuno-staining pattern was observed for the ChgA 17-38 peptide antibodies that overlap with the ChgA 29-42 sequence. Moreover, sera from diabetic NOD mice showed elevated titers of autoantibodies to the ChgA 29-42 peptide. These findings indicate that peptides from the N-terminal region of ChgA are able to induce cellular and humoral immune responses in NOD mice.


Subject(s)
Autoantigens/immunology , Chromogranin A/immunology , Epitope Mapping/methods , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Peptide Fragments/immunology , Amino Acid Sequence , Animals , Autoantibodies/immunology , Chromogranin A/chemistry , Chromogranin A/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Immunohistochemistry , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
15.
Indian J Med Res ; 138(5): 591-4, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24434314

ABSTRACT

Following the discovery of interleukin (IL)-17 producing T helper (Th17) cells as a distinct lineage of CD4+ T helper cells it became clear that these cells play an important role in the host defense against extracellular fungal and bacterial pathogens and participate in the pathogenesis of multiple inflammatory and autoimmune disorders. Depending on the microenvironment, Th17 cells can alter their differentiation programme ultimately giving rise to either protective or pro-inflammatory pathogenic cells. We found that besides the conventional in vitro protocol for Th17 differentiation by transforming growth factor-beta (TGF-ß) plus IL-6 cytokines, a combination of IL-23 plus IL-6 can also induce Th17 cells. The Th17 cells induced by IL-23 plus IL-6 (termed as effector Th17, Teff17 cells) are pathogenic upon adoptive transfer into non-obese diabetic (NOD) mice contributing to the development of type 1 diabetes (T1D) while cells induced by TGF-ß plus IL-6 (termed as regulatory T cells, Treg17 cells) are non pathogenic and regulatory, and suppressed the pathogenic T cells in T1D. These cells differentially expressed a number of cytokines where Teff17 cells exhibited an increase in granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-22 whereas Treg17 cells demonstrated increased expression of IL-21 and immunosuppressive cytokine IL-10. Differentiation of Th17 cells is controlled by a transcription factor, RORγT although these cells also express variable levels of T-bet and FoxP3 transcription factors. This points to a dual functional role of Th17 subsets in autoimmune diseases particularly T1D. We suggest that similar to conventional regulatory T cells (Treg), induction of regulatory Treg17 cells could play an important role in modulating and preventing certain autoimmune diseases.


Subject(s)
Autoimmune Diseases/immunology , Cell Differentiation/genetics , Diabetes Mellitus, Type 1/immunology , Interleukin-17/metabolism , Th17 Cells/metabolism , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Lineage/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Mice , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
16.
Cell Regen ; 2(1): 2, 2013.
Article in English | MEDLINE | ID: mdl-25408874

ABSTRACT

BACKGROUND: In Type 1 diabetes, the insulin-producing ß-cells within the pancreatic islets of Langerhans are destroyed. We showed previously that immunotherapy with Bacillus Calmette-Guerin (BCG) or complete Freund's adjuvant (CFA) of non-obese diabetic (NOD) mice can prevent disease process and pancreatic ß-cell loss. This was associated with increased islet Regenerating (Reg) genes expression, and elevated IL-22-producing Th17 T-cells in the pancreas. RESULTS: We hypothesized that IL-22 was responsible for the increased Reg gene expression in the pancreas. We therefore quantified the Reg1, Reg2, and Reg3δ (INGAP) mRNA expression in isolated pre-diabetic NOD islets treated with IL-22. We measured IL-22, and IL-22 receptor(R)-α mRNA expression in the pancreas and spleen of pre-diabetic and diabetic NOD mice. Our results showed: 1) Reg1 and Reg2 mRNA abundance to be significantly increased in IL-22-treated islets in vitro; 2) IL-22 mRNA expression in the pre-diabetic mouse pancreas increased with time following CFA treatment; 3) a reduced expression of IL-22Rα following CFA treatment; 4) a down-regulation in Reg1 and Reg2 mRNA expression in the pancreas of pre-diabetic mice injected with an IL-22 neutralizing antibody; and 5) an increased islet ß-cell DNA synthesis in vitro in the presence of IL-22. CONCLUSIONS: We conclude that IL-22 may contribute to the regeneration of ß-cells by up-regulating Regenerating Reg1 and Reg2 genes in the islets.

17.
Innovations (Phila) ; 7(1): 23-32, 2012.
Article in English | MEDLINE | ID: mdl-22576032

ABSTRACT

OBJECTIVE: The aim of this study was to compare the early systemic inflammatory response of the Resting Heart System (RHS; Medtronic, Minneapolis, MN USA), a miniaturized cardiopulmonary bypass (CPB) system, with two groups using a standard extracorporeal circulation system during on-pump coronary artery bypass grafting (CABG) surgery. METHODS: A total of 60 consecutive patients requiring CABG were prospectively randomized to undergo on-pump CABG using conventional CPB without cardiotomy suction (group A), conventional CPB with cardiotomy suction (group B), or the RHS (group C). Blood samples were collected at five time points: immediately before CPB, 30 minutes into CPB, immediately at the end of CPB, 30 minutes post-CPB, and 1 hour post-CPB. Inflammation was analyzed by changes in (a) levels of plasma proteins, including inflammatory cytokines (interleukin-6 [IL-6], IL-10, and tumor necrosis factor-α), chemokines (IL-8, monokine induced by interferon-γ, monocyte chemotactic protein-1, regulated on activation normal T cell expressed and secreted, and interferon-inducible protein-10), and acute phase proteins (C-reactive protein and complement protein 3); (b) biochemical variables (cardiac troponin I, hematocrit, and immunoglobulin G); and (c) cell numbers (leukocytes, neutrophils, and thrombocytes). RESULTS: The RHS showed more delayed secretion of the cytokines tumor necrosis factor-α and IL-10, chemokines monokine induced by interferon-γ (P < 0.001); IL-8, and interferon-inducible protein-10; and complement protein 3 than conventional CPB systems did. Median thrombocyte numbers were higher in the RHS group. Levels of cardiac troponin I, monocyte chemotactic protein-1, and IL-6 were lower in both the RHS and conventional CPB without suction than with suction. Levels of C-reactive protein and regulated on activation normal T cell expressed and secreted, plus leukocyte and neutrophil numbers, were similar in all groups. CONCLUSIONS: The Medtronic RHS may induce less systemic inflammation than conventional CPB systems, particularly when cardiotomy suction was used, but it did not result in improved clinical benefit.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Cardiopulmonary Bypass/methods , Coronary Artery Bypass/adverse effects , Coronary Artery Bypass/methods , Extracorporeal Circulation/methods , Inflammation/diagnosis , Postoperative Complications/diagnosis , Aged , Aged, 80 and over , Cytokines/blood , Female , Humans , Inflammation/blood , Inflammation/etiology , Male , Postoperative Complications/blood , Postoperative Complications/etiology , Prospective Studies
19.
J Interferon Cytokine Res ; 31(10): 711-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21851268

ABSTRACT

Juvenile or type 1 diabetes (T1D) involves autoimmune-mediated destruction of insulin-producing ß cells in the islets of Langerhans in the pancreas. Lack of insulin prevents the absorption and metabolism of glucose throughout the body by interfering with cell signaling. Cytokines have been shown to play a key role in ß cell destruction and regulation of autoimmunity in T1D. The multiple roles of cytokines in T1D pathogenesis, regulation, and regeneration of ß cells presents both promise and challenge for their use in immunotherapy. We found that mycobacterial adjuvants induce various regulatory T cells in the non-obese diabetic (NOD) mouse model of T1D. Cytokines produced by these cells not only regulate innate and adaptive immunity but also prevent the development of diabetes and partially restored normoglycemia in diabetic NOD mice. We discovered that adjuvant immunotherapy upregulated Regenerating (Reg) genes in the islets and induced interleukin 22 (IL-22)-producing Th17 cells. IL-22 is known to upregulate Reg gene expression in islets and could potentially induce regeneration of ß cells and prevent their apoptosis. Therefore, cytokines both induce and regulate T1D and have the potential to regenerate and preserve insulin-producing ß cells in the islets.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Immunomodulation , Insulin-Secreting Cells/immunology , Regeneration/immunology , Adaptive Immunity , Animals , Apoptosis/immunology , Autoimmunity , Diabetes Mellitus, Type 1/pathology , Humans , Immunity, Innate , Insulin-Secreting Cells/pathology , Interleukins/immunology , Mice , Mice, Inbred NOD , Th17 Cells/immunology , Th17 Cells/pathology , Interleukin-22
20.
J Immunol ; 186(7): 3831-5, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21357258

ABSTRACT

Mechanistic and therapeutic insights in autoimmune diabetes would benefit from a more complete identification of relevant autoantigens. BDC2.5 TCR transgenic NOD mice express transgenes for TCR Vα1 and Vß4 chains from the highly diabetogenic BDC2.5 CD4(+) T cell clone, which recognizes pancreatic ß cell membrane Ags presented by NOD I-A(g7) MHC class II molecules. The antigenic epitope of BDC2.5 TCR is absent in ß cells that do not express chromogranin A (ChgA) protein. However, characterization of the BDC2.5 epitope in ChgA has given inconclusive results. We have now identified a ChgA29-42 peptide within vasostatin-1, an N-terminal natural derivative of ChgA as the BDC2.5 TCR epitope. Having the necessary motif for binding to I-A(g7), it activates BDC2.5 T cells and induces an IFN-γ response. More importantly, adoptive transfer of naive BDC2.5 splenocytes activated with ChgA29-42 peptide transferred diabetes into NOD/SCID mice.


Subject(s)
Chromogranin A/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Epitopes, T-Lymphocyte/immunology , Peptide Fragments/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Cell Proliferation , Cells, Cultured , Chromogranin A/administration & dosage , Chromogranin A/metabolism , Diabetes Mellitus, Type 1/pathology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/metabolism , Humans , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...