Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Front Endocrinol (Lausanne) ; 14: 1176566, 2023.
Article in English | MEDLINE | ID: mdl-37334284

ABSTRACT

Introduction: During the development of Autoimmune Diabetes (AD) an autoimmune attack against the Peripheral Nervous System occurs. To gain insight into this topic, analyses of Dorsal Root Ganglia (DRG) from Non-Obese Diabetic (NOD) mice were carried out. Methods: Histopathological analysis by electron and optical microscopy in DRG samples, and mRNA expression analyzes by the microarray technique in DRG and blood leukocyte samples from NOD and C57BL/6 mice were performed. Results: The results showed the formation of cytoplasmic vacuoles in DRG cells early in life that could be related to a neurodegenerative process. In view of these results, mRNA expression analyses were conducted to determine the cause and/or the molecules involved in this suspected disorder. The results showed that DRG cells from NOD mice have alterations in the transcription of a wide range of genes, which explain the previously observed alterations. In addition, differences in the transcription genes in white blood cells were also detected. Discussion: Taken together, these results indicate that functional defects are not only seen in beta cells but also in DRG in NOD mice. These results also indicate that these defects are not a consequence of the autoimmune process that takes place in NOD mice and suggest that they may be involved as triggers for its development.


Subject(s)
Diabetes Mellitus, Type 1 , Mice , Animals , Mice, Inbred NOD , Diabetes Mellitus, Type 1/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Mice, Inbred C57BL , Gene Expression , RNA, Messenger/metabolism
3.
Front Immunol ; 12: 634797, 2021.
Article in English | MEDLINE | ID: mdl-33664748

ABSTRACT

Background: Pancreatic islets are exposed to strong pro-apoptotic stimuli: inflammation and hyperglycemia, during the progression of the autoimmune diabetes (T1D). We found that the Cdk11(Cyclin Dependent Kinase 11) is downregulated by inflammation in the T1D prone NOD (non-obese diabetic) mouse model. The aim of this study is to determine the role of CDK11 in the pathogenesis of T1D and to assess the hierarchical relationship between CDK11 and Cyclin D3 in beta cell viability, since Cyclin D3, a natural ligand for CDK11, promotes beta cell viability and fitness in front of glucose. Methods: We studied T1D pathogenesis in NOD mice hemideficient for CDK11 (N-HTZ), and, in N-HTZ deficient for Cyclin D3 (K11HTZ-D3KO), in comparison to their respective controls (N-WT and K11WT-D3KO). Moreover, we exposed pancreatic islets to either pro-inflammatory cytokines in the presence of increasing glucose concentrations, or Thapsigargin, an Endoplasmic Reticulum (ER)-stress inducing agent, and assessed apoptotic events. The expression of key ER-stress markers (Chop, Atf4 and Bip) was also determined. Results: N-HTZ mice were significantly protected against T1D, and NS-HTZ pancreatic islets exhibited an impaired sensitivity to cytokine-induced apoptosis, regardless of glucose concentration. However, thapsigargin-induced apoptosis was not altered. Furthermore, CDK11 hemideficiency did not attenuate the exacerbation of T1D caused by Cyclin D3 deficiency. Conclusions: This study is the first to report that CDK11 is repressed in T1D as a protection mechanism against inflammation-induced apoptosis and suggests that CDK11 lies upstream Cyclin D3 signaling. We unveil the CDK11/Cyclin D3 tandem as a new potential intervention target in T1D.


Subject(s)
Apoptosis/drug effects , Blood Glucose/metabolism , Cyclin-Dependent Kinases/metabolism , Cytokines/pharmacology , Diabetes Mellitus, Type 1/enzymology , Inflammation/enzymology , Insulin-Secreting Cells/drug effects , Protein Serine-Threonine Kinases/physiology , Activating Transcription Factor 4/metabolism , Animals , Autoimmunity/drug effects , Cyclin D3/genetics , Cyclin D3/metabolism , Cyclin-Dependent Kinases/genetics , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Heat-Shock Proteins/metabolism , Inflammation/blood , Inflammation/genetics , Inflammation/pathology , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Thapsigargin/pharmacology , Tissue Culture Techniques , Transcription Factor CHOP/metabolism
4.
Cancer Res ; 81(4): 968-985, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33361391

ABSTRACT

Liver X receptors (LXR) are transcription factors from the nuclear receptor family that are activated by oxysterols and synthetic high-affinity agonists. In this study, we assessed the antitumor effects of synthetic LXR agonist TO901317 in a murine model of syngeneic Lewis Lung carcinoma. Treatment with TO901317 inhibited tumor growth in wild-type, but not in LXR-deficient mice, indicating that the antitumor effects of the agonist depends on functional LXR activity in host cells. Pharmacologic activation of the LXR pathway reduced the intratumoral abundance of regulatory T cells (Treg) and the expression of the Treg-attracting chemokine Ccl17 by MHCIIhigh tumor-associated macrophages (TAM). Moreover, gene expression profiling indicated a broad negative impact of the LXR agonist on other mechanisms used by TAM for the maintenance of an immunosuppressive environment. In studies exploring the macrophage response to GM-CSF or IL4, activated LXR repressed IRF4 expression, resulting in subsequent downregulation of IRF4-dependent genes including Ccl17. Taken together, this work reveals the combined actions of the LXR pathway in the control of TAM responses that contribute to the antitumoral effects of pharmacologic LXR activation. Moreover, these data provide new insights for the development of novel therapeutic options for the treatment of cancer. SIGNIFICANCE: This study reveals unrecognized roles of LXR in the transcriptional control of the tumor microenvironment and suggests use of a synthetic LXR agonist as a novel therapeutic strategy to stimulate antitumor activity.


Subject(s)
Benzoates/pharmacology , Benzylamines/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes, Regulatory/drug effects , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Liver X Receptors/agonists , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , RAW 264.7 Cells , T-Lymphocytes, Regulatory/pathology , Transcriptome/drug effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
5.
Front Immunol ; 10: 1732, 2019.
Article in English | MEDLINE | ID: mdl-31428087

ABSTRACT

Previous studies indicate that B-lymphocytes play a key role activating diabetogenic T-lymphocytes during the development of autoimmune diabetes. Recently, two transgenic NOD mouse models were generated: the NOD-PerIg and the 116C-NOD mice. In NOD-PerIg mice, B-lymphocytes acquire an activated proliferative phenotype and support accelerated autoimmune diabetes development. In contrast, in 116C-NOD mice, B-lymphocytes display an anergic-like phenotype delaying autoimmune diabetes onset and decreasing disease incidence. The present study further evaluates the T- and B-lymphocyte phenotype in both models. In islet-infiltrating B-lymphocytes (IIBLs) from 116C-NOD mice, the expression of H2-Kd and H2-Ag7 is decreased, whereas that of BAFF, BAFF-R, and TACI is increased. In contrast, IIBLs from NOD-PerIg show an increase in CD86 and FAS expression. In addition, islet-infiltrating T-lymphocytes (IITLs) from NOD-PerIg mice exhibit an increase in PD-1 expression. Moreover, proliferation assays indicate a high capacity of B-lymphocytes from NOD-PerIg mice to secrete high amounts of cytokines and induce T-lymphocyte activation compared to 116C B-lymphocytes. This functional variability between 116C and PerIg B-lymphocytes ultimately results in differences in the ability to shape T-lymphocyte phenotype. These results support the role of B-lymphocytes as key regulators of T-lymphocytes in autoimmune diabetes and provide essential information on the phenotypic characteristics of the T- and B-lymphocytes involved in the autoimmune response in autoimmune diabetes.


Subject(s)
B-Lymphocyte Subsets/immunology , Diabetes Mellitus, Type 1/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen Presentation , Autoimmunity , Clonal Anergy , Cytokines/blood , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Female , Gene Expression Profiling , Genes, Immunoglobulin , Immunophenotyping , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocyte Activation , Lymphopoiesis , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/pathology
6.
PLoS One ; 14(3): e0212431, 2019.
Article in English | MEDLINE | ID: mdl-30822308

ABSTRACT

Swine influenza viruses (SIVs), the causal agents of swine influenza, are not only important to control due to the economic losses in the swine industry, but also can be pandemic pathogens. Vaccination is one of the most relevant strategies to control and prevent influenza infection. Current human vaccines against influenza induce strain-specific immunity and annual update is required due to the virus antigenic shift phenomena. Previously, our group has reported the use of conserved hemagglutinin peptides (HA-peptides) derived from H1-influenza virus as a potential multivalent vaccine candidate. Immunization of swine with these HA-peptides elicited antibodies that recognized and neutralized heterologous influenza viruses in vitro and demonstrated strong hemagglutination-inhibiting activity. In the present work, we cloned one HA-peptide (named NG34) into a plasmid fused with cytotoxic T lymphocyte-associated antigen (CTLA4) which is a molecule that modifies T cell activation and with an adjuvant activity interfering with the adaptive immune response. The resulting plasmid, named pCMV-CTLA4-Ig-NG34, was administered twice to animals employing a needle-free delivery approach. Two studies were carried out to test the efficacy of pCMV-CTLA4-Ig-NG34 as a potential swine influenza vaccine, one in seronegative and another in seropositive pigs against SIV. The second one was aimed to evaluate whether pCMV-CTLA4-Ig-NG34 vaccination would overcome maternally derived antibodies (MDA). After immunization, all animals were intranasally challenged with an H3N2 influenza strain. A complete elimination or significant reduction in the viral shedding was observed within the first week after the challenge in the vaccinated animals from both studies. In addition, no challenged heterologous virus load was detected in the airways of vaccinated pigs. Overall, it is suggested that the pCMV-CTLA4-Ig-NG34 vaccine formulation could potentially be used as a multivalent vaccine against influenza viruses.


Subject(s)
Abatacept , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines , Orthomyxoviridae Infections , Peptides , Swine Diseases , Vaccines, DNA , Virus Shedding , Abatacept/genetics , Abatacept/immunology , Abatacept/pharmacology , Animals , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Madin Darby Canine Kidney Cells , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Peptides/genetics , Peptides/immunology , Peptides/pharmacology , Plasmids/genetics , Plasmids/immunology , Plasmids/pharmacology , Swine , Swine Diseases/genetics , Swine Diseases/immunology , Swine Diseases/prevention & control , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology , Virus Shedding/drug effects , Virus Shedding/genetics , Virus Shedding/immunology
7.
Sci Rep ; 8(1): 8106, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802270

ABSTRACT

Type 1 diabetes can be overcome by regulatory T cells (Treg) in NOD mice yet an efficient method to generate and maintain antigen-specific Treg is difficult to come by. Here, we devised a combination therapy of peptide/MHC tetramers and IL-2/anti-IL-2 monoclonal antibody complexes to generate antigen-specific Treg and maintain them over extended time periods. We first optimized treatment protocols conceived to obtain an improved islet-specific Treg/effector T cell ratio that led to the in vivo expansion and activation of these Treg as well as to an improved suppressor function. Optimized protocols were applied to treatment for testing diabetes prevention in NOD mice as well as in an accelerated T cell transfer model of T1D. The combined treatment led to robust protection against diabetes, and in the NOD model, to a close to complete prevention of insulitis. Treatment was accompanied with increased secretion of IL-10, detectable in total splenocytes and in Foxp3- CD4 T cells. Our data suggest that a dual protection mechanism takes place by the collaboration of Foxp3+ and Foxp3- regulatory cells. We conclude that antigen-specific Treg are an important target to improve current clinical interventions against this disease.


Subject(s)
Antibodies, Monoclonal/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Histocompatibility Antigens/chemistry , Interleukin-2/immunology , Peptides/chemistry , T-Lymphocytes, Regulatory/immunology , Amino Acid Sequence , Animals , Diabetes Mellitus, Type 1/drug therapy , Female , Mice , Peptides/pharmacology , Phenotype , Protein Multimerization , Protein Structure, Quaternary , T-Lymphocytes, Regulatory/drug effects
8.
Front Immunol ; 8: 2001, 2017.
Article in English | MEDLINE | ID: mdl-29403481

ABSTRACT

Autoreactive CD8+ and CD4+ T cells have been assigned independent key roles in the destruction of insulin-producing beta cells resulting in type 1 diabetes. Although CD4 help for the generation of efficient CD8+ T cell responses in lymphoid tissue has been extensively described, whether these two cell populations cooperate in islet destruction in situ remains unclear. By using intravital 2-photon microscopy in a mouse model of diabetes, we visualized both effector T cell populations in the pancreas during disease onset. CD4+ T helper cells displayed a much higher arrest in the exocrine tissue than islet-specific CD8+ T cells. This increased arrest was major histocompatibility complex (MHC) class II-dependent and locally correlated with antigen-presenting cell recruitment. CD8+ T cells deprived of continued CD4 help specifically in the pancreas, through blocking MHC class II recognition, failed to maintain optimal effector functions, which contributed to hamper diabetes progression. Thus, we provide novel insight in the cellular mechanisms regulating effector T cell functionality in peripheral tissues with important implications for immunotherapies.

9.
Diabetes ; 65(7): 1977-1987, 2016 07.
Article in English | MEDLINE | ID: mdl-26961115

ABSTRACT

While the autoimmune destruction of pancreatic ß-cells underlying type 1 diabetes (1D) development is ultimately mediated by T-cells in NOD mice and also likely humans, B-lymphocytes play an additional key pathogenic role. It appears expression of plasma membrane bound immunoglobulin (Ig) molecules that efficiently capture ß-cell antigens allows autoreactive B-lymphocytes bypassing normal tolerance induction processes to be the subset of antigen presenting cells most efficiently activating diabetogenic T-cells. NOD mice transgenically expressing Ig molecules recognizing antigens that are (insulin) or not (hen egg lysozyme; HEL) expressed by ß-cells have proven useful in dissecting the developmental basis of diabetogenic B-lymphocytes. However, these transgenic Ig specificities were originally selected for their ability to recognize insulin or HEL as foreign, rather than autoantigens. Thus, we generated and characterized NOD mice transgenically expressing an Ig molecule representative of a large proportion of naturally occurring islet-infiltrating B-lymphocytes in NOD mice recognizing the neuronal antigen peripherin. Transgenic peripherin autoreactive B-lymphocytes infiltrate NOD pancreatic islets, acquire an activated proliferative phenotype, and potently support accelerated T1D development. These results support the concept of neuronal autoimmunity as a pathogenic feature of T1D, and targeting such responses could ultimately provide an effective disease intervention approach.

10.
Nature ; 530(7591): 434-40, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26886799

ABSTRACT

Regulatory T cells hold promise as targets for therapeutic intervention in autoimmunity, but approaches capable of expanding antigen-specific regulatory T cells in vivo are currently not available. Here we show that systemic delivery of nanoparticles coated with autoimmune-disease-relevant peptides bound to major histocompatibility complex class II (pMHCII) molecules triggers the generation and expansion of antigen-specific regulatory CD4(+) T cell type 1 (TR1)-like cells in different mouse models, including mice humanized with lymphocytes from patients, leading to resolution of established autoimmune phenomena. Ten pMHCII-based nanomedicines show similar biological effects, regardless of genetic background, prevalence of the cognate T-cell population or MHC restriction. These nanomedicines promote the differentiation of disease-primed autoreactive T cells into TR1-like cells, which in turn suppress autoantigen-loaded antigen-presenting cells and drive the differentiation of cognate B cells into disease-suppressing regulatory B cells, without compromising systemic immunity. pMHCII-based nanomedicines thus represent a new class of drugs, potentially useful for treating a broad spectrum of autoimmune conditions in a disease-specific manner.


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen-Presenting Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD11 Antigens/immunology , Cell Differentiation , Cytokines/immunology , Female , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Nanomedicine , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Organ Specificity , Prevalence , Solubility , T-Lymphocytes, Regulatory/cytology
12.
Vaccines (Basel) ; 3(3): 579-96, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26350596

ABSTRACT

Cholera toxin subunit B (CTB) is the nontoxic portion of cholera toxin. Its affinity to the monosialotetrahexosylganglioside (GM1) that is broadly distributed in a variety of cell types including epithelial cells of the gut and antigen presenting cells, macrophages, dendritic cells, and B cells, allows its optimal access to the immune system. CTB can easily be expressed on its own in a variety of organisms, and several approaches can be used to couple it to antigens, either by genetic fusion or by chemical manipulation, leading to strongly enhanced immune responses to the antigens. In autoimmune diseases, CTB has the capacity to evoke regulatory responses and to thereby dampen autoimmune responses, in several but not all animal models. It remains to be seen whether the latter approach translates to success in the clinic, however, the versatility of CTB to manipulate immune responses in either direction makes this protein a promising adjuvant for vaccine development.

13.
PLoS One ; 10(6): e0127057, 2015.
Article in English | MEDLINE | ID: mdl-26039878

ABSTRACT

INTRODUCTION: The development of new therapies to induce self-tolerance has been an important medical health challenge in type 1 diabetes. An ideal immunotherapy should inhibit the autoimmune attack, avoid systemic side effects and allow ß-cell regeneration. Based on the immunomodulatory effects of apoptosis, we hypothesized that apoptotic mimicry can help to restore tolerance lost in autoimmune diabetes. OBJECTIVE: To generate a synthetic antigen-specific immunotherapy based on apoptosis features to specifically reestablish tolerance to ß-cells in type 1 diabetes. METHODS: A central event on the surface of apoptotic cells is the exposure of phosphatidylserine, which provides the main signal for efferocytosis. Therefore, phosphatidylserine-liposomes loaded with insulin peptides were generated to simulate apoptotic cells recognition by antigen presenting cells. The effect of antigen-specific phosphatidylserine-liposomes in the reestablishment of peripheral tolerance was assessed in NOD mice, the spontaneous model of autoimmune diabetes. MHC class II-peptide tetramers were used to analyze the T cell specific response after treatment with phosphatidylserine-liposomes loaded with peptides. RESULTS: We have shown that phosphatidylserine-liposomes loaded with insulin peptides induce tolerogenic dendritic cells and impair autoreactive T cell proliferation. When administered to NOD mice, liposome signal was detected in the pancreas and draining lymph nodes. This immunotherapy arrests the autoimmune aggression, reduces the severity of insulitis and prevents type 1 diabetes by apoptotic mimicry. MHC class II tetramer analysis showed that peptide-loaded phosphatidylserine-liposomes expand antigen-specific CD4+ T cells in vivo. The administration of phosphatidylserine-free liposomes emphasizes the importance of phosphatidylserine in the modulation of antigen-specific CD4+ T cell expansion. CONCLUSIONS: We conclude that this innovative immunotherapy based on the use of liposomes constitutes a promising strategy for autoimmune diseases.


Subject(s)
Autoantigens/immunology , Autoimmunity , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Phosphatidylserines/therapeutic use , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cytokines/metabolism , Dendritic Cells/immunology , Immunotherapy , Injections, Intraperitoneal , Insulin/therapeutic use , Liposomes , Mice, Inbred NOD , Phenotype
14.
Proc Natl Acad Sci U S A ; 111(33): E3405-14, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25092329

ABSTRACT

Type 1 diabetes is an autoimmune condition caused by the lymphocyte-mediated destruction of the insulin-producing ß cells in pancreatic islets. We aimed to identify final molecular entities targeted by the autoimmune assault on pancreatic ß cells that are causally related to ß cell viability. Here, we show that cyclin D3 is targeted by the autoimmune attack on pancreatic ß cells in vivo. Cyclin D3 is down-regulated in a dose-dependent manner in ß cells by leukocyte infiltration into the islets of the nonobese diabetic (NOD) type 1 diabetes-prone mouse model. Furthermore, we established a direct in vivo causal link between cyclin D3 expression levels and ß-cell fitness and viability in the NOD mice. We found that changes in cyclin D3 expression levels in vivo altered the ß-cell apoptosis rates, ß-cell area homeostasis, and ß-cell sensitivity to glucose without affecting ß-cell proliferation in the NOD mice. Cyclin D3-deficient NOD mice exhibited exacerbated diabetes and impaired glucose responsiveness; conversely, transgenic NOD mice overexpressing cyclin D3 in ß cells exhibited mild diabetes and improved glucose responsiveness. Overexpression of cyclin D3 in ß cells of cyclin D3-deficient mice rescued them from the exacerbated diabetes observed in transgene-negative littermates. Moreover, cyclin D3 overexpression protected the NOD-derived insulinoma NIT-1 cell line from cytokine-induced apoptosis. Here, for the first time to our knowledge, cyclin D3 is identified as a key molecule targeted by autoimmunity that plays a nonredundant, protective, and cell cycle-independent role in ß cells against inflammation-induced apoptosis and confers metabolic fitness to these cells.


Subject(s)
Cell Cycle , Cyclin D3/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Islets of Langerhans/pathology , Animals , Apoptosis , Cytokines/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Mice , Mice, Inbred NOD
15.
J Immunol ; 192(7): 3080-90, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24610011

ABSTRACT

Autoreactive B cells are essential for the pathogenesis of type 1 diabetes. The genesis and dynamics of autoreactive B cells remain unknown. In this study, we analyzed the immune response in the NOD mouse model to the neuronal protein peripherin (PRPH), a target Ag of islet-infiltrating B cells. PRPH autoreactive B cells recognized a single linear epitope of this protein, in contrast to the multiple epitope recognition commonly observed during autoreactive B cell responses. Autoantibodies to this epitope were also detected in the disease-resistant NOR and C57BL/6 strains. To specifically detect the accumulation of these B cells, we developed a novel approach, octameric peptide display, to follow the dynamics and localization of anti-PRPH B cells during disease progression. Before extended insulitis was established, anti-PRPH B cells preferentially accumulated in the peritoneum. Anti-PRPH B cells were likewise detected in C57BL/6 mice, albeit at lower frequencies. As disease unfolded in NOD mice, anti-PRPH B cells invaded the islets and increased in number at the peritoneum of diabetic but not prediabetic mice. Isotype-switched B cells were only detected in the peritoneum. Anti-PRPH B cells represent a heterogeneous population composed of both B1 and B2 subsets. In the spleen, anti-PRPH B cell were predominantly in the follicular subset. Therefore, anti-PRPH B cells represent a heterogeneous population that is generated early in life but proliferates as diabetes is established. These findings on the temporal and spatial progression of autoreactive B cells should be relevant for our understanding of B cell function in diabetes pathogenesis.


Subject(s)
B-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Peripherins/immunology , Amino Acid Sequence , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Blotting, Western , Cell Line, Tumor , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Disease Progression , Epitope Mapping/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Female , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Microscopy, Fluorescence , Molecular Sequence Data , Peripherins/genetics , Peripherins/metabolism , Peritoneum/immunology , Peritoneum/metabolism , Protein Isoforms/immunology , Spleen/immunology , Spleen/metabolism
16.
Eur J Immunol ; 43(11): 2969-79, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23925934

ABSTRACT

The cholera toxin B subunit (CTB) has been used as adjuvant to improve oral vaccine delivery in type 1 diabetes. The effect of CTB/peptide formulations on Ag-specific CD4(+) T cells has remained largely unexplored. Here, using tetramer analysis, we investigated how oral delivery of CTB fused to two CD4(+) T-cell epitopes, the BDC-2.5 T-cell 2.5 mi mimotope and glutamic acid decarboxylase (GAD) 286-300, affected diabetogenic CD4(+) T cells in nonobese diabetic (NOD) mice. When administered i.p., CTB-2.5 mi activated 2.5 mi(+) T cells and following intragastric delivery generated Ag-specific Foxp3(+) Treg and Th2 cells. While 2.5 mi(+) and GAD-specific T cells were tolerized in diabetes-resistant NODxB6.Foxp3(EGFP) F1 and nonobese resistant (NOR) mice, this did not occur in NOD mice. This indicated that NOD mice had a recessive genetic resistance to induce oral tolerance to both CTB-fused epitopes. In contrast to NODxB6.Foxp3(EGFP) F1 mice, oral treatment in NOD mice lead to strong 2.5 mi(+) T-cell activation and the sequestration of these cells to the effector-memory pool. Oral treatment of NOD mice with CTB-2.5 mi failed to prevent diabetes. These findings underline the importance of investigating the effect of oral vaccine formulations on diabetogenic T cells as in selected cases they may have counterproductive consequences in human patients.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cholera Toxin/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , Peptide Fragments/immunology , Recombinant Fusion Proteins/immunology , Administration, Oral , Animals , CD4-Positive T-Lymphocytes/immunology , Cholera Toxin/administration & dosage , Cholera Vaccines/immunology , Glutamate Decarboxylase/immunology , Immune Tolerance/immunology , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Peptide Fragments/administration & dosage
17.
RNA Biol ; 10(5): 708-15, 2013 May.
Article in English | MEDLINE | ID: mdl-23392250

ABSTRACT

Prokaryotic immunity against foreign nucleic acids mediated by clustered, regularly interspaced, short palindromic repeats (CRISPR) depends on the expression of the CRISPR-associated (Cas) proteins and the formation of small CRISPR RNAs (crRNAs). The crRNA-loaded Cas ribonucleoprotein complexes convey the specific recognition and inactivation of target nucleic acids. In E. coli K12, the maturation of crRNAs and the interference with target DNA is performed by the Cascade complex. The transcription of the Cascade operon is tightly repressed through H-NS-dependent inhibition of the Pcas promoter. Elevated levels of the LysR-type regulator LeuO induce the Pcas promoter and concomitantly activate the CRISPR-mediated immunity against phages. Here, we show that the Pcas promoter can also be induced by constitutive expression of the regulator BglJ. This activation is LeuO-dependent as heterodimers of BglJ and RcsB activate leuO transcription. Each transcription factor, LeuO or BglJ, induced the transcription of the Cascade genes to comparable amounts. However, the maturation of the crRNAs was activated in LeuO but not in BglJ-expressing cells. Studies on CRISPR promoter activities, transcript stabilities, crRNA processing and Cascade protein levels were performed to answer the question why crRNA maturation is defective in BglJ-expressing cells. Our results demonstrate that the activation of Cascade gene transcription is necessary but not sufficient to turn on the CRISPR-mediated immunity and suggest a more complex regulation of the type I-E CRISPR-Cas system in E. coli.


Subject(s)
CRISPR-Associated Proteins/genetics , Escherichia coli K12/genetics , Escherichia coli Proteins/genetics , Operon , RNA, Bacterial/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/metabolism , Escherichia coli K12/chemistry , Escherichia coli K12/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Promoter Regions, Genetic , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Trans-Activators/chemistry , Transcription Factors/chemistry
18.
Mol Microbiol ; 83(6): 1109-23, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22295907

ABSTRACT

The LysR-type transcription factor LeuO is involved in regulation of pathogenicity determinants and stress responses in Enterobacteriaceae, and acts as antagonist of the global repressor H-NS. Expression of the leuO gene is repressed by H-NS, and it is upregulated in stationary phase and under amino acid starvation conditions. Here, we show that the heterodimer of the FixJ/NarL-type transcription regulators RcsB and BglJ strongly activates expression of leuO and that RcsB-BglJ regulates additional loci. Activation of leuO by RcsB-BglJ is independent of the Rcs phosphorelay system. RcsB-BglJ binds to the leuO promoter region and activates one of two leuO promoters mapped in vivo. Moreover, LeuO antagonizes activation of leuO by RcsB-BglJ and acts as negative autoregulator in vivo and in vitro. Further, the H-NS paralogue StpA causes repression of leuO in addition to H-NS. Together, our data suggest a complex arrangement of regulatory elements and they indicate a feedback control mechanism of leuO expression.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Fimbriae Proteins/metabolism , Gene Expression Regulation, Bacterial , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Virulence Factors/metabolism , Base Sequence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/antagonists & inhibitors , Fimbriae Proteins/antagonists & inhibitors , Fimbriae Proteins/genetics , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Sequence Data , Operon , Promoter Regions, Genetic , Protein Binding , Trans-Activators/genetics , Transcriptional Activation , Virulence Factors/genetics
19.
Soc Sci Med ; 74(1): 58-66, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22122910

ABSTRACT

In this paper we estimate long-run effects of fetal exposure to the 1918/19 influenza pandemic for a European country. Using data from the 1970 Swiss census, we find that the male 1919 cohort that had a strongly increased likelihood of fetal exposure to the pandemic performs significantly worse in terms of educational attainment and has a lower chance of marriage than the surrounding cohorts. Further, we find similar results when we in addition use regional differences in influenza severity to test for the impact of influenza on later-life outcomes. A set of robustness tests confirm our findings.


Subject(s)
Infectious Disease Transmission, Vertical , Influenza, Human/transmission , Prenatal Exposure Delayed Effects , Aged , Aged, 80 and over , Censuses , Cohort Studies , Disease Outbreaks , Educational Status , Female , Humans , Influenza, Human/complications , Influenza, Human/epidemiology , Male , Marriage , Middle Aged , Models, Theoretical , Pregnancy , Switzerland/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL