Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cytokine ; 150: 155770, 2022 02.
Article in English | MEDLINE | ID: mdl-34839177

ABSTRACT

Signal transducer and activator of transcription 5 (STAT5) plays an important role in regulating gene expression in response to cytokines of the common (γc) chain family. In this capacity, STAT5 promotes CD8+ effector and memory T cell survival and regulatory T cell development. However, its function in conventional CD4+ T cells is less clear. In this study, the requirement of intact STAT5 signaling for CD4+ effector and memory T cell generation and maintenance was investigated by using DO11.10 TCR transgenic T cells that are genetically deficient in STAT5A or B, as well as by transducing DO11 T cells with a dominant-negative STAT5 to temporally block STAT5 function. We found that the presence of STAT5A or B alone was sufficient for primary CD4+ effector T cell generation, but not for establishing a long-lived memory cell population. Similarly, blocking STAT5 signaling during priming did not prevent initial T cell activation, but inhibited the generation of memory cells. Surprisingly, blocking STAT5 post-priming did not impact the long-term survival of CD4+ memory T cells in vivo. Mechanistically, intact STAT5B, but not STAT5A, was required for IL-7Rα re-expression in activated T cells, which is an important cytokine receptor for CD4+ memory generation. These data show that fully functional STAT5 is essential to deliver an early, non-redundant signal for memory programming during the primary CD4+ T cell response, while partial STAT5 signaling is sufficient for effector differentiation. Our results have implications for the precise use of STAT5 inhibitors to timely inhibit memory T cell responses.


Subject(s)
Immunologic Memory , STAT5 Transcription Factor , CD4-Positive T-Lymphocytes , Lymphocyte Activation , STAT5 Transcription Factor/metabolism , Signal Transduction/genetics
2.
Front Immunol ; 12: 635767, 2021.
Article in English | MEDLINE | ID: mdl-33815387

ABSTRACT

Despite many decades of investigation uncovering the autoimmune mechanisms underlying Type 1 Diabetes (T1D), translating these findings into effective therapeutics has proven extremely challenging. T1D is caused by autoreactive T cells that become inappropriately activated and kill the ß cells in the pancreas, resulting in insulin insufficiency and hyperglycemia. A large body of evidence supports the idea that the unchecked activation and expansion of autoreactive T cells in T1D is due to defects in immunosuppressive regulatory T cells (Tregs) that are critical for maintaining peripheral tolerance to islet autoantigens. Hence, repairing these Treg deficiencies is a much sought-after strategy to treat the disease. To accomplish this goal in the most precise, effective and safest way possible, restored Treg functions will need to be targeted towards suppressing the autoantigen-specific immune responses only and/or be localized in the pancreas. Here we review the most recent developments in designing Treg therapies that go beyond broad activation or expansion of non-specific polyclonal Treg populations. We focus on two cutting-edge strategies namely ex vivo generation of optimized Tregs for re-introduction in T1D patients vs direct in situ stimulation and restoration of endogenous Treg function.


Subject(s)
Adoptive Transfer , Autoimmunity , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes, Regulatory/transplantation , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Phenotype , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
3.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-31990682

ABSTRACT

Community-acquired pneumonia is a widespread disease with significant morbidity and mortality. Alveolar macrophages are tissue-resident lung cells that play a crucial role in innate immunity against bacteria that cause pneumonia. We hypothesized that alveolar macrophages display adaptive characteristics after resolution of bacterial pneumonia. We studied mice 1 to 6 months after self-limiting lung infections with Streptococcus pneumoniae, the most common cause of bacterial pneumonia. Alveolar macrophages, but not other myeloid cells, recovered from the lung showed long-term modifications of their surface marker phenotype. The remodeling of alveolar macrophages was (a) long-lasting (still observed 6 months after infection), (b) regionally localized (observed only in the affected lobe after lobar pneumonia), and (c) associated with macrophage-dependent enhanced protection against another pneumococcal serotype. Metabolomic and transcriptomic profiling revealed that alveolar macrophages of mice that recovered from pneumonia had new baseline activities and altered responses to infection that better resembled those of adult humans. The enhanced lung protection after mild and self-limiting bacterial respiratory infections includes a profound remodeling of the alveolar macrophage pool that is long-lasting; compartmentalized; and manifest across surface receptors, metabolites, and both resting and stimulated transcriptomes.


Subject(s)
Macrophages, Alveolar/immunology , Pneumonia, Pneumococcal/immunology , Animals , Cell Differentiation , Disease Models, Animal , Immunity, Innate , Lung/immunology , Mice , Myeloid Cells/cytology , Myeloid Cells/immunology
4.
PLoS One ; 14(3): e0214379, 2019.
Article in English | MEDLINE | ID: mdl-30908554

ABSTRACT

Autoantigen-specific methods to prevent and treat Type 1 Diabetes (T1D) carry high hopes to permanently cure this disease, but have largely failed in clinical trials. One suggested approach to increase the efficacy of islet antigen-specific vaccination is to combine it with a modulator of the T cell response, with the goal of reducing effector differentiation and promoting regulatory T cells (Tregs). Here we asked if addition of antibodies that block the IL-7/IL-7Rα pathway altered the T cell response to islet antigen vaccination and prevented T1D in non-obese diabetic (NOD) mice. Anti-IL-7Rα monoclonal antibodies (mAbs) reduced the numbers of islet antigen-specific T cells generated after vaccination with islet peptides and alum. However, addition of anti-IL-7Rα antibodies to peptide/alum vaccination unexpectedly increased non-specific IFN-γ, IL-2 and IL-10 cytokine production and did not result in improved prevention of T1D onset. In a second approach, we used a conjugate vaccine to deliver islet autoantigens, using Keyhole Limpet Hemocyanin (KLH) as a carrier. Islet antigen-KLH vaccination led to a significant expansion of antigen-specific Tregs and delayed diabetes onset in NOD mice. These outcomes were not further improved by addition of anti-IL-7Rα antibodies. To the contrary, blocking IL-7Rα during vaccination led to non-specific cytokine production and reduced the efficacy of a KLH-conjugated vaccine to prevent T1D. Our study thus revealed that adding anti-IL-7Rα antibodies during autoantigen immunization did not improve the efficacy of such vaccinations to prevent T1D, despite altering some aspects of the T cell response in a potentially advantageous way. Further refinement of this approach will be required to separate the beneficial from the adverse effects of anti-IL-7Rα antibodies to treat autoimmune disease.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Autoantigens/administration & dosage , Cytokines/metabolism , Diabetes Mellitus, Type 1/immunology , Interleukin-7 Receptor alpha Subunit/immunology , Animals , Antibodies, Monoclonal/pharmacology , Autoantigens/immunology , Cell Proliferation , Diabetes Mellitus, Type 1/drug therapy , Disease Models, Animal , Female , Immunization , Immunotherapy , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...