Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
1.
Sci Immunol ; 9(97): eadp1139, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39058762

ABSTRACT

Type I and III interferons (IFNs) are robustly induced during infections and protect cells against viral infection. Both type I and III IFNs are also produced at low levels in the thymus at steady state; however, their role in T cell development and immune tolerance is unclear. Here, we found that both type I and III IFNs were constitutively produced by a very small number of AIRE+ murine thymic epithelial cells, independent of microbial stimulation. Antigen-presenting cells were highly responsive to thymic IFNs, and IFNs were required for the activation and maturation of thymic type 1 conventional dendritic cells, macrophages, and B cells. Loss of IFN sensing led to reduced regulatory T cell selection, reduced T cell receptor (TCR) repertoire diversity, and enhanced autoreactive T cell responses to self-antigens expressed during peripheral IFN signaling. Thus, constitutive exposure to IFNs in the thymus is required for generating a tolerant and diverse TCR repertoire.


Subject(s)
Interferons , Mice, Inbred C57BL , Thymus Gland , Animals , Thymus Gland/immunology , Mice , Interferons/immunology , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology
2.
Nat Rev Immunol ; 24(2): 103-117, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37464188

ABSTRACT

The thymus is an evolutionarily conserved organ that supports the development of T cells. Not only does the thymic environment support the rearrangement and expression of diverse T cell receptors but also provides a unique niche for the selection of appropriate T cell clones. Thymic selection ensures that the repertoire of available T cells is both useful (being MHC-restricted) and safe (being self-tolerant). The unique antigen-presentation features of the thymus ensure that the display of self-antigens is optimal to induce tolerance to all types of self-tissue. MHC class-specific functions of CD4+ T helper cells, CD8+ killer T cells and CD4+ regulatory T cells are also established in the thymus. Finally, the thymus provides signals for the development of several minor T cell subsets that promote immune and tissue homeostasis. This Review provides an introductory-level overview of our current understanding of the sophisticated thymic selection mechanisms that ensure T cells are useful and safe.


Subject(s)
CD4-Positive T-Lymphocytes , Thymus Gland , Humans , T-Lymphocyte Subsets , Self Tolerance , T-Lymphocytes, Cytotoxic
3.
Cell Rep ; 42(11): 113323, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37889750

ABSTRACT

Intestinal colonization by antigenically foreign microbes necessitates expanded peripheral immune tolerance. Here we show commensal microbiota prime expansion of CD4 T cells unified by the Kruppel-like factor 2 (KLF2) transcriptional regulator and an essential role for KLF2+ CD4 cells in averting microbiota-driven intestinal inflammation. CD4 cells with commensal specificity in secondary lymphoid organs and intestinal tissues are enriched for KLF2 expression, and distinct from FOXP3+ regulatory T cells or other differentiation lineages. Mice with conditional KLF2 deficiency in T cells develop spontaneous rectal prolapse and intestinal inflammation, phenotypes overturned by eliminating microbiota or reconstituting with donor KLF2+ cells. Activated KLF2+ cells selectively produce IL-10, and eliminating IL-10 overrides their suppressive function in vitro and protection against intestinal inflammation in vivo. Together with reduced KLF2+ CD4 cell accumulation in Crohn's disease, a necessity for the KLF2+ subpopulation of T regulatory type 1 (Tr1) cells in sustaining commensal tolerance is demonstrated.


Subject(s)
CD4-Positive T-Lymphocytes , Microbiota , Mice , Animals , Interleukin-10/metabolism , T-Lymphocytes, Regulatory , Transcription Factors/metabolism , Inflammation/metabolism , Kruppel-Like Transcription Factors/metabolism
4.
Curr Opin Immunol ; 84: 102389, 2023 10.
Article in English | MEDLINE | ID: mdl-37738858

ABSTRACT

Interferons (IFNs) are a family of proteins that are generated in response to viral infection and induce an antiviral response in many cell types. The COVID-19 pandemic revealed that patients with inborn errors of type-I IFN immunity were more prone to severe infections, but also found that many patients with severe COVID-19 had anti-IFN autoantibodies that led to acquired defects in type-I IFN immunity. These findings revealed the previously unappreciated finding that central immune tolerance to IFN is essential to immune health. Further evidence has also highlighted the importance of IFN within the thymus and its impact on T-cell development. This review will highlight what is known of IFN's role in T-cell development, T-cell central tolerance, and the impact of IFN on the thymus.


Subject(s)
COVID-19 , Interferon Type I , Virus Diseases , Humans , Interferons , Pandemics , Thymus Gland
5.
Nat Rev Immunol ; 23(10): 697, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37528192
6.
Proc Natl Acad Sci U S A ; 120(9): e2220120120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36802427

ABSTRACT

The activation of thymic B cells is critical for their licensing as antigen presenting cells and resulting ability to mediate T cell central tolerance. The processes leading to licensing are still not fully understood. By comparing thymic B cells to activated Peyer's patch B cells at steady state, we found that thymic B cell activation starts during the neonatal period and is characterized by TCR/CD40-dependent activation, followed by immunoglobulin class switch recombination (CSR) without forming germinal centers. Transcriptional analysis also demonstrated a strong interferon signature, which was not apparent in the periphery. Thymic B cell activation and CSR were primarily dependent on type III IFN signaling, and loss of type III IFN receptor in thymic B cells resulted in reduced thymocyte regulatory T cell (Treg) development. Finally, from TCR deep sequencing, we estimate that licensed B cells induce development of a substantial fraction of the Treg cell repertoire. Together, these findings reveal the importance of steady-state type III IFN in generating licensed thymic B cells that induce T cell tolerance to activated B cells.


Subject(s)
Interferon Lambda , T-Lymphocytes, Regulatory , Humans , Infant, Newborn , Thymus Gland , Thymocytes , Receptors, Antigen, T-Cell
7.
Methods Mol Biol ; 2580: 283-292, 2023.
Article in English | MEDLINE | ID: mdl-36374464

ABSTRACT

Intra-thymic injection is a powerful tool for adoptive transfer of cells, cellular tag reagents for tracking recent thymic emigrants (RTEs), or other substances directly into the thymus. The traditional approach developed decades ago requires an invasive surgery to open the thoracic cavity and visualize the thymus. Subsequently, a technique was developed requiring only a small skin incision needed to identify the precise injection site. Nevertheless, both techniques require surgical intervention, and this can lead to elevated animal stress levels and pain which necessitates analgesic medication administration. Here we describe a less invasive technique allowing in situ visualization and transfer of cell suspensions or substances into the thymus via an ultrasound-guided intra-thymic injection approach.


Subject(s)
T-Lymphocytes , Thymus Gland , Animals , Cell Movement , Adoptive Transfer , Ultrasonography, Interventional
8.
Front Immunol ; 13: 874863, 2022.
Article in English | MEDLINE | ID: mdl-35874700

ABSTRACT

The immunosuppressive regulatory T (Treg) cells exert emerging effects on adipose tissue homeostasis and systemic metabolism. However, the metabolic regulation and effector mechanisms of Treg cells in coping with obesogenic insults are not fully understood. We have previously established an indispensable role of the O-linked N-Acetylglucosamine (O-GlcNAc) signaling in maintaining Treg cell identity and promoting Treg suppressor function, via STAT5 O-GlcNAcylation and activation. Here, we investigate the O-GlcNAc transferase (OGT)-STAT5 axis in driving the immunomodulatory function of Treg cells for metabolic homeostasis. Treg cell-specific OGT deficiency renders mice more vulnerable to high-fat diet (HFD)-induced adiposity and insulin resistance. Conversely, constitutive STAT5 activation in Treg cells confers protection against adipose tissue expansion and impaired glucose and insulin metabolism upon HFD feeding, in part by suppressing adipose lipid uptake and redistributing systemic iron storage. Treg cell function can be augmented by targeting the OGT-STAT5 axis to combat obesity and related metabolic disorders.


Subject(s)
Insulin Resistance , N-Acetylglucosaminyltransferases , STAT5 Transcription Factor , T-Lymphocytes, Regulatory , Acetylglucosamine/metabolism , Animals , Iron/metabolism , Mice , N-Acetylglucosaminyltransferases/metabolism , Obesity/metabolism , STAT5 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/metabolism
10.
Nat Immunol ; 23(7): 1042-1051, 2022 07.
Article in English | MEDLINE | ID: mdl-35637352

ABSTRACT

The thymus contains a diversity of dendritic cells (DCs) that exist in defined locations and have different antigen-processing and -presenting features. This suggests that they play nonredundant roles in mediating thymocyte selection. In an effort to eliminate SIRPα+ classic DC2 subsets, we discovered that a substantial proportion expresses the surface lectin, CD301b, in the thymus. These cells resemble the CD301b+ type 2 immune response promoting DCs that are present in the skin-draining lymph nodes. Transcriptional and phenotypic comparison to other DC subsets in the thymus revealed that thymic CD301b+ cDCs represent an activated state that exhibits enhanced antigen processing and presentation. Furthermore, a CD301b+ cDC2 subset demonstrated a type 2 cytokine signature and required steady-state interleukin-4 receptor signaling. Selective ablation of CD301b+ cDC2 subsets impaired clonal deletion without affecting regulatory T cells (Treg cells). The T cell receptor α repertoire sequencing confirmed that a cDC2 subset promotes deletion of conventional T cells with minimal effect on Treg cell selection. Together, these findings suggest that cytokine-induced activation of DCs in the thymus substantially enforces central tolerance.


Subject(s)
Clonal Deletion , Dendritic Cells , Antigen Presentation , Cytokines , Lymphocyte Activation , Thymus Gland
11.
Immunity ; 55(4): 623-638.e5, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35385697

ABSTRACT

The epithelium is an integral component of mucosal barrier and host immunity. Following helminth infection, the intestinal epithelial cells secrete "alarmin" cytokines, such as interleukin-25 (IL-25) and IL-33, to initiate the type 2 immune responses for helminth expulsion and tolerance. However, it is unknown how helminth infection and the resulting cytokine milieu drive epithelial remodeling and orchestrate alarmin secretion. Here, we report that epithelial O-linked N-Acetylglucosamine (O-GlcNAc) protein modification was induced upon helminth infections. By modifying and activating the transcription factor STAT6, O-GlcNAc transferase promoted the transcription of lineage-defining Pou2f3 in tuft cell differentiation and IL-25 production. Meanwhile, STAT6 O-GlcNAcylation activated the expression of Gsdmc family genes. The membrane pore formed by GSDMC facilitated the unconventional secretion of IL-33. GSDMC-mediated IL-33 secretion was indispensable for effective anti-helminth immunity and contributed to induced intestinal inflammation. Protein O-GlcNAcylation can be harnessed for future treatment of type 2 inflammation-associated human diseases.


Subject(s)
Alarmins , Intestinal Mucosa , Acylation , Alarmins/immunology , Anthelmintics/immunology , Biomarkers, Tumor , Cytokines , DNA-Binding Proteins , Helminthiasis/immunology , Humans , Hyperplasia , Inflammation , Interleukin-33 , Intestinal Mucosa/immunology , Mebendazole , N-Acetylglucosaminyltransferases/immunology , Pore Forming Cytotoxic Proteins , STAT6 Transcription Factor/immunology
12.
Immunity ; 55(1): 98-114.e5, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34932944

ABSTRACT

Elevated gene expression of the costimulatory receptor Icos is a hallmark of CD8+ tissue-resident memory (Trm) T cells. Here, we examined the contribution of ICOS in Trm cell differentiation. Upon transfer into WT mice, Icos-/- CD8+ T cells exhibited defective Trm generation but produced recirculating memory populations normally. ICOS deficiency or ICOS-L blockade compromised establishment of CD8+ Trm cells but not their maintenance. ICOS ligation during CD8+ T cell priming did not determine Trm induction; rather, effector CD8+ T cells showed reduced Trm differentiation after seeding into Icosl-/- mice. IcosYF/YF CD8+ T cells were compromised in Trm generation, indicating a critical role for PI3K signaling. Modest transcriptional changes in the few Icos-/- Trm cells suggest that ICOS-PI3K signaling primarily enhances the efficiency of CD8+ T cell tissue residency. Thus, local ICOS signaling promotes production of Trm cells, providing insight into the contribution of costimulatory signals in the generation of tissue-resident populations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Memory T Cells/immunology , Adoptive Transfer , Animals , Antibodies, Blocking/metabolism , Cell Differentiation , Cells, Cultured , Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
14.
Nat Commun ; 12(1): 2308, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33863906

ABSTRACT

Conventional T cells are selected by peptide-MHC expressed by cortical epithelial cells in the thymus, and not by cortical thymocytes themselves that do not express MHC I or MHC II. Instead, cortical thymocytes express non-peptide presenting MHC molecules like CD1d and MR1, and promote the selection of PLZF+ iNKT and MAIT cells, respectively. Here, we report an inducible class-I transactivator mouse that enables the expression of peptide presenting MHC I molecules in different cell types. We show that MHC I expression in DP thymocytes leads to expansion of peptide specific PLZF+ innate-like (PIL) T cells. Akin to iNKT cells, PIL T cells differentiate into three functional effector subsets in the thymus, and are dependent on SAP signaling. We demonstrate that PIL and NKT cells compete for a narrow niche, suggesting that the absence of peptide-MHC on DP thymocytes facilitates selection of non-peptide specific lymphocytes.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Immunity, Innate , Thymocytes/immunology , Thymus Gland/immunology , Animals , Cell Differentiation/immunology , Histocompatibility Antigens Class I/immunology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Mucosal-Associated Invariant T Cells/immunology , Natural Killer T-Cells/immunology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Thymocytes/metabolism , Thymus Gland/cytology
15.
Eur J Immunol ; 51(5): 1080-1088, 2021 05.
Article in English | MEDLINE | ID: mdl-33521937

ABSTRACT

TCRαß+ CD8α+ CD8ß- intestinal intraepithelial lymphocytes (CD8αα IEL) are gut T cells that maintain barrier surface homeostasis. Most CD8αα IEL are derived from thymic precursors (IELp) through a mechanism referred to as clonal diversion. In this model, self-reactive thymocytes undergo deletion in the presence of CD28 costimulation, but in its absence undergo diversion to the IEL fate. While previous reports showed that IELp were largely ß2m dependent, the APC that drive the development of these cells are poorly defined. We found that both CD80 and CD86 restrain IELp development, and conventional DCs play a prominent role. We sought to define a CD80/86 negative, MHCI positive APC that supports the development to the IEL lineage. Chimera studies showed that MHCI needs to be expressed on hematopoietic APC for selection. As thymic hematopoietic APC are heterogeneous in their expression of MHCI and costimulatory molecules, we identified four thymic APC types that were CD80/86neg/low and MHCI+ . However, selective depletion of ß2m in individual APC suggested functional redundancy. Thus, while hematopoietic APC play a critical role in clonal diversion, no single APC subset is specialized to promote the CD8αα IEL fate.


Subject(s)
Clonal Selection, Antigen-Mediated , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Lymphopoiesis , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/metabolism , Thymus Gland/cytology , Animals , Biomarkers , Cell Differentiation , Genes, MHC Class I , Immunophenotyping , Intraepithelial Lymphocytes/cytology , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Mice , Precursor Cells, T-Lymphoid/cytology , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism
16.
Hepatology ; 74(2): 704-722, 2021 08.
Article in English | MEDLINE | ID: mdl-33609303

ABSTRACT

BACKGROUND AND AIMS: Nonalcoholic steatohepatitis is rapidly becoming the leading cause of liver failure and indication for liver transplantation. Hepatic inflammation is a key feature of NASH but the immune pathways involved in this process are poorly understood. B lymphocytes are cells of the adaptive immune system that are critical regulators of immune responses. However, the role of B cells in the pathogenesis of NASH and the potential mechanisms leading to their activation in the liver are unclear. APPROACH AND RESULTS: In this study, we report that NASH livers accumulate B cells with elevated pro-inflammatory cytokine secretion and antigen-presentation ability. Single-cell and bulk RNA sequencing of intrahepatic B cells from mice with NASH unveiled a transcriptional landscape that reflects their pro-inflammatory function. Accordingly, B-cell deficiency ameliorated NASH progression, and adoptively transferring B cells from NASH livers recapitulates the disease. Mechanistically, B-cell activation during NASH involves signaling through the innate adaptor myeloid differentiation primary response protein 88 (MyD88) as B cell-specific deletion of MyD88 reduced hepatic T cell-mediated inflammation and fibrosis, but not steatosis. In addition, activation of intrahepatic B cells implicates B cell-receptor signaling, delineating a synergy between innate and adaptive mechanisms of antigen recognition. Furthermore, fecal microbiota transplantation of human NAFLD gut microbiotas into recipient mice promoted the progression of NASH by increasing the accumulation and activation of intrahepatic B cells, suggesting that gut microbial factors drive the pathogenic function of B cells during NASH. CONCLUSION: Our findings reveal that a gut microbiota-driven activation of intrahepatic B cells leads to hepatic inflammation and fibrosis during the progression of NASH through innate and adaptive immune mechanisms.


Subject(s)
B-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , Liver/pathology , Non-alcoholic Fatty Liver Disease/immunology , Adaptive Immunity , Animals , B-Lymphocytes/metabolism , Disease Models, Animal , Fecal Microbiota Transplantation , Feces/microbiology , Humans , Immunity, Innate , Liver/cytology , Liver/immunology , Lymphocyte Activation , Male , Mice , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Non-alcoholic Fatty Liver Disease/pathology , RNA-Seq , Signal Transduction/immunology , Single-Cell Analysis
17.
J Exp Med ; 217(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32687575

ABSTRACT

CD8αα intraepithelial lymphocytes (IELs) are abundant T cells that protect the gut epithelium. Their thymic precursors (IELps) include PD-1+ type A and Tbet+ type B populations, which differ in their antigen-receptor specificities. To better understand CD8αα IEL ontogeny, we performed "time-stamp" fate mapping experiments and observed that it seeds the intestine predominantly during a narrow time window in early life. Adoptively transferred IELps parked better in the intestines of young mice than in adults. In young mice, both type A and type B IELps had an S1PR1+ and α4ß7+ emigration- and mucosal-homing competent phenotype, while this was restricted to type A IELps in adults. Only CD8αα IELs established in early life were enriched in cells bearing type B IELp TCR usage. Together, our results suggest that the young intestine facilitates CD8αα IEL establishment and that early IELs are distinct from IELs established after this initial wave. These data provide novel insight into the ontogeny of CD8αα IELs.


Subject(s)
CD8 Antigens/immunology , Cell Movement/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , Intraepithelial Lymphocytes/immunology , Thymocytes/immunology , Animals , CD8 Antigens/genetics , Cell Movement/genetics , Mice , Mice, Transgenic
18.
Science ; 367(6475)2020 01 17.
Article in English | MEDLINE | ID: mdl-31949051

ABSTRACT

Negative checkpoint regulators (NCRs) temper the T cell immune response to self-antigens and limit the development of autoimmunity. Unlike all other NCRs that are expressed on activated T lymphocytes, V-type immunoglobulin domain-containing suppressor of T cell activation (VISTA) is expressed on naïve T cells. We report an unexpected heterogeneity within the naïve T cell compartment in mice, where loss of VISTA disrupted the major quiescent naïve T cell subset and enhanced self-reactivity. Agonistic VISTA engagement increased T cell tolerance by promoting antigen-induced peripheral T cell deletion. Although a critical player in naïve T cell homeostasis, the ability of VISTA to restrain naïve T cell responses was lost under inflammatory conditions. VISTA is therefore a distinctive NCR of naïve T cells that is critical for steady-state maintenance of quiescence and peripheral tolerance.


Subject(s)
B7 Antigens/physiology , Membrane Proteins/physiology , Peripheral Tolerance/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , B7 Antigens/genetics , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Tolerance/genetics , Receptors, Antigen, T-Cell/physiology
19.
Proc Natl Acad Sci U S A ; 116(44): 22262-22268, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611396

ABSTRACT

Interleukin-4 (IL-4) is produced by a unique subset of invariant natural killer T (iNKT) cells (NKT2) in the thymus in the steady state, where it conditions CD8+ T cells to become "memory-like" among other effects. However, the signals that cause NKT2 cells to constitutively produce IL-4 remain poorly defined. Using histocytometry, we observed IL-4-producing NKT2 cells localized to the thymic medulla, suggesting that medullary signals might instruct NKT2 cells to produce IL-4. Moreover, NKT2 cells receive and require T cell receptor (TCR) stimulation for continuous IL-4 production in the steady state, since NKT2 cells lost IL-4 production when intrathymically transferred into CD1d-deficient recipients. In bone marrow chimeric recipients, only hematopoietic, not stromal, antigen-presenting cells (APCs), provided such stimulation. Furthermore, using different Cre-recombinase transgenic mouse strains to specifically target CD1d deficiency to various APCs, together with the use of diphtheria toxin receptor (DTR) transgenic mouse strains to deplete various APCs, we found that macrophages were the predominant cell to stimulate NKT2 IL-4 production. Thus, NKT2 cells appear to encounter and require different activating ligands for selection in the cortex and activation in the medulla.


Subject(s)
Interleukin-4/metabolism , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Thymus Gland/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, CD1/genetics , Antigens, CD1/metabolism , Cells, Cultured , Interleukin-4/genetics , Lymphocyte Activation , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Thymus Gland/cytology
20.
J Immunol ; 203(4): 844-852, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31324724

ABSTRACT

Programmed death-1 (PD-1) inhibits T and B cell function upon ligand binding. PD-1 blockade revolutionized cancer treatment, and although numerous patients respond, some develop autoimmune-like symptoms or overt autoimmunity characterized by autoantibody production. PD-1 inhibition accelerates autoimmunity in mice, but its role in regulating germinal centers (GC) is controversial. To address the role of PD-1 in the GC reaction in type 1 diabetes, we used tetramers to phenotype insulin-specific CD4+ T and B cells in NOD mice. PD-1 or PD-L1 deficiency, and PD-1 but not PD-L2 blockade, unleashed insulin-specific T follicular helper CD4+ T cells and enhanced their survival. This was concomitant with an increase in GC B cells and augmented insulin autoantibody production. The effect of PD-1 blockade on the GC was reduced when mice were treated with a mAb targeting the insulin peptide:MHC class II complex. This work provides an explanation for autoimmune side effects following PD-1 pathway inhibition and suggests that targeting the self-peptide:MHC class II complex might limit autoimmunity arising from checkpoint blockade.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , B7-H1 Antigen/immunology , Diabetes Mellitus, Experimental/immunology , Female , Germinal Center/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD
SELECTION OF CITATIONS
SEARCH DETAIL