Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Host Microbe ; 31(2): 213-227.e9, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36603588

ABSTRACT

Diet and commensals can affect the development of autoimmune diseases like type 1 diabetes (T1D). However, whether dietary interventions are microbe-mediated was unclear. We found that a diet based on hydrolyzed casein (HC) as a protein source protects non-obese diabetic (NOD) mice in conventional and germ-free (GF) conditions via improvement in the physiology of insulin-producing cells to reduce autoimmune activation. The addition of gluten (a cereal protein complex associated with celiac disease) facilitates autoimmunity dependent on microbial proteolysis of gluten: T1D develops in GF animals monocolonized with Enterococcus faecalis harboring secreted gluten-digesting proteases but not in mice colonized with protease deficient bacteria. Gluten digestion by E. faecalis generates T cell-activating peptides and promotes innate immunity by enhancing macrophage reactivity to lipopolysaccharide (LPS). Gnotobiotic NOD Toll4-negative mice monocolonized with E. faecalis on an HC + gluten diet are resistant to T1D. These findings provide insights into strategies to develop dietary interventions to help protect humans against autoimmunity.


Subject(s)
Diabetes Mellitus, Type 1 , Microbiota , Mice , Animals , Humans , Diabetes Mellitus, Type 1/prevention & control , Glutens , Mice, Inbred NOD , Proteolysis , Diet
2.
Nat Commun ; 13(1): 5901, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36202870

ABSTRACT

Production of a functional peripheral T cell compartment typically involves massive expansion of the bone marrow progenitors that seed the thymus. There are two main phases of expansion during T cell development, following T lineage commitment of double-negative (DN) 2 cells and after successful rearrangement and selection for functional TCRß chains in DN3 thymocytes, which promotes the transition of DN4 cells to the DP stage. The signals driving the expansion of DN2 thymocytes are well studied. However, factors regulating the proliferation and survival of DN4 cells remain poorly understood. Here, we uncover an unexpected link between the transcription factor Zfp335 and control of cGAS/STING-dependent cell death in post-ß-selection DN4 thymocytes. Zfp335 controls survival by sustaining expression of Ankle2, which suppresses cGAS/STING-dependent cell death. Together, this study identifies Zfp335 as a key transcription factor regulating the survival of proliferating post-ß-selection thymocytes and demonstrates a key role for the cGAS/STING pathway in driving apoptosis of developing T cells.


Subject(s)
Apoptosis , Membrane Proteins/metabolism , Thymocytes , Animals , Apoptosis/genetics , Cell Differentiation , Mice , Mice, Inbred C57BL , Nucleotidyltransferases , Thymocytes/metabolism , Thymus Gland/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Nat Commun ; 13(1): 6483, 2022 10 29.
Article in English | MEDLINE | ID: mdl-36309495

ABSTRACT

Glioblastoma (GBM) is notorious for its immunosuppressive tumor microenvironment (TME) and is refractory to immune checkpoint blockade (ICB). Here, we identify calmodulin-dependent kinase kinase 2 (CaMKK2) as a driver of ICB resistance. CaMKK2 is highly expressed in pro-tumor cells and is associated with worsened survival in patients with GBM. Host CaMKK2, specifically, reduces survival and promotes ICB resistance. Multimodal profiling of the TME reveals that CaMKK2 is associated with several ICB resistance-associated immune phenotypes. CaMKK2 promotes exhaustion in CD8+ T cells and reduces the expansion of effector CD4+ T cells, additionally limiting their tumor penetrance. CaMKK2 also maintains myeloid cells in a disease-associated microglia-like phenotype. Lastly, neuronal CaMKK2 is required for maintaining the ICB resistance-associated myeloid phenotype, is deleterious to survival, and promotes ICB resistance. Our findings reveal CaMKK2 as a contributor to ICB resistance and identify neurons as a driver of immunotherapeutic resistance in GBM.


Subject(s)
Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , CD8-Positive T-Lymphocytes , Tumor Microenvironment , Immunosuppression Therapy , Neurons/pathology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics
4.
NPJ Breast Cancer ; 7(1): 58, 2021 May 24.
Article in English | MEDLINE | ID: mdl-34031428

ABSTRACT

ID proteins are helix-loop-helix (HLH) transcriptional regulators frequently overexpressed in cancer. ID proteins inhibit basic-HLH transcription factors often blocking differentiation and sustaining proliferation. A small-molecule, AGX51, targets ID proteins for degradation and impairs ocular neovascularization in mouse models. Here we show that AGX51 treatment of cancer cell lines impairs cell growth and viability that results from an increase in reactive oxygen species (ROS) production upon ID degradation. In mouse models, AGX51 treatment suppresses breast cancer colonization in the lung, regresses the growth of paclitaxel-resistant breast tumors when combined with paclitaxel and reduces tumor burden in sporadic colorectal neoplasia. Furthermore, in cells and mice, we fail to observe acquired resistance to AGX51 likely the result of the inability to mutate the binding pocket without loss of ID function and efficient degradation of the ID proteins. Thus, AGX51 is a first-in-class compound that antagonizes ID proteins, shows strong anti-tumor effects and may be further developed for the management of multiple cancers.

5.
Proc Natl Acad Sci U S A ; 117(45): 28212-28220, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33106431

ABSTRACT

Somatic mutations are major genetic contributors to cancers and many other age-related diseases. Many disease-causing somatic mutations can initiate clonal growth prior to the appearance of any disease symptoms, yet experimental models that can be used to examine clonal abnormalities are limited. We describe a mosaic analysis system with Cre or Tomato (MASCOT) for tracking mutant cells and demonstrate its utility for modeling clonal hematopoiesis. MASCOT can be induced to constitutively express either Cre-GFP or Tomato for lineage tracing of a mutant and a reference group of cells simultaneously. We conducted mosaic analysis to assess functions of the Id3 and/or Tet2 gene in hematopoietic cell development and clonal hematopoiesis. Using Tomato-positive cells as a reference population, we demonstrated the high sensitivity of this system for detecting cell-intrinsic phenotypes during short-term or long-term tracking of hematopoietic cells. Long-term tracking of Tet2 mutant or Tet2/Id3 double-mutant cells in our MASCOT model revealed a dynamic shift from myeloid expansion to lymphoid expansion and subsequent development of lymphoma. This work demonstrates the utility of the MASCOT method in mosaic analysis of single or combined mutations, making the system suitable for modeling somatic mutations identified in humans.


Subject(s)
Integrases/genetics , Models, Genetic , Mutation/genetics , Solanum lycopersicum/genetics , Animals , Clonal Hematopoiesis/genetics , Genetic Techniques , Lymphoma/genetics , Mice , Mice, Transgenic , Mosaicism , Sequence Analysis, DNA
6.
J Immunol ; 201(7): 1907-1917, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30127089

ABSTRACT

In both NOD mice and humans, the development of type 1 diabetes (T1D) is dependent in part on autoreactive CD8+ T cells recognizing pancreatic ß cell peptides presented by often quite common MHC class I variants. Studies in NOD mice previously revealed that the common H2-Kd and/or H2-Db class I molecules expressed by this strain aberrantly lose the ability to mediate the thymic deletion of pathogenic CD8+ T cell responses through interactions with T1D susceptibility genes outside the MHC. A gene(s) mapping to proximal chromosome 7 was previously shown to be an important contributor to the failure of the common class I molecules expressed by NOD mice to mediate the normal thymic negative selection of diabetogenic CD8+ T cells. Using an inducible model of thymic negative selection and mRNA transcript analyses, we initially identified an elevated Nfkbid expression variant as a likely NOD-proximal chromosome 7 region gene contributing to impaired thymic deletion of diabetogenic CD8+ T cells. CRISPR/Cas9-mediated genetic attenuation of Nfkbid expression in NOD mice resulted in improved negative selection of autoreactive diabetogenic AI4 and NY8.3 CD8+ T cells. These results indicated that allelic variants of Nfkbid contribute to the efficiency of intrathymic deletion of diabetogenic CD8+ T cells. However, although enhancing thymic deletion of pathogenic CD8+ T cells, ablating Nfkbid expression surprisingly accelerated T1D onset that was associated with numeric decreases in both regulatory T and B lymphocytes in NOD mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chromosomes, Human, Pair 7/genetics , Diabetes Mellitus, Type 1/immunology , I-kappa B Proteins/genetics , Thymus Gland/immunology , Alleles , Animals , Autoantigens/immunology , Cell Differentiation , Cells, Cultured , Clonal Deletion , Disease Models, Animal , Disease Susceptibility , Humans , I-kappa B Proteins/metabolism , Mice , Mice, Inbred NOD , Polymorphism, Genetic
7.
Diabetes ; 67(5): 923-935, 2018 05.
Article in English | MEDLINE | ID: mdl-29472249

ABSTRACT

Improved mouse models for type 1 diabetes (T1D) therapy development are needed. T1D susceptibility is restored to normally resistant NOD.ß2m-/- mice transgenically expressing human disease-associated HLA-A*02:01 or HLA-B*39:06 class I molecules in place of their murine counterparts. T1D is dependent on pathogenic CD8+ T-cell responses mediated by these human class I variants. NOD.ß2m-/--A2.1 mice were previously used to identify ß-cell autoantigens presented by this human class I variant to pathogenic CD8+ T cells and for testing therapies to attenuate such effectors. However, NOD.ß2m-/- mice also lack nonclassical MHC I family members, including FcRn, required for antigen presentation, and maintenance of serum IgG and albumin, precluding therapies dependent on these molecules. Hence, we used CRISPR/Cas9 to directly ablate the NOD H2-Kd and H2-Db classical class I variants either individually or in tandem (cMHCI-/-). Ablation of the H2-Ag7 class II variant in the latter stock created NOD mice totally lacking in classical murine MHC expression (cMHCI/II-/-). NOD-cMHCI-/- mice retained nonclassical MHC I molecule expression and FcRn activity. Transgenic expression of HLA-A2 or -B39 restored pathogenic CD8+ T-cell development and T1D susceptibility to NOD-cMHCI-/- mice. These next-generation HLA-humanized NOD models may provide improved platforms for T1D therapy development.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Disease Models, Animal , HLA-A2 Antigen/genetics , HLA-B Antigens/genetics , Mice , beta 2-Microglobulin/genetics , Animals , CRISPR-Cas Systems , Diabetes Mellitus, Type 1/therapy , Histocompatibility Antigens Class I/genetics , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic
8.
J Immunol ; 199(11): 3757-3770, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29055002

ABSTRACT

In NOD mice and also likely humans, B lymphocytes play an important role as APC-expanding autoreactive T cell responses ultimately causing type 1 diabetes (T1D). Currently, humans at high future T1D risk can only be identified at late prodromal stages of disease indicated by markers such as insulin autoantibodies. When commenced in already insulin autoantibody+ NOD mice, continuous BAFFR-Fc treatment alone or in combination with anti-CD20 (designated combo therapy) inhibited T1D development. Despite eliciting broader B lymphocyte depletion, continuous combo therapy afforded no greater T1D protection than did BAFFR-Fc alone. As previously observed, late disease stage-initiated anti-CD20 monotherapy did not inhibit T1D, and in this study was additionally found to be associated with development of drug-blocking Abs. Promisingly, NOD mice given transient late disease stage BAFFR-Fc monotherapy were rendered T1D resistant. However, combo treatment abrogated the protective effect of transient BAFFR-Fc monotherapy. NOD mice receiving transient BAFF blockade were characterized by an enrichment of regulatory B lymphocytes that inhibit T1D development through IL-10 production, but this population is sensitive to deletion by anti-CD20 treatment. B lymphocytes from transient BAFFR-Fc-treated mice suppressed T cell proliferation to a greater extent than did those from controls. Proportions of B lymphocytes expressing CD73, an ecto-enzyme operating in a pathway converting proinflammatory ATP to anti-inflammatory adenosine, were also temporarily increased by transient BAFFR-Fc treatment, but not anti-CD20 therapy. These collective studies indicate transient BAFFR-Fc-mediated B lymphocyte depletion elicits long-term T1D protection by enriching regulatory B lymphocytes that are deleted by anti-CD20 cotherapy.


Subject(s)
B-Cell Activating Factor/antagonists & inhibitors , B-Lymphocytes, Regulatory/immunology , Diabetes Mellitus, Type 1/immunology , Immunoglobulin Fc Fragments/therapeutic use , Immunotherapy/methods , Rituximab/therapeutic use , T-Lymphocytes/immunology , Animals , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/therapeutic use , Cell Proliferation , Cells, Cultured , Combined Modality Therapy , Humans , Immunoglobulin Fc Fragments/genetics , Immunosuppression Therapy , Interleukin-10/metabolism , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD
9.
Comp Med ; 67(4): 335-343, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28830580

ABSTRACT

Segmented filamentous bacterium (SFB) a gram-positive, anaerobic, and intestinal commensal organism directly influences the development of Th17 helper cells in the small intestine of mice. In NOD mice, SFB colonization interferes with the development of type 1 diabetes (T1D), a T-cell-mediated autoimmune disease, suggesting that SFB may influence Th17 cells to inhibit Th1 populations associated with the anti-ß-cell immune response. This effect is a serious concern for investigators who use NOD mice for diabetes research because the expected incidence of disease decreases markedly when they are colonized by SFB. A room housing mice for T1D studies at The Jackson Laboratory was determined by fecal PCR testing to have widespread SFB colonization of multiple NOD strains after a steady decline in the incidence of T1D was noted. Rederivation of all NOD-related mouse strains was not feasible; therefore an alternative treatment using antibiotics to eliminate SFB from colonized mice was undertaken. After antibiotic treatment, soiled bedding from NOD mouse strains housed in SFB-free high-health-status production barrier rooms was used to reintroduce the gastrointestinal microbiota. Over the past 16 mo since treating the mice and disinfecting the mouse room, regular PCR testing has shown that no additional SFB colonization of mice has occurred, and the expected incidence of T1D has been reestablished in the offspring of treated mice.


Subject(s)
Ampicillin/pharmacology , Anti-Bacterial Agents/pharmacology , Diabetes Mellitus, Type 1/microbiology , Gastrointestinal Microbiome/drug effects , Gram-Positive Bacteria/drug effects , Gram-Positive Bacterial Infections/veterinary , Intestines/drug effects , Animal Husbandry/methods , Animals , Decontamination/methods , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Environmental Monitoring/methods , Feces/microbiology , Genetic Predisposition to Disease , Gram-Positive Bacteria/classification , Gram-Positive Bacteria/immunology , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/microbiology , Host-Pathogen Interactions , Intestines/immunology , Intestines/microbiology , Mice, Inbred NOD , Phenotype , Th1 Cells/immunology , Th1 Cells/microbiology , Th17 Cells/immunology , Th17 Cells/microbiology , Time Factors
10.
J Immunol ; 198(11): 4255-4267, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28461573

ABSTRACT

B lymphocytes play a key role in type 1 diabetes (T1D) development by serving as a subset of APCs preferentially supporting the expansion of autoreactive pathogenic T cells. As a result of their pathogenic importance, B lymphocyte-targeted therapies have received considerable interest as potential T1D interventions. Unfortunately, the B lymphocyte-directed T1D interventions tested to date failed to halt ß cell demise. IgG autoantibodies marking humans at future risk for T1D indicate that B lymphocytes producing them have undergone the affinity-maturation processes of class switch recombination and, possibly, somatic hypermutation. This study found that CRISPR/Cas9-mediated ablation of the activation-induced cytidine deaminase gene required for class switch recombination/somatic hypermutation induction inhibits T1D development in the NOD mouse model. The activation-induced cytidine deaminase protein induces genome-wide DNA breaks that, if not repaired through RAD51-mediated homologous recombination, result in B lymphocyte death. Treatment with the RAD51 inhibitor 4,4'-diisothiocyanatostilbene-2, 2'-disulfonic acid also strongly inhibited T1D development in NOD mice. The genetic and small molecule-targeting approaches expanded CD73+ B lymphocytes that exert regulatory activity suppressing diabetogenic T cell responses. Hence, an initial CRISPR/Cas9-mediated genetic modification approach has identified the AID/RAD51 axis as a target for a potentially clinically translatable pharmacological approach that can block T1D development by converting B lymphocytes to a disease-inhibitory CD73+ regulatory state.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Carrier Proteins/antagonists & inhibitors , Cytidine Deaminase/antagonists & inhibitors , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Lymphocyte Activation , Nuclear Proteins/antagonists & inhibitors , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , 5'-Nucleotidase/immunology , Animals , Autoantibodies/immunology , CRISPR-Cas Systems , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA-Binding Proteins , Diabetes Mellitus, Experimental , Immunoglobulin Class Switching , Mice , Mice, Inbred NOD , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins , Somatic Hypermutation, Immunoglobulin
11.
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.

SELECTION OF CITATIONS
SEARCH DETAIL
...