Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Stem Cell Reports ; 16(11): 2607-2616, 2021 11 09.
Article En | MEDLINE | ID: mdl-34678207

PBX1 regulates the balance between self-renewal and differentiation of hematopoietic stem cells and maintains proto-oncogenic transcriptional pathways in early progenitors. Its increased expression was found in myeloproliferative neoplasm (MPN) patients bearing the JAK2V617F mutation. To investigate if PBX1 contributes to MPN, and to explore its potential as therapeutic target, we generated the JP mouse strain, in which the human JAK2 mutation is induced in the absence of PBX1. Typical MPN features, such as thrombocythemia and granulocytosis, did not develop without PBX1, while erythrocytosis, initially displayed by JP mice, gradually resolved over time; splenic myeloid metaplasia and in vitro cytokine independent growth were absent upon PBX1 inactivation. The aberrant transcriptome in stem/progenitor cells from the MPN model was reverted by the absence of PBX1, demonstrating that PBX1 controls part of the molecular pathways deregulated by the JAK2V617F mutation. Modulation of the PBX1-driven transcriptional program might represent a novel therapeutic approach.


Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/metabolism , Myeloproliferative Disorders/genetics , Neoplasms/genetics , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Animals , Disease Models, Animal , Disease Progression , Gene Expression Profiling/methods , Humans , Mice, Knockout , Mice, Transgenic , Mutation , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Neoplasms/metabolism , Neoplasms/pathology , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , RNA-Seq/methods , Signal Transduction/genetics
2.
Front Immunol ; 12: 669943, 2021.
Article En | MEDLINE | ID: mdl-34211466

Major Histocompatibility Complex (MHC) class II (MHCII) deficiency (MHCII-D), also known as Bare Lymphocyte Syndrome (BLS), is a rare combined immunodeficiency due to mutations in genes regulating expression of MHCII molecules. MHCII deficiency results in impaired cellular and humoral immune responses, leading to severe infections and autoimmunity. Abnormal cross-talk with developing T cells due to the absence of MHCII expression likely leads to defects in thymic epithelial cells (TEC). However, the contribution of TEC alterations to the pathogenesis of this primary immunodeficiency has not been well characterized to date, in particular in regard to immune dysregulation. To this aim, we have performed an in-depth cellular and molecular characterization of TEC in this disease. We observed an overall perturbation of thymic structure and function in both MHCII-/- mice and patients. Transcriptomic and proteomic profiling of murine TEC revealed several alterations. In particular, we demonstrated that impairment of lymphostromal cross-talk in the thymus of MHCII-/- mice affects mTEC maturation and promiscuous gene expression and causes defects of central tolerance. Furthermore, we observed peripheral tolerance impairment, likely due to defective Treg cell generation and/or function and B cell tolerance breakdown. Overall, our findings reveal disease-specific TEC defects resulting in perturbation of central tolerance and limiting the potential benefits of hematopoietic stem cell transplantation in MHCII deficiency.


Epithelial Cells/immunology , Histocompatibility Antigens Class II/immunology , Immune Tolerance , Severe Combined Immunodeficiency/immunology , Thymus Gland/immunology , Adolescent , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Case-Control Studies , Child , Child, Preschool , Disease Models, Animal , Epithelial Cells/metabolism , Europe , Female , Hematopoietic Stem Cell Transplantation , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Homeodomain Proteins/genetics , Humans , Infant , Male , Mice, Inbred C57BL , Mice, Knockout , North America , Proteome , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/metabolism , Severe Combined Immunodeficiency/surgery , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thymocytes , Thymus Gland/metabolism , Transcriptome , Young Adult
3.
J Allergy Clin Immunol ; 146(5): 1165-1179.e11, 2020 11.
Article En | MEDLINE | ID: mdl-32311393

BACKGROUND: Severe early-onset erythroderma and gut inflammation, with massive tissue infiltration of oligoclonal activated T cells are the hallmark of Omenn syndrome (OS). OBJECTIVE: The impact of altered gut homeostasis in the cutaneous manifestations of OS remains to be clarified. METHODS: We analyzed a cohort of 15 patients with OS and the 129Sv/C57BL/6 knock-in Rag2R229Q/R229Q (Rag2R229Q) mouse model. Homing phenotypes of circulating lymphocytes were analyzed by flow cytometry. Inflammatory cytokines and chemokines were examined in the sera by ELISA and in skin biopsies by immunohistochemistry and in situ RNA hybridization. Experimental colitis was induced in mice by dextran sulfate sodium salt. RESULTS: We show that memory/activated T cells from patients with OS and from the Rag2R229Q mouse model of OS abundantly express the skin homing receptors cutaneous lymphocyte associated antigen and CCR4 (Ccr4), associated with high levels of chemokine C-C motif ligands 17 and 22. Serum levels of LPS are also elevated. A broad Th1/Th2/Th17 inflammatory signature is detected in the periphery and in the skin. Increased Tlr4 expression in the skin of Rag2R229Q mice is associated with enhanced cutaneous inflammation on local and systemic administration of LPS. Likewise, boosting colitis in Rag2R229Q mice results in increased frequency of Ccr4+ splenic T cells and worsening of skin inflammation, as indicated by epidermal thickening, enhanced epithelial cell activation, and dermal infiltration by Th1 effector T cells. CONCLUSIONS: These results support the existence of an interplay between gut and skin that can sustain skin inflammation in OS.


Dermatitis/immunology , Inflammation/immunology , Intestines/immunology , Severe Combined Immunodeficiency/immunology , Skin/pathology , Th1 Cells/immunology , Tight Junctions/pathology , Animals , Cohort Studies , DNA-Binding Proteins/genetics , Disease Models, Animal , Gastrointestinal Microbiome , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Receptors, CCR4/metabolism
4.
J Bone Miner Res ; 34(11): 2133-2148, 2019 11.
Article En | MEDLINE | ID: mdl-31295380

Controlling oxidative stress through the activation of antioxidant pathways is crucial in bone homeostasis, and impairments of the cellular defense systems involved contribute to the pathogenesis of common skeletal diseases. In this work we focused on the dipeptidyl peptidase 3 (DPP3), a poorly investigated ubiquitous zinc-dependent exopeptidase activating the Keap1-Nrf2 antioxidant pathway. We showed Dpp3 expression in bone and, to understand its role in this compartment, we generated a Dpp3 knockout (KO) mouse model and specifically investigated the skeletal phenotype. Adult Dpp3 KO mice showed a mild growth defect, a significant increase in bone marrow cellularity, and bone loss mainly caused by increased osteoclast activity. Overall, in the mouse model, lack of DPP3 resulted in sustained oxidative stress and in alterations of bone microenvironment favoring the osteoclast compared to the osteoblast lineage. Accordingly, in vitro studies revealed that Dpp3 KO osteoclasts had an inherent increased resorptive activity and ROS production, which on the other hand made them prone to apoptosis. Moreover, absence of DPP3 augmented bone loss after estrogen withdrawal in female mice, further supporting its relevance in the framework of bone pathophysiology. Overall, we show a nonredundant role for DPP3 in the maintenance of bone homeostasis and propose that DPP3 might represent a possible new osteoimmunological player and a marker of human bone loss pathology. © 2019 American Society for Bone and Mineral Research.


Bone Resorption , Cellular Microenvironment , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/deficiency , Osteoclasts , Oxidative Stress , Signal Transduction , Animals , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology
5.
J Autoimmun ; 98: 132-148, 2019 03.
Article En | MEDLINE | ID: mdl-30704941

Primary immunodeficiencies (PIDs) are inherited disorders of the immune system, associated with a considerable increase in susceptibility to infections. PIDs can also predispose to malignancy, inflammation and autoimmunity. There is increasing awareness that some aspects of the immune dysregulation in PIDs may be linked to intestinal microbiota. Indeed, the gut microbiota and its metabolites have been shown to influence immune functions and immune homeostasis both locally and systemically. Recent studies have indicated that genetic defects causing PIDs lead to perturbations in the conventional mechanisms underlying homeostasis in the gut, resulting in poor immune surveillance at the intestinal barrier, which associates with altered intestinal permeability and bacterial translocation. Consistently, a substantial proportion of PID patients presents with clinically challenging IBD-like pathology. Here, we describe the current body of literature reporting on dysbiosis of the gut microbiota in different PIDs and how this can be either the result or cause of immune dysregulation. Further, we report how infections in PIDs enhance pathobionts colonization and speculate how, in turn, pathobionts may be responsible for increased disease susceptibility and secondary infections in these patients. The potential relationship between the microbial composition in the intestine and other sites, such as the oral cavity and skin, is also highlighted. Finally, we provide evidence, in preclinical models of PIDs, for the efficacy of microbiota manipulation to ameliorate disease complications, and suggest that the potential use of dietary intervention to correct dysbiotic flora in PID patients may hold promise.


Dysbiosis/microbiology , Immunologic Deficiency Syndromes/microbiology , Microbiota/immunology , Animals , Autoimmunity , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Humans , Immune System , Immunity , Immunologic Deficiency Syndromes/immunology , Rare Diseases
6.
J Allergy Clin Immunol ; 142(3): 928-941.e8, 2018 09.
Article En | MEDLINE | ID: mdl-29241731

BACKGROUND: Omenn syndrome (OS) is a rare severe combined immunodeficiency associated with autoimmunity and caused by defects in lymphoid-specific V(D)J recombination. Most patients carry hypomorphic mutations in recombination-activating gene (RAG) 1 or 2. Hematopoietic stem cell transplantation is the standard treatment; however, gene therapy (GT) might represent a valid alternative, especially for patients lacking a matched donor. OBJECTIVE: We sought to determine the efficacy of lentiviral vector (LV)-mediated GT in the murine model of OS (Rag2R229Q/R229Q) in correcting immunodeficiency and autoimmunity. METHODS: Lineage-negative cells from mice with OS were transduced with an LV encoding the human RAG2 gene and injected into irradiated recipients with OS. Control mice underwent transplantation with wild-type or OS-untransduced lineage-negative cells. Immunophenotyping, T-dependent and T-independent antigen challenge, immune spectratyping, autoantibody detection, and detailed tissue immunohistochemical analyses were performed. RESULTS: LV-mediated GT allowed immunologic reconstitution, although it was suboptimal compared with that seen in wild-type bone marrow (BM)-transplanted OS mice in peripheral blood and hematopoietic organs, such as the BM, thymus, and spleen. We observed in vivo variability in the efficacy of GT correlating with the levels of transduction achieved. Immunoglobulin levels and T-cell repertoire normalized, and gene-corrected mice responded properly to challenges in vivo. Autoimmune manifestations, such as skin infiltration and autoantibodies, dramatically improved in GT mice with a vector copy number/genome higher than 1 in the BM and 2 in the thymus. CONCLUSIONS: Our data show that LV-mediated GT for patients with OS significantly ameliorates the immunodeficiency, even in an inflammatory environment.


DNA-Binding Proteins/genetics , Genetic Therapy , Lentivirus/genetics , Severe Combined Immunodeficiency/therapy , Animals , Autoimmunity , B-Lymphocytes/immunology , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/therapy , Lymphocyte Count , Male , Mice, Inbred C57BL , Mice, Transgenic , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology
7.
Gut Microbes ; 7(6): 503-511, 2016 11.
Article En | MEDLINE | ID: mdl-27575988

Hypomorphic Rag mutations in humans cause Omenn Syndrome (OS) a severe immunodeficiency associated with autoimmune-like manifestations mediated by oligoclonal activated T and B cells. The clinical and immunological spectrum of OS presentation is extremely broad. However, the role played by environmental triggers in the disease pathogenesis remains largely unknown. We have recently shown in a murine model that gut microbiota has a substantial role in determining the distinctive immune dysregulation of OS. Here, we describe how dysbiosis and loss of T cell tolerance to commensals influence the expression of autoimmunity at the barrier site and beyond, and the disease hallmark hyper-IgE. We discuss how commensal antigens and gut-derived pathogenic T cells could potentially modulate skin immunity to determine cutaneous degenerations in OS. These mechanisms may have broader implications for a deeper understanding of the role of gut microbes in influencing barriers integrity and host physiology.


Autoimmunity , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Animals , B-Lymphocytes/immunology , DNA-Binding Proteins/immunology , Homeodomain Proteins/immunology , Humans , Immunoglobulin E/immunology , T-Lymphocytes/immunology
8.
Mol Ther ; 24(11): 1898-1912, 2016 Nov.
Article En | MEDLINE | ID: mdl-27506451

Duchenne muscular dystrophy is an inherited fatal genetic disease characterized by mutations in dystrophin gene, causing membrane fragility leading to myofiber necrosis and inflammatory cell recruitment in dystrophic muscles. The resulting environment enriched in proinflammatory cytokines, like IFN-γ and TNF-α, determines the transformation of myofiber constitutive proteasome into the immunoproteasome, a multisubunit complex involved in the activation of cell-mediate immunity. This event has a fundamental role in producing peptides for antigen presentation by MHC class I, for the immune response and also for cytokine production and T-cell differentiation. Here, we characterized for the first time the presence of T-lymphocytes activated against revertant dystrophin epitopes, in the animal model of Duchenne muscular dystrophy, the mdx mice. Moreover, we specifically blocked i-proteasome subunit LMP7, which was up-regulated in dystrophic skeletal muscles, and we demonstrated the rescue of the dystrophin expression and the amelioration of the dystrophic phenotype. The i-proteasome blocking lowered myofiber MHC class I expression and self-antigen presentation to T cells, thus reducing the specific antidystrophin T cell response, the muscular cell infiltrate, and proinflammatory cytokine production, together with muscle force recovery. We suggest that i-proteasome inhibition should be considered as new promising therapeutic approach for Duchenne muscular dystrophy pathology.


Immunoproteins/antagonists & inhibitors , Muscular Dystrophy, Duchenne/drug therapy , Proteasome Inhibitors/administration & dosage , T-Lymphocytes/immunology , Animals , Cell Differentiation , Disease Models, Animal , Genetic Therapy , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/immunology , Oligopeptides/administration & dosage , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology
9.
J Exp Med ; 213(3): 355-75, 2016 Mar 07.
Article En | MEDLINE | ID: mdl-26926994

Omenn syndrome (OS) is caused by hypomorphic Rag mutations and characterized by a profound immunodeficiency associated with autoimmune-like manifestations. Both in humans and mice, OS is mediated by oligoclonal activated T and B cells. The role of microbial signals in disease pathogenesis is debated. Here, we show that Rag2(R229Q) knock-in mice developed an inflammatory bowel disease affecting both the small bowel and colon. Lymphocytes were sufficient for disease induction, as intestinal CD4 T cells with a Th1/Th17 phenotype reproduced the pathological picture when transplanted into immunocompromised hosts. Moreover, oral tolerance was impaired in Rag2(R229Q) mice, and transfer of wild-type (WT) regulatory T cells ameliorated bowel inflammation. Mucosal immunoglobulin A (IgA) deficiency in the gut resulted in enhanced absorption of microbial products and altered composition of commensal communities. The Rag2(R229Q) microbiota further contributed to the immunopathology because its transplant into WT recipients promoted Th1/Th17 immune response. Consistently, long-term dosing of broad-spectrum antibiotics (ABXs) in Rag2(R229Q) mice ameliorated intestinal and systemic autoimmunity by diminishing the frequency of mucosal and circulating gut-tropic CCR9(+) Th1 and Th17 T cells. Remarkably, serum hyper-IgE, a hallmark of the disease, was also normalized by ABX treatment. These results indicate that intestinal microbes may play a critical role in the distinctive immune dysregulation of OS.


Autoimmunity , DNA-Binding Proteins/metabolism , Gastrointestinal Microbiome , Inflammation/immunology , Inflammation/pathology , Adoptive Transfer , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Autoimmunity/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bacterial Load/drug effects , Bacterial Translocation/drug effects , Colitis/immunology , Colitis/pathology , DNA-Binding Proteins/deficiency , Gastrointestinal Microbiome/drug effects , Immune Tolerance/drug effects , Immunoglobulin E/metabolism , Immunophenotyping , Inflammation/microbiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Tropism/drug effects
10.
Immunity ; 41(5): 789-801, 2014 Nov 20.
Article En | MEDLINE | ID: mdl-25464855

Microbial colonization of the gut induces the development of gut-associated lymphoid tissue (GALT). The molecular mechanisms that regulate GALT function and result in gut-commensal homeostasis are poorly defined. T follicular helper (Tfh) cells in Peyer's patches (PPs) promote high-affinity IgA responses. Here we found that the ATP-gated ionotropic P2X7 receptor controls Tfh cell numbers in PPs. Lack of P2X7 in Tfh cells enhanced germinal center reactions and high-affinity IgA secretion and binding to commensals. The ensuing depletion of mucosal bacteria resulted in reduced systemic translocation of microbial components, lowering B1 cell stimulation and serum IgM concentrations. Mice lacking P2X7 had increased susceptibility to polymicrobial sepsis, which was rescued by Tfh cell depletion or administration of purified IgM. Thus, regulation of Tfh cells by P2X7 activity is important for mucosal colonization, which in turn results in IgM serum concentrations necessary to protect the host from bacteremia.


Intestinal Mucosa/immunology , Peyer's Patches/immunology , Receptors, Purinergic P2X7/immunology , Symbiosis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adenosine Triphosphate/metabolism , Animals , B-Lymphocytes/immunology , Bacteremia/immunology , Genetic Predisposition to Disease , Germinal Center/immunology , Humans , Immunoglobulin A/immunology , Immunoglobulin M/blood , Intestinal Mucosa/microbiology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology , Peyer's Patches/cytology , Receptors, Purinergic P2X7/genetics , Sepsis/immunology , Sepsis/microbiology
...