Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Int J Mol Sci ; 25(7)2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38612860

ABSTRACT

Bacterial and viral respiratory tract infections are the most common infectious diseases, leading to worldwide morbidity and mortality. In the past 10 years, the importance of lung microbiota emerged in the context of pulmonary diseases, although the mechanisms by which it impacts the intestinal environment have not yet been fully identified. On the contrary, gut microbial dysbiosis is associated with disease etiology or/and development in the lung. In this review, we present an overview of the lung microbiome modifications occurring during respiratory infections, namely, reduced community diversity and increased microbial burden, and of the downstream consequences on host-pathogen interaction, inflammatory signals, and cytokines production, in turn affecting the disease progression and outcome. Particularly, we focus on the role of the gut-lung bidirectional communication in shaping inflammation and immunity in this context, resuming both animal and human studies. Moreover, we discuss the challenges and possibilities related to novel microbial-based (probiotics and dietary supplementation) and microbial-targeted therapies (antibacterial monoclonal antibodies and bacteriophages), aimed to remodel the composition of resident microbial communities and restore health. Finally, we propose an outlook of some relevant questions in the field to be answered with future research, which may have translational relevance for the prevention and control of respiratory infections.


Subject(s)
Bacteriophages , Microbiota , Respiratory Tract Infections , Animals , Humans , Antibodies, Monoclonal, Humanized , Lung
2.
EMBO Mol Med ; 15(3): e16244, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36533294

ABSTRACT

Duchenne muscular dystrophy (DMD) is a progressive severe muscle-wasting disease caused by mutations in DMD, encoding dystrophin, that leads to loss of muscle function with cardiac/respiratory failure and premature death. Since dystrophic muscles are sensed by infiltrating inflammatory cells and gut microbial communities can cause immune dysregulation and metabolic syndrome, we sought to investigate whether intestinal bacteria support the muscle immune response in mdx dystrophic murine model. We highlighted a strong correlation between DMD disease features and the relative abundance of Prevotella. Furthermore, the absence of gut microbes through the generation of mdx germ-free animal model, as well as modulation of the microbial community structure by antibiotic treatment, influenced muscle immunity and fibrosis. Intestinal colonization of mdx mice with eubiotic microbiota was sufficient to reduce inflammation and improve muscle pathology and function. This work identifies a potential role for the gut microbiota in the pathogenesis of DMD.


Subject(s)
Microbiota , Muscular Dystrophy, Duchenne , Animals , Mice , Dystrophin/genetics , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Dysbiosis , Muscular Dystrophy, Duchenne/genetics , Immune System/metabolism , Immune System/pathology , Disease Models, Animal
3.
Int J Mol Sci ; 23(23)2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36498987

ABSTRACT

Muscle wasting is a major pathological feature observed in Duchenne muscular dystrophy (DMD) and is the result of the concerted effects of inflammation, oxidative stress and cell senescence. The inducible form of proteasome, or immunoproteasome (IP), is involved in all the above mentioned processes, regulating antigen presentation, cytokine production and immune cell response. IP inhibition has been previously shown to dampen the altered molecular, histological and functional features of 3-month-old mdx mice, the animal model for DMD. In this study, we described the role of ONX-0914, a selective inhibitor of the PSMB8 subunit of immunoproteasome, in ameliorating the pathological traits that could promote muscle wasting progression in older, 9-month-old mdx mice. ONX-0914 reduces the number of macrophages and effector memory T cells in muscle and spleen, while increasing the number of regulatory T cells. It modulates inflammatory markers both in skeletal and cardiac muscle, possibly counteracting heart remodeling and hypertrophy. Moreover, it buffers oxidative stress by improving mitochondrial efficiency. These changes ultimately lead to a marked decrease of fibrosis and, potentially, to more controlled myofiber degeneration/regeneration cycles. Therefore, ONX-0914 is a promising molecule that may slow down muscle mass loss, with relatively low side effects, in dystrophic patients with moderate to advanced disease.


Subject(s)
Muscle, Skeletal , Muscular Dystrophy, Duchenne , Mice , Animals , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myocardium/metabolism , Macrophages/metabolism , Disease Models, Animal
4.
Int J Mol Sci ; 23(19)2022 Oct 06.
Article in English | MEDLINE | ID: mdl-36233146

ABSTRACT

Cellular senescence is characterized by irreversible cell cycle arrest in response to different triggers and an inflammatory secretome. Although originally described in fibroblasts and cell types of solid organs, cellular senescence affects most tissues with advancing age, including the lymphoid tissue, causing chronic inflammation and dysregulation of both innate and adaptive immune functions. Besides its normal occurrence, persistent microbial challenge or pathogenic microorganisms might also accelerate the activation of cellular aging, inducing the premature senescence of immune cells. Therapeutic strategies counteracting the detrimental effects of cellular senescence are being developed. Their application to target immune cells might have the potential to improve immune dysfunctions during aging and reduce the age-dependent susceptibility to infections. In this review, we discuss how immune senescence influences the host's ability to resolve more common infections in the elderly and detail the different markers proposed to identify such senescent cells; the mechanisms by which infectious agents increase the extent of immune senescence are also reviewed. Finally, available senescence therapeutics are discussed in the context of their effects on immunity and against infections.


Subject(s)
Aging , Cellular Senescence , Aged , Aging/pathology , Humans , Inflammation
5.
Nat Commun ; 12(1): 2099, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833239

ABSTRACT

In Duchenne muscular dystrophy (DMD), sarcolemma fragility and myofiber necrosis produce cellular debris that attract inflammatory cells. Macrophages and T-lymphocytes infiltrate muscles in response to damage-associated molecular pattern signalling and the release of TNF-α, TGF-ß and interleukins prevent skeletal muscle improvement from the inflammation. This immunological scenario was extended by the discovery of a specific response to muscle antigens and a role for regulatory T cells (Tregs) in muscle regeneration. Normally, autoimmunity is avoided by autoreactive T-lymphocyte deletion within thymus, while in the periphery Tregs monitor effector T-cells escaping from central regulatory control. Here, we report impairment of thymus architecture of mdx mice together with decreased expression of ghrelin, autophagy dysfunction and AIRE down-regulation. Transplantation of dystrophic thymus in recipient nude mice determine the up-regulation of inflammatory/fibrotic markers, marked metabolic breakdown that leads to muscle atrophy and loss of force. These results indicate that involution of dystrophic thymus exacerbates muscular dystrophy by altering central immune tolerance.


Subject(s)
Immune Tolerance/immunology , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Muscular Dystrophy, Animal/pathology , Thymus Gland/pathology , Animals , Autophagy/physiology , Ghrelin/biosynthesis , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Nude , Muscular Dystrophy, Duchenne/pathology , T-Lymphocytes/transplantation , T-Lymphocytes, Regulatory/immunology , Thymus Gland/transplantation , Transcription Factors/biosynthesis , AIRE Protein
7.
J Allergy Clin Immunol ; 146(5): 1165-1179.e11, 2020 11.
Article in English | MEDLINE | ID: mdl-32311393

ABSTRACT

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.


Subject(s)
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
9.
J Bone Miner Res ; 34(11): 2133-2148, 2019 11.
Article in English | MEDLINE | ID: mdl-31295380

ABSTRACT

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.


Subject(s)
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
10.
Stem Cells ; 37(7): 876-887, 2019 07.
Article in English | MEDLINE | ID: mdl-30895693

ABSTRACT

In spite of the progress in gene editing achieved in recent years, a subset of genetic diseases involving structural chromosome abnormalities, including aneuploidies, large deletions and complex rearrangements, cannot be treated with conventional gene therapy approaches. We have previously devised a strategy, dubbed chromosome transplantation (CT), to replace an endogenous mutated chromosome with an exogenous normal one. To establish a proof of principle for our approach, we chose as disease model the chronic granulomatous disease (CGD), an X-linked severe immunodeficiency due to abnormalities in CYBB (GP91) gene, including large genomic deletions. We corrected the gene defect by CT in induced pluripotent stem cells (iPSCs) from a CGD male mouse model. The Hprt gene of the endogenous X chromosome was inactivated by CRISPR/Cas9 technology thus allowing the exploitation of the hypoxanthine-aminopterin-thymidine selection system to introduce a normal donor X chromosome by microcell-mediated chromosome transfer. X-transplanted clones were obtained, and diploid XY clones which spontaneously lost the endogenous X chromosome were isolated. These cells were differentiated toward the myeloid lineage, and functional granulocytes producing GP91 protein were obtained. We propose the CT approach to correct iPSCs from patients affected by other X-linked diseases with large deletions, whose treatment is still unsatisfactory. Stem Cells 2019;37:876-887.


Subject(s)
Chromosomes, Mammalian , Genetic Therapy/methods , Granulocytes/metabolism , Granulomatous Disease, Chronic/therapy , Hypoxanthine Phosphoribosyltransferase/genetics , Induced Pluripotent Stem Cells/metabolism , NADPH Oxidase 2/genetics , Aminopterin/metabolism , Aminopterin/pharmacology , Animals , Base Sequence , CRISPR-Cas Systems , Cell Differentiation , Clone Cells , Culture Media/chemistry , Disease Models, Animal , Gene Editing/methods , Granulocytes/cytology , Granulocytes/drug effects , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/pathology , Humans , Hypoxanthine/metabolism , Hypoxanthine/pharmacology , Hypoxanthine Phosphoribosyltransferase/deficiency , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Male , Mice , NADPH Oxidase 2/deficiency , Proof of Concept Study , Sequence Deletion , Thioguanine/metabolism , Thioguanine/pharmacology , Thymidine/metabolism , Thymidine/pharmacology , X Chromosome/chemistry , X Chromosome/metabolism
11.
J Autoimmun ; 98: 132-148, 2019 03.
Article in English | MEDLINE | ID: mdl-30704941

ABSTRACT

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.


Subject(s)
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
12.
J Allergy Clin Immunol ; 142(3): 928-941.e8, 2018 09.
Article in English | MEDLINE | ID: mdl-29241731

ABSTRACT

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.


Subject(s)
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
13.
Hepatology ; 67(5): 1970-1985, 2018 05.
Article in English | MEDLINE | ID: mdl-29105104

ABSTRACT

Hepatocellular carcinoma (HCC) is a frequent neoplasia and a leading cause of inflammation-related cancer mortality. Despite that most HCCs arise from persistent inflammatory conditions, pathways linking chronic inflammation to cancer development are still incompletely elucidated. We dissected the role of adaptive immunity in the Mdr2 knockout (Mdr2-/- ) mouse, a model of inflammation-associated cancer, in which ablation of adaptive immunity has been induced genetically (Rag2-/- Mdr2-/- and µMt-Mdr2-/- mice) or with in vivo treatments using lymphocyte-specific depleting antibodies (anti-CD20 or anti-CD4/CD8). We found that activated B and T lymphocytes, secreting fibrogenic tumor necrosis factor alpha (TNFα) and other proinflammatory cytokines, infiltrated liver of the Mdr2-/- mice during chronic fibrosing cholangitis. Lymphocyte ablation, in the Rag2-/- Mdr2-/- and µMt-Mdr2-/- mice, strongly suppressed hepatic stellate cell (HSC) activation and extracellular matrix deposition, enhancing HSC transition to cellular senescence. Moreover, lack of lymphocytes changed the intrahepatic metabolic/oxidative state, resulting in skewed macrophage polarization toward an anti-inflammatory M2 phenotype. Remarkably, hepatocarcinogenesis was significantly suppressed in the Rag2-/- Mdr2-/- mice, correlating with reduced TNFα/NF-κB (nuclear factor kappa B) pathway activation. Ablation of CD20+ B cells, but not of CD4+ /CD8+ T cells, in Mdr2-/- mice, promoted senescence-mediated fibrosis resolution and inhibited the protumorigenic TNFα/NF-κB pathway. Interestingly, presence of infiltrating B cells correlated with increased tumor aggressiveness and reduced disease-free survival in human HCC. CONCLUSION: Adaptive immunity sustains liver fibrosis (LF) and favors HCC growth in chronic injury, by modulating innate components of inflammation and limiting the extent of HSC senescence. Therapies designed for B-cell targeting may be an effective strategy in LF. (Hepatology 2018;67:1970-1985).


Subject(s)
B-Lymphocytes/immunology , Carcinogenesis/immunology , Carcinoma, Hepatocellular/immunology , Liver Cirrhosis/immunology , Liver Neoplasms/immunology , ATP Binding Cassette Transporter, Subfamily B/genetics , Adaptive Immunity/immunology , Animals , Carcinogenesis/pathology , Cell Culture Techniques , Cellular Senescence/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Immunohistochemistry , Liver/immunology , Liver/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , ATP-Binding Cassette Sub-Family B Member 4
14.
Gut Microbes ; 7(6): 503-511, 2016 11.
Article in English | MEDLINE | ID: mdl-27575988

ABSTRACT

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.


Subject(s)
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
15.
Mol Ther ; 24(11): 1898-1912, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27506451

ABSTRACT

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.


Subject(s)
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
16.
J Exp Med ; 213(3): 355-75, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26926994

ABSTRACT

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.


Subject(s)
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
17.
J Clin Invest ; 125(10): 3941-51, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26368308

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is an X-linked immunodeficiency characterized by microthrombocytopenia, eczema, and high susceptibility to developing tumors and autoimmunity. Recent evidence suggests that B cells may be key players in the pathogenesis of autoimmunity in WAS. Here, we assessed whether WAS protein deficiency (WASp deficiency) affects the establishment of B cell tolerance by testing the reactivity of recombinant antibodies isolated from single B cells from 4 WAS patients before and after gene therapy (GT). We found that pre-GT WASp-deficient B cells were hyperreactive to B cell receptor stimulation (BCR stimulation). This hyperreactivity correlated with decreased frequency of autoreactive new emigrant/transitional B cells exiting the BM, indicating that the BCR signaling threshold plays a major role in the regulation of central B cell tolerance. In contrast, mature naive B cells from WAS patients were enriched in self-reactive clones, revealing that peripheral B cell tolerance checkpoint dysfunction is associated with impaired suppressive function of WAS regulatory T cells. The introduction of functional WASp by GT corrected the alterations of both central and peripheral B cell tolerance checkpoints. We conclude that WASp plays an important role in the establishment and maintenance of B cell tolerance in humans and that restoration of WASp by GT is able to restore B cell tolerance in WAS patients.


Subject(s)
B-Lymphocytes/immunology , Genetic Therapy , Genetic Vectors/therapeutic use , Immune Tolerance , Wiskott-Aldrich Syndrome Protein/therapeutic use , Wiskott-Aldrich Syndrome/therapy , Adult , Amino Acid Sequence , Bone Marrow/pathology , Child , Child, Preschool , Clonal Deletion , Clone Cells/immunology , Humans , Lentivirus/genetics , Male , Molecular Sequence Data , Receptors, Antigen, B-Cell/immunology , Recombinant Fusion Proteins , T-Lymphocytes, Regulatory/immunology , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/genetics
18.
J Immunol ; 194(9): 4144-53, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25825446

ABSTRACT

The immune and the skeletal system are tightly interconnected, and B lymphocytes are uniquely endowed with osteo-interactive properties. In this context, receptor activator of NF-κB (RANK) ligand (RANKL) plays a pivotal role in lymphoid tissue formation and bone homeostasis. Although murine models lacking RANK or RANKL show defects in B cell number, the role of the RANKL-RANK axis on B physiology is still a matter of debate. In this study, we have characterized in detail B cell compartment in Rankl(-/-) mice, finding a relative expansion of marginal zone B cells, B1 cells, and plasma cells associated with increased Ig serum levels, spontaneous germinal center formation, and hyperresponse to CD40 triggering. Such abnormalities were associated with an increased frequency of regulatory B cells and augmented B cell-derived IL-10 production. Remarkably, in vivo IL-10-R blockade reduced T cell-triggered plasma cell differentiation and restrained the expansion of regulatory B cells. These data point to a novel role of the RANKL-RANK axis in the regulation of B cell homeostasis and highlight an unexpected link between IL-10 CD40 signaling and the RANKL pathway.


Subject(s)
B-Lymphocytes/immunology , Interleukin-10/immunology , RANK Ligand/deficiency , RANK Ligand/immunology , Animals , Mice , Mice, Knockout
19.
J Int AIDS Soc ; 17(4 Suppl 3): 19717, 2014.
Article in English | MEDLINE | ID: mdl-25397463

ABSTRACT

INTRODUCTION: We aimed to assess any factors associated with dysplasia regression and with HPV clearance in a cohort of HIV+ patients, with particular focus on cART and gender. METHODS: Asymptomatic HIV+ patients of the San Paolo Infectious Disease (SPID) cohort who underwent anoscopy/gynaecological evaluation were enrolled. Anal/cervical brushing were analyzed for: HPV-PCR detection/genotyping (HR-HPV), cytologic abnormalities (Bethesda System 2001: LSIL-HSIL). Demographics and HIV-related parameters were evaluated at baseline. Activated CD8+/CD38+ lymphocytes were measured (flow citometry). Patients were examined at baseline (T0) and at 12-18 months visit (T1). HPV clearance was defined as negativisation of HPV at T1; SIL regression (SIL-R) and progression (SIL-P) were defined as change from HSIL/LSIL to a lower-grade/absence of dysplasia and as change from absence of HSIL/LSIL to a higher-grade dysplasia at T1, respectively. Mann Whitney test, Chi-square test and multivariate logistic regression were used. RESULTS: A total of 189 patients were examined, 60 (32%) were women. One hundred fifty patients (79%) were HPV+, 113 (75%) harboured HR-HPV; 103 (68%) showed LSIL/HSIL at T0 (32% of women and 65% of men) (all were HPV-positive). No differences in demographics and HIV-related markers were found between patients with SIL-P (33, 41%) and patients with SIL-R (47, 59%). HPV+ patients who cleared HPV (28, 18%) were found to be more frequently female, heterosexual infected, more frequently on cART and with lower Log10 HIV-RNA and lower levels of CD8+/CD38+ % compared with HPV persistence group (Table 1). CONCLUSIONS: Close follow-up of HPV and SIL should be promoted particularly in men and in untreated individuals. We cannot exclude behavioural variables linked to risky sex and reinfection.

SELECTION OF CITATIONS
SEARCH DETAIL
...