Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Mol Ther ; 32(6): 1658-1671, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38532630

ABSTRACT

Base editing of hematopoietic stem/progenitor cells (HSPCs) is an attractive strategy for treating immunohematologic diseases. However, the feasibility of using adenine-base-edited HSPCs for treating X-linked severe combined immunodeficiency (SCID-X1), the influence of dose-response relationships on immune cell generation, and the potential risks have not been demonstrated in vivo. Here, a humanized SCID-X1 mouse model was established, and 86.67% ± 2.52% (n = 3) of mouse hematopoietic stem cell (HSC) pathogenic mutations were corrected, with no single-guide-RNA (sgRNA)-dependent off-target effects detected. Analysis of peripheral blood over 16 weeks post-transplantation in mice with different immunodeficiency backgrounds revealed efficient immune cell generation following transplantation of different amounts of modified HSCs. Therefore, a large-scale infusion of gene-corrected HSCs within a safe range can achieve rapid, stable, and durable immune cell regeneration. Tissue-section staining further demonstrated the restoration of immune organ tissue structures, with no tumor formation in multiple organs. Collectively, these data suggest that base-edited HSCs are a potential therapeutic approach for SCID-X1 and that a threshold infusion dose of gene-corrected cells is required for immune cell regeneration. This study lays a theoretical foundation for the clinical application of base-edited HSCs in treating SCID-X1.


Subject(s)
Adenine , B-Lymphocytes , Disease Models, Animal , Gene Editing , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , T-Lymphocytes , X-Linked Combined Immunodeficiency Diseases , Animals , Mice , Hematopoietic Stem Cells/metabolism , X-Linked Combined Immunodeficiency Diseases/therapy , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Humans , Adenine/analogs & derivatives , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Mice, SCID , Genetic Therapy/methods , CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems
2.
Sci Rep ; 11(1): 18397, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526536

ABSTRACT

Cryptococcosis is an opportunistic disease caused by the fungus Cryptococcus neoformans and Cryptococcus gattii. It starts as a pulmonary infection that can spread to other organs, such as the brain, leading to the most serious occurrence of the disease, meningoencephalitis. The humoral response has already been described in limiting the progression of cryptococcosis where the B-1 cell seems to be responsible for producing natural IgM antibodies, crucial for combating fungal infections. The role of the B-1 cell in C. neoformans infection has been initially described, however the role of the humoral response of B-1 cells has not yet been evaluated during C. gattii infections. In the present study we tried to unravel this issue using XID mice, a murine model deficient in the Btk protein which compromises the development of B-1 lymphocytes. We use the XID mice compared to BALB/c mice that are sufficient for the B-1 population during C. gattii infection. Our model of chronic lung infection revealed that XID mice, unlike the sufficient group of B-1, had early mortality with significant weight loss, in addition to reduced levels of IgM and IgG specific to GXM isolated from the capsule of C. neoformans. In addition to this, we observed an increased fungal load in the blood and in the brain. We described an increase in the capsular size of C. gattii and the predominant presence of cytokines with a Th2 profile was also observed in these animals. Thus, the present study strongly points to a higher susceptibility of the XID mouse to C. gattii, which suggests that the presence of B-1 cells and anti-GXM antibodies is fundamental during the control of infection by C. gattii.


Subject(s)
Cryptococcosis/etiology , Cryptococcus gattii , Disease Susceptibility/immunology , Immunocompromised Host , X-Linked Combined Immunodeficiency Diseases/complications , Animals , Biomarkers , Colony Count, Microbial , Cryptococcosis/metabolism , Cryptococcus gattii/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Mice , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology
3.
Front Immunol ; 12: 696350, 2021.
Article in English | MEDLINE | ID: mdl-34248995

ABSTRACT

X-linked severe combined immunodeficiency (X-SCID) is caused by mutations of IL2RG, the gene encoding the interleukin common gamma chain (IL-2Rγ or γc) of cytokine receptors for interleukin (IL)-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Hypomorphic mutations of IL2RG may cause combined immunodeficiencies with atypical clinical and immunological presentations. Here, we report a clinical, immunological, and functional characterization of a missense mutation in exon 1 (c.115G>A; p. Asp39Asn) of IL2RG in a 7-year-old boy. The patient suffered from recurrent sinopulmonary infections and refractory eczema. His total lymphocyte counts have remained normal despite skewed T cell subsets, with a pronounced serum IgE elevation. Surface expression of IL-2Rγ was reduced on his lymphocytes. Signal transducer and activator of transcription (STAT) phosphorylation in response to IL-2, IL-4, and IL-7 showed a partially preserved receptor function. T-cell proliferation in response to mitogens and anti-CD3/anti-CD28 monoclonal antibodies was significantly reduced. Further analysis revealed a decreased percentage of CD4+ T cells capable of secreting IFN-γ, but not IL-4 or IL-17. Studies on the functional consequences of IL-2Rγ variants are important to get more insight into the pathogenesis of atypical phenotypes which may lay the ground for novel therapeutic strategies.


Subject(s)
Interleukin Receptor Common gamma Subunit/genetics , Job Syndrome/genetics , Mutation, Missense , X-Linked Combined Immunodeficiency Diseases/genetics , Cell Proliferation , Child , Genetic Predisposition to Disease , Humans , Interleukin Receptor Common gamma Subunit/metabolism , Job Syndrome/diagnosis , Job Syndrome/immunology , Lymphocyte Activation , Male , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/immunology
4.
Front Immunol ; 12: 644687, 2021.
Article in English | MEDLINE | ID: mdl-33959125

ABSTRACT

Mutations of the interleukin 2 receptor γ chain (IL2RG) result in the most common form of severe combined immunodeficiency (SCID), which is characterized by severe and persistent infections starting in early life with an absence of T cells and natural killer cells, normal or elevated B cell counts and hypogammaglobulinemia. SCID is commonly fatal within the first year of life, unless the immune system is reconstituted by hematopoietic stem cell transplantation (HSCT) or gene therapy. We herein describe a male infant with X-linked severe combined immunodeficiency (X-SCID) diagnosed at 5 months of age. Genetic testing revealed a novel C to G missense mutation in exon 1 resulting in a 3' splice site disruption with premature stop codon and aberrant IL2 receptor signaling. Following the diagnosis of X-SCID, the patient subsequently underwent a TCRαß/CD19-depleted haploidentical HSCT. Post transplantation the patient presented with early CD8+ T cell recovery with the majority of T cells (>99%) being non-donor T cells. Genetic analysis of CD4+ and CD8+ T cells revealed a spontaneous 14 nucleotide insertion at the mutation site resulting in a novel splice site and restoring the reading frame although defective IL2RG function was still demonstrated. In conclusion, our findings describe a spontaneous second-site mutation in IL2RG as a novel cause of somatic mosaicism and early T cell recovery following haploidentical HSCT.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation , Mutation , X-Linked Combined Immunodeficiency Diseases , Allografts , Humans , Infant , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Male , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/therapy
5.
Blood ; 138(12): 1019-1033, 2021 09 23.
Article in English | MEDLINE | ID: mdl-33876203

ABSTRACT

Sterile alpha motif (SAM) and Src homology-3 (SH3) domain-containing 3 (SASH3), also called SH3-containing lymphocyte protein (SLY1), is a putative adaptor protein that is postulated to play an important role in the organization of signaling complexes and propagation of signal transduction cascades in lymphocytes. The SASH3 gene is located on the X-chromosome. Here, we identified 3 novel SASH3 deleterious variants in 4 unrelated male patients with a history of combined immunodeficiency and immune dysregulation that manifested as recurrent sinopulmonary, cutaneous, and mucosal infections and refractory autoimmune cytopenias. Patients exhibited CD4+ T-cell lymphopenia, decreased T-cell proliferation, cell cycle progression, and increased T-cell apoptosis in response to mitogens. In vitro T-cell differentiation of CD34+ cells and molecular signatures of rearrangements at the T-cell receptor α (TRA) locus were indicative of impaired thymocyte survival. These patients also manifested neutropenia and B-cell and natural killer (NK)-cell lymphopenia. Lentivirus-mediated transfer of the SASH3 complementary DNA-corrected protein expression, in vitro proliferation, and signaling in SASH3-deficient Jurkat and patient-derived T cells. These findings define a new type of X-linked combined immunodeficiency in humans that recapitulates many of the abnormalities reported in mice with Sly1-/- and Sly1Δ/Δ mutations, highlighting an important role of SASH3 in human lymphocyte function and survival.


Subject(s)
Chromosomes, Human, X/genetics , Mutation , X-Linked Combined Immunodeficiency Diseases/genetics , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Child, Preschool , Chromosomes, Human, X/immunology , Genetic Loci , Humans , Jurkat Cells , Killer Cells, Natural/immunology , Lymphopenia/genetics , Lymphopenia/immunology , Male , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , X-Linked Combined Immunodeficiency Diseases/immunology
6.
Front Immunol ; 12: 726406, 2021.
Article in English | MEDLINE | ID: mdl-35069520

ABSTRACT

X-linked moesin associated immunodeficiency (X-MAID) is a primary immunodeficiency disease in which patients suffer from profound lymphopenia leading to recurrent infections. The disease is caused by a single point mutation leading to a R171W amino acid change in the protein moesin (moesinR171W). Moesin is a member of the ERM family of proteins, which reversibly link the cortical actin cytoskeleton to the plasma membrane. Here, we describe a novel mouse model with global expression of moesinR171W that recapitulates multiple facets of patient disease, including severe lymphopenia. Further analysis reveals that these mice have diminished numbers of thymocytes and bone marrow precursors. X-MAID mice also exhibit systemic inflammation that is ameliorated by elimination of mature lymphocytes through breeding to a Rag1-deficient background. The few T cells in the periphery of X-MAID mice are highly activated and have mostly lost moesinR171W expression. In contrast, single-positive (SP) thymocytes do not appear activated and retain high expression levels of moesinR171W. Analysis of ex vivo CD4 SP thymocytes reveals defects in chemotactic responses and reduced migration on integrin ligands. While chemokine signaling appears intact, CD4 SP thymocytes from X-MAID mice are unable to polarize and rearrange cytoskeletal elements. This mouse model will be a valuable tool for teasing apart the complexity of the immunodeficiency caused by moesinR171W, and will provide new insights into how the actin cortex regulates lymphocyte function.


Subject(s)
Cell Movement/immunology , Microfilament Proteins/deficiency , T-Lymphocytes/immunology , X-Linked Combined Immunodeficiency Diseases/immunology , Animals , Cell Movement/genetics , Disease Models, Animal , Mice , Mice, Knockout , Microfilament Proteins/immunology , X-Linked Combined Immunodeficiency Diseases/genetics
8.
Front Immunol ; 11: 608653, 2020.
Article in English | MEDLINE | ID: mdl-33329605

ABSTRACT

X-linked severe immunodeficiency disease (SCID-X1) is an inherited, rare, and life-threating disease. The genetic origin is a defect in the interleukin 2 receptor γ chain (IL2RG) gene and patients are classically characterized by absence of T and NK cells, as well as presence of partially-functional B cells. Without any treatment the disease is usually lethal during the first year of life. The treatment of choice for these patients is hematopoietic stem cell transplantation, with an excellent survival rate (>90%) if an HLA-matched sibling donor is available. However, when alternative donors are used, the success and survival rates are often lower. Gene therapy has been developed as an alternative treatment initially using γ-retroviral vectors to correct the defective γ chain in the absence of pre-conditioning treatment. The results were highly promising in SCID-X1 infants, showing long-term T-cell recovery and clinical benefit, although NK and B cell recovery was less robust. However, some infants developed T-cell acute lymphoblastic leukemia after the gene therapy, due to vector-mediated insertional mutagenesis. Consequently, considerable efforts have been made to develop safer vectors. The most recent clinical trials using lentiviral vectors together with a low-dose pre-conditioning regimen have demonstrated excellent sustained T cell recovery, but also B and NK cells, in both children and adults. This review provides an overview about the different gene therapy approaches used over the last 20 years to treat SCID-X1 patients, particularly focusing on lymphoid immune reconstitution, as well as the developments that have improved the process and outcomes.


Subject(s)
Immune Reconstitution/immunology , X-Linked Combined Immunodeficiency Diseases/immunology , Animals , B-Lymphocytes/immunology , Genetic Therapy/methods , Humans , Killer Cells, Natural/immunology , T-Lymphocytes/immunology
9.
Clin Immunol ; 219: 108573, 2020 10.
Article in English | MEDLINE | ID: mdl-32814211

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency disorder (PID) characterized by microthrombocytopenia, bloody diarrhea, eczema, recurrent infections, and a high incidence of autoimmunity and malignancy. OBJECTIVE: To investigate the mechanism of thrombocytopenia and infections in four boys of consanguineous parents from Lebanon. METHODS: Patient gDNA was studied using Next Generation Sequencing and Sanger Sequencing. Protein expression was determined by immunoblotting, and mRNA expression by semi-quantitative RT-PCR. F-actin polymerization and cellular proliferation were assayed by flow cytometry. RESULTS: We identified a threonine to a methionine change at position 45 (T45M) of the WAS protein (WASp) that abolished protein expression and disturbed F-actin polymerization and T cell proliferation, but not B cell proliferation. In addition, the levels of the WAS-interacting protein (WIP) were significantly decreased in the patients. CONCLUSION: The mutation identified severely destabilizes WASp and affects the downstream signaling events important for T cell function, but not B cell function. It was previously known that the stability of WASp depends on WIP. In this manuscript, we report that the stability of WIP also depends on WASp. Finally, it is important to suspect X-linked PIDs even in consanguineous families. CLINICAL IMPLICATIONS: The patients are above the optimal age for transplant in WAS, and it is difficult to identify one or more donors for four patients, therefore, they represent ideal candidates for gene therapy or interleukin-2 therapy.


Subject(s)
Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/genetics , X-Linked Combined Immunodeficiency Diseases/genetics , B-Lymphocytes/immunology , Child , Child, Preschool , Consanguinity , Humans , Lebanon , Male , Mutation , Siblings , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome/immunology , X-Linked Combined Immunodeficiency Diseases/immunology
10.
EBioMedicine ; 59: 102961, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32841837

ABSTRACT

BACKGOUND: The potential of a single progenitor cell to establish and maintain long-term protective T-cell immunity in humans is unknown. For genetic disorders disabling T-cell immunity, somatic reversion was shown to support limited T-cell development attenuating the clinical phenotype. However, the cases reported so far deteriorated over time leaving unanswered the important question of long-term activity of revertant precursors and the robustness of the resulting T-cell system. METHODS: We applied TCRß-CDR3 sequencing and mass cytometry on serial samples of a now 18 year-old SCIDX1 patient with somatic reversion to analyse the longitudinal diversification and stability of a T-cell system emerging from somatic gene rescue. FINDINGS: We detected close to 105 individual CDR3ß sequences in the patient. Blood samples of equal size contained about 10-fold fewer unique CDR3ß sequences compared to healthy donors, indicating a surprisingly broad repertoire. Despite dramatic expansions and contractions of individual clonotypes representing up to 30% of the repertoire, stable diversity indices revealed that these transient clonal distortions did not cause long-term repertoire imbalance. Phenotypically, the T-cell system did not show evidence for progressive exhaustion. Combined with immunoglobulin substitution, the limited T-cell system in this patient supported an unremarkable clinical course over 18 years. INTERPRETATION: Genetic correction in the appropriate cell type, in our patient most likely in a T-cell biased self-renewing hematopoietic progenitor, can yield a diverse T-cell system that provides long-term repertoire stability, does not show evidence for progressive exhaustion and is capable of providing protective and regulated T-cell immunity for at least two decades. FUNDING: DFG EH 145/9-1, DFG SCHW 432/4-1 and the German Research Foundation under Germany's Excellence Strategy-EXC-2189-Project ID: 390939984.


Subject(s)
Cell Differentiation/genetics , Lymphopoiesis/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adolescent , Biomarkers , Case-Control Studies , Child , Clonal Evolution/genetics , Female , Flow Cytometry , Genetic Testing , Humans , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , T-Lymphocyte Subsets/cytology , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology
11.
Dis Model Mech ; 13(8)2020 08 27.
Article in English | MEDLINE | ID: mdl-32651192

ABSTRACT

Model animals are indispensable for the study of human diseases, and in general, of complex biological processes. The Syrian hamster is an important model animal for infectious diseases, behavioral science and metabolic science, for which more experimental tools are becoming available. Here, we describe the generation and characterization of an interleukin-2 receptor subunit gamma (Il2rg) knockout (KO) Syrian hamster strain. In humans, mutations in IL2RG can result in a total failure of T and natural killer (NK) lymphocyte development and nonfunctional B lymphocytes (X-linked severe combined immunodeficiency; XSCID). Therefore, we sought to develop a non-murine model to study XSCID and the infectious diseases associated with IL2RG deficiency. We demonstrated that the Il2rg KO hamsters have a lymphoid compartment that is greatly reduced in size and diversity, and is impaired in function. As a result of the defective adaptive immune response, Il2rg KO hamsters developed a more severe human adenovirus infection and cleared virus less efficiently than immune competent wild-type hamsters. Because of this enhanced virus replication, Il2rg KO hamsters developed more severe adenovirus-induced liver pathology than wild-type hamsters. This novel hamster strain will provide researchers with a new tool to investigate human XSCID and its related infections.


Subject(s)
Adaptive Immunity , Adenovirus Infections, Human/virology , Adenoviruses, Human/pathogenicity , Immunocompromised Host , Interleukin Receptor Common gamma Subunit/genetics , X-Linked Combined Immunodeficiency Diseases/genetics , A549 Cells , Adenovirus Infections, Human/genetics , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/metabolism , Adenoviruses, Human/growth & development , Animals , Animals, Genetically Modified , Disease Models, Animal , Female , Gene Knockout Techniques , HEK293 Cells , Host-Pathogen Interactions , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Liver/immunology , Liver/metabolism , Liver/virology , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/virology , Male , Mesocricetus/genetics , Viral Load , Virus Replication , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/metabolism
13.
Sci Rep ; 10(1): 9957, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561775

ABSTRACT

Severe immunodeficient mice are widely used to examine human and animal cells behaviour in vivo. However, mice are short-lived and small in size; while large animals require specific large-scale equipment. Rabbits are also commonly employed as experimental models and are larger than mice or rats, easy to handle, and suitable for long-term observational and pre-clinical studies. Herein, we sought to develop and maintain stable strains of rabbits with X-linked severe combined immunodeficiency (X-SCID) via the CRISPR/Cas9 system targeting Il2rg. Consequently, X-SCID rabbits presented immunodeficient phenotypes including the loss of T and B cells and hypoplasia of the thymus. Further, these rabbits exhibited a higher success rate with engraftments upon allogeneic transplantation of skin tissue than did wild type controls. X-SCID rabbits could be stably maintained for a minimum of four generations. These results indicate that X-SCID rabbits are effective animals for use in a non-rodent model of severe immunodeficiency.


Subject(s)
CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , X-Linked Combined Immunodeficiency Diseases/genetics , Animals , B-Lymphocytes/immunology , CRISPR-Cas Systems/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , Female , Gene Knockout Techniques/methods , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Rabbits , Skin/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , X-Linked Combined Immunodeficiency Diseases/immunology
14.
Front Immunol ; 11: 415, 2020.
Article in English | MEDLINE | ID: mdl-32265911

ABSTRACT

Introduction: Severe Combined Immunodeficiency (SCID) is a life-threatening immunodeficiency caused by several pathogenic genetic variants, and it is characterized by profound defects in T-cell numbers and immune function. First performed in the late 1960's, hematopoietic stem cell transplantation remains the standard treatment for most cases of SCID. There is a growing number of post-transplant SCID patients, and it is imperative to assess the long-term outcomes of these patients. We have reported here the longest follow-up of a post-transplant SCID patient who, to our knowledge, bears the first gamma chain (γc) variant to show intact IL-21 signaling. Case Presentation: The patient presented at 5 months of age with recurrent thrush and Pneumocystis jiroveci pneumonia. In 1971, at the age of 11 months, he received an unconditioned, matched, related donor transplant comprising whole, unprocessed bone marrow. He is now 48 years old without significant illness and has never required immunoglobulin replacement. He exhibits T-dependent vaccine responses. He does suffer from chronic warts and bacterial infections that have worsened in recent years. We confirmed a known pathogenic variant in the IL2RG gene showing a hemizygous variant NM_000206.2:c.675C>A, resulting in p.Ser225Arg. His chimerism studies revealed donor T cells, host B cells, host myeloid cells, and mixed NK cells. Lymphocyte enumeration revealed normal numbers and distribution of B cells. The host B cells carry the pathogenic variant in IL2RG, but, when stimulated with IL-21, they demonstrated intact, γc-dependent signaling. Conclusions: Even with host B cells, reconstitution with donor T cells can be sufficient to allow over four decades of survival when B-cell function is intact. Our case demonstrates that satisfactory B-cell function can arise as a consequence of both intact IL-21 signaling due to a hypomorphic γc variant, and close HLA matching with the donor to allow for effective T-cell help.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Transplantation , Graft Survival/immunology , Immune Reconstitution , X-Linked Combined Immunodeficiency Diseases/surgery , Allografts , Chromosomes, Human, X/genetics , Corneal Transplantation , Female , Genes, X-Linked , Graft Rejection , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Humans , Infections/etiology , Interleukin Receptor Common gamma Subunit/genetics , Living Donors , Lymphocyte Count , Male , Middle Aged , Mutation, Missense , Point Mutation , Recurrence , Siblings , Warts/etiology , X-Linked Combined Immunodeficiency Diseases/immunology
15.
Cell Mol Gastroenterol Hepatol ; 10(1): 83-100, 2020.
Article in English | MEDLINE | ID: mdl-32017983

ABSTRACT

BACKGROUND & AIMS: Organ-level research using an animal model lacking Il2rg, the gene responsible for X-linked severe combined immunodeficiency (X-SCID), is clinically unavailable and would be a powerful tool to gain deeper insights into the symptoms of patients with X-SCID. METHODS: We used an X-SCID animal model, which was first established in our group by the deletion of Il2rg gene in pigs, to understand the clinical signs from multiple perspectives based on pathology, immunology, microbiology, and nutrition. We also treated the X-SCID pigs with bone marrow transplantation (BMT) for mimicking a current therapeutic treatment for patients with X-SCID and investigated the effect at the organ-level. Moreover, the results were confirmed using serum and fecal samples collected from patients with X-SCID. RESULTS: We demonstrated that X-SCID pigs completely lacked Peyer's patches (PPs) and IgA production in the small intestine, but possessed some dysfunctional intestinal T and B cells. Another novel discovery was that X-SCID pigs developed a heterogeneous intestinal microflora and possessed abnormal plasma metabolites, indicating that X-SCID could be an immune disorder that affects various in vivo functions. Importantly, the organogenesis of PPs in X-SCID pigs was not promoted by BMT. Although a few isolated lymphoid follicles developed in the small intestine of BMT-treated X-SCID pigs, there was no evidence that they contributed to IgA production and microflora formation. Consistently, most patients with X-SCID who received BMT possessed abnormal intestinal immune and microbial environments regardless of the presence of sufficient serum IgG. CONCLUSIONS: These results indicate that the current BMT therapies for patients with X-SCID may be insufficient to induce the organogenesis of intestinal lymphoid tissues that are associated with numerous functions in vivo.


Subject(s)
Bone Marrow Transplantation , Interleukin Receptor Common gamma Subunit/genetics , Intestinal Mucosa/growth & development , Peyer's Patches/growth & development , X-Linked Combined Immunodeficiency Diseases/therapy , Adolescent , Adult , Animals , Animals, Genetically Modified , Child , Child, Preschool , Disease Models, Animal , Female , Gastrointestinal Microbiome/immunology , Gene Knockout Techniques , Humans , Immunity, Mucosal , Immunoglobulin G/blood , Immunoglobulin G/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Organogenesis/genetics , Organogenesis/immunology , Peyer's Patches/immunology , Swine , Treatment Outcome , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/pathology
16.
J Clin Immunol ; 40(2): 299-309, 2020 02.
Article in English | MEDLINE | ID: mdl-31865525

ABSTRACT

Variants in MAGT1 have been identified as the cause of an immune deficiency termed X-linked immunodeficiency with magnesium defect, Epstein-Barr virus (EBV) infection and neoplasia (XMEN) disease. Here, we describe 2 cases of XMEN disease due to novel mutations in MAGT1, one of whom presented with classical features of XMEN disease and another who presented with a novel phenotype including probable CNS vasculitis, HHV-8 negative multicentric Castelman disease and severe molluscum contagiosum, thus highlighting the clinical diversity that may be seen in this condition. Peripheral blood immunophenotyping of these 2 patients, together with an additional 4 XMEN patients, revealed reduced NKG2D expression, impaired CD28 expression on CD8+ T cells, CD4+ T cell lymphopenia, an inverted CD4:CD8 ratio and decreased memory B cells. In addition, we showed for the first time alterations to the CD8+ T cell memory compartment, reduced CD56hi NK cells, MAIT and iNKT cells, as well as compromised differentiation of naïve CD4+ T cells into IL-21-producing Tfh-type cells in vitro. Both patients were treated with supplemental magnesium with limited benefit. However, one patient has undergone allogeneic haematopoietic stem cell transplant, with full donor chimerism and immune reconstitution. These results expand our understanding of the clinical and immunological phenotype in XMEN disease, adding to the current literature, which we further discuss here.


Subject(s)
Cation Transport Proteins/genetics , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/physiology , Leukocytes, Mononuclear/immunology , Neoplasms/genetics , X-Linked Combined Immunodeficiency Diseases/genetics , Adult , Cell Differentiation , Child , Chimerism , Epstein-Barr Virus Infections/immunology , Hematopoietic Stem Cell Transplantation , Humans , Immunologic Memory , Immunophenotyping , Lymphopenia , Magnesium/metabolism , Male , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasms/immunology , X-Linked Combined Immunodeficiency Diseases/immunology
17.
J Clin Invest ; 130(1): 507-522, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31714901

ABSTRACT

X-linked immunodeficiency with magnesium defect, EBV infection, and neoplasia (XMEN) disease are caused by deficiency of the magnesium transporter 1 (MAGT1) gene. We studied 23 patients with XMEN, 8 of whom were EBV naive. We observed lymphadenopathy (LAD), cytopenias, liver disease, cavum septum pellucidum (CSP), and increased CD4-CD8-B220-TCRαß+ T cells (αßDNTs), in addition to the previously described features of an inverted CD4/CD8 ratio, CD4+ T lymphocytopenia, increased B cells, dysgammaglobulinemia, and decreased expression of the natural killer group 2, member D (NKG2D) receptor. EBV-associated B cell malignancies occurred frequently in EBV-infected patients. We studied patients with XMEN and patients with autoimmune lymphoproliferative syndrome (ALPS) by deep immunophenotyping (32 immune markers) using time-of-flight mass cytometry (CyTOF). Our analysis revealed that the abundance of 2 populations of naive B cells (CD20+CD27-CD22+IgM+HLA-DR+CXCR5+CXCR4++CD10+CD38+ and CD20+CD27-CD22+IgM+HLA-DR+CXCR5+CXCR4+CD10-CD38-) could differentially classify XMEN, ALPS, and healthy individuals. We also performed glycoproteomics analysis on T lymphocytes and show that XMEN disease is a congenital disorder of glycosylation that affects a restricted subset of glycoproteins. Transfection of MAGT1 mRNA enabled us to rescue proteins with defective glycosylation. Together, these data provide new clinical and pathophysiological foundations with important ramifications for the diagnosis and treatment of XMEN disease.


Subject(s)
Autoimmune Lymphoproliferative Syndrome/immunology , Magnesium Deficiency/immunology , X-Linked Combined Immunodeficiency Diseases/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Autoimmune Lymphoproliferative Syndrome/genetics , Autoimmune Lymphoproliferative Syndrome/pathology , CD4-CD8 Ratio , Cation Transport Proteins/genetics , Cation Transport Proteins/immunology , Female , Glycosylation , Humans , Magnesium Deficiency/genetics , Magnesium Deficiency/pathology , Male , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/pathology
18.
Parasite Immunol ; 41(9): e12661, 2019 09.
Article in English | MEDLINE | ID: mdl-31267529

ABSTRACT

Evaluating the histopathological and morphometric changes caused by Leishmania (Leishmania) infantum chagasi infection either in the presence or absence of B-1 cells. Wild-type Balb/c and XID mice were used. Half of XID mice received B-1 cells adoptive transfer (XID + B1). Five animals from each group were infected (Balb/c I, XID I and XID + B1 I), totalizing six groups (n = 5). After 45 days of infection, the ileum was collected for histological processing and analysis. After infection, the XID animals showed an increase in the thickness of the intestinal layers, in the depth and width of the crypt and in the villi width. However, the Balb/c I group showed a reduction in almost all these parameters, whereas the villi width was increased. The villi height decreased in the infected XID animals; however, it was increased in the XID + B1 I group. Leishmania (L) infantum chagasiinfection caused a decrease in the number of Paneth cells; however, their area was increased. Finally, goblet cells and enterocytes presented different change profiles among groups. This study showed that the parasite infection causes structural and histopathological alterations in the intestine. These changes might be influenced by the absence of B-1 cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Leishmania infantum/physiology , Leishmaniasis, Visceral/pathology , Adoptive Transfer , Animals , B-Lymphocyte Subsets/pathology , Female , Immunity, Innate , Intestines/cytology , Intestines/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Mice , Mice, Inbred BALB C , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/parasitology , X-Linked Combined Immunodeficiency Diseases/pathology
20.
Immunity ; 50(4): 832-850, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995502

ABSTRACT

The common cytokine receptor γ chain, γc, is a component of the receptors for interleukin-2 (IL-2), IL-4, IL-7, IL-9, IL-15, and IL-21. Mutation of the gene encoding γc results in X-linked severe combined immunodeficiency in humans, and γc family cytokines collectively regulate development, proliferation, survival, and differentiation of immune cells. Here, we review the basic biology of these cytokines, highlighting mechanisms of signaling and gene regulation that have provided insights for immunodeficiency, autoimmunity, allergic diseases, and cancer. Moreover, we discuss how studies of this family stimulated the development of JAK3 inhibitors and present an overview of current strategies targeting these pathways in the clinic, including novel antibodies, antagonists, and partial agonists. The diverse roles of these cytokines on a range of immune cells have important therapeutic implications.


Subject(s)
Cytokines/classification , Interleukin Receptor Common gamma Subunit/genetics , Multigene Family/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Cytokines/genetics , Cytokines/immunology , Evolution, Molecular , Gene Expression Regulation , Genetic Therapy , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/therapy , Janus Kinase 3/antagonists & inhibitors , Janus Kinases/antagonists & inhibitors , Janus Kinases/physiology , Lymphocyte Subsets/immunology , Mice , Molecular Targeted Therapy , Multigene Family/genetics , Neoplasms/genetics , Neoplasms/immunology , Protein Subunits , STAT Transcription Factors/physiology , Signal Transduction , Translational Research, Biomedical , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...