Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Science ; 376(6599): eabm6380, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35587511

ABSTRACT

The molecular basis of interindividual clinical variability upon infection with Staphylococcus aureus is unclear. We describe patients with haploinsufficiency for the linear deubiquitinase OTULIN, encoded by a gene on chromosome 5p. Patients suffer from episodes of life-threatening necrosis, typically triggered by S. aureus infection. The disorder is phenocopied in patients with the 5p- (Cri-du-Chat) chromosomal deletion syndrome. OTULIN haploinsufficiency causes an accumulation of linear ubiquitin in dermal fibroblasts, but tumor necrosis factor receptor-mediated nuclear factor κB signaling remains intact. Blood leukocyte subsets are unaffected. The OTULIN-dependent accumulation of caveolin-1 in dermal fibroblasts, but not leukocytes, facilitates the cytotoxic damage inflicted by the staphylococcal virulence factor α-toxin. Naturally elicited antibodies against α-toxin contribute to incomplete clinical penetrance. Human OTULIN haploinsufficiency underlies life-threatening staphylococcal disease by disrupting cell-intrinsic immunity to α-toxin in nonleukocytic cells.


Subject(s)
Bacterial Toxins , Cri-du-Chat Syndrome , Endopeptidases , Haploinsufficiency , Hemolysin Proteins , Staphylococcal Infections , Staphylococcus aureus , Bacterial Toxins/immunology , Cri-du-Chat Syndrome/genetics , Cri-du-Chat Syndrome/immunology , Endopeptidases/genetics , Haploinsufficiency/genetics , Haploinsufficiency/immunology , Hemolysin Proteins/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Cellular/genetics , Necrosis , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/pathology
2.
Nat Commun ; 13(1): 931, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177641

ABSTRACT

Koolen-de Vries syndrome (KdVS) is a rare disorder caused by haploinsufficiency of KAT8 regulatory NSL complex subunit 1 (KANSL1), which is characterized by intellectual disability, heart failure, hypotonia, and congenital malformations. To date, no effective treatment has been found for KdVS, largely due to its unknown pathogenesis. Using siRNA screening, we identified KANSL1 as an essential gene for autophagy. Mechanistic study shows that KANSL1 modulates autophagosome-lysosome fusion for cargo degradation via transcriptional regulation of autophagosomal gene, STX17. Kansl1+/- mice exhibit impairment in the autophagic clearance of damaged mitochondria and accumulation of reactive oxygen species, thereby resulting in defective neuronal and cardiac functions. Moreover, we discovered that the FDA-approved drug 13-cis retinoic acid can reverse these mitophagic defects and neurobehavioral abnormalities in Kansl1+/- mice by promoting autophagosome-lysosome fusion. Hence, these findings demonstrate a critical role for KANSL1 in autophagy and indicate a potentially viable therapeutic strategy for KdVS.


Subject(s)
Abnormalities, Multiple/genetics , Intellectual Disability/genetics , Mitophagy/genetics , Nuclear Proteins/genetics , Abnormalities, Multiple/drug therapy , Abnormalities, Multiple/immunology , Abnormalities, Multiple/pathology , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/pathology , Cerebral Cortex/cytology , Cerebral Cortex/pathology , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 17/immunology , Disease Models, Animal , Female , Haploinsufficiency/immunology , HeLa Cells , Humans , Intellectual Disability/drug therapy , Intellectual Disability/immunology , Intellectual Disability/pathology , Isotretinoin/pharmacology , Isotretinoin/therapeutic use , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Mice , Mice, Transgenic , Mitophagy/drug effects , Mitophagy/immunology , Neurons , Nuclear Proteins/metabolism , Primary Cell Culture
3.
Cancer Treat Res Commun ; 28: 100437, 2021.
Article in English | MEDLINE | ID: mdl-34425470

ABSTRACT

PURPOSE: PAX5 haploinsufficiency promoting tumorigenesis is related to immune escape. But the mechanisms of PAX5 mutations inducing tumor immune escape have not been clarified. Our aim was to study how PAX5 haploinsufficiency influences effector CD8 + T cells in tumor microenvironment. METHODS: We estimated the proportions of 22 immune cell types and the expressions of immune inhibitory-related molecules based on gene expression profiles (GEPs) from children's B- acute lymphoblastic leukemia(B-ALL) with PAX5 mutations by CIBERSORT, an established algorithm. We constructed the PAX5 haplodeletion A20 cell lines, built allografted A20 tumor models and evaluated the effect of PAX5 haplodeletion on immune inhibitory-related molecules in the tumor microenvironment (TME). RESULTS: Our results indicated the percentages of T cells in bone marrow of children's B-ALL with PAX5 mutations were not statistically different from that in bone marrow of B-ALL without PAX5 mutations, except for T follicular helper (Tfh) cells. But a variety of up-regulated immune inhibitory-related molecules in bone marrow of children's B- ALL with PAX5 mutations were identified. By different approaches, we found that several immune inhibitory-related molecules of CD8+ T cells in TME of PAX5 haplodeletion clones such as TIM3, NR4A1 and BATF, were increased significantly compared with that of PAX5 wild type control. The IFN-ɤ of CD8+ T cells in TME of PAX5 haplodeletion tumors was decreased significantly compared with that of PAX5 wild type control. CONCLUSION: Our study showed that PAX5 haploinsufficiency induced CD8+ T cells dysfunction or exhaustion by high expression of TIM3, NR4A1 and BATF in the CD8+ T cells of TME.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Haploinsufficiency/immunology , PAX5 Transcription Factor/immunology , Tumor Escape/immunology , Humans
4.
Clin Immunol ; 229: 108779, 2021 08.
Article in English | MEDLINE | ID: mdl-34116213

ABSTRACT

CTLA4-haploinsufficiency is a complex disease of immune dysregulation presenting with a broad spectrum of clinical manifestations. CTLA4-Fc fusion proteins such as abatacept have been described to alleviate immune dysregulation in several adult cases of CTLA4-haploinsufficiency. However, until now only few cases of pediatric CTLA4-haploinsufficiency treated with abatacept have been described. Here we present two pediatric cases of severe CTLA4-haploinsufficiency refractory to conventional immunosuppressive therapies that responded rapidly to treatment with abatacept. No side effects were observed during a follow-up period of 7-15 months. While one patient has successfully undergone HSCT the second patient continues to receive abatacept. Our cases demonstrate safe medium-term use of abatacept in the pediatric population.


Subject(s)
Abatacept/therapeutic use , CTLA-4 Antigen/deficiency , Immunosuppressive Agents/therapeutic use , Adolescent , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Female , Haploinsufficiency/genetics , Haploinsufficiency/immunology , Hematopoietic Stem Cell Transplantation , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/therapy , Male , Mutation, Missense , T-Lymphocytes, Regulatory/immunology
5.
J Exp Med ; 218(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-33970190

ABSTRACT

DNMT3A encodes an enzyme that carries out de novo DNA methylation, which is essential for the acquisition of cellular identity and specialized functions during cellular differentiation. DNMT3A is the most frequently mutated gene in age-related clonal hematopoiesis. As such, mature immune cells harboring DNMT3A mutations can be readily detected in elderly persons. Most DNMT3A mutations associated with clonal hematopoiesis are heterozygous and predicted to cause loss of function, indicating that haploinsufficiency is the predominant pathogenic mechanism. Yet, the impact of DNMT3A haploinsufficiency on the function of mature immune cells is poorly understood. Here, we demonstrate that DNMT3A haploinsufficiency impairs the gain of DNA methylation at decommissioned enhancers, while simultaneously and unexpectedly impairing DNA demethylation of newly activated enhancers in mature human myeloid cells. The DNA methylation defects alter the activity of affected enhancers, leading to abnormal gene expression and impaired immune response. These findings provide insights into the mechanism of immune dysfunction associated with clonal hematopoiesis and acquired DNMT3A mutations.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Haploinsufficiency/genetics , Immune System/immunology , Regulatory Sequences, Nucleic Acid/genetics , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/immunology , DNA Methylation/immunology , DNA Methyltransferase 3A , Gene Expression/genetics , Gene Expression/immunology , Haploinsufficiency/immunology , Humans , Mutation/genetics , Mutation/immunology , Regulatory Sequences, Nucleic Acid/immunology
6.
J Clin Immunol ; 41(6): 1241-1249, 2021 08.
Article in English | MEDLINE | ID: mdl-33855675

ABSTRACT

PURPOSE: IKAROS, encoded by IKZF1, is a member of the IKAROS family of zinc-finger transcription factors playing critical roles in lymphocyte development, differentiation, and tumor suppression. Several studies demonstrated that IKZF1 mutations affecting DNA binding or homo-/hetero-dimerization are mostly associated with common variable immunodeficiency, combined immunodeficiency, or hematologic manifestations. Herein we report a likely de novo, nonsense IKZF1 mutation (p.C182*) in a baby with low T cell receptor excision circles (TREC) identified by newborn screening testing for severe combined immunodeficiency. The patient also presented a profound B cell deficiency at birth. METHODS: Genetic, functional, immunologic, and clinical outcome data associated with this patient and her mutation were evaluated. RESULTS: Mutant p.C182* was detected in the cytoplasm of the patient's primary cells, in contrast to wild type (WT) IKAROS protein, only detected in the nucleus. Functional in vitro assessments revealed that p.C182* was less stable than WT IKAROS protein and failed to bind to its target DNA binding sequence and dimerize with WT IKAROS protein, resulting in impaired pericentromeric targeting and transcriptional repression by means of haploinsufficiency. During follow-up, while a spontaneous recovery of TREC and T cells was observed, B cells improved but not to sustained normal ranges. CONCLUSIONS: Patients with IKAROS-associated diseases can present with SCID-like TREC values through newborn screening testing. IKZF1 mutations should be added to the low TREC differential, although spontaneous recovery has to be considered.


Subject(s)
Haploinsufficiency/genetics , Ikaros Transcription Factor/genetics , Mutation/genetics , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/genetics , B-Lymphocytes/immunology , Common Variable Immunodeficiency/genetics , Common Variable Immunodeficiency/immunology , DNA/genetics , HEK293 Cells , Haploinsufficiency/immunology , Humans , Ikaros Transcription Factor/immunology , Infant, Newborn , Neonatal Screening/methods , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology
7.
Clin Exp Immunol ; 205(1): 28-43, 2021 07.
Article in English | MEDLINE | ID: mdl-33788257

ABSTRACT

Cytotoxic T lymphocyte antigen 4 (CTLA-4) haploinsufficiency (CHAI) and lipopolysaccharide-responsive beige-like anchor (LRBA) deficiency (LATAIE) are newly identified inborn errors of immunity with shared molecular pathomechanisms and clinical manifestations. In this review, we aimed to provide differential comparisons regarding demographic, clinical, immunological and molecular characteristics between these two similar conditions. A literature search was conducted in PubMed, Web of Science and Scopus databases and included studies were systematically evaluated. Overall, 434 (222 CHAI and 212 LATAIE) patients were found in 101 eligible studies. The CHAI patients were mainly reported from North America and western Europe, while LATAIE patients were predominantly from Asian countries. In CHAI, positive familial history (P < 0·001) and in LATAIE, consanguineous parents (P < 0·001) were more common. In CHAI patients the rates of granulomas (P < 0·001), malignancies (P = 0·001), atopy (P = 0·001), cutaneous disorders (P < 0·001) and neurological (P = 0·002) disorders were higher, while LATAIE patients were more commonly complicated with life-threatening infections (P = 0·002), pneumonia (P = 0·006), ear, nose and throat disorders (P < 0·001), organomegaly (P = 0·023), autoimmune enteropathy (P = 0·038) and growth failure (P < 0·001). Normal lymphocyte subsets and immunoglobulins except low serum levels of CD9+ B cells (14·0 versus 38·4%, P < 0·001), natural killer (NK) cells (21 versus 41·1%, P < 0·001), immunoglobulin (Ig)G (46·9 versus 41·1%, P = 0·291) and IgA (54·5 versus 44·7%, P = 0·076) were found in the majority of CHAI and LATAIE patients, respectively. The most frequent biological immunosuppressive agents prescribed for CHAI and LATAIE patients were rituximab and abatacept, respectively. Further investigations into the best conditioning and treatment regimens pre- and post-transplantation are required to improve the survival rate of transplanted CHAI and LATAIE patients.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Haploinsufficiency/genetics , Haploinsufficiency/immunology , Humans , Immunoglobulins/immunology , Immunosuppressive Agents/immunology , Lymphocytes/immunology
8.
FEBS J ; 288(2): 640-662, 2021 01.
Article in English | MEDLINE | ID: mdl-32386462

ABSTRACT

Nuclear factor 'κ-light-chain-enhancer' of activated B cells (NF-κB) signaling is a signaling pathway used by most immune cells to promote immunostimulatory functions. Recent studies have indicated that regulatory T cells (Treg) differentially integrate TCR-derived signals, thereby maintaining their suppressive features. However, the role of NF-κB signaling in the activation of human peripheral blood (PB) Treg has not been fully elucidated so far. We show that the activity of the master transcription factor forkhead box protein 3 (FOXP3) attenuates p65 phosphorylation and nuclear translocation of the NF-κB proteins p50, p65, and c-Rel following activation in human Treg. Using pharmacological and genetic inhibition of canonical NF-κB signaling in FOXP3-transgenic T cells and PB Treg from healthy donors as well as Treg from a patient with a primary NFKB1 haploinsufficiency, we validate that Treg activation and suppressive capacity is independent of NF-κB signaling. Additionally, repression of residual NF-κB signaling in Treg further enhances interleukin-10 (IL-10) production. Blockade of NF-κB signaling can be exploited for the generation of in vitro induced Treg (iTreg) with enhanced suppressive capacity and functional stability. In this respect, dual blockade of mammalian target of rapamycin (mTOR) and NF-κB signaling was accompanied by enhanced expression of the transcription factors FOXP1 and FOXP3 and demethylation of the Treg-specific demethylated region compared to iTreg generated under mTOR blockade alone. Thus, we provide first insights into the role of NF-κB signaling in human Treg. These findings could lead to strategies for the selective manipulation of Treg and the generation of improved iTreg for cellular therapy.


Subject(s)
Forkhead Transcription Factors/immunology , Haploinsufficiency/immunology , NF-kappa B p50 Subunit/immunology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/immunology , Transcription Factor RelA/immunology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/immunology , Cell Nucleus/drug effects , Cell Nucleus/immunology , Cell Nucleus/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Lymphocyte Activation , NF-kappa B p50 Subunit/deficiency , NF-kappa B p50 Subunit/genetics , Phosphorylation/drug effects , Primary Cell Culture , Repressor Proteins/genetics , Repressor Proteins/immunology , Signal Transduction , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Thiazoles/pharmacology , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics
9.
Clin Immunol ; 216: 108441, 2020 07.
Article in English | MEDLINE | ID: mdl-32335289

ABSTRACT

Haploinsufficiency of A20 (HA20) causes inflammatory disease resembling Behçet's disease; many cases have been reported, including some that are complicated with autoimmune diseases. This study aims to clarify the immunophenotype of patients with HA20 by analyzing lymphocyte subsets using multicolor flow cytometry. The patients with HA20 previously diagnosed in a nationwide survey were compared by their cell subpopulations. In total, 27 parameters including regulatory T cells (Tregs), double-negative T cells (DNTs), and follicular helper T cells (TFHs) were analyzed and compared with the reference values in four age groups: 0-1, 2-6, 7-19, and ≥20 years. The Tregs of patients with HA20 tended to increase in tandem with age-matched controls at all ages. In addition, patients ≥20 years had increased DNTs compared with controls, whereas TFHs significantly increased in younger patients. In HA20 patients, the increase in DNTs and TFHs may contribute to the development of autoimmune diseases.


Subject(s)
Haploinsufficiency/immunology , Adolescent , Adult , Autoimmune Diseases/immunology , Behcet Syndrome/immunology , Child , Child, Preschool , Female , Flow Cytometry/methods , Humans , Immunophenotyping/methods , Infant , Male , Phenotype , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Young Adult
11.
Front Immunol ; 10: 998, 2019.
Article in English | MEDLINE | ID: mdl-31156616

ABSTRACT

CTLA-4 is essential for immune tolerance. Heterozygous CTLA4 mutations cause immune dysregulation evident in defective regulatory T cells with low levels of CTLA-4 expression. Biallelic mutations in LRBA also result in immune dysregulation with low levels of CTLA-4 and clinical presentation indistinguishable from CTLA-4 haploinsufficiency. CTLA-4 has become an immunotherapy target whereby its blockade with a monoclonal antibody has resulted in improved survival in advanced melanoma patients, amongst other malignancies. However, this therapeutic manipulation can result in autoimmune/inflammatory complications reminiscent of those seen in genetic defects affecting the CTLA-4 pathway. Despite efforts made to understand and establish disease genotype/phenotype correlations in CTLA-4-haploinsufficiency and LRBA-deficiency, such relationships remain elusive. There is currently no specific immunological marker to assess the degree of CTLA-4 pathway disruption or its relationship with clinical manifestations. Here we compare three different patient groups with disturbances in the CTLA-4 pathway-CTLA-4-haploinsufficiency, LRBA-deficiency, and ipilimumab-treated melanoma patients. Assessment of CTLA4 mRNA expression in these patient groups demonstrated an inverse correlation between the CTLA4 message and degree of CTLA-4 pathway disruption. CTLA4 mRNA levels from melanoma patients under therapeutic CTLA-4 blockade (ipilimumab) were increased compared to patients with either CTLA4 or LRBA mutations that were clinically stable with abatacept treatment. In summary, we show that increased CTLA4 mRNA levels correlate with the degree of CTLA-4 pathway disruption, suggesting that CTLA4 mRNA levels may be a quantifiable surrogate for altered CTLA-4 expression.


Subject(s)
CTLA-4 Antigen/physiology , Haploinsufficiency/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Autoimmune Diseases/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/genetics , Humans , Ipilimumab/therapeutic use , Melanoma/drug therapy , Melanoma/immunology , Mutation , Signal Transduction/physiology , T-Lymphocytes, Regulatory/immunology
14.
Am J Med Genet A ; 173(6): 1514-1520, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28432740

ABSTRACT

3q27.2-qter deletion syndromes feature an overlapping set of terminal and interstitial deletions with variable congenital malformations. Diamond-Blackfan anemia (DBA) is etiologically heterogeneous disorder in which one cause is dominant mutations of the RPL35A gene on 3q29. We report a child with a 3q27.2-qter deletion that contains the RPL35A gene. She had clinical and laboratory features consistent with DBA and as well, an unexplained immunodeficiency disorder. Given these unusual findings, we reviewed other patients in the literature with overlapping genomic deletions. In addition, we evaluated our patient for the immunodeficiency disorder, RIDDLE syndrome, due to recessive mutations in the RNF168 gene on 3q29. A PubMed search for case reports of 3q27.2-qter overlapping deletions was performed. To determine if RPL35A was in the deletion region, the chromosomal regions reported were mapped to genomic regions using the UCSC Genome Browser. We identified 85 overlapping deletions, of which six included the RPL35A gene and all should be had DBA. Interestingly, none of the reported cases had immunodeficiency. To evaluate RIDDLE syndrome (radiosensitivity, immunodeficiency, dysmorphic features, and learning difficulties), we sequenced the remaining RNF168 gene and examined her fibroblast culture for a DNA double strand break repair deficiency. These results were normal, indicating that the immunodeficiency is unlikely to result from a RNF168 deficiency. We show that RPL35A haploinsufficiency is a cause of DBA and we report a novel case with 3q27.2-qter deletion and immunodeficiency. The etiology for the immunodeficiency remains unsolved and could be caused by an unknown gene effect or consequent to the DBA phenotype.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Craniofacial Abnormalities/genetics , Haploinsufficiency/genetics , Immunologic Deficiency Syndromes/genetics , Learning Disabilities/genetics , Ribosomal Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Anemia, Diamond-Blackfan/immunology , Anemia, Diamond-Blackfan/pathology , Child , Chromosomes, Human, Pair 3/genetics , Craniofacial Abnormalities/immunology , Craniofacial Abnormalities/physiopathology , DNA Breaks, Double-Stranded , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Deletion , Haploinsufficiency/immunology , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/physiopathology , Learning Disabilities/immunology , Learning Disabilities/physiopathology , Phenotype , Primary Cell Culture , Primary Immunodeficiency Diseases
15.
Annu Rev Genomics Hum Genet ; 17: 245-72, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27362340

ABSTRACT

Autoinflammatory diseases are inborn disorders of the innate immune system characterized by episodes of systemic inflammation that are mediated largely by myeloid cells. The field of autoinflammatory diseases has been established since 1999, following the identification of the first genes underlying periodic fever syndromes. This review focuses on developments that have transformed the field in the last two years. We discuss three newly described monogenic autoinflammatory diseases [deficiency of adenosine deaminase 2 (DADA2), a subtype of macrophage activation syndrome (MAS), and stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy (SAVI)], discuss the possibilities of somatic mosaicism and digenic inheritance, and give an update on new concepts in pathways involved in familial Mediterranean fever (FMF). Finally, the new monogenic autoinflammatory disease haploinsufficiency of A20 (HA20) underscores the placement of monogenic diseases in the firmament of common autoinflammatory phenotypes. The advances in the last two years have shed light on the pathophysiology of several autoinflammatory diseases and have elucidated new pathways that play a role in innate immunity.


Subject(s)
Adenosine Deaminase/genetics , Familial Mediterranean Fever/genetics , Inflammation/genetics , Intercellular Signaling Peptides and Proteins/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Adenosine Deaminase/immunology , Familial Mediterranean Fever/immunology , Familial Mediterranean Fever/pathology , Haploinsufficiency/genetics , Haploinsufficiency/immunology , Humans , Immunity, Innate/genetics , Inflammation/immunology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology
17.
J Hematol Oncol ; 8: 94, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26219558

ABSTRACT

Bcl11b is a transcription factor important for T cell development and also a tumor-suppressor gene that is hemizygously inactivated in ~10% human T cell acute lymphoblastic leukemia (T-ALL) and several murine T-ALL models, including ATM(-/-) thymic lymphomas. Here we report that heterozygous loss of Bcl11b (Bcl11b(+/-)) unexpectedly reduced lethal thymic lymphoma in ATM(-/-) mice by suppressing lymphoma progression, but not initiation. The suppression was associated with a T cell-mediated immune response in ATM(-/-)Bcl11b(+/-) mice, revealing a haploid insufficient function of Bcl11b in immune modulation against lymphoma and offering an explanation for the complex relationship between Bcl11b status with T-ALL prognosis.


Subject(s)
Haploinsufficiency/immunology , Lymphoma, T-Cell/immunology , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Disease Progression , Gene Expression Regulation , Humans , Lymphoma, T-Cell/pathology , Mice
18.
Blood Cells Mol Dis ; 55(1): 3-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25976459

ABSTRACT

Heparan sulfate (HS) proteoglycans on stromal and hematopoietic stem/progenitor cells (HSPC) help form the stem cell niche, co-localize molecules that direct stem cell fate, and modulate HSPC homing and retention. Inhibition of HS function mobilizes marrow HSPC. In vitro, HSPC maintenance is influenced by stromal HS structure and concentration. Because inhibition of HS activity or synthesis may be developed for HSPC transplantation, it is important to examine if systemic HS deficiency influences hematopoiesis in vivo. In a transgenic mouse model of HS haploinsufficiency, we examined endogenous hematopoiesis and engraftment of allogeneic bone marrow. Endogenous hematopoiesis was normal except gender-specific alterations in peripheral blood monocyte and platelet counts. Donor engraftment was achieved in all mice following myeloablative irradiation, but HS deficiency in the stromal microenvironment, on HSPC, or both (the 3 test conditions), was associated with a trend towards lower donor engraftment percentage in the bone marrow. Following non-myeloablative irradiation, competitive engraftment was achieved in 22% of mice in the test conditions, vs 50% of control animals (P = 0.03). HS deficiency did not re-direct donor engraftment from bone marrow to spleen or liver. Normal HS levels in the stromal microenvironment and HSPC are required for HSPC engraftment following non-myeloablative conditioning.


Subject(s)
Bone Marrow Transplantation , Graft Survival/genetics , Haploinsufficiency/immunology , Hematopoiesis/genetics , Heparitin Sulfate/genetics , N-Acetylglucosaminyltransferases/genetics , Animals , Blood Platelets/immunology , Blood Platelets/pathology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Count , Female , Gene Expression , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Heparitin Sulfate/deficiency , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/pathology , N-Acetylglucosaminyltransferases/deficiency , Sex Factors , Stem Cell Niche , Transplantation Chimera , Transplantation Conditioning , Transplantation, Homologous , Whole-Body Irradiation
19.
Immunobiology ; 220(5): 624-5, 2015 May.
Article in English | MEDLINE | ID: mdl-25638260

ABSTRACT

The inhibitory anti-CTLA-4 antibody, ipilimumab, dramatically improved survival in a subgroup of metastatic melanoma patients. The majority, however, suffered autoimmune-related adverse events (irAEs), sometimes pathognomonic of acute graft-versus-host-disease (GVHD). This implies that the CTLA-4 blockade is not tumor specific. We make a risky but testable prediction: anti-CTLA-4 therapy may have mechanism similar to that occurring in inherited human CTLA-4 haploinsufficiency. If so, a therapeutic paradigm shift is required. The task is not desperately trying to put the genie back in the bottle by immune-suppressive treatments, but harnessing the immense forces liberated by the anti-CTLA-4 antibody blockade by pretargeting or dose adjustment.


Subject(s)
Antibodies, Blocking/therapeutic use , Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/prevention & control , CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy/methods , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Animals , Antibodies, Blocking/adverse effects , Antibodies, Monoclonal/adverse effects , Autoimmune Diseases/etiology , CTLA-4 Antigen/genetics , Clinical Protocols , Haploinsufficiency/immunology , Homeostasis , Humans , Immunotherapy/adverse effects , Ipilimumab , Melanoma/complications , Melanoma/immunology , Mice , Mice, Knockout , Neoplasm Metastasis , Skin Neoplasms/complications , Skin Neoplasms/immunology
20.
J Immunol ; 194(6): 2551-60, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25667416

ABSTRACT

The link between autoimmune diseases and primary immunodeficiency syndromes has been increasingly appreciated. Immunologic evaluation of a young man with autoimmune enterocolopathy and unexplained infections revealed evidence of immunodeficiency, including IgG subclass deficiency, impaired Ag-induced lymphocyte proliferation, reduced cytokine production by CD8(+) T lymphocytes, and decreased numbers of NK cells. Genetic evaluation identified haploinsufficiency of NFAT5, a transcription factor regulating immune cell function and cellular adaptation to hyperosmotic stress, as a possible cause of this syndrome. Inhibition or deletion of NFAT5 in normal human and murine cells recapitulated several of the immune deficits identified in the patient. These results provide evidence of a primary immunodeficiency disorder associated with organ-specific autoimmunity linked to NFAT5 deficiency.


Subject(s)
Autoimmune Diseases/immunology , Gastrointestinal Diseases/immunology , Haploinsufficiency/immunology , Immunologic Deficiency Syndromes/immunology , Transcription Factors/immunology , Animals , Autoimmune Diseases/diagnosis , Autoimmune Diseases/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , DNA Mutational Analysis , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/genetics , Gene Expression/immunology , Haploinsufficiency/genetics , Humans , Immunoblotting , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Jurkat Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Mice, 129 Strain , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL