Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Immunol ; 41: 561-585, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37126418

ABSTRACT

Infection with SARS-CoV-2 results in clinical outcomes ranging from silent or benign infection in most individuals to critical pneumonia and death in a few. Genetic studies in patients have established that critical cases can result from inborn errors of TLR3- or TLR7-dependent type I interferon immunity, or from preexisting autoantibodies neutralizing primarily IFN-α and/or IFN-ω. These findings are consistent with virological studies showing that multiple SARS-CoV-2 proteins interfere with pathways of induction of, or response to, type I interferons. They are also congruent with cellular studies and mouse models that found that type I interferons can limit SARS-CoV-2 replication in vitro and in vivo, while their absence or diminution unleashes viral growth. Collectively, these findings point to insufficient type I interferon during the first days of infection as a general mechanism underlying critical COVID-19 pneumonia, with implications for treatment and directions for future research.


Subject(s)
COVID-19 , Interferon Type I , Mice , Humans , Animals , Interferons/pharmacology , SARS-CoV-2
2.
Nat Immunol ; 21(12): 1528-1539, 2020 12.
Article in English | MEDLINE | ID: mdl-33020661

ABSTRACT

Mutations that impact immune cell migration and result in immune deficiency illustrate the importance of cell movement in host defense. In humans, loss-of-function mutations in DOCK8, a guanine exchange factor involved in hematopoietic cell migration, lead to immunodeficiency and, paradoxically, allergic disease. Here, we demonstrate that, like humans, Dock8-/- mice have a profound type 2 CD4+ helper T (TH2) cell bias upon pulmonary infection with Cryptococcus neoformans and other non-TH2 stimuli. We found that recruited Dock8-/-CX3CR1+ mononuclear phagocytes are exquisitely sensitive to migration-induced cell shattering, releasing interleukin (IL)-1ß that drives granulocyte-macrophage colony-stimulating factor (GM-CSF) production by CD4+ T cells. Blocking IL-1ß, GM-CSF or caspase activation eliminated the type-2 skew in mice lacking Dock8. Notably, treatment of infected wild-type mice with apoptotic cells significantly increased GM-CSF production and TH2 cell differentiation. This reveals an important role for cell death in driving type 2 signals during infection, which may have implications for understanding the etiology of type 2 CD4+ T cell responses in allergic disease.


Subject(s)
Guanine Nucleotide Exchange Factors/deficiency , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Biomarkers , Caspases/metabolism , Cell Movement/genetics , Cell Movement/immunology , Cytokines/genetics , Cytokines/metabolism , Disease Susceptibility , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunophenotyping , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Signal Transduction
3.
Cell ; 159(7): 1578-90, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525876

ABSTRACT

Proteasomes and lysosomes constitute the major cellular systems that catabolize proteins to recycle free amino acids for energy and new protein synthesis. Tripeptidyl peptidase II (TPPII) is a large cytosolic proteolytic complex that functions in tandem with the proteasome-ubiquitin protein degradation pathway. We found that autosomal recessive TPP2 mutations cause recurrent infections, autoimmunity, and neurodevelopmental delay in humans. We show that a major function of TPPII in mammalian cells is to maintain amino acid levels and that TPPII-deficient cells compensate by increasing lysosome number and proteolytic activity. However, the overabundant lysosomes derange cellular metabolism by consuming the key glycolytic enzyme hexokinase-2 through chaperone-mediated autophagy. This reduces glycolysis and impairs the production of effector cytokines, including IFN-γ and IL-1ß. Thus, TPPII controls the balance between intracellular amino acid availability, lysosome number, and glycolysis, which is vital for adaptive and innate immunity and neurodevelopmental health.


Subject(s)
Adaptive Immunity , Aminopeptidases/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Glycolysis , Immunity, Innate , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Proteolysis , Serine Endopeptidases/metabolism , Amino Acid Sequence , Aminopeptidases/chemistry , Animals , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/chemistry , Female , Humans , Immunologic Deficiency Syndromes/immunology , Lysosomes/metabolism , Male , Models, Molecular , Molecular Sequence Data , Pedigree , Sequence Alignment , Serine Endopeptidases/chemistry
4.
Article in English | MEDLINE | ID: mdl-39233015

ABSTRACT

BACKGROUND: DOCK8 deficiency is a primary immunodeficiency in which allogeneic hematopoietic cell transplantation (HCT) represents the only known cure. We tested the ability of a busulfan-based regimen to achieve reliable engraftment and high levels of donor chimerism with acceptable toxicity in a prospective clinical trial in DOCK8 deficiency. OBJECTIVES: To both evaluate the ability of HCT to reverse the clinical phenotype and to correct the immunologic abnormalities by 1 year post HCT. METHODS: We conducted a prospective HCT trial for recipients with DOCK8 deficiency. Subjects were recruited from October 5, 2010, to December 30, 2022. Donor sources included fully matched related and unrelated donors and haploidentical donors. The reduced toxicity, myeloablative conditioning regimen contained no serotherapy. Graft-versus-host disease (GVHD) prophylaxis included either a calcineurin inhibitor with methotrexate or post-HCT cyclophosphamide (PT/Cy) followed by tacrolimus and mycophenolate mofetil. The trial was later amended to study PT/Cy in all patients. (Pilot Study of Reduced-Intensity Hematopoietic Stem Cell Transplant of DOCK8 [NCT01176006].) RESULTS: Thirty-six subjects, both children and adults (median age 16.4 years), underwent HCT for DOCK8 deficiency. Most patients, 33 of 36 (92%), achieved full (≥98%) donor chimerism in whole blood as early as day +30. With a median potential follow-up of 7.4 years, 29 (80.6%) were alive with no evidence of new DOCK8 deficiency-related complications. PT/Cy was effective in reducing the risk of acute GVHD in patients who had received matched unrelated donor and haploidentical transplants, but it was associated with transient delays in immune-reconstitution and hemorrhagic cystitis. CONCLUSIONS: A busulfan-based HCT regimen using PT/Cy for GVHD prophylaxis and a broad range of donor types and hematopoietic cell sources were well tolerated, leading to the reversal of the clinical immunophenotype.

5.
Immunol Rev ; 287(1): 9-19, 2019 01.
Article in English | MEDLINE | ID: mdl-30565250

ABSTRACT

DOCK8 immunodeficiency syndrome (DIDS) is a progressive combined immunodeficiency that can be distinguished from other combined immunodeficiencies or hyperimmunoglobulinemia E syndromes in featuring (a) profound susceptibility to virus infections of the skin, with associated skin cancers, and (b) severe food allergies. The DOCK8 locus has many repetitive sequence elements that predispose to the generation of large germline deletions as well as recombination-mediated somatic DNA repair. Residual DOCK8 protein contributes to the variable disease phenotype. The severe virus infections of the skin, and probably also VZV-associated vasculopathy, reflect an important function of DOCK8, which is normally required to maintain lymphocyte shape integrity as the cells migrate through dense tissues. Loss of DOCK8 also causes immune deficits through other mechanisms including a milder generalized cell survival defect and skewing of T helper cell subsets. Recent work has uncovered the roles for DOCK8 in dendritic cell responses that can also help explain the virus susceptibility, as well as in regulatory T cells that might help explain autoimmunity in a minority of patients. Fortunately, hematopoietic stem cell transplantation cures the eczema and infection susceptibility of DIDS, but not necessarily the other disease manifestations including food allergies.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Immunologic Deficiency Syndromes/immunology , T-Lymphocytes/immunology , Clustered Regularly Interspaced Short Palindromic Repeats , Disease Susceptibility , Eczema , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Neoplasms, Squamous Cell , Pneumonia , Vasculitis, Central Nervous System
6.
J Clin Immunol ; 42(1): 119-129, 2022 01.
Article in English | MEDLINE | ID: mdl-34657245

ABSTRACT

Rare, biallelic loss-of-function mutations in DOCK8 result in a combined immune deficiency characterized by severe and recurrent cutaneous infections, eczema, allergies, and susceptibility to malignancy, as well as impaired humoral and cellular immunity and hyper-IgE. The advent of next-generation sequencing technologies has enabled the rapid molecular diagnosis of rare monogenic diseases, including inborn errors of immunity. These advances have resulted in the implementation of gene-guided treatments, such as hematopoietic stem cell transplant for DOCK8 deficiency. However, putative disease-causing variants revealed by next-generation sequencing need rigorous validation to demonstrate pathogenicity. Here, we report the eventual diagnosis of DOCK8 deficiency in a consanguineous family due to a novel homozygous intronic deletion variant that caused aberrant exon splicing and subsequent loss of expression of DOCK8 protein. Remarkably, the causative variant was not initially detected by clinical whole-genome sequencing but was subsequently identified and validated by combining advanced genomic analysis, RNA-seq, and flow cytometry. This case highlights the need to adopt multipronged confirmatory approaches to definitively solve complex genetic cases that result from variants outside protein-coding exons and conventional splice sites.


Subject(s)
Job Syndrome , Consanguinity , Guanine Nucleotide Exchange Factors/genetics , Homozygote , Humans , Job Syndrome/diagnosis , Job Syndrome/genetics , Mutation/genetics , Exome Sequencing
7.
Hum Genet ; 140(9): 1299-1312, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34185153

ABSTRACT

Genetic defects of innate immunity impairing intestinal bacterial sensing are linked to the development of Inflammatory Bowel Disease (IBD). Although much evidence supports a role of the intestinal virome in gut homeostasis, most studies focus on intestinal viral composition rather than on host intestinal viral sensitivity. To demonstrate the association between the development of Very Early Onset IBD (VEOIBD) and variants in the IFIH1 gene which encodes MDA5, a key cytosolic sensor for viral nucleic acids. Whole exome sequencing (WES) was performed in two independent cohorts of children with VEOIBD enrolled in Italy (n = 18) and USA (n = 24). Luciferase reporter assays were employed to assess MDA5 activity. An enrichment analysis was performed on IFIH1 comparing 42 VEOIBD probands with 1527 unrelated individuals without gastrointestinal or immunological issues. We identified rare, likely loss-of-function (LoF), IFIH1 variants in eight patients with VEOIBD from a combined cohort of 42 children. One subject, carrying a homozygous truncating variant resulting in complete LoF, experienced neonatal-onset, pan-gastrointestinal, IBD-like enteropathy plus multiple infectious episodes. The remaining seven subjects, affected by VEOIBD without immunodeficiency, were carriers of one LoF variant in IFIH1. Among these, two patients also carried a second hypomorphic variant, with partial function apparent when MDA5 was weakly stimulated. Furthermore, IFIH1 variants were significantly enriched in children with VEOIBD as compared to controls (p = 0.007). Complete and partial MDA5 deficiency is associated with VEOIBD with variable penetrance and expressivity, suggesting a role for impaired intestinal viral sensing in IBD pathogenesis.


Subject(s)
Inflammatory Bowel Diseases/genetics , Interferon-Induced Helicase, IFIH1/genetics , Loss of Function Mutation , Child, Preschool , Female , Humans , Infant , Italy , Male , Whole Genome Sequencing
8.
J Biol Chem ; 294(37): 13638-13656, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31337704

ABSTRACT

Magnesium transporter 1 (MAGT1) critically mediates magnesium homeostasis in eukaryotes and is highly-conserved across different evolutionary branches. In humans, loss-of-function mutations in the MAGT1 gene cause X-linked magnesium deficiency with Epstein-Barr virus (EBV) infection and neoplasia (XMEN), a disease that has a broad range of clinical and immunological consequences. We have previously shown that EBV susceptibility in XMEN is associated with defective expression of the antiviral natural-killer group 2 member D (NKG2D) protein and abnormal Mg2+ transport. New evidence suggests that MAGT1 is the human homolog of the yeast OST3/OST6 proteins that form an integral part of the N-linked glycosylation complex, although the exact contributions of these perturbations in the glycosylation pathway to disease pathogenesis are still unknown. Using MS-based glycoproteomics, along with CRISPR/Cas9-KO cell lines, natural killer cell-killing assays, and RNA-Seq experiments, we now demonstrate that humans lacking functional MAGT1 have a selective deficiency in both immune and nonimmune glycoproteins, and we identified several critical glycosylation defects in important immune-response proteins and in the expression of genes involved in immunity, particularly CD28. We show that MAGT1 function is partly interchangeable with that of the paralog protein tumor-suppressor candidate 3 (TUSC3) but that each protein has a different tissue distribution in humans. We observed that MAGT1-dependent glycosylation is sensitive to Mg2+ levels and that reduced Mg2+ impairs immune-cell function via the loss of specific glycoproteins. Our findings reveal that defects in protein glycosylation and gene expression underlie immune defects in an inherited disease due to MAGT1 deficiency.


Subject(s)
Cation Transport Proteins/metabolism , Magnesium Deficiency/genetics , Neoplasms/genetics , Cation Transport Proteins/genetics , Epstein-Barr Virus Infections/genetics , Glycoproteins/metabolism , Glycosylation , HEK293 Cells , Homeostasis , Humans , Killer Cells, Natural/metabolism , Magnesium/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
9.
Curr Opin Pediatr ; 31(6): 815-820, 2019 12.
Article in English | MEDLINE | ID: mdl-31693592

ABSTRACT

PURPOSE OF REVIEW: Studying primary immunodeficiencies (PIDs) provides insights into human antiviral immunity in the natural infectious environment. This review describes new PIDs with genetic defects that impair innate antiviral responses. RECENT FINDINGS: New genetic defects in the interferon (IFN) signaling pathway include IFNAR1 deficiency, which causes uncontrolled infections with measles-mumps-rubella or yellow fever vaccines, and possibly also cytomegalovirus (CMV); and IRF9 deficiency, which results in influenza virus susceptibility. Genetic defects in several pattern recognition receptors include MDA5 deficiency, which impairs viral RNA sensing and confers human rhinovirus susceptibility; RNA polymerase III haploinsufficiency, which impairs sensing of A:T-rich virus DNA and confers VZV susceptibility; and TLR3 deficiency, which causes HSV-1 encephalitis (HSE) or influenza virus pneumonitis. Defects in RNA metabolism, such as that caused by Debranching enzyme 1 deficiency, can cause virus meningoencephalitis. Finally, defects in host restriction factors for virus replication, such as in CIB1 deficiency, contribute to uncontrolled ß-HPV infections. SUMMARY: Several new PIDs highlight the role of type I/III IFN signaling pathway, virus sensors, and host virus restriction factors in human antiviral immunity.


Subject(s)
Antiviral Agents/therapeutic use , Immunity, Innate/drug effects , Influenza, Human , Interferon Type I/genetics , Primary Immunodeficiency Diseases/drug therapy , Humans , Interferon Type I/deficiency , Signal Transduction
10.
J Clin Immunol ; 36(5): 441-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27207373

ABSTRACT

Since the discovery of the genetic basis of DOCK8 immunodeficiency syndrome (DIDS) in 2009, several hundred patients worldwide have been reported, validating and extending the initial clinical descriptions. Importantly, the beneficial role of hematopoietic stem cell transplantation for this disease has emerged, providing impetus for improved diagnosis. Additionally, several groups have further elucidated the biological functions of DOCK8 in the immune system that help explain disease pathogenesis. Here, we summarize these recent developments.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Guanine Nucleotide Exchange Factors/genetics , Hematopoietic Stem Cell Transplantation , Immunologic Deficiency Syndromes/immunology , Natural Killer T-Cells/immunology , Animals , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Immunologic Memory , Lymphopenia , Mutation/genetics
11.
J Allergy Clin Immunol ; 133(6): 1667-75, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24797421

ABSTRACT

BACKGROUND: Autosomal recessive loss-of-function mutations in dedicator of cytokinesis 8 (DOCK8) cause a combined immunodeficiency characterized by atopy, recurrent infections, and cancer susceptibility. A genotype-phenotype explanation for the variable disease expression is lacking. OBJECTIVE: We investigated whether reversions contributed to the variable disease expression. METHODS: Patients followed at the National Institutes of Health's Clinical Center were studied. We performed detailed genetic analyses and intracellular flow cytometry to detect DOCK8 protein expression within lymphocyte subsets. RESULTS: We identified 17 of 34 DOCK8-deficient patients who had germline mutations with variable degrees of reversion caused by somatic repair. Somatic repair of the DOCK8 mutations resulted from second-site mutation, original-site mutation, gene conversion, and intragenic crossover. Higher degrees of reversion were associated with recombination-mediated repair. DOCK8 expression was restored primarily within antigen-experienced T cells or natural killer cells but less so in naive T or B cells. Several patients exhibited multiple different repair events. Patients who had reversions were older and had less severe allergic disease, although infection susceptibility persisted. No patients were cured without hematopoietic cell transplantation. CONCLUSIONS: In patients with DOCK8 deficiency, only certain combinations of germline mutations supported secondary somatic repair. Those patients had an ameliorated disease course with longer survival but still had fatal complications or required hematopoietic cell transplantation. These observations support the concept that some DOCK8-immunodeficient patients have mutable mosaic genomes that can modulate disease phenotype over time.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Immunologic Deficiency Syndromes/genetics , Mutation , Phenotype , Adolescent , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Child , Child, Preschool , DNA Mutational Analysis , DNA Repair , Genotype , Germ-Line Mutation , Guanine Nucleotide Exchange Factors/metabolism , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/mortality , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
J Allergy Clin Immunol ; 133(5): 1400-9, 1409.e1-5, 2014 May.
Article in English | MEDLINE | ID: mdl-24589341

ABSTRACT

BACKGROUND: Identifying genetic syndromes that lead to significant atopic disease can open new pathways for investigation and intervention in allergy. OBJECTIVE: We sought to define a genetic syndrome of severe atopy, increased serum IgE levels, immune deficiency, autoimmunity, and motor and neurocognitive impairment. METHODS: Eight patients from 2 families with similar syndromic features were studied. Thorough clinical evaluations, including brain magnetic resonance imaging and sensory evoked potentials, were performed. Peripheral lymphocyte flow cytometry, antibody responses, and T-cell cytokine production were measured. Whole-exome sequencing was performed to identify disease-causing mutations. Immunoblotting, quantitative RT-PCR, enzymatic assays, nucleotide sugar, and sugar phosphate analyses, along with matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry of glycans, were used to determine the molecular consequences of the mutations. RESULTS: Marked atopy and autoimmunity were associated with increased T(H)2 and T(H)17 cytokine production by CD4(+) T cells. Bacterial and viral infection susceptibility were noted along with T-cell lymphopenia, particularly of CD8(+) T cells, and reduced memory B-cell numbers. Apparent brain hypomyelination resulted in markedly delayed evoked potentials and likely contributed to neurologic abnormalities. Disease segregated with novel autosomal recessive mutations in a single gene, phosphoglucomutase 3 (PGM3). Although PGM3 protein expression was variably diminished, impaired function was demonstrated by decreased enzyme activity and reduced uridine diphosphate-N-acetyl-D-glucosamine, along with decreased O- and N-linked protein glycosylation in patients' cells. These results define a new congenital disorder of glycosylation. CONCLUSIONS: Autosomal recessive hypomorphic PGM3 mutations underlie a disorder of severe atopy, immune deficiency, autoimmunity, intellectual disability, and hypomyelination.


Subject(s)
Autoimmune Diseases/genetics , Cognition Disorders/genetics , Common Variable Immunodeficiency/genetics , Genetic Diseases, Inborn/genetics , Hypersensitivity/genetics , Mutation , Phosphoglucomutase/genetics , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Child , Child, Preschool , Cognition Disorders/enzymology , Cognition Disorders/immunology , Cognition Disorders/pathology , Common Variable Immunodeficiency/enzymology , Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/pathology , Family , Female , Genetic Diseases, Inborn/enzymology , Genetic Diseases, Inborn/immunology , Genetic Diseases, Inborn/pathology , Humans , Hypersensitivity/enzymology , Hypersensitivity/immunology , Hypersensitivity/pathology , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Male , Pedigree , Phosphoglucomutase/immunology , Phosphoglucomutase/metabolism , Th17 Cells/enzymology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/enzymology , Th2 Cells/immunology , Th2 Cells/pathology , Young Adult
13.
Science ; 385(6715): eadd8947, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39298586

ABSTRACT

Humans with monogenic inborn errors responsible for extreme disease phenotypes can reveal essential physiological pathways. We investigated germline mutations in GNAI2, which encodes Gαi2, a key component in heterotrimeric G protein signal transduction usually thought to regulate adenylyl cyclase-mediated cyclic adenosine monophosphate (cAMP) production. Patients with activating Gαi2 mutations had clinical presentations that included impaired immunity. Mutant Gαi2 impaired cell migration and augmented responses to T cell receptor (TCR) stimulation. We found that mutant Gαi2 influenced TCR signaling by sequestering the guanosine triphosphatase (GTPase)-activating protein RASA2, thereby promoting RAS activation and increasing downstream extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)-AKT S6 signaling to drive cellular growth and proliferation.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2 , Germ-Line Mutation , Receptors, Antigen, T-Cell , T-Lymphocytes , ras GTPase-Activating Proteins , Humans , Cell Movement/genetics , Cell Proliferation , GTP-Binding Protein alpha Subunit, Gi2/genetics , Immunity/genetics , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , ras GTPase-Activating Proteins/genetics , ras Proteins/metabolism , ras Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Pedigree
14.
J Biol Chem ; 286(45): 38913-23, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21940623

ABSTRACT

Dendritic Cells (DCs) play an important role in the initiation of the immune response by migrating to regional lymph nodes and presenting antigen processed at the inflammatory site to antigen-specific naïve T cells. Prostaglandin E2 (PGE2) has been reported to play an essential role in DC migration. We reported previously that PGE2 induces matrix metalloproteinase 9 (MMP-9) expression in DCs and that PGE2-induced MMP-9 is required for DC migration in vivo and in vitro. In this study, we investigated the signaling mechanisms involved in PGE2-induced MMP-9 expression in DCs. We show that PGE2-induced MMP-9 expression is mediated primarily through the EP2/EP4 → cAMP → protein kinase A (PKA)/PI3K → ERK signaling pathway, leading to c-Fos expression, and through JNK-mediated activation of c-Jun in a PKA/PI3K/ERK-independent manner. EP2 and EP4 receptor agonists, as well as cAMP analogs, mimic the up-regulation of MMP-9 by PGE2. PKA, PI3K, and ERK inhibitors abolished PGE2- and cAMP-induced c-Fos and MMP-9 up-regulation, and ERK activation was required for the binding of activator protein 1 (AP-1) transcription factor to the MMP-9 promoter. Our results describe a new molecular mechanism for the effect of PGE2 on MMP-9 production in DCs that could lead to future therapeutic approaches using ERK inhibitors to regulate DC migration.


Subject(s)
Dendritic Cells/metabolism , Dinoprostone/metabolism , MAP Kinase Signaling System/physiology , Matrix Metalloproteinase 9/biosynthesis , Transcription Factor AP-1/metabolism , Up-Regulation/physiology , Animals , Cyclic AMP/genetics , Cyclic AMP/metabolism , Dendritic Cells/cytology , Dinoprostone/pharmacology , Enzyme Induction/drug effects , Enzyme Induction/physiology , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 9/genetics , Mice , Protein Kinases/genetics , Protein Kinases/metabolism , Transcription Factor AP-1/genetics , Up-Regulation/drug effects
15.
J Clin Immunol ; 32(4): 698-708, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22476911

ABSTRACT

We describe seven Turkish children with DOCK8 deficiency who have not been previously reported. Three patients presented with typical features of recurrent or severe cutaneous viral infections, atopic dermatitis, and recurrent respiratory or gastrointestinal tract infections. However, four patients presented with other features. Patient 1-1 featured sclerosing cholangitis and colitis; patient 2-1, granulomatous soft tissue lesion and central nervous system involvement, with primary central nervous system lymphoma found on follow-up; patient 3-1, a fatal metastatic leiomyosarcoma; and patient 4-2 showed no other symptoms initially besides atopic dermatitis. Similar to other previously reported Turkish patients, but in contrast to patients of non-Turkish ethnicity, the patients' lymphopenia was primarily restricted to CD4(+) T cells. Patients had homozygous mutations in DOCK8 that altered splicing, introduced premature terminations, destabilized protein, or involved large deletions within the gene. Genotyping of remaining family members showed that DOCK8 deficiency is a fully penetrant, autosomal recessive disease. In our patients, bone marrow transplantation resulted in rapid improvement followed by disappearance of viral skin lesions, including lesions resembling epidermodysplasia verruciformis, atopic dermatitis, and recurrent infections. Particularly for patients who feature unusual clinical manifestations, immunological testing, in conjunction with genetic testing, can prove invaluable in diagnosing DOCK8 deficiency and providing potentially curative treatment.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Severe Combined Immunodeficiency/genetics , Adolescent , Bone Marrow Transplantation , Child , Child, Preschool , Dermatitis, Atopic/genetics , Female , Genotype , Humans , Immunoglobulins, Intravenous/therapeutic use , Lymphopenia/genetics , Lymphopenia/therapy , Male , Sequence Deletion , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/therapy , Turkey
16.
N Engl J Med ; 361(21): 2046-55, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19776401

ABSTRACT

BACKGROUND: Recurrent sinopulmonary and cutaneous viral infections with elevated serum levels of IgE are features of some variants of combined immunodeficiency. The genetic causes of these variants are unknown. METHODS: We collected longitudinal clinical data on 11 patients from eight families who had recurrent sinopulmonary and cutaneous viral infections. We performed comparative genomic hybridization arrays and targeted gene sequencing. Variants with predicted loss-of-expression mutations were confirmed by means of a quantitative reverse-transcriptase-polymerase-chain-reaction assay and immunoblotting. We evaluated the number and function of lymphocytes with the use of in vitro assays and flow cytometry. RESULTS: Patients had recurrent otitis media, sinusitis, and pneumonias; recurrent Staphylococcus aureus skin infections with otitis externa; recurrent, severe herpes simplex virus or herpes zoster infections; extensive and persistent infections with molluscum contagiosum; and human papillomavirus infections. Most patients had severe atopy with anaphylaxis; several had squamous-cell carcinomas, and one had T-cell lymphoma-leukemia. Elevated serum IgE levels, hypereosinophilia, low numbers of T cells and B cells, low serum IgM levels, and variable IgG antibody responses were common. Expansion in vitro of activated CD8 T cells was impaired. Novel homozygous or compound heterozygous deletions and point mutations in the gene encoding the dedicator of cytokinesis 8 protein (DOCK8) led to the absence of DOCK8 protein in lymphocytes. CONCLUSIONS: Autosomal recessive DOCK8 deficiency is associated with a novel variant of combined immunodeficiency.


Subject(s)
Gene Deletion , Guanine Nucleotide Exchange Factors/genetics , Severe Combined Immunodeficiency/genetics , CD8-Positive T-Lymphocytes/physiology , Female , Genes, Recessive , Guanine Nucleotide Exchange Factors/metabolism , Humans , Immunoglobulins/blood , Longitudinal Studies , Lymphocyte Activation , Male , Pedigree , Respiratory Tract Infections/genetics , Respiratory Tract Infections/immunology , Severe Combined Immunodeficiency/immunology , Skin Diseases, Infectious/genetics , Skin Diseases, Infectious/immunology
17.
J Exp Med ; 219(8)2022 08 01.
Article in English | MEDLINE | ID: mdl-35708626

ABSTRACT

Recessive or dominant inborn errors of type I interferon (IFN) immunity can underlie critical COVID-19 pneumonia in unvaccinated adults. The risk of COVID-19 pneumonia in unvaccinated children, which is much lower than in unvaccinated adults, remains unexplained. In an international cohort of 112 children (<16 yr old) hospitalized for COVID-19 pneumonia, we report 12 children (10.7%) aged 1.5-13 yr with critical (7 children), severe (3), and moderate (2) pneumonia and 4 of the 15 known clinically recessive and biochemically complete inborn errors of type I IFN immunity: X-linked recessive TLR7 deficiency (7 children) and autosomal recessive IFNAR1 (1), STAT2 (1), or TYK2 (3) deficiencies. Fibroblasts deficient for IFNAR1, STAT2, or TYK2 are highly vulnerable to SARS-CoV-2. These 15 deficiencies were not found in 1,224 children and adults with benign SARS-CoV-2 infection without pneumonia (P = 1.2 × 10-11) and with overlapping age, sex, consanguinity, and ethnicity characteristics. Recessive complete deficiencies of type I IFN immunity may underlie ∼10% of hospitalizations for COVID-19 pneumonia in children.


Subject(s)
COVID-19 , Interferon Type I , Pneumonia , Adult , COVID-19/genetics , Child , Humans , Inheritance Patterns , SARS-CoV-2
18.
J Exp Med ; 218(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34726731

ABSTRACT

Enterovirus (EV) infection rarely results in life-threatening infection of the central nervous system. We report two unrelated children with EV30 and EV71 rhombencephalitis. One patient carries compound heterozygous TLR3 variants (loss-of-function F322fs2* and hypomorphic D280N), and the other is homozygous for an IFIH1 variant (loss-of-function c.1641+1G>C). Their fibroblasts respond poorly to extracellular (TLR3) or intracellular (MDA5) poly(I:C) stimulation. The baseline (TLR3) and EV-responsive (MDA5) levels of IFN-ß in the patients' fibroblasts are low. EV growth is enhanced at early and late time points of infection in TLR3- and MDA5-deficient fibroblasts, respectively. Treatment with exogenous IFN-α2b before infection renders both cell lines resistant to EV30 and EV71, whereas post-infection treatment with IFN-α2b rescues viral susceptibility fully only in MDA5-deficient fibroblasts. Finally, the poly(I:C) and viral phenotypes of fibroblasts are rescued by the expression of WT TLR3 or MDA5. Human TLR3 and MDA5 are critical for cell-intrinsic immunity to EV, via the control of baseline and virus-induced type I IFN production, respectively.


Subject(s)
Encephalitis, Viral/immunology , Enterovirus Infections/immunology , Interferon-Induced Helicase, IFIH1/genetics , Toll-Like Receptor 3/genetics , Cells, Cultured , Child, Preschool , Encephalitis, Viral/genetics , Enterovirus/drug effects , Enterovirus/physiology , Enterovirus Infections/genetics , Female , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/virology , Humans , Infant , Interferon alpha-2/pharmacology , Interferon-Induced Helicase, IFIH1/immunology , Interferon-beta/immunology , Interferon-beta/metabolism , Loss of Function Mutation , Male , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/immunology , Poly I-C/pharmacology , Rhombencephalon/virology , Toll-Like Receptor 3/immunology , Virus Replication/drug effects
19.
Mol Cell Biol ; 27(12): 4551-65, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17438135

ABSTRACT

GATA factors orchestrate hematopoiesis via multistep transcriptional mechanisms, but the interrelationships and importance of individual steps are poorly understood. Using complementation analysis with GATA-1-null cells and mice containing a hypomorphic allele of the chromatin remodeler BRG1, we dissected the pathway from GATA-1 binding to cofactor recruitment, chromatin loop formation, and transcriptional activation. Analysis of GATA-1-mediated activation of the beta-globin locus, in which GATA-1 assembles dispersed complexes at the promoters and the distal locus control region (LCR), revealed molecular intermediates, including GATA-1-independent and GATA-1-containing LCR subcomplexes, both defective in promoting loop formation. An additional intermediate consisted of an apparently normal LCR complex and a promoter complex with reduced levels of total RNA polymerase II (Pol II) and Pol II phosphorylated at serine 5 of the carboxy-terminal domain. Reduced BRG1 activity solely compromised Pol II and serine 5-phosphorylated Pol II occupancy at the promoter, phenocopying the LCR-deleted mouse. These studies defined a hierarchical order of GATA-1-triggered events at a complex locus and establish a novel mechanism of long-range gene regulation.


Subject(s)
Cell Differentiation , Chromatin/metabolism , GATA1 Transcription Factor/physiology , Gene Expression Regulation , Hematopoietic System/physiology , Alleles , Animals , Cell Line , Chromatin/chemistry , Chromatin Immunoprecipitation , DNA Helicases/genetics , DNA Helicases/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/genetics , Gene Deletion , Globins/genetics , Locus Control Region , Mice , Mice, Knockout , Models, Genetic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Temperature , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL