Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Article in English | MEDLINE | ID: mdl-33634762

ABSTRACT

BACKGROUND: Dedicator of Cytokinesis 8 (DOCK8) deficiency, the most frequent cause of autosomal recessive hyper immunoglobulin (Ig)E syndrome, is a rare combined immunodeficiency. OBJECTIVE: In this study, we report seven patients, with consanguineous parents, with five novel variants within the DOCK8 gene. METHODS: For genetic analysis, we performed Whole Exome Sequencing (WES) or targeted sequencing by means of Next-generation sequencing (NGS) for some of the patients. For others, Sanger sequencing, Fluorescence-activated cell sorting (FACS), or polymerase chain reaction (PCR) were used. RESULTS: We report five novel variants within the DOCK8 gene: three deletions (deletion of exons 4-12, 24-30, and 22-27), one frameshift (LRG_196:g.189315dup;p.(Leu1052Profs*7)), and a splice region variant (LRG_196t1:c.741+5G>T). Patients presented with skin lesions, food allergy, candidiasis, otitis, recurrent respiratory infections, short stature, aortic aneurism, gynecomastia, and coarse facial features. Patients had leukocytosis, eosinophilia, lymphopenia, and monocytosis, elevated IgE, IgG, IgA, reduced IgM and IgA levels. Patients had a low percentage of CD3+ and CD4+ cells and a high percentage of CD19+, CD27+CD19+, and recent thymic emigrants T cells. The percentage of natural killer cells was increased in one of the patients while it was decreased in another patient. One patient died due to disseminated intravascular coagulation after hematopoietic stem cell transplantation. CONCLUSION: We reported novel variants within the DOCK8 gene and highlighted the risk of aneurysms in these patients, which have been rarely reported in these patients.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Job Syndrome/genetics , Adolescent , Child , Child, Preschool , Consanguinity , DNA Mutational Analysis , Female , Guanine Nucleotide Exchange Factors/deficiency , Humans , Iran , Job Syndrome/immunology , Job Syndrome/pathology , Male , Mutation , Pedigree , Exome Sequencing
2.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Article in English | MEDLINE | ID: mdl-34172578

ABSTRACT

Job syndrome is a rare genetic disorder caused by STAT3 mutations and primarily characterized by immune dysfunction along with comorbid skeleton developmental abnormalities including osteopenia, recurrent fracture of long bones, and scoliosis. So far, there is no definitive cure for the skeletal defects in Job syndrome, and treatments are limited to management of clinical symptoms only. Here, we have investigated the molecular mechanism whereby Stat3 regulates skeletal development and osteoblast differentiation. We showed that removing Stat3 function in the developing limb mesenchyme or osteoprogenitor cells in mice resulted in shortened and bow limbs with multiple fractures in long bones that resembled the skeleton symptoms in the Job Syndrome. However, Stat3 loss did not alter chondrocyte differentiation and hypertrophy in embryonic development, while osteoblast differentiation was severely reduced. Genome-wide transcriptome analyses as well as biochemical and histological studies showed that Stat3 loss resulted in down-regulation of Wnt/ß-catenin signaling. Restoration of Wnt/ß-catenin signaling by injecting BIO, a small molecule inhibitor of GSK3, or crossing with a Lrp5 gain of function (GOF) allele, rescued the bone reduction phenotypes due to Stat3 loss to a great extent. These studies uncover the essential functions of Stat3 in maintaining Wnt/ß-catenin signaling in early mesenchymal or osteoprogenitor cells and provide evidence that bone defects in the Job Syndrome are likely caused by Wnt/ß-catenin signaling reduction due to reduced STAT3 activities in bone development. Enhancing Wnt/ß-catenin signaling could be a therapeutic approach to reduce bone symptoms of Job syndrome patients.


Subject(s)
Bone and Bones/pathology , Job Syndrome/metabolism , Job Syndrome/pathology , Mesenchymal Stem Cells/metabolism , STAT3 Transcription Factor/deficiency , Wnt Signaling Pathway , Alleles , Animals , Cartilage/pathology , Cell Differentiation , Embryo, Mammalian/pathology , Extremities/pathology , Gene Deletion , Humans , Integrases/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Mesenchymal Stem Cells/pathology , Mesoderm/embryology , Mice, Transgenic , Osteoblasts/pathology , Osteogenesis
3.
J Clin Invest ; 130(8): 4167-4181, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32369445

ABSTRACT

There are more than 7000 described rare diseases, most lacking specific treatment. Autosomal-dominant hyper-IgE syndrome (AD-HIES, also known as Job's syndrome) is caused by mutations in STAT3. These patients present with immunodeficiency accompanied by severe nonimmunological features, including skeletal, connective tissue, and vascular abnormalities, poor postinfection lung healing, and subsequent pulmonary failure. No specific therapies are available for these abnormalities. Here, we investigated underlying mechanisms in order to identify therapeutic targets. Histological analysis of skin wounds demonstrated delayed granulation tissue formation and vascularization during skin-wound healing in AD-HIES patients. Global gene expression analysis in AD-HIES patient skin fibroblasts identified deficiencies in a STAT3-controlled transcriptional network regulating extracellular matrix (ECM) remodeling and angiogenesis, with hypoxia-inducible factor 1α (HIF-1α) being a major contributor. Consistent with this, histological analysis of skin wounds and coronary arteries from AD-HIES patients showed decreased HIF-1α expression and revealed abnormal organization of the ECM and altered formation of the coronary vasa vasorum. Disease modeling using cell culture and mouse models of angiogenesis and wound healing confirmed these predicted deficiencies and demonstrated therapeutic benefit of HIF-1α-stabilizing drugs. The study provides mechanistic insights into AD-HIES pathophysiology and suggests potential treatment options for this rare disease.


Subject(s)
Extracellular Matrix/metabolism , Job Syndrome/metabolism , Neovascularization, Physiologic , Skin/metabolism , Wound Healing , Wounds and Injuries/metabolism , Animals , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Job Syndrome/genetics , Job Syndrome/pathology , Male , Mice , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Skin/blood supply , Skin/pathology , Wounds and Injuries/genetics , Wounds and Injuries/pathology
4.
Stem Cell Res ; 41: 101586, 2019 12.
Article in English | MEDLINE | ID: mdl-31707214

ABSTRACT

Autosomal dominant Hyper IgE syndrome (AD-HIES), a rare immune deficiency affecting fewer than one per million people, is caused by heterozygous deleterious mutations in STAT3. STAT3 signaling plays crucial roles in basic cellular functions affecting broad aspects of cellular homeostasis. Accordingly, in addition to immunological deficits, patients experience severe multisystem non-immunological features. Human induced pluripotent stem cells (hiPSC) are well established as in vivo disease models for various human pathologies. We describe the generation of iPSC from three AD-HIES patients. These iPSCs express pluripotency markers, differentiate into three germ layers, have normal karyotype and similar genome identity to parental cells.


Subject(s)
Cell Differentiation , Fibroblasts/pathology , Induced Pluripotent Stem Cells/pathology , Job Syndrome/genetics , Job Syndrome/pathology , Mutation , STAT3 Transcription Factor/genetics , Cells, Cultured , Fibroblasts/metabolism , Genes, Dominant , Humans , Induced Pluripotent Stem Cells/metabolism
5.
Proc Natl Acad Sci U S A ; 116(33): 16463-16472, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31346092

ABSTRACT

Heterozygous in-frame mutations in coding regions of human STAT3 underlie the only known autosomal dominant form of hyper IgE syndrome (AD HIES). About 5% of familial cases remain unexplained. The mutant proteins are loss-of-function and dominant-negative when tested following overproduction in recipient cells. However, the production of mutant proteins has not been detected and quantified in the cells of heterozygous patients. We report a deep intronic heterozygous STAT3 mutation, c.1282-89C>T, in 7 relatives with AD HIES. This mutation creates a new exon in the STAT3 complementary DNA, which, when overexpressed, generates a mutant STAT3 protein (D427ins17) that is loss-of-function and dominant-negative in terms of tyrosine phosphorylation, DNA binding, and transcriptional activity. In immortalized B cells from these patients, the D427ins17 protein was 2 kDa larger and 4-fold less abundant than wild-type STAT3, on mass spectrometry. The patients' primary B and T lymphocytes responded poorly to STAT3-dependent cytokines. These findings are reminiscent of the impaired responses of leukocytes from other patients with AD HIES due to typical STAT3 coding mutations, providing further evidence for the dominance of the mutant intronic allele. These findings highlight the importance of sequencing STAT3 introns in patients with HIES without candidate variants in coding regions and essential splice sites. They also show that AD HIES-causing STAT3 mutant alleles can be dominant-negative even if the encoded protein is produced in significantly smaller amounts than wild-type STAT3.


Subject(s)
DNA-Binding Proteins/genetics , Job Syndrome/genetics , RNA Splice Sites/genetics , STAT3 Transcription Factor/genetics , Adult , Alleles , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Child, Preschool , Exons/genetics , Female , Gene Expression Regulation/genetics , Heterozygote , Humans , Job Syndrome/pathology , Loss of Function Mutation/genetics , Male , Middle Aged , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
6.
BMC Med Genet ; 20(1): 114, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31242861

ABSTRACT

BACKGROUND: Dedicator of cytokinesis 8 (DOCK8) deficiency (MIM #243700) is a rare disease, leads to a combined primary immunodeficiency (PID), and accounts for the autosomal recessive-hyper immunoglobulin E syndrome (AR-HIES). DOCK8 deficiency status characterizes by recurrent infections, atopy, and risk of cancer. Lymphoproliferative disease complicating PID, is difficult to diagnose. Our aim is to present a rare case of PID, and to the best of our knowledge, she is the first case of DOCK8 deficiency from Iraq. The genetic diagnosis was carried out in Japan using dried blood spot-based DNA transfer and whole-exome sequencing. CASE PRESENTATION: An 11-year-old Iraqi girl, of double first-cousin-parents, had a history of severe eczema, food allergy, and repeated infections. She presented with a jaw mass, bilateral cervical and axillary lymphadenopathy, and immunoglobulin (Ig) assays of 20, 3.3 and 1.7-fold above maximum normal level for age of IgE, IgA and IgG, respectively, along with a low IgM, eosinophilia and lymphopenia. Based on the jaw mass biopsy, non-Hodgkin lymphoma was suggested in Iraq, whereas histopathological re-evaluation in Japan revealed the diagnosis of a polyclonal reactive proliferation spectrum of lymphoproliferative disorders/plasmacytic hyperplasia, complicating PID. Whole-exome sequencing supported the diagnosis of PID by identifying a homozygous DOCK8 mutation with previously reported pathogenicity (NM_203447:c.3332delT, p.Phe1113Leufs*2), that may be attributed to consanguinity. CONCLUSIONS: International collaboration using an effective DNA transportation technique and next-generation sequencing was the key to pinpoint the diagnosis of DOCK8 deficiency. Our case asserted that careful pathogenetic evaluation, in an advanced setting, was crucial for ruling out the neoplastic process. Pediatricians in areas with a high prevalence of consanguinity marriage should have a high index of suspicion of DOCK8 deficiency in patients with recalcitrant eczema, and frequent respiratory and skin infectious episodes.


Subject(s)
Exome Sequencing/methods , Genetic Association Studies , Genetic Predisposition to Disease , Guanine Nucleotide Exchange Factors/genetics , Job Syndrome/genetics , Mutation , Antibodies/blood , Child , Consanguinity , DNA/blood , Eosinophilia/immunology , Female , Homozygote , Humans , Iraq , Japan , Jaw/pathology , Job Syndrome/diagnostic imaging , Job Syndrome/immunology , Job Syndrome/pathology , Lymphopenia/immunology , Lymphoproliferative Disorders/genetics , Pedigree
9.
Sci Rep ; 8(1): 16719, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30425284

ABSTRACT

In hyper-IgE syndromes (HIES), a group of primary immunodeficiencies clinically overlapping with atopic dermatitis, early diagnosis is crucial to initiate appropriate therapy and prevent irreversible complications. Identification of underlying gene defects such as in DOCK8 and STAT3 and corresponding molecular testing has improved diagnosis. Yet, in a child and her newborn sibling with HIES phenotype molecular diagnosis was misleading. Extensive analyses driven by the clinical phenotype identified an intronic homozygous DOCK8 variant c.4626 + 76 A > G creating a novel splice site as disease-causing. While the affected newborn carrying the homozygous variant had no expression of DOCK8 protein, in the index patient molecular diagnosis was compromised due to expression of altered and wildtype DOCK8 transcripts and DOCK8 protein as well as defective STAT3 signaling. Sanger sequencing of lymphocyte subsets revealed that somatic alterations and reversions revoked the predominance of the novel over the canonical splice site in the index patient explaining DOCK8 protein expression, whereas defective STAT3 responses in the index patient were explained by a T cell phenotype skewed towards central and effector memory T cells. Hence, somatic alterations and skewed immune cell phenotypes due to selective pressure may compromise molecular diagnosis and need to be considered with unexpected clinical and molecular findings.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Introns/genetics , Job Syndrome/genetics , Mutation , RNA Splice Sites/genetics , Base Sequence , Child, Preschool , Computational Biology , Female , Gene Expression Regulation/genetics , Humans , Infant , Job Syndrome/pathology , Molecular Diagnostic Techniques , Pregnancy , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics
11.
J Clin Invest ; 128(8): 3595-3604, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30035749

ABSTRACT

Autosomal dominant hyper IgE syndrome (AD-HIES), or Job's syndrome, is a primary immune deficiency caused by dominant-negative mutations in STAT3. Recurrent Staphylococcus aureus skin abscesses are a defining feature of this syndrome. A widely held hypothesis that defects in peripheral Th17 differentiation confer this susceptibility has never been directly evaluated. To assess the cutaneous immune response in AD-HIES, we induced suction blisters in healthy volunteers (HVs) and patients with AD-HIES and then challenged the wound with lethally irradiated bacteria. We show that cutaneous production of IL-17A and IL-17F was normal in patients with AD-HIES. Overproduction of TNF-α differentiated the responses in AD-HIES from HVs. This was associated with reduced IL-10 family signaling in blister-infiltrating cells and defective epithelial cell function. Mouse models of AD-HIES recapitulated these aberrant epithelial responses to S. aureus and involved defective epithelial-to-mesenchymal transition (EMT) rather than a failure of bacterial killing. Defective responses in mouse models of AD-HIES and primary keratinocyte cultures from patients with AD-HIES could be reversed by TNF-α blockade and by drugs with reported modulatory effects on EMT. Our results identify these as potential therapeutic approaches in patients with AD-HIES suffering S. aureus infections.


Subject(s)
Epithelial Cells/immunology , Furunculosis/immunology , Job Syndrome/immunology , Keratinocytes/immunology , Staphylococcus aureus/immunology , Tumor Necrosis Factor-alpha/immunology , Adult , Animals , Disease Models, Animal , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/immunology , Female , Furunculosis/genetics , Furunculosis/pathology , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Job Syndrome/genetics , Job Syndrome/pathology , Keratinocytes/pathology , Male , Mice , Mice, Transgenic , Tumor Necrosis Factor-alpha/genetics
12.
Sci Rep ; 8(1): 6956, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29725107

ABSTRACT

Complete tyrosine kinase 2 (TYK2) deficiency has been previously described in patients with primary immunodeficiency diseases. The patients were infected with various pathogens, including mycobacteria and/or viruses, and one of the patients developed hyper-IgE syndrome. A detailed immunological investigation of these patients revealed impaired responses to type I IFN, IL-10, IL-12 and IL-23, which are associated with increased susceptibility to mycobacterial and/or viral infections. Herein, we report a recessive partial TYK2 deficiency in two siblings who presented with T-cell lymphopenia characterized by low naïve CD4+ T-cell counts and who developed Epstein-Barr virus (EBV)-associated B-cell lymphoma. Targeted exome-sequencing of the siblings' genomes demonstrated that both patients carried novel compound heterozygous mutations (c.209_212delGCTT/c.691C > T, p.Cys70Serfs*21/p.Arg231Trp) in the TYK2. The TYK2 protein levels were reduced by 35% in the T cells of the patient. Unlike the response under complete TYK2 deficiency, the patient's T cells responded normally to type I IFN, IL-6, IL-10 and IL-12, whereas the cells displayed an impaired response to IL-23. Furthermore, the level of STAT1 was low in the cells of the patient. These studies reveal a new clinical entity of a primary immunodeficiency with T-cell lymphopenia that is associated with compound heterozygous TYK2 mutations in the patients.


Subject(s)
Immunologic Deficiency Syndromes/genetics , Job Syndrome/genetics , Lymphopenia/genetics , Mutation , TYK2 Kinase/deficiency , Adolescent , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/pathology , Female , Herpesvirus 4, Human/isolation & purification , Heterozygote , Humans , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/pathology , Job Syndrome/complications , Job Syndrome/pathology , Lymphoma, B-Cell/complications , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Lymphopenia/complications , Lymphopenia/pathology , Male , Primary Immunodeficiency Diseases , Siblings , T-Lymphocytes/pathology , TYK2 Kinase/genetics
13.
J Pediatr Hematol Oncol ; 40(6): e380-e382, 2018 08.
Article in English | MEDLINE | ID: mdl-28902078

ABSTRACT

BACKGROUND: Hyper-immunoglobulin E syndrome (HIES) is a rare primary immunodeficiency disease characterized by recurrent infections and elevated levels of serum immunoglobulin E, usually over 2000 IU/mL. Recurrent and chronic infection of the epidermis and squamous epithelium may also be a cause of squamous cell carcinoma (SCC). SCC is rare with HIES. CASE REPORT: A 17-year-old male patient who was diagnosed as HIES was admitted with purulent right ear discharge. The patient had a history of eczema starting from the age of 7 months and a history of recurrent middle ear infection starting from the age of 5. Biopsy specimens were taken from the lesion in the external auditory canal, and the lesion was reported as SCC. CONSLUSION: Patients with autosomal recessive HIES are at an increased risk for infections and malignancies. SCC should be considered in the differential diagnosis of the patients presenting with recurrent middle ear infections and immunodeficiency.


Subject(s)
Carcinoma, Squamous Cell , Ear Neoplasms , Job Syndrome , Skin Neoplasms , Adolescent , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Ear Neoplasms/diagnosis , Ear Neoplasms/genetics , Ear Neoplasms/pathology , Humans , Job Syndrome/diagnosis , Job Syndrome/genetics , Job Syndrome/pathology , Male , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics , Skin Neoplasms/pathology
14.
J Clin Immunol ; 37(7): 695-700, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28803389

ABSTRACT

OBJECTIVE: STAT 3 deficiency (autosomal dominant hyper immunoglobulin E syndrome (AD-HIES)) is a primary immunodeficiency disorder with multi-organ involvement caused by dominant negative signal transducer and activator of transcription gene 3 (STAT3) mutations. We sought to describe the gastrointestinal (GI) manifestations of this disease. METHODS: Seventy subjects aged five to 60 years with a molecular diagnosis of AD-HIES were evaluated at the National Institutes of Health (NIH). Data collection involved a GI symptom questionnaire and retrospective chart review. RESULTS: In our cohort of 70 subjects, we found that 60% had GI symptoms (42/70). The most common manifestations were gastroesophageal reflux disease (GERD) observed in 41%, dysphagia in 31%, and abdominal pain in 24%. The most serious complications were food impaction in 13% and colonic perforation in 6%. Diffuse esophageal wall thickening in 74%, solid stool in the right colon in 50% (12/24), and hiatal hernia in 26% were the most prevalent radiologic findings. Esophagogastroduodenoscopy (EGD) demonstrated esophageal tortuosity in 35% (8/23), esophageal ulceration in 17% (4/23), esophageal strictures requiring dilation in 9% (2/23), and gastric ulceration in 17% (4/23). Esophageal eosinophilic infiltration was an unexpected histologic finding seen in 65% (11/17). CONCLUSION: The majority of AD-HIES subjects develop GI manifestations as part of their disease. Most notable are the symptoms and radiologic findings of GI dysmotility, as well as significant eosinophilic infiltration, concerning for a secondary eosinophilic esophagitis. These findings suggest that the STAT3 pathway may be implicated in a new mechanism for the pathogenesis of several GI disorders.


Subject(s)
Gastrointestinal Diseases/etiology , Job Syndrome/complications , STAT3 Transcription Factor/deficiency , Adolescent , Adult , Biopsy , Child , Child, Preschool , Colonoscopy , Eosinophils/immunology , Female , Gastrointestinal Diseases/blood , Gastrointestinal Diseases/diagnostic imaging , Gastrointestinal Diseases/pathology , Gastrointestinal Tract/diagnostic imaging , Gastrointestinal Tract/pathology , Gastrointestinal Tract/surgery , Humans , Immunoglobulin E/blood , Job Syndrome/blood , Job Syndrome/diagnostic imaging , Job Syndrome/pathology , Leukocyte Count , Male , Middle Aged , Surveys and Questionnaires , Tomography, X-Ray Computed , Ultrasonography , Young Adult
15.
J Neurovirol ; 23(4): 632-636, 2017 08.
Article in English | MEDLINE | ID: mdl-28643229

ABSTRACT

We, herein, report a 23-year-old male with a rare inherited immunodeficiency disease, hyperimmunoglobulin IgE syndrome (HIES), who developed progressive multifocal leukoencephalopathy (PML) and lymphoma simultaneously. Primary immunodeficiency of the patient has remained undiagnosed until adulthood. PML is a severe demyelinating disease of the central nervous system caused by John Cunningham virus. HIES is a rare, inherited immunodeficiency characterized by high serum levels of IgE, recurrent staphylococcal infection, eczema, and hypereosinophilia. PML may accompany primary immunodeficiency syndromes, but the association with HIES is exceedingly rare. We discuss the imaging findings, medical management, and a review of related literature on primary immunodeficiency cases complicating with PML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Eczema/diagnosis , Hypereosinophilic Syndrome/diagnosis , Job Syndrome/diagnosis , Leukoencephalopathy, Progressive Multifocal/diagnosis , Lymphoma/diagnosis , Eczema/drug therapy , Eczema/immunology , Eczema/pathology , Fatal Outcome , Humans , Hypereosinophilic Syndrome/drug therapy , Hypereosinophilic Syndrome/immunology , Hypereosinophilic Syndrome/pathology , Immunoglobulin E/blood , JC Virus/isolation & purification , JC Virus/pathogenicity , Job Syndrome/drug therapy , Job Syndrome/immunology , Job Syndrome/pathology , Leukoencephalopathy, Progressive Multifocal/drug therapy , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/pathology , Lymphoma/drug therapy , Lymphoma/immunology , Lymphoma/pathology , Male , Treatment Failure , Young Adult
16.
Blood ; 128(26): 3061-3072, 2016 12 29.
Article in English | MEDLINE | ID: mdl-27799162

ABSTRACT

Autosomal dominant hyper-IgE syndrome (AD-HIES) is caused by dominant-negative mutations in STAT3; however, the molecular basis for mutant STAT3 allele dysfunction is unclear and treatment remains supportive. We hypothesized that AD-HIES mutations decrease STAT3 protein stability and that mutant STAT3 activity can be improved by agents that increase chaperone protein activity. We used computer modeling to characterize the effect of STAT3 mutations on protein stability. We measured STAT3 protein half-life (t1/2) and determined levels of STAT3 phosphorylated on tyrosine (Y) 705 (pY-STAT3) and mRNA levels of STAT3 gene targets in Epstein-Barr virus-transformed B (EBV) cells, human peripheral blood mononuclear cells (PBMCs), and mouse splenocytes incubated without or with chaperone protein modulators-HSF1A, a small-molecule TRiC modulator, or geranylgeranylacetone (GGA), a drug that upregulates heat shock protein (HSP) 70 and HSP90. Computer modeling predicted that 81% of AD-HIES mutations are destabilizing. STAT3 protein t1/2 in EBV cells from AD-HIES patients with destabilizing STAT3 mutations was markedly reduced. Treatment of EBV cells containing destabilizing STAT3 mutations with either HSF1A or GGA normalized STAT3 t1/2, increased pY-STAT3 levels, and increased mRNA levels of STAT3 target genes up to 79% of control. In addition, treatment of human PBMCs or mouse splenocytes containing destabilizing STAT3 mutations with either HSF1A or GGA increased levels of cytokine-activated pY-STAT3 within human CD4+ and CD8+ T cells and numbers of IL-17-producing CD4+ mouse splenocytes, respectively. Thus, most AD-HIES STAT3 mutations are destabilizing; agents that modulate chaperone protein function improve STAT3 stability and activity in T cells and may provide a specific treatment.


Subject(s)
Job Syndrome/metabolism , STAT3 Transcription Factor/metabolism , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cytokines/pharmacology , DNA-Binding Proteins/metabolism , Diterpenes/pharmacology , Half-Life , Heat Shock Transcription Factors , Herpesvirus 4, Human/physiology , Humans , Interleukin-17/metabolism , Job Syndrome/pathology , Mice , Models, Molecular , Mutant Proteins/metabolism , Mutation/genetics , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Stability/drug effects , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/genetics , Spleen/pathology , Transcription Factors/metabolism
17.
Sci Rep ; 6: 33274, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27633343

ABSTRACT

Bacterial resistance against classical antibiotics is a growing problem and the development of new antibiotics is limited. Thus, novel alternatives to antibiotics are warranted. Antimicrobial peptides (AMPs) are effector molecules of innate immunity that can be induced by several compounds, including vitamin D and phenyl-butyrate (PBA). Utilizing a luciferase based assay, we recently discovered that the histone deacetylase inhibitor Entinostat is a potent inducer of the CAMP gene encoding the human cathelicidin LL-37. Here we investigate a mechanism for the induction and also find that Entinostat up-regulates human ß-defensin 1. Analysis of the CAMP promoter sequence revealed binding sites for the transcription factors STAT3 and HIF-1α. By using short hairpin RNA and selective inhibitors, we found that both transcription factors are involved in Entinostat-induced expression of LL-37. However, only HIF-1α was found to be recruited to the CAMP promoter, suggesting that Entinostat activates STAT3, which promotes transcription of CAMP by increasing the expression of HIF-1α. Finally, we provide in vivo relevance to our findings by showing that Entinostat-elicited LL-37 expression was impaired in macrophages from a patient with a STAT3-mutation. Combined, our findings support a role for STAT3 and HIF-1α in the regulation of LL-37 expression.


Subject(s)
Benzamides/pharmacology , Cathelicidins/genetics , Histone Deacetylase Inhibitors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Job Syndrome/genetics , Pyridines/pharmacology , STAT3 Transcription Factor/genetics , Antimicrobial Cationic Peptides , Cathelicidins/agonists , Cathelicidins/metabolism , Genes, Reporter , HEK293 Cells , HT29 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/agonists , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Job Syndrome/immunology , Job Syndrome/pathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Luciferases/genetics , Luciferases/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/agonists , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcriptional Activation
19.
Molecules ; 21(6)2016 Jun 10.
Article in English | MEDLINE | ID: mdl-27294900

ABSTRACT

BACKGROUND: Atopic eczema is a common childhood disease associated with high IgE and eosinophilia. We characterized the clinical features associated with hyper-IgE (defined as IgE > 2000 IU/L) in eczema. METHODS: Nottingham Eczema Severity Score (NESS), family and personal history of atopy, skin prick test (SPT) for common food and aeroallergens, highest serum IgE ever and eosinophil counts were evaluated in 330 children eczema patients. Childhood-NESS (NESS performed at <10 years of age) and adolescent-NESS (NESS performed at >10 years of age) were further analyzed. RESULTS: IgE correlated with NESS (spearman coefficient 0.35, p < 0.001) and eosinophil percentage (spearman coefficient 0.56, p = 0.001). Compared with IgE ≤ 2000IU/L (n = 167), patients with hyper-IgE (n = 163) were associated with male gender (p = 0.002); paternal atopy (p = 0.026); personal history of atopic rhinitis (p = 0.016); asthma (p < 0.001); dietary avoidance (p < 0.001); use of wet wrap (p < 0.001); traditional Chinese medicine use (TCM, p < 0.001); immunomodulant use (azathioprine or cyclosporine, p < 0.001); skin prick sensitization by dust mites (p < 0.001), cats (p = 0.012), dogs (p = 0.018), food (p = 0.002); eosinophilia (p < 0.001); more severe disease during childhood (p < 0.0001) and during adolescence (p < 0.0001), but not onset age of eczema or maternal atopy. Logistic regression showed that hyper-IgE was associated with personal history of asthma (exp(B) = 5.12, p = 0.002) and eczema severity during childhood and adolescence (p < 0.001). For patients <10 years of age, dust mite sensitization (p = 0.008) was associated with hyper-IgE. For patients >10years of age, food allergen sensitization was associated with hyper-IgE (p = 0.008). CONCLUSIONS: Hyper-IgE is independently associated with asthma, more severe atopy and more severe eczema during childhood and adolescence. IgE > 2000 IU/L may be a tool to aid prognostication of this chronic relapsing dermatologic disease and its progression to asthma.


Subject(s)
Asthma/blood , Eczema/blood , Immunoglobulin E/blood , Job Syndrome/blood , Allergens/immunology , Asthma/immunology , Asthma/pathology , Child , Child, Preschool , China , Eczema/pathology , Eosinophils/immunology , Female , Humans , Hypersensitivity, Immediate/blood , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/pathology , Immunoglobulin E/immunology , Job Syndrome/immunology , Job Syndrome/pathology , Logistic Models , Male , Prognosis
20.
Indian J Pediatr ; 83(10): 1104-8, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27226025

ABSTRACT

OBJECTIVES: To assess the utility of T helper17 (TH17) cell enumeration vis-à-vis National Institutes of Health (NIH) scoring in Hyper IgE syndrome (HIES). METHODS: Clinical phenotypes of Hyper IgE syndrome patients with and without STAT3 mutation were analysed and correlated with absolute eosinophil count, serum IgE levels and TH17 cell numbers in 19 patients with clinically suspected HIES and compared with healthy controls (n = 20). RESULTS: The difference in serum IgE between patients with and without STAT3 mutation and healthy controls was statistically significant (p < 0.05). Six patients had NIH score > 40; of which 4 were positive for STAT3 pathogenic variants, whereas two patients in the group with no identifiable STAT3 pathogenic variant had NIH score > 40. NIH score had sensitivity of 80 % and specificity of 87.5 % to detect cases with STAT3 pathogenic variants. TH17 cells were markedly low in all cases with STAT3 pathogenic variants. Among patients without STAT3 pathogenic variants, none had low TH17 cell numbers. CONCLUSIONS: Low TH17 cell numbers together with NIH scores can be a better indicator for presence of STAT3 mutations.


Subject(s)
Job Syndrome/pathology , Mutation , STAT3 Transcription Factor/genetics , Child, Preschool , Humans , Job Syndrome/genetics , Phenotype , Th17 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...