Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Imaging Sci Dent ; 53(1): 69-75, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37006794

ABSTRACT

Purpose: Maxillofacial trauma predominantly affects young adults between 20 and 40 years of age. Although radioprotection is a legal requirement, the significant potential of dose reduction in computed tomography (CT) is still underused in the clinical routine. The objective of this study was to evaluate whether maxillofacial fractures can be reliably detected and classified using ultra-low-dose CT. Materials and Methods: CT images of 123 clinical cases with maxillofacial fractures were classified by two readers using the AOCOIAC software and compared with the corresponding results from post-treatment images. In group 1, consisting of 97 patients with isolated facial trauma, pre-treatment CT images at different dose levels (volumetric computed tomography dose index: ultra-low dose, 2.6 mGy; low dose, <10 mGy; and regular dose, <20 mGy) were compared with post-treatment cone-beam computed tomography (CBCT). In group 2, consisting of 31 patients with complex midface fractures, pre-treatment shock room CT images were compared with post-treatment CT at different dose levels or CBCT. All images were presented in random order and classified by 2 readers blinded to the clinical results. All cases with an unequal classification were re-evaluated. Results: In both groups, ultra-low-dose CT had no clinically relevant effect on fracture classification. Fourteen cases in group 2 showed minor differences in the classification code, which were no longer obvious after comparing the images directly to each other. Conclusion: Ultra-low-dose CT images allowed the correct diagnosis and classification of maxillofacial fractures. These results might lead to a substantial reconsideration of current reference dose levels.

2.
J Exp Med ; 219(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-36094518

ABSTRACT

Human cells homozygous for rare loss-of-expression (LOE) TYK2 alleles have impaired, but not abolished, cellular responses to IFN-α/ß (underlying viral diseases in the patients) and to IL-12 and IL-23 (underlying mycobacterial diseases). Cells homozygous for the common P1104A TYK2 allele have selectively impaired responses to IL-23 (underlying isolated mycobacterial disease). We report three new forms of TYK2 deficiency in six patients from five families homozygous for rare TYK2 alleles (R864C, G996R, G634E, or G1010D) or compound heterozygous for P1104A and a rare allele (A928V). All these missense alleles encode detectable proteins. The R864C and G1010D alleles are hypomorphic and loss-of-function (LOF), respectively, across signaling pathways. By contrast, hypomorphic G996R, G634E, and A928V mutations selectively impair responses to IL-23, like P1104A. Impairment of the IL-23-dependent induction of IFN-γ is the only mechanism of mycobacterial disease common to patients with complete TYK2 deficiency with or without TYK2 expression, partial TYK2 deficiency across signaling pathways, or rare or common partial TYK2 deficiency specific for IL-23 signaling.


Subject(s)
Job Syndrome , TYK2 Kinase , Humans , Interferon-gamma/metabolism , Interleukin-23 , Job Syndrome/genetics , TYK2 Kinase/deficiency , TYK2 Kinase/genetics , TYK2 Kinase/metabolism
3.
Cancers (Basel) ; 14(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35158803

ABSTRACT

Transcriptional regulation of the actin-bundling protein and tumor marker Fascin is highly diverse depending on cell and tumor type. Previously, we discovered that the viral oncoprotein Tax-1 of human T-cell leukemia virus type 1 (HTLV-1) considerably enhances Fascin expression in T-cells, depending on classical NF-κB signaling. In this study, we asked if the non-oncogenic Tax-2 of the related HTLV-2 is still able to induce Fascin by using luciferase assays, immunoblot, and qPCR. We found that Tax-2 only slightly induces Fascin expression compared to Tax-1; however, both Tax-1 and Tax-2 comparably activated a 1.6 kb fragment in the human Fascin promoter including Tax-responsive elements. Furthermore, we identified a link between Tax-induced activity of the alternative NF-κB pathway and Fascin induction. While treatment with the second mitochondria-derived activator of caspases (SMAC)-mimetic AZD5582, a compound known to robustly activate alternative NF-κB signaling, did not induce Fascin, combination of AZD5582 with activation of classical NF-κB signaling by Tax-2 significantly induced Fascin expression. In conclusion, our data demonstrate that both classical and alternative NF-κB activity are necessary for strong Fascin induction by the viral Tax oncoproteins, thus, shedding new light on the regulation of Fascin in T-cells and during viral transformation.

4.
Int J Mol Sci ; 22(24)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34948391

ABSTRACT

The human T-cell leukemia virus type 1 (HTLV-1)-encoded transactivator and oncoprotein Tax-1 is essential for HTLV-1 replication. We recently found that Tax-1 interacts with transcription elongation factor for RNA polymerase II 2, ELL2, which enhances Tax-1-mediated transactivation of the HTLV-1 promotor. Here, we characterize the Tax-1:ELL2 interaction and its impact on viral transactivation by confocal imaging, co-immunoprecipitation, and luciferase assays. We found that Tax-1 and ELL2 not only co-precipitate, but also co-localize in dot-like structures in the nucleus. Tax-1:ELL2 complex formation occurred independently of Tax-1 point mutations, which are crucial for post translational modifications (PTMs) of Tax-1, suggesting that these PTMs are irrelevant for Tax-1:ELL2 interaction. In contrast, Tax-1 deletion mutants lacking either N-terminal (aa 1-37) or C-terminal regions (aa 150-353) of Tax-1 were impaired in interacting with ELL2. Contrary to Tax-1, the related, non-oncogenic Tax-2B from HTLV-2B did not interact with ELL2. Finally, we found that ELL2-R1 (aa 1-353), which carries an RNA polymerase II binding domain, and ELL2-R3 (aa 515-640) are sufficient to interact with Tax-1; however, only ELL2-truncations expressing R1 could enhance Tax-1-mediated transactivation of the HTLV-1 promoter. Together, this study identifies domains in Tax-1 and ELL2 being required for Tax-1:ELL2 complex formation and for viral transactivation.


Subject(s)
HTLV-I Infections/metabolism , Human T-lymphotropic virus 1/physiology , Trans-Activators/metabolism , Transcriptional Elongation Factors/metabolism , Viral Proteins/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , Host-Pathogen Interactions , Human T-lymphotropic virus 1/genetics , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Point Mutation , Protein Interaction Maps , Trans-Activators/genetics , Transcriptional Activation , Viral Proteins/genetics
5.
J Exp Med ; 218(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34160550

ABSTRACT

We have described a child suffering from Mendelian susceptibility to mycobacterial disease (MSMD) due to autosomal recessive, complete T-bet deficiency, which impairs IFN-γ production by innate and innate-like adaptive, but not mycobacterial-reactive purely adaptive, lymphocytes. Here, we explore the persistent upper airway inflammation (UAI) and blood eosinophilia of this patient. Unlike wild-type (WT) T-bet, the mutant form of T-bet from this patient did not inhibit the production of Th2 cytokines, including IL-4, IL-5, IL-9, and IL-13, when overexpressed in T helper 2 (Th2) cells. Moreover, Herpesvirus saimiri-immortalized T cells from the patient produced abnormally large amounts of Th2 cytokines, and the patient had markedly high plasma IL-5 and IL-13 concentrations. Finally, the patient's CD4+ αß T cells produced most of the Th2 cytokines in response to chronic stimulation, regardless of their antigen specificities, a phenotype reversed by the expression of WT T-bet. T-bet deficiency thus underlies the excessive production of Th2 cytokines, particularly IL-5 and IL-13, by CD4+ αß T cells, causing blood eosinophilia and UAI. The MSMD of this patient results from defective IFN-γ production by innate and innate-like adaptive lymphocytes, whereas the UAI and eosinophilia result from excessive Th2 cytokine production by adaptive CD4+ αß T lymphocytes.


Subject(s)
Cytokines/metabolism , Pneumonia/immunology , T-Box Domain Proteins/deficiency , Th2 Cells/immunology , Animals , Cytokines/blood , Epigenesis, Genetic , Epitopes/immunology , Female , Humans , Immunologic Memory , Male , Mice, Inbred C57BL , Mutation/genetics , Pedigree , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Respiratory Hypersensitivity/blood , Respiratory Hypersensitivity/immunology , Sequence Analysis, RNA , Single-Cell Analysis , T-Box Domain Proteins/genetics
6.
Retrovirology ; 17(1): 30, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32912211

ABSTRACT

BACKGROUND: Human T-cell leukemia virus type 1 (HTLV-1) infects primarily CD4+ T-lymphocytes and evoques severe diseases, predominantly Adult T-Cell Leukemia/ Lymphoma (ATL/L) and HTLV-1-associated Myelopathy/ Tropical Spastic Paraparesis (HAM/TSP). The viral transactivator of the pX region (Tax) is important for initiating malignant transformation, and deregulation of the major signaling pathway nuclear factor of kappa B (NF-κB) by Tax represents a hallmark of HTLV-1 driven cancer. RESULTS: Here we found that Tax mutants which are defective in NF-κB signaling showed diminished protein expression levels compared to Tax wildtype in T-cells, whereas Tax transcript levels were comparable. Strikingly, constant activation of NF-κB signaling by the constitutive active mutant of inhibitor of kappa B kinase (IKK2, IKK-ß), IKK2-EE, rescued protein expression of the NF-κB defective Tax mutants M22 and K1-10R and even increased protein levels of Tax wildtype in various T-cell lines while Tax transcript levels were only slightly affected. Using several Tax expression constructs, an increase of Tax protein occurred independent of Tax transcripts and independent of the promoter used. Further, Tax and M22 protein expression were strongly enhanced by 12-O-Tetradecanoylphorbol-13-Acetate [TPA; Phorbol 12-myristate 13-acetate (PMA)]/ ionomycin, inducers of NF-κB and cytokine signaling, but not by tumor necrosis factor alpha (TNF-α). On the other hand, co-expression of Tax with a dominant negative inhibitor of κB, IκBα-DN, or specific inhibition of IKK2 by the compound ACHP, led to a vast decrease in Tax protein levels to some extent independent of Tax transcripts in transiently transfected and Tax-transformed T-cells. Cycloheximide chase experiments revealed that co-expression of IKK2-EE prolongs the half-life of M22, and constant repression of NF-κB signaling by IκBα-DN strongly reduces protein stability of Tax wildtype suggesting that NF-κB activity is required for Tax protein stability. Finally, protein expression of Tax and M22 could be recovered by NH4Cl and PYR-41, inhibitors of the lysosome and the ubiquitin-activating enzyme E1, respectively. CONCLUSIONS: Together, these findings suggest that Tax's capability to induce NF-κB is critical for protein expression and stabilization of Tax itself. Overall, identification of this novel positive feedback loop between Tax and NF-κB in T-cells improves our understanding of Tax-driven transformation.


Subject(s)
Feedback, Physiological , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/physiology , NF-kappa B p50 Subunit/metabolism , Gene Expression Regulation , Gene Products, tax/genetics , Humans , Ionomycin/pharmacology , Jurkat Cells , Mutation , NF-kappa B p50 Subunit/genetics , Protein Stability , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology
7.
J Clin Invest ; 130(6): 3158-3171, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32163377

ABSTRACT

Mendelian susceptibility to mycobacterial disease (MSMD) is characterized by a selective predisposition to clinical disease caused by the Bacille Calmette-Guérin (BCG) vaccine and environmental mycobacteria. The known genetic etiologies of MSMD are inborn errors of IFN-γ immunity due to mutations of 15 genes controlling the production of or response to IFN-γ. Since the first MSMD-causing mutations were reported in 1996, biallelic mutations in the genes encoding IFN-γ receptor 1 (IFN-γR1) and IFN-γR2 have been reported in many patients of diverse ancestries. Surprisingly, mutations of the gene encoding the IFN-γ cytokine itself have not been reported, raising the remote possibility that there might be other agonists of the IFN-γ receptor. We describe 2 Lebanese cousins with MSMD, living in Kuwait, who are both homozygous for a small deletion within the IFNG gene (c.354_357del), causing a frameshift that generates a premature stop codon (p.T119Ifs4*). The mutant allele is loss of expression and loss of function. We also show that the patients' herpesvirus Saimiri-immortalized T lymphocytes did not produce IFN-γ, a phenotype that can be rescued by retrotransduction with WT IFNG cDNA. The blood T and NK lymphocytes from these patients also failed to produce and secrete detectable amounts of IFN-γ. Finally, we show that human IFNG has evolved under stronger negative selection than IFNGR1 or IFNGR2, suggesting that it is less tolerant to heterozygous deleterious mutations than IFNGR1 or IFNGR2. This may account for the rarity of patients with autosomal-recessive, complete IFN-γ deficiency relative to patients with complete IFN-γR1 and IFN-γR2 deficiencies.


Subject(s)
Base Sequence , Genetic Diseases, Inborn , Homozygote , Interferon-gamma/deficiency , Mycobacterium Infections , Sequence Deletion , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/immunology , Humans , Mycobacterium Infections/genetics , Mycobacterium Infections/immunology , Mycobacterium bovis/immunology , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Interferon gamma Receptor
8.
Clin Cancer Res ; 26(13): 3259-3270, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32060103

ABSTRACT

PURPOSE: If routine diagnostics are inconclusive, neurologic deterioration and death of patients with brain cancer are attributed to tumor or therapy. Therefore, diagnosing symptoms of encephalopathy caused by human cytomegalovirus (HCMV) reactivation remains uncommon. We investigated the role of HCMV reactivation in neurologic decline and clinical outcome after the start of radiochemotherapy. EXPERIMENTAL DESIGN: HCMV analyses and extended MRI studies including additional independent retrospective neuroradiologic evaluation were performed at predetermined intervals and in case of sudden neurologic decline for 118 adult patients: 63 histologically proven high-grade gliomas, 55 with brain metastases. Immunophenotyping from simultaneously taken whole-blood samples was carried out to detect immune cells serving as prognostic marker for HCMV-associated complications. Symptomatic viremia and overall survival (OS) were the endpoints. RESULTS: Twenty-four percent (28/118) of all patients (12/44 glioblastoma, 3/13 anaplastic astrocytoma; 8/31 non-small cell lung cancer (NSCLC), 13/24 other brain metastases) developed HCMV-viremia during or within 4 weeks after radiotherapy; 21 of 28 patients experienced concurrent major neurologic decline, reversible by antiviral treatment. Identified by immunophenotyping, pretherapeutically low basophil counts predicted a high-risk for HCMV-associated encephalopathy (glioblastoma: P = 0.002, NSCLC: P = 0.02). Median OS was substantially reduced after HCMV-associated encephalopathy without MRI signs of tumor progression [glioblastoma: 99 vs. 570 days (calculated 1-year OS: 22% vs. 69%; P = 0.01) and NSCLC: 47 vs. 219 days (calculated 1-year OS: 0% vs. 32%; P = 0.02)]. CONCLUSIONS: For patients with brain cancer, HCMV reactivation after the start of radiochemotherapy is a frequent risk for cognitively detrimental but treatable encephalopathy and premature death. Routinely performed HCMV diagnostics, assessing basophil counts and study-based anti-viral regimens, are necessary to combat this hidden threat.See related commentary by Lawler et al., p. 3077.


Subject(s)
Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adult , Carcinoma, Non-Small-Cell Lung/therapy , Chemoradiotherapy/adverse effects , Cytomegalovirus , Humans , Retrospective Studies
9.
Sci Immunol ; 3(30)2018 12 21.
Article in English | MEDLINE | ID: mdl-30578351

ABSTRACT

Hundreds of patients with autosomal recessive, complete IL-12p40 or IL-12Rß1 deficiency have been diagnosed over the last 20 years. They typically suffer from invasive mycobacteriosis and, occasionally, from mucocutaneous candidiasis. Susceptibility to these infections is thought to be due to impairments of IL-12-dependent IFN-γ immunity and IL-23-dependent IL-17A/IL-17F immunity, respectively. We report here patients with autosomal recessive, complete IL-12Rß2 or IL-23R deficiency, lacking responses to IL-12 or IL-23 only, all of whom, unexpectedly, display mycobacteriosis without candidiasis. We show that αß T, γδ T, B, NK, ILC1, and ILC2 cells from healthy donors preferentially produce IFN-γ in response to IL-12, whereas NKT cells and MAIT cells preferentially produce IFN-γ in response to IL-23. We also show that the development of IFN-γ-producing CD4+ T cells, including, in particular, mycobacterium-specific TH1* cells (CD45RA-CCR6+), is dependent on both IL-12 and IL-23. Last, we show that IL12RB1, IL12RB2, and IL23R have similar frequencies of deleterious variants in the general population. The comparative rarity of symptomatic patients with IL-12Rß2 or IL-23R deficiency, relative to IL-12Rß1 deficiency, is, therefore, due to lower clinical penetrance. There are fewer symptomatic IL-23R- and IL-12Rß2-deficient than IL-12Rß1-deficient patients, not because these genetic disorders are rarer, but because the isolated absence of IL-12 or IL-23 is, in part, compensated by the other cytokine for the production of IFN-γ, thereby providing some protection against mycobacteria. These experiments of nature show that human IL-12 and IL-23 are both required for optimal IFN-γ-dependent immunity to mycobacteria, both individually and much more so cooperatively.


Subject(s)
Immunity, Innate/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-23/immunology , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium/immunology , Humans , Interleukin-12/deficiency , Interleukin-12/genetics , Interleukin-23/deficiency , Interleukin-23/genetics , Pedigree
10.
Sci Immunol ; 3(30)2018 12 21.
Article in English | MEDLINE | ID: mdl-30578352

ABSTRACT

Inherited IL-12Rß1 and TYK2 deficiencies impair both IL-12- and IL-23-dependent IFN-γ immunity and are rare monogenic causes of tuberculosis, each found in less than 1/600,000 individuals. We show that homozygosity for the common TYK2 P1104A allele, which is found in about 1/600 Europeans and between 1/1000 and 1/10,000 individuals in regions other than East Asia, is more frequent in a cohort of patients with tuberculosis from endemic areas than in ethnicity-adjusted controls (P = 8.37 × 10-8; odds ratio, 89.31; 95% CI, 14.7 to 1725). Moreover, the frequency of P1104A in Europeans has decreased, from about 9% to 4.2%, over the past 4000 years, consistent with purging of this variant by endemic tuberculosis. Surprisingly, we also show that TYK2 P1104A impairs cellular responses to IL-23, but not to IFN-α, IL-10, or even IL-12, which, like IL-23, induces IFN-γ via activation of TYK2 and JAK2. Moreover, TYK2 P1104A is properly docked on cytokine receptors and can be phosphorylated by the proximal JAK, but lacks catalytic activity. Last, we show that the catalytic activity of TYK2 is essential for IL-23, but not IL-12, responses in cells expressing wild-type JAK2. In contrast, the catalytic activity of JAK2 is redundant for both IL-12 and IL-23 responses, because the catalytically inactive P1057A JAK2, which is also docked and phosphorylated, rescues signaling in cells expressing wild-type TYK2. In conclusion, homozygosity for the catalytically inactive P1104A missense variant of TYK2 selectively disrupts the induction of IFN-γ by IL-23 and is a common monogenic etiology of tuberculosis.


Subject(s)
Interferon-gamma/immunology , Interleukin-23/immunology , Mutation, Missense/genetics , TYK2 Kinase/genetics , Tuberculosis/immunology , Cells, Cultured , Homozygote , Humans , Interleukin-23/deficiency , TYK2 Kinase/immunology
11.
Front Immunol ; 9: 2400, 2018.
Article in English | MEDLINE | ID: mdl-30386345

ABSTRACT

Serine/threonine kinase 4 (STK4) deficiency is an autosomal recessive genetic condition that leads to primary immunodeficiency (PID) typically characterized by lymphopenia, recurrent infections and Epstein Barr Virus (EBV) induced lymphoproliferation and -lymphoma. State-of-the-art treatment regimens consist of prevention or treatment of infections, immunoglobulin substitution (IVIG) and restoration of the immune system by hematopoietic stem cell transplantation. Here, we report on two patients from two consanguineous families of Turkish (patient P1) and Moroccan (patient P2) decent, with PID due to homozygous STK4 mutations. P1 harbored a previously reported frameshift (c.1103 delT, p.M368RfsX2) and P2 a novel splice donor site mutation (P2; c.525+2 T>G). Both patients presented in childhood with recurrent infections, CD4 lymphopenia and dysregulated immunoglobulin levels. Patient P1 developed a highly malignant B cell lymphoma at the age of 10 years and a second, independent Hodgkin lymphoma 5 years later. To our knowledge she is the first STK4 deficient case reported who developed lymphoma in the absence of detectable EBV or other common viruses. Lymphoma development may be due to the lacking tumor suppressive function of STK4 or the perturbed immune surveillance due to the lack of CD4+ T cells. Our data should raise physicians' awareness of [1] lymphoma proneness of STK4 deficient patients even in the absence of EBV infection and [2] possibly underlying STK4 deficiency in pediatric patients with a history of recurrent infections, CD4 lymphopenia and lymphoma and unknown genetic make-up. Patient P2 experienced recurrent otitis in childhood, but when she presented at the age of 14, she showed clinical and immunological characteristics similar to patients suffering from Autoimmune Lymphoproliferative Syndrome (ALPS): elevated DNT cell number, non-malignant lymphadenopathy and hepatosplenomegaly, hematolytic anemia, hypergammaglobulinemia. Also patient P1 presented with ALPS-like features (lymphadenopathy, elevated DNT cell number and increased Vitamin B12 levels) and both were initially clinically diagnosed as ALPS-like. Closer examination of P2, however, revealed active EBV infection and genetic testing identified a novel STK4 mutation. None of the patients harbored typically ALPS-associated mutations of the Fas receptor mediated apoptotic pathway and Fas-mediated apoptosis was not affected. The presented case reports extend the clinical spectrum of STK4 deficiency.


Subject(s)
Autoimmune Lymphoproliferative Syndrome/etiology , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human , Immunologic Deficiency Syndromes/etiology , Lymphoma/etiology , Phenotype , Protein Serine-Threonine Kinases/deficiency , Autoimmune Lymphoproliferative Syndrome/diagnosis , Case-Control Studies , Computational Biology/methods , DNA Mutational Analysis , Epstein-Barr Virus Infections/virology , Female , Humans , Immunologic Deficiency Syndromes/diagnosis , Intracellular Signaling Peptides and Proteins , Lymphoma/diagnosis , Male , Mutation , Pedigree , Exome Sequencing
12.
Sci Immunol ; 3(24)2018 06 15.
Article in English | MEDLINE | ID: mdl-29907690

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is a central regulator of immune homeostasis. STAT3 levels are strictly controlled, and STAT3 impairment contributes to several diseases including the monogenic autosomal-dominant hyper-immunoglobulin E (IgE) syndrome (AD-HIES). We investigated patients of four consanguineous families with an autosomal-recessive disorder resembling the phenotype of AD-HIES, with symptoms of immunodeficiency, recurrent infections, skeletal abnormalities, and elevated IgE. Patients presented with reduced STAT3 expression and diminished T helper 17 cell numbers, in absence of STAT3 mutations. We identified two distinct homozygous nonsense mutations in ZNF341, which encodes a zinc finger transcription factor. Wild-type ZNF341 bound to and activated the STAT3 promoter, whereas the mutant variants showed impaired transcriptional activation, partly due to nuclear translocation failure. In summary, nonsense mutations in ZNF341 account for the STAT3-like phenotype in four autosomal-recessive kindreds. Thus, ZNF341 is a previously unrecognized regulator of immune homeostasis.


Subject(s)
Immunocompetence/genetics , Job Syndrome/genetics , STAT3 Transcription Factor/genetics , Th17 Cells/immunology , Transcription Factors/genetics , Adolescent , Adult , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Nucleus/metabolism , Child , Codon, Nonsense , Consanguinity , Exons/genetics , Female , Genes, Recessive/genetics , Genes, Recessive/immunology , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Infant , Job Syndrome/blood , Job Syndrome/immunology , Male , Pedigree , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/immunology , Th17 Cells/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Young Adult , Zinc Fingers/genetics
13.
Sci Immunol ; 3(24)2018 06 15.
Article in English | MEDLINE | ID: mdl-29907691

ABSTRACT

Heterozygosity for human signal transducer and activator of transcription 3 (STAT3) dominant-negative (DN) mutations underlies an autosomal dominant form of hyper-immunoglobulin E syndrome (HIES). We describe patients with an autosomal recessive form of HIES due to loss-of-function mutations of a previously uncharacterized gene, ZNF341 ZNF341 is a transcription factor that resides in the nucleus, where it binds a specific DNA motif present in various genes, including the STAT3 promoter. The patients' cells have low basal levels of STAT3 mRNA and protein. The autoinduction of STAT3 production, activation, and function by STAT3-activating cytokines is strongly impaired. Like patients with STAT3 DN mutations, ZNF341-deficient patients lack T helper 17 (TH17) cells, have an excess of TH2 cells, and have low memory B cells due to the tight dependence of STAT3 activity on ZNF341 in lymphocytes. Their milder extra-hematopoietic manifestations and stronger inflammatory responses reflect the lower ZNF341 dependence of STAT3 activity in other cell types. Human ZNF341 is essential for the STAT3 transcription-dependent autoinduction and sustained activity of STAT3.


Subject(s)
Gene Expression Regulation/immunology , Job Syndrome/genetics , STAT3 Transcription Factor/genetics , Transcription Factors/genetics , Transcription, Genetic/immunology , Adolescent , Adult , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Nucleus/metabolism , Consanguinity , Cytokines/immunology , Cytokines/metabolism , Exons/genetics , Female , Genes, Recessive/genetics , Genes, Recessive/immunology , Homozygote , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Job Syndrome/blood , Job Syndrome/immunology , Loss of Function Mutation , Lymphocyte Count , Male , Middle Aged , Pedigree , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Exome Sequencing , Young Adult , Zinc Fingers/genetics
14.
Curr Protoc Immunol ; 115: 7.21C.1-7.21C.12, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27801513

ABSTRACT

Human T cells can be transformed and expanded with herpesvirus saimiri (HVS). HVS-transformed T cells from patients have facilitated the study of a broad range of primary immunodeficiencies (PID) in which T-cell development or function is altered. However, the utility of HVS-transformed T cells for genetic studies has been limited by technical challenges in the expression of exogenous genes, including wild-type or mutant alleles. A novel, gamma retrovirus-based method for the simple and reliable transduction, purification, and study of HVS-transformed T cells is described. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Genetic Vectors/genetics , Herpesvirus 2, Saimiriine/genetics , T-Lymphocytes/metabolism , Transduction, Genetic/methods , Transgenes , Animals , Cell Line, Transformed , Gene Order , Gene Transfer Techniques , Humans , Plasmids/genetics , Retroviridae/genetics
15.
J Exp Med ; 213(11): 2413-2435, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27647349

ABSTRACT

Combined immunodeficiency (CID) refers to inborn errors of human T cells that also affect B cells because of the T cell deficit or an additional B cell-intrinsic deficit. In this study, we report six patients from three unrelated families with biallelic loss-of-function mutations in RLTPR, the mouse orthologue of which is essential for CD28 signaling. The patients have cutaneous and pulmonary allergy, as well as a variety of bacterial and fungal infectious diseases, including invasive tuberculosis and mucocutaneous candidiasis. Proportions of circulating regulatory T cells and memory CD4+ T cells are reduced. Their CD4+ T cells do not respond to CD28 stimulation. Their CD4+ T cells exhibit a "Th2" cell bias ex vivo and when cultured in vitro, contrasting with the paucity of "Th1," "Th17," and T follicular helper cells. The patients also display few memory B cells and poor antibody responses. This B cell phenotype does not result solely from the T cell deficiency, as the patients' B cells fail to activate NF-κB upon B cell receptor (BCR) stimulation. Human RLTPR deficiency is a CID affecting at least the CD28-responsive pathway in T cells and the BCR-responsive pathway in B cells.


Subject(s)
Alleles , B-Lymphocytes/immunology , Microfilament Proteins/genetics , Mutation/genetics , T-Lymphocytes/immunology , Adolescent , Adult , Base Sequence , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Child , Child, Preschool , Dimerization , Female , HEK293 Cells , Humans , Immunologic Memory , Immunophenotyping , Leukocytes/pathology , Male , NF-kappa B/metabolism , Pedigree , Phenotype , Receptors, Antigen, B-Cell , Signal Transduction , Th17 Cells/immunology , Th2 Cells/immunology , Young Adult
17.
J Clin Immunol ; 36(7): 684-92, 2016 10.
Article in English | MEDLINE | ID: mdl-27473539

ABSTRACT

Hereditary defects in several genes have been shown to disturb the normal immune response to EBV and to give rise to severe EBV-induced lymphoproliferation in the recent years. Nevertheless, in many patients, the molecular basis of fatal EBV infection still remains unclear. The Fanconi anemia-associated protein 24 (FAAP24) plays a dual role in DNA repair. By association with FANCM as component of the FA core complex, it recruits the FA core complex to damaged DNA. Additionally, FAAP24 has been shown to evoke ATR-mediated checkpoint responses independently of the FA core complex. By whole exome sequencing, we identified a homozygous missense mutation in the FAAP24 gene (cC635T, pT212M) in two siblings of a consanguineous Turkish family who died from an EBV-associated lymphoproliferative disease after infection with a variant EBV strain, expressing a previously unknown EBNA2 allele.In order to analyze the functionality of the variant FAAP24 allele, we used herpes virus saimiri-transformed patient T cells to test endogenous cellular FAAP24 functions that are known to be important in DNA damage control. We saw an impaired FANCD2 monoubiquitination as well as delayed checkpoint responses, especially affecting CHK1 phosphorylation in patient samples in comparison to healthy controls. The phenotype of this FAAP24 mutation might have been further accelerated by an EBV strain that harbors an EBNA2 allele with enhanced activities compared to the prototype laboratory strain B95.8. This is the first report of an FAAP24 loss of function mutation found in human patients with EBV-associated lymphoproliferation.


Subject(s)
DNA-Binding Proteins/genetics , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/genetics , Mutation , Siblings , Amino Acid Substitution , Cell Cycle , Codon , Consanguinity , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins , Fatal Outcome , Female , Genotype , Homozygote , Humans , Lymphocyte Count , Lymphoproliferative Disorders/virology , Male , Pedigree , Phenotype , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Sister Chromatid Exchange , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Ubiquitination , Exome Sequencing
18.
Neuro Oncol ; 18(12): 1664-1672, 2016 12.
Article in English | MEDLINE | ID: mdl-27286796

ABSTRACT

BACKGROUND: Neurological decline during radio(chemo)therapy of the brain is often attributed to disease progression or side effects of radiotherapy. Diagnosis of opportunistic neurotropic infections such as cytomegalovirus (CMV) infections is uncommon, even though high-grade gliomas and some brain metastases are known to contain CMV particles. We prospectively examined the frequency of CMV encephalopathy during radiotherapy of the brain. METHODS: Fifty patients requiring whole-brain radiotherapy for brain metastases (n = 27) or local radio(chemo)therapy of the brain for high-grade gliomas (n = 23) were observed in the prospective observational GLIO-CMV-01 study. MRIs and blood samples were obtained before, halfway through, and at the end of radiotherapy. MRIs were screened for disease progression or increased intracranial pressure. Blood was tested for anti-CMV immunoglobulin (Ig)M, anti-CMV IgG, and CMV DNA. RESULTS: Thirty-two of 50 (64%) patients were positive for anti-CMV IgG before radio(chemo)therapy. Fifteen of those 32 (48%) developed viremia during or up to 28 days after treatment. Thirteen of those 15 (87%) required treatment for CMV-associated encephalopathy. MRIs were negative for disease progression, edema, or bleeding. None of the patients negative for anti-CMV IgG developed viremia, suggesting a reactivation rather than a primary infection.In the group at risk consisting of anti-CMV IgG+ patients, age >65 (P = .004) and the amount of dexamethasone taken during radio(chemo)therapy (P = .004) were associated with an increased risk for CMV-associated encephalopathy. One hundred and fifty days after the start of radio(chemo)therapy, survival was 74% (14/19) (no encephalopathy) versus 54% (7/13) (encephalopathy) (odds ratio, 0.42; 95% CI, 0.03-1.86; P = .25). CONCLUSION: CMV reactivation frequently causes encephalopathy during radio(chemo)therapy of the brain. The unexpected high incidence of this infection makes it highly clinically relevant for every treating physician.


Subject(s)
Brain Diseases/epidemiology , Brain Neoplasms/epidemiology , Brain Neoplasms/radiotherapy , Brain/virology , Cytomegalovirus Infections/epidemiology , Aged , Brain Diseases/etiology , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Chemoradiotherapy/adverse effects , Cytomegalovirus Infections/complications , Female , Humans , Male , Middle Aged , Opportunistic Infections/complications , Opportunistic Infections/epidemiology , Prospective Studies , Survival Analysis
19.
J Exp Med ; 212(10): 1641-62, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26304966

ABSTRACT

Autosomal recessive, complete TYK2 deficiency was previously described in a patient (P1) with intracellular bacterial and viral infections and features of hyper-IgE syndrome (HIES), including atopic dermatitis, high serum IgE levels, and staphylococcal abscesses. We identified seven other TYK2-deficient patients from five families and four different ethnic groups. These patients were homozygous for one of five null mutations, different from that seen in P1. They displayed mycobacterial and/or viral infections, but no HIES. All eight TYK2-deficient patients displayed impaired but not abolished cellular responses to (a) IL-12 and IFN-α/ß, accounting for mycobacterial and viral infections, respectively; (b) IL-23, with normal proportions of circulating IL-17(+) T cells, accounting for their apparent lack of mucocutaneous candidiasis; and (c) IL-10, with no overt clinical consequences, including a lack of inflammatory bowel disease. Cellular responses to IL-21, IL-27, IFN-γ, IL-28/29 (IFN-λ), and leukemia inhibitory factor (LIF) were normal. The leukocytes and fibroblasts of all seven newly identified TYK2-deficient patients, unlike those of P1, responded normally to IL-6, possibly accounting for the lack of HIES in these patients. The expression of exogenous wild-type TYK2 or the silencing of endogenous TYK2 did not rescue IL-6 hyporesponsiveness, suggesting that this phenotype was not a consequence of the TYK2 genotype. The core clinical phenotype of TYK2 deficiency is mycobacterial and/or viral infections, caused by impaired responses to IL-12 and IFN-α/ß. Moreover, impaired IL-6 responses and HIES do not appear to be intrinsic features of TYK2 deficiency in humans.


Subject(s)
Job Syndrome/etiology , TYK2 Kinase/deficiency , Adolescent , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Interferon-gamma/metabolism , Interleukin-10/pharmacology , Interleukin-12/metabolism , Interleukin-12/pharmacology , Interleukin-23/pharmacology , Interleukin-6/pharmacology , Job Syndrome/complications , Job Syndrome/genetics , Leukocytes/drug effects , Leukocytes/metabolism , Male , Mutation , Mycobacterium Infections/etiology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , TYK2 Kinase/genetics , TYK2 Kinase/metabolism , Virus Diseases/etiology , Young Adult
20.
Haematologica ; 100(9): 1189-98, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26113417

ABSTRACT

Autoimmune lymphoproliferative syndrome is frequently caused by mutations in genes involved in the Fas death receptor pathway, but for 20-30% of patients the genetic defect is unknown. We observed that treatment of healthy T cells with interleukin-12 induces upregulation of Fas ligand and Fas ligand-dependent apoptosis. Consistently, interleukin-12 could not induce apoptosis in Fas ligand-deficient T cells from patients with autoimmune lymphoproliferative syndrome. We hypothesized that defects in the interleukin-12 signaling pathway may cause a similar phenotype as that caused by mutations of the Fas ligand gene. To test this, we analyzed 20 patients with autoimmune lymphoproliferative syndrome of unknown cause by whole-exome sequencing. We identified a homozygous nonsense mutation (c.698G>A, p.R212*) in the interleukin-12/interleukin-23 receptor-component IL12RB1 in one of these patients. The mutation led to IL12RB1 protein truncation and loss of cell surface expression. Interleukin-12 and -23 signaling was completely abrogated as demonstrated by deficient STAT4 phosphorylation and interferon γ production. Interleukin-12-mediated expression of membrane-bound and soluble Fas ligand was lacking and basal expression was much lower than in healthy controls. The patient presented with the classical symptoms of autoimmune lymphoproliferative syndrome: chronic non-malignant, non-infectious lymphadenopathy, splenomegaly, hepatomegaly, elevated numbers of double-negative T cells, autoimmune cytopenias, and increased levels of vitamin B12 and interleukin-10. Sanger sequencing and whole-exome sequencing excluded the presence of germline or somatic mutations in genes known to be associated with the autoimmune lymphoproliferative syndrome. Our data suggest that deficient regulation of Fas ligand expression by regulators such as the interleukin-12 signaling pathway may be an alternative cause of autoimmune lymphoproliferative syndrome-like disease.


Subject(s)
Autoimmune Lymphoproliferative Syndrome/immunology , Codon, Nonsense , Fas Ligand Protein/immunology , Gene Expression Regulation/immunology , Receptors, Interleukin-12/immunology , Signal Transduction/immunology , Apoptosis/genetics , Apoptosis/immunology , Autoimmune Lymphoproliferative Syndrome/genetics , Caspase 10/genetics , Caspase 10/immunology , Caspase 8/genetics , Caspase 8/immunology , Cell Line, Transformed , Fas Ligand Protein/genetics , Female , Humans , Interleukin-12/genetics , Interleukin-12/immunology , Male , Receptors, Interleukin-12/genetics , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/immunology , Signal Transduction/genetics , fas Receptor/genetics , fas Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...