Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Allergy Clin Immunol ; 153(5): 1392-1405, 2024 May.
Article in English | MEDLINE | ID: mdl-38280573

ABSTRACT

BACKGROUND: Ataxia telangiectasia (AT) is characterized by cerebellar ataxia, telangiectasia, immunodeficiency, and increased cancer susceptibility and is caused by mutations in the ataxia telangiectasia mutated (ATM) gene. The immunodeficiency comprises predominantly immunoglobulin deficiency, mainly IgA and IgG2, with a variable severity. So far, the exact mechanisms underlying the immunoglobulin deficiency, especially the variable severity, remain unelucidated. OBJECTIVE: We characterized the clinical impact of immunoglobulin deficiencies in AT and elucidated their mechanisms in AT. METHODS: We analyzed long-term immunoglobulin levels, immunophenotyping, and survival time in our cohort (n = 87, median age 16 years; maximum 64 years). Somatic hypermutation and class-switch junctions in B cells were analyzed by next-generation sequencing. Furthermore, an in vitro class-switching induction assay was performed, followed by RNA sequencing, to assess the effect of ATM inhibition. RESULTS: Only the hyper-IgM AT phenotype significantly worsened survival time, while IgA or IgG2 deficiencies did not. The immunoglobulin levels showed predominantly decreased IgG2 and IgA. Moreover, flow cytometric analysis demonstrated reduced naive B and T lymphocytes and a deficiency of class-switched IgG2 and IgA memory B cells. Somatic hypermutation frequencies were lowered in IgA- and IgG2-deficient patients, indicating hampered germinal center reaction. In addition, the microhomology of switch junctions was elongated, suggesting alternative end joining during class-switch DNA repair. The in vitro class switching and proliferation were negatively affected by ATM inhibition. RNA sequencing analysis showed that ATM inhibitor influenced expression of germinal center reaction genes. CONCLUSION: Immunoglobulin deficiency in AT is caused by disturbed development of class-switched memory B cells. ATM deficiency affects both germinal center reaction and choice of DNA-repair pathway in class switching.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Ataxia Telangiectasia , B-Lymphocytes , Immunoglobulin Class Switching , Humans , Ataxia Telangiectasia/immunology , Ataxia Telangiectasia/genetics , Adult , Adolescent , Male , Female , Middle Aged , Child , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , B-Lymphocytes/immunology , Young Adult , Aged , Somatic Hypermutation, Immunoglobulin , Child, Preschool , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood
2.
Hum Genet ; 143(1): 19-33, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37994973

ABSTRACT

CD58 plays roles in cell adhesion and co-stimulation with antigen presentation from major histocompatibility complex class II on antigen-presenting cells to T-cell antigen receptors on naïve T cells. CD58 reportedly contributes to the development of various human autoimmune diseases. Recently, genome-wide association studies (GWASs) identified CD58 as a susceptibility locus for autoimmune diseases such as systemic lupus erythematosus (SLE), multiple sclerosis (MS), and primary biliary cholangitis (PBC). However, the primary functional variant and molecular mechanisms of susceptibility to autoimmune diseases in the CD58 locus were not clarified. Here, rs10924104, located in the ZNF35-binding motif within the gene expression regulatory motif, was identified as the primary functional variant for SLE, MS, and PBC among genetic variants showing stronger linkage disequilibrium (LD) with GWAS-lead variants in the CD58 locus. Expression-quantitative trait locus (e-QTL) data for each distinct blood cell type and in vitro functional analysis using the CRISPR/Cas9 system corroborated the functional role of rs10924104 in the upregulation of CD58 transcription by the disease-risk allele. Additionally, the strength of disease susceptibility observed in the CD58 locus could be accounted for by the strength of LD between rs10924104 and each GWAS-lead variant. In conclusion, the present study demonstrated for the first time the existence of a shared autoimmune disease-related primary functional variant (i.e., rs10924104) that regulates the expression of CD58. Clarifying the molecular mechanism of disease susceptibility derived from such a shared genetic background is important for understanding human autoimmune diseases and human immunology.


Subject(s)
Autoimmune Diseases , Lupus Erythematosus, Systemic , Multiple Sclerosis , Humans , Autoimmune Diseases/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Lupus Erythematosus, Systemic/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Polymorphism, Single Nucleotide , CD58 Antigens/metabolism
3.
Clin Genet ; 105(1): 72-76, 2024 01.
Article in English | MEDLINE | ID: mdl-37526414

ABSTRACT

KDM4B (MIM*609765, NM_015015.3, formerly JMJD2B) encodes a histone demethylase and regulates gene expression via demethylation, mainly of H3K9 tri-methylation. Heterozygous KDM4B loss-of-function variants cause autosomal dominant intellectual developmental disorder 65 (MIM#619320), which is characterized by global developmental delay, intellectual disability, language and gross motor delays, structural brain anomalies, characteristic facial features, and clinodactyly. Although the majority of reported patients have de novo pathogenic variants, some patients inherit pathogenic variants from affected parents. To our knowledge, only 23 patients with heterozygous KDM4B variants have been reported to date, and there are no reports of patients with biallelic KDM4B pathogenic variants. Herein, we report a female patient with a biallelic KDM4B frameshift variant (NM_015015.3: c.1384_1394delinsGGG, p.(Leu462Glyfs*43)) located at exon 12 of 23 protein-coding exons, which is thought to be subject to nonsense-mediated mRNA decay and no protein production. She presented developmental and language delays and a hypotonic and characteristic face. The patient's phenotype was more obvious than that of her mother, who is heterozygous for the same variant. Although declining birth rate (embryonic lethality in male mice) in homozygous knockout mice has been demonstrated, our report suggests that homozygous KDM4B frameshift variants can be viable in humans at least female.


Subject(s)
Intellectual Disability , Language Development Disorders , Humans , Male , Female , Animals , Mice , Frameshift Mutation/genetics , Exons , Phenotype , Intellectual Disability/genetics , Intellectual Disability/pathology , Language Development Disorders/genetics , Jumonji Domain-Containing Histone Demethylases/genetics
4.
Hum Genomics ; 16(1): 46, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36271469

ABSTRACT

BACKGROUND: Ligation of CD28 with ligands such as CD80 or CD86 provides a critical second signal alongside antigen presentation by class II major histocompatibility complex expressed on antigen-presenting cells through the T cell antigen receptor for naïve T cell activation. A number of studies suggested that CD28 plays an important role in the pathogenesis of various human diseases. Recent genome-wide association studies (GWASs) identified CD28 as a susceptibility locus for lymphocyte and eosinophil counts, multiple sclerosis, ulcerative colitis, celiac disease, rheumatoid arthritis, asthma, and primary biliary cholangitis. However, the primary functional variant and molecular mechanisms of disease susceptibility in this locus remain to be elucidated. This study aimed to identify the primary functional variant from thousands of genetic variants in the CD28 locus and elucidate its functional effect on the CD28 molecule. RESULTS: Among the genetic variants exhibiting stronger linkage disequilibrium (LD) with all GWAS-lead variants in the CD28 locus, rs2013278, located in the Rbfox binding motif related to splicing regulation, was identified as a primary functional variant related to multiple immunological traits. Relative endogenous expression levels of CD28 splicing isoforms (CD28i and CD28Δex2) compared with full-length CD28 in allele knock-in cell lines generated using CRISPR/Cas9 were directly regulated by rs2013278 (P < 0.05). Although full-length CD28 protein expressed on Jurkat T cells showed higher binding affinity for CD80/CD86, both CD28i and CD28Δex2 encoded loss-of-function isoforms. CONCLUSION: The present study demonstrated for the first time that CD28 has a shared disease-related primary functional variant (i.e., rs2013278) that regulates the CD28 alternative splicing that generates loss-of-function isoforms. They reduce disease risk by inducing anergy of effector T cells that over-react to autoantigens and allergens.


Subject(s)
CD28 Antigens , Genome-Wide Association Study , Humans , CD28 Antigens/genetics , CD28 Antigens/metabolism , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , Protein Isoforms/genetics , Autoantigens
6.
Horm Res Paediatr ; 94(11-12): 448-455, 2021.
Article in English | MEDLINE | ID: mdl-34706368

ABSTRACT

Coats plus syndrome is an autosomal recessive multisystemic and pleiotropic disorder affecting the eyes, brain, bone, and gastrointestinal tract, usually caused by compound heterozygous variants of the conserved telomere maintenance component 1 gene (CTC1), involved in telomere homeostasis and replication. So far, most reported patients are compound heterozygous for a truncating mutation and a missense variant. The phenotype is believed to result from telomere dysfunction, with accumulation of DNA damage, cellular senescence, and stem cell depletion. Here, we report a 23-year-old female with prenatal and postnatal growth retardation, microcephaly, osteopenia, recurrent fractures, intracranial calcification, leukodystrophy, parenchymal brain cysts, bicuspid aortic valve, and primary ovarian failure. She carries a previously reported maternally inherited pathogenic variant in exon 5 (c.724_727del, p.(Lys242Leufs*41)) and a novel, paternally inherited splice site variant (c.1617+5G>T; p.(Lys480Asnfs*17)) in intron 9. CTC1 transcript analysis showed that the latter resulted in skipping of exon 9. A trace of transcripts was normally spliced resulting in the presence of a low level of wild-type CTC1 transcripts. We speculate that ovarian failure is caused by telomere shortening or chromosome cohesion failure in oocytes and granulosa cells, with early decrease in follicular reserve. This is the first patient carrying 2 truncating CTC1 variants and the first presenting primary ovarian failure.


Subject(s)
Calcinosis , Central Nervous System Cysts , Leukoencephalopathies , Ataxia/genetics , Ataxia/pathology , Brain Neoplasms , Calcinosis/genetics , Central Nervous System Cysts/genetics , Central Nervous System Cysts/pathology , Female , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Muscle Spasticity , Mutation , Retinal Diseases , Seizures , Telomere-Binding Proteins/genetics
7.
Trends Immunol ; 42(4): 350-365, 2021 04.
Article in English | MEDLINE | ID: mdl-33663955

ABSTRACT

ATM is often dubbed the master regulator of the DNA double stranded break (DSB) response. Since proper induction and repair of DNA DSBs forms the core of immunological diversity, it is surprising that patients with ataxia telangiectasia generally have a mild immunodeficiency in contrast to other DSB repair syndromes. In this review, we address this discrepancy by delving into the functions of ATM in DSB repair and cell cycle control and translate these to adaptive immunity. We conclude that ATM, despite its myriad functions, is not an absolute requirement for acquiring sufficient levels of immunological diversity to prevent severe viral and opportunistic infections. There is, however, a more clinically pronounced antibody deficiency in ataxia telangiectasia due to disturbed class switch recombination.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Adaptive Immunity , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Immunoglobulin Class Switching
8.
Pediatr Neurol ; 113: 26-32, 2020 12.
Article in English | MEDLINE | ID: mdl-32980744

ABSTRACT

BACKGROUND: We aimed to expand the number of currently known pathogenic PNKP mutations, to study the phenotypic spectrum, including radiological characteristics and genotype-phenotype correlations, and to assess whether immunodeficiency and increased cancer risk are part of the DNA repair disorder caused by mutations in the PNKP gene. METHODS: We evaluated nine patients with PNKP mutations. A neurological history and examination was obtained. All patients had undergone neuroimaging and genetic testing as part of the prior diagnostic process. Laboratory measurements included potential biomarkers, and, in the context of a DNA repair disorder, we performed a detailed immunologic evaluation, including B cell repertoire analysis. RESULTS: We identified three new mutations in the PNKP gene and confirm the phenotypic spectrum of PNKP-associated disease, ranging from microcephaly, seizures, and developmental delay to ataxia with oculomotor apraxia type 4. Irrespective of the phenotype, alpha-fetoprotein is a biochemical marker and increases with age and progression of the disease. On neuroimaging, (progressive) cerebellar atrophy was a universal feature. No clinical signs of immunodeficiency were present, and immunologic assessment was unremarkable. One patient developed cancer, but this was attributed to a concurrent von Hippel-Lindau mutation. CONCLUSIONS: Immunodeficiency and cancer predisposition do not appear to be part of PNKP-associated disease, contrasting many other DNA repair disorders. Furthermore, our study illustrates that the previously described syndromes microcephaly, seizures, and developmental delay, and ataxia with oculomotor apraxia type 4, represent the extremes of an overlapping spectrum of disease. Cerebellar atrophy and elevated serum alpha-fetoprotein levels are early diagnostic findings across the entire phenotypical spectrum.


Subject(s)
DNA Repair Enzymes/genetics , Immunologic Deficiency Syndromes/epidemiology , Microcephaly/genetics , Mutation/genetics , Neoplasms/epidemiology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Spinocerebellar Ataxias/congenital , Adolescent , Child , Child, Preschool , Cohort Studies , Developmental Disabilities/genetics , Female , Genetic Association Studies , Humans , Male , Netherlands , Phenotype , Seizures/genetics , Spinocerebellar Ataxias/genetics , Young Adult
9.
J Exp Med ; 217(11)2020 11 02.
Article in English | MEDLINE | ID: mdl-32865561

ABSTRACT

The autosomal recessive immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome is a genetically heterogeneous disorder. Despite the identification of the underlying gene defects, it is unclear how mutations in any of the four known ICF genes cause a primary immunodeficiency. Here we demonstrate that loss of ZBTB24 in B cells from mice and ICF2 patients affects nonhomologous end-joining (NHEJ) during immunoglobulin class-switch recombination and consequently impairs immunoglobulin production and isotype balance. Mechanistically, we found that ZBTB24 associates with poly(ADP-ribose) polymerase 1 (PARP1) and stimulates its auto-poly(ADP-ribosyl)ation. The zinc-finger in ZBTB24 binds PARP1-associated poly(ADP-ribose) chains and mediates the PARP1-dependent recruitment of ZBTB24 to DNA breaks. Moreover, through its association with poly(ADP-ribose) chains, ZBTB24 protects them from degradation by poly(ADP-ribose) glycohydrolase (PARG). This facilitates the poly(ADP-ribose)-dependent assembly of the LIG4/XRCC4 complex at DNA breaks, thereby promoting error-free NHEJ. Thus, we uncover ZBTB24 as a regulator of PARP1-dependent NHEJ and class-switch recombination, providing a molecular basis for the immunodeficiency in ICF2 syndrome.


Subject(s)
DNA End-Joining Repair/genetics , Face/abnormalities , Immunoglobulin Class Switching/genetics , Mutation , Primary Immunodeficiency Diseases/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , B-Lymphocytes/immunology , DNA Breaks , Face/pathology , HEK293 Cells , Humans , Immunoglobulin Switch Region , Mice , Poly (ADP-Ribose) Polymerase-1/metabolism , Primary Immunodeficiency Diseases/blood , Primary Immunodeficiency Diseases/pathology , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transfection
10.
G Ital Dermatol Venereol ; 155(5): 537-541, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32618442

ABSTRACT

Blau Syndrome, or early-onset sarcoidosis, is hereditary juvenile-onset systemic granulomatosis. Clinical symptoms appear before the age of four years and mainly affect the skin, joints, and eyes. The symptoms are progressive and cause severe complications, such as joint destruction and blindness. Although tumor necrosis factor alpha (TNFα) antagonists are effective for controlling some of the symptoms of Blau Syndrome, there is no specific curative treatment. Heterozygous mutations in nucleotide-binding oligomerization domain 2 (NOD2) were identified as the cause of Blau Syndrome onset. NOD2 is an intracellular pathogen recognition receptor, the ligand of which is muramyl dipeptide (MDP) found in bacterial cell walls. Upon binding to MDP, NOD2 activates the NF-κB pathway, which leads to upregulation of proinflammatory cytokines. However, the detailed molecular mechanisms by which disease associated NOD2 mutations lead to autoinflammation and granuloma formation are still unclear. To clarify the relationship between disease associated NOD2 mutations and the inflammatory response, we established induced pluripotent stem (iPS) cells from Blau Syndrome patients. Functional analyses using these iPS cells suggested that IFNγ is a critical mediator of the inflammatory manifestations in this disease. This experimental finding is supported by the clinical observation that bacillus Calmette-Guesrin (BCG) vaccination is sometimes associated with disease onset, since IFNγ is a major cytokine associated with BCG-mediated immune responses. Further investigation of NOD2 signaling and accumulation of clinical cases are essential to elucidate the mechanisms of Blau Syndrome and develop an effective treatment for patients.


Subject(s)
Arthritis/genetics , Mutation , Nod2 Signaling Adaptor Protein/genetics , Sarcoidosis/genetics , Synovitis/genetics , Uveitis/genetics , Arthritis/diagnosis , Humans , Sarcoidosis/diagnosis , Synovitis/diagnosis , Uveitis/diagnosis
11.
Sci Transl Med ; 12(551)2020 07 08.
Article in English | MEDLINE | ID: mdl-32641488

ABSTRACT

Atopic dermatitis (AD) is commonly associated with colonization by Staphylococcus aureus in the affected skin. To understand the role of S. aureus in the development of AD, we performed whole-genome sequencing of S. aureus strains isolated from the cheek skin of 268 Japanese infants 1 and 6 months after birth. About 45% of infants were colonized with S. aureus at 1 month regardless of AD outcome. In contrast, skin colonization by S. aureus at 6 months of age increased the risk of developing AD. Acquisition of dysfunctional mutations in the S. aureus Agr quorum-sensing (QS) system was primarily observed in strains from 6-month-old infants who did not develop AD. Expression of a functional Agr system in S. aureus was required for epidermal colonization and the induction of AD-like inflammation in mice. Thus, retention of functional S. aureus agr virulence during infancy is associated with pathogen skin colonization and the development of AD.


Subject(s)
Dermatitis, Atopic , Eczema , Animals , Mice , Skin , Staphylococcus/genetics , Staphylococcus aureus , Virulence
12.
J Allergy Clin Immunol ; 141(1): 339-349.e11, 2018 01.
Article in English | MEDLINE | ID: mdl-28587749

ABSTRACT

BACKGROUND: Blau syndrome, or early-onset sarcoidosis, is a juvenile-onset systemic granulomatosis associated with a mutation in nucleotide-binding oligomerization domain 2 (NOD2). The underlying mechanisms of Blau syndrome leading to autoinflammation are still unclear, and there is currently no effective specific treatment for Blau syndrome. OBJECTIVES: To elucidate the mechanisms of autoinflammation in patients with Blau syndrome, we sought to clarify the relation between disease-associated mutant NOD2 and the inflammatory response in human samples. METHODS: Blau syndrome-specific induced pluripotent stem cell (iPSC) lines were established. The disease-associated NOD2 mutation of iPSCs was corrected by using a CRISPR-Cas9 system to precisely evaluate the in vitro phenotype of iPSC-derived cells. We also introduced the same NOD2 mutation into a control iPSC line. These isogenic iPSCs were then differentiated into monocytic cell lineages, and the statuses of nuclear factor κB pathway and proinflammatory cytokine secretion were investigated. RESULTS: IFN-γ acted as a priming signal through upregulation of NOD2. In iPSC-derived macrophages with mutant NOD2, IFN-γ treatment induced ligand-independent nuclear factor κB activation and proinflammatory cytokine production. RNA sequencing analysis revealed distinct transcriptional profiles of mutant macrophages both before and after IFN-γ treatment. Patient-derived macrophages demonstrated a similar IFN-γ-dependent inflammatory response. CONCLUSIONS: Our data support the significance of ligand-independent autoinflammation in the pathophysiology of Blau syndrome. Our comprehensive isogenic disease-specific iPSC panel provides a useful platform for probing therapeutic and diagnostic clues for the treatment of patients with Blau syndrome.


Subject(s)
Arthritis/etiology , Arthritis/metabolism , Interferon-gamma/metabolism , Macrophages/metabolism , Pluripotent Stem Cells/metabolism , Synovitis/etiology , Synovitis/metabolism , Uveitis/etiology , Uveitis/metabolism , Cell Lineage/genetics , Cytokines/metabolism , DNA Mutational Analysis , Exons , Gene Targeting , Genetic Loci , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Inflammation Mediators/metabolism , Interferon-gamma/genetics , Ligands , Macrophages/immunology , Male , Mutation , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/genetics , Phenotype , Pluripotent Stem Cells/cytology , Sarcoidosis
SELECTION OF CITATIONS
SEARCH DETAIL
...