Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Med Genet A ; 194(6): e63562, 2024 06.
Article in English | MEDLINE | ID: mdl-38337186

ABSTRACT

Biallelic pathogenic variants in RMRP, the gene encoding the RNA component of RNase mitochondrial RNA processing enzyme complex, have been reported in individuals with cartilage hair hypoplasia (CHH). CHH is prevalent in Finnish and Amish populations due to a founder pathogenic variant, n.71A > G. Based on the manifestations in the Finnish and Amish individuals, the hallmarks of CHH are prenatal-onset growth failure, metaphyseal dysplasia, hair hypoplasia, immunodeficiency, and other extraskeletal manifestations. Herein, we report six Japanese individuals with CHH from four families. All probands presented with moderate short stature with mild metaphyseal dysplasia or brachydactyly. One of them had hair hypoplasia and the other immunodeficiency. By contrast, the affected siblings of two families showed only mild short stature. We also reviewed all previously reported 13 Japanese individuals. No n.71A > G allele was detected. The proportions of Japanese versus Finnish individuals were 0% versus 70% for birth length < -2.0 SD, 84% versus 100% for metaphyseal dysplasia and 26% versus 88% for hair hypoplasia. Milder manifestations in the Japanese individuals may be related to the difference of genotypes. The mildest form of CHH phenotypes is mild short stature without overt skeletal alteration or extraskeletal manifestation and can be termed "RMRP-related short stature".


Subject(s)
Hair , Osteochondrodysplasias , Adolescent , Child , Child, Preschool , Female , Humans , Male , Alleles , Dwarfism/genetics , Dwarfism/pathology , East Asian People , Genotype , Hair/abnormalities , Hair/pathology , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Hirschsprung Disease/diagnosis , Japan/epidemiology , Mutation/genetics , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Osteochondrodysplasias/congenital , Pedigree , Phenotype , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/pathology , RNA, Long Noncoding/genetics
2.
J Pediatr Hematol Oncol ; 46(7): e541-e543, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39196641

ABSTRACT

Encephalocraniocutaneous lipomatosis (ECCL) is a rare genetic condition with well-described skin, ocular, and central nervous system findings. Several case reports have been documented demonstrating the presence of low-grade gliomas in patients with ECCL and the association with certain FGFR1 mutations. We report on a case of diffuse low-grade glioma, mitogen activated protein kinase pathway altered in a patient with ECCL, who was found to have a distinct FGFR1 mutation.


Subject(s)
Lipomatosis , Neurocutaneous Syndromes , Humans , Neurocutaneous Syndromes/pathology , Neurocutaneous Syndromes/genetics , Neurocutaneous Syndromes/complications , Lipomatosis/pathology , Lipomatosis/genetics , Lipomatosis/complications , Receptor, Fibroblast Growth Factor, Type 1/genetics , Male , Mutation , Glioma/pathology , Glioma/genetics , Glioma/complications , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/complications , Eye Diseases/pathology , Eye Diseases/etiology , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/pathology , Primary Immunodeficiency Diseases/genetics , Female
3.
Blood ; 137(20): 2770-2784, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33512478

ABSTRACT

Dendritic cells (DCs) encompass several cell subsets that collaborate to initiate and regulate immune responses. Proper DC localization determines their function and requires the tightly controlled action of chemokine receptors. All DC subsets express CXCR4, but the genuine contribution of this receptor to their biology has been overlooked. We addressed this question using natural CXCR4 mutants resistant to CXCL12-induced desensitization and harboring a gain of function that cause the warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome (WS), a rare immunodeficiency associated with high susceptibility to the pathogenesis of human papillomavirus (HPV). We report a reduction in the number of circulating plasmacytoid DCs (pDCs) in WHIM patients, whereas that of conventional DCs is preserved. This pattern was reproduced in an original mouse model of WS, enabling us to show that the circulating pDC defect can be corrected upon CXCR4 blockade and that pDC differentiation and function are preserved, despite CXCR4 dysfunction. We further identified proper CXCR4 signaling as a critical checkpoint for Langerhans cell and DC migration from the skin to lymph nodes, with corollary alterations of their activation state and tissue inflammation in a model of HPV-induced dysplasia. Beyond providing new hypotheses to explain the susceptibility of WHIM patients to HPV pathogenesis, this study shows that proper CXCR4 signaling establishes a migration threshold that controls DC egress from CXCL12-containing environments and highlights the critical and subset-specific contribution of CXCR4 signal termination to DC biology.


Subject(s)
Dendritic Cells/physiology , Inflammation/pathology , Primary Immunodeficiency Diseases/physiopathology , Receptors, CXCR4/physiology , Warts/physiopathology , Alphapapillomavirus/genetics , Animals , Benzylamines/pharmacology , Cell Count , Cell Differentiation , Chemokine CXCL12/physiology , Chemotaxis , Cyclams/pharmacology , Dendritic Cells/classification , Epidermis/pathology , Female , Gene Knock-In Techniques , Genes, Viral , Humans , Inflammation/metabolism , Langerhans Cells/physiology , Lymphoid Tissue/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Organ Specificity , Parabiosis , Primary Immunodeficiency Diseases/blood , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/pathology , Recombinant Proteins/metabolism , Warts/blood , Warts/genetics , Warts/pathology
4.
Hum Mol Genet ; 29(19): 3197-3210, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32916696

ABSTRACT

The most distal 2 kb region in the majority of human subtelomeres contains CpG-rich promoters for TERRA, a long non-coding RNA. When the function of the de novo DNA methyltransferase DNMT3B is disrupted, as in ICF1 syndrome, subtelomeres are abnormally hypomethylated, subtelomeric heterochromatin acquires open chromatin characteristics, TERRA is highly expressed, and telomeres shorten rapidly. In this study, we explored whether the regulation of subtelomeric epigenetic characteristics by DNMT3B is conserved between humans and mice. Studying the DNA sequence of the distal 30 kb of the majority of murine q-arm subtelomeres indicated that these regions are relatively CpG-poor and do not contain TERRA promoters similar to those present in humans. Despite the lack of human-like TERRA promoters, we clearly detected TERRA expression originating from at least seven q-arm subtelomeres, and at higher levels in mouse pluripotent stem cells in comparison with mouse embryonic fibroblasts (MEFs). However, these differences in TERRA expression could not be explained by differential methylation of CpG islands present in the TERRA-expressing murine subtelomeres. To determine whether Dnmt3b regulates the expression of TERRA in mice, we characterized subtelomeric methylation and associated telomeric functions in cells derived from ICF1 model mice. Littermate-derived WT and ICF1 MEFs demonstrated no significant differences in subtelomeric DNA methylation, chromatin modifications, TERRA expression levels, telomere sister chromatid exchange or telomere length. We conclude that the epigenetic characteristics of murine subtelomeres differ substantially from their human counterparts and that TERRA transcription in mice is regulated by factors others than Dnmt3b.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Face/abnormalities , Fibroblasts/pathology , Primary Immunodeficiency Diseases/pathology , Telomere/physiology , Transcription Factors/metabolism , Animals , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/genetics , Face/pathology , Fibroblasts/metabolism , Humans , Mice , Primary Immunodeficiency Diseases/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription, Genetic , DNA Methyltransferase 3B
5.
Stem Cells ; 39(4): 389-402, 2021 04.
Article in English | MEDLINE | ID: mdl-33400834

ABSTRACT

Hematopoietic stem cells (HSCs) are the progenitor cells that give rise to the diverse repertoire of all immune cells. As they differentiate, HSCs yield a series of cell states that undergo gradual commitment to become mature blood cells. Studies of hematopoiesis in murine models have provided critical insights about the lineage relationships among stem cells, progenitors, and mature cells, and these have guided investigations of the molecular basis for these distinct developmental stages. Primary immune deficiencies are caused by inborn errors of immunity that result in immune dysfunction and subsequent susceptibility to severe and recurrent infection(s). Over the last decade there has been a dramatic increase in the number and depth of the molecular, cellular, and clinical characterization of such genetically defined causes of immune dysfunction. Patients harboring inborn errors of immunity thus represent a unique resource to improve our understanding of the multilayered and complex mechanisms underlying lymphocyte development in humans. These breakthrough discoveries not only enable significant advances in the diagnosis of such rare and complex conditions but also provide substantial improvement in the development of personalized treatments. Here, we will discuss the clinical, cellular, and molecular phenotypes, and treatments of selected inborn errors of immunity that impede, either intrinsically or extrinsically, the development of B- or T-cells at different stages.


Subject(s)
Gene Expression Regulation/immunology , Hematopoietic Stem Cells/immunology , Lymphopoiesis/immunology , Precision Medicine/methods , Primary Immunodeficiency Diseases/genetics , Animals , Cell Differentiation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Genetic Therapy/methods , Hematopoiesis/genetics , Hematopoiesis/immunology , Hematopoietic Stem Cells/cytology , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/immunology , Janus Kinase 3/genetics , Janus Kinase 3/immunology , Lymphocytes/immunology , Lymphocytes/pathology , Lymphopoiesis/genetics , Mice , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/immunology , Primary Immunodeficiency Diseases/pathology , Primary Immunodeficiency Diseases/therapy
6.
Am J Med Genet A ; 185(10): 2873-2877, 2021 10.
Article in English | MEDLINE | ID: mdl-34037310

ABSTRACT

Trichohepatoenteric syndrome (THES) is a very rare autosomal recessive genetic disorder, which is characterized by intractable diarrhea during infancy, dysmorphic features, immunodeficiency, and a failure to thrive. There are still significant difficulties for patients and clinicians in terms of the management of THES, even though its molecular basis has been uncovered in the last decade. In this article, we have presented two cases relating to siblings that have been diagnosed with the condition. Concerning one of the patients, we described a novel variation (c.2114 + 5G > A) in the TTC37 gene and a mild clinical course; meanwhile, the other one was clinically diagnosed with THES at 17 years of age, but they had seizures and died suddenly. These cases expand the spectrum of clinical findings in relation to THES.


Subject(s)
Carrier Proteins/genetics , Diarrhea, Infantile/genetics , Failure to Thrive/genetics , Fetal Growth Retardation/genetics , Hair Diseases/genetics , Malabsorption Syndromes/genetics , Microvilli/pathology , Mucolipidoses/genetics , Adolescent , Diarrhea, Infantile/complications , Diarrhea, Infantile/diagnosis , Diarrhea, Infantile/pathology , Facies , Failure to Thrive/complications , Failure to Thrive/diagnosis , Failure to Thrive/pathology , Female , Fetal Growth Retardation/diagnosis , Fetal Growth Retardation/pathology , Genetic Predisposition to Disease , Hair Diseases/complications , Hair Diseases/diagnosis , Hair Diseases/pathology , Humans , Infant , Malabsorption Syndromes/complications , Malabsorption Syndromes/diagnosis , Malabsorption Syndromes/pathology , Male , Microvilli/genetics , Mucolipidoses/complications , Mucolipidoses/diagnosis , Mucolipidoses/pathology , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/pathology , Siblings
7.
Am J Hematol ; 96(9): 1077-1086, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34000087

ABSTRACT

The differential diagnosis of marrow failure (MF) is crucial in the diagnostic work-up, since genetic forms require specific care. We retrospectively studied all patients with single/multi-lineage MF evaluated in a single-center to identify the type and incidence of underlying molecular defects. The diepoxybutane test was used to screen Fanconi Anemia. Other congenital MFs have been searched using Sanger and/or Next Generation Sequencing analysis, depending on the available tools over the years. Between 2009-2019, 97 patients (aged 0-32 years-median 5) with single-lineage (29%) or multilineage (68%) MF were evaluated. Fifty-three (54%) and 28 (29%) were diagnosed with acquired and congenital MF, respectively. The remaining 16 (17%), with trilinear (n=9) and monolinear (n=7) MF, were found to have an underlying primary immunodeficiency (PID) and showed clinical and biochemical signs of immune-dysregulation in 10/16 (62%) and in 14/16 (87%) of cases, respectively. Clinical signs were also found in 22/53 (41%) and 8/28 (28%) patients with idiopathic and classical cMF, respectively. Eight out of 16 PIDs patients were successfully transplanted, four received immunosuppression, two did not require treatment, and the remaining two died. We show that patients with single/multi-lineage MF may have underlying PIDs in a considerable number of cases and that MF may represent a relevant clinical sign in patients with PIDs, thus widening their clinical phenotype. An accurate immunological work-up should be performed in all patients with MF, and PID-related genes should be considered when screening MF in order to identify disorders that may receive targeted treatments and/or appropriate conditioning regimens before transplant.


Subject(s)
Bone Marrow Failure Disorders/genetics , Bone Marrow/pathology , Primary Immunodeficiency Diseases/genetics , Adolescent , Adult , Bone Marrow/metabolism , Bone Marrow Failure Disorders/pathology , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Primary Immunodeficiency Diseases/pathology , Retrospective Studies , Young Adult
8.
Nucleic Acids Res ; 47(16): 8388-8398, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31226215

ABSTRACT

ZBTB24, encoding a protein of the ZBTB family of transcriptional regulators, is one of four known genes-the other three being DNMT3B, CDCA7 and HELLS-that are mutated in immunodeficiency, centromeric instability and facial anomalies (ICF) syndrome, a genetic disorder characterized by DNA hypomethylation and antibody deficiency. The molecular mechanisms by which ZBTB24 regulates gene expression and the biological functions of ZBTB24 are poorly understood. Here, we identified a 12-bp consensus sequence [CT(G/T)CCAGGACCT] occupied by ZBTB24 in the mouse genome. The sequence is present at multiple loci, including the Cdca7 promoter region, and ZBTB24 binding is mostly associated with gene activation. Crystallography and DNA-binding data revealed that the last four of the eight zinc fingers (ZFs) (i.e. ZF5-8) in ZBTB24 confer specificity of DNA binding. Two ICF missense mutations have been identified in the ZBTB24 ZF domain, which alter zinc-binding cysteine residues. We demonstrated that the corresponding C382Y and C407G mutations in mouse ZBTB24 abolish specific DNA binding and fail to induce Cdca7 expression. Our analyses indicate and suggest a structural basis for the sequence specific recognition by a transcription factor centrally important for the pathogenesis of ICF syndrome.


Subject(s)
Cell Cycle Proteins/genetics , Face/abnormalities , Genome , Mutation, Missense , Nuclear Proteins/genetics , Primary Immunodeficiency Diseases/genetics , Repressor Proteins/chemistry , Transcription Factors/chemistry , Zinc Fingers/genetics , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Face/pathology , Gene Expression , Genetic Loci , Genetic Vectors , Humans , Mice , Models, Molecular , Nuclear Proteins/metabolism , Nucleotide Motifs , Primary Immunodeficiency Diseases/metabolism , Primary Immunodeficiency Diseases/pathology , Promoter Regions, Genetic , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Int J Mol Sci ; 22(3)2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33572538

ABSTRACT

Inborn errors of immunity (IEI) are a group of disorders that are mostly caused by genetic mutations affecting immune host defense and immune regulation. Although IEI present with a wide spectrum of clinical features, in about one third of them various degrees of gastrointestinal (GI) involvement have been described and for some IEI the GI manifestations represent the main and peculiar clinical feature. The microbiome plays critical roles in the education and function of the host's innate and adaptive immune system, and imbalances in microbiota-immunity interactions can contribute to intestinal pathogenesis. Microbial dysbiosis combined to the impairment of immunosurveillance and immune dysfunction in IEI, may favor mucosal permeability and lead to inflammation. Here we review how immune homeostasis between commensals and the host is established in the gut, and how these mechanisms can be disrupted in the context of primary immunodeficiencies. Additionally, we highlight key aspects of the first studies on gut microbiome in patients affected by IEI and discuss how gut microbiome could be harnessed as a therapeutic approach in these diseases.


Subject(s)
Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Host Microbial Interactions , Primary Immunodeficiency Diseases/microbiology , Adaptive Immunity , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Homeostasis , Humans , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/pathology
10.
Bull Exp Biol Med ; 171(3): 367-369, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34302206

ABSTRACT

The phenomenon of multiple primary malignant tumors (MPMT) is characterized by the presence of several primary neoplasms in the same patient. An experimental model of MPMT with one dominating tumor was developed. Female BALB/c nude mice received simultaneous subcutaneous inoculation of Guerin's carcinoma (5×105 tumor cells in 0.5 ml saline) and B16/F10 melanoma (0.5 ml suspension diluted 1:20 with saline). Control females received transplantation of either melanoma or carcinoma alone in the same doses and volumes. In animals with MPMT model, tumors appeared 3-fold faster than after isolated transplantation of melanoma or Guerin's carcinoma and were larger by 7.5 and 2.2 times, respectively; the survival of mice with MPMT was lower. Guerin's carcinoma in the MPMT model metastasized to melanoma and almost completely suppressed its growth. Thus, a MPMT model was created with carcinoma suppressing the malignant growth of melanoma.


Subject(s)
Carcinoma/pathology , Melanoma, Experimental/pathology , Neoplasms, Multiple Primary/pathology , Primary Immunodeficiency Diseases/pathology , Urologic Neoplasms/pathology , Animals , Carcinoma/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Injections, Subcutaneous , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplasms, Multiple Primary/immunology , Primary Immunodeficiency Diseases/immunology , Rats , Time Factors , Tumor Burden , Urologic Neoplasms/immunology
11.
Bull Exp Biol Med ; 171(6): 770-773, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34705181

ABSTRACT

The experimental model of synchronous multiple primary malignant tumors (MPMT) was created. B16/F10 melanoma (0.5 ml of suspension diluted 1:20 in saline) and sarcoma 45 (0.5 million tumor cells in 0.5 ml saline) were simultaneously subcutaneously inoculated to male BALB/c nude mice. In the model of synchronous MPMT, the tumors appeared faster by 2.4 times and had greater volumes: melanoma by 2.2 times and sarcoma by 3.2 times; melanoma metastasized into sarcoma in 71.4% cases; the survival of mice with MPMT was lower. The altered dynamics of malignant growth in the MPMT model is based on the mutual influence of tumors, which results in the exchange of "structural information".


Subject(s)
Lung Neoplasms/pathology , Melanoma, Experimental/pathology , Neoplasm Transplantation/methods , Neoplasms, Multiple Primary/pathology , Primary Immunodeficiency Diseases/pathology , Skin Neoplasms/pathology , Animals , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/immunology , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Rats , Skin Neoplasms/genetics , Skin Neoplasms/immunology
12.
Curr Opin Hematol ; 27(1): 11-17, 2020 01.
Article in English | MEDLINE | ID: mdl-31652152

ABSTRACT

PURPOSE OF REVIEW: WHIM syndrome (warts, hypogammaglobulinemia, immunodeficiency, myelokathexis, or WHIMs) is a very rare autosomal dominant immunodeficiency disorder attributable to mutations in CXCR4. We reviewed clinical manifestations in 24 patients in 9 families to expand understanding of this syndrome. RECENT FINDINGS: Warts, cellulitis and respiratory infections are common in patients with WHIMs. Less commonly these patients have congenital heart disease, human papilloma virus-associated malignancies (cervical and vulvular) and lymphomas. Hearing loss because of recurrent otitis media is another important complication. Treatment with granulocyte colony-stimulating factor is controversial; this review indicates that it is effective to prevent and treat infections based upon long-term observations of patients enrolled in the Severe Chronic Neutropenia International Registry. Understanding the natural history and diversity of this syndrome are important for ongoing clinical trials of novel agents to treat WHIMs. SUMMARY: WHIM syndrome has diverse manifestations; some features occur consistently in almost all patients, for example, neutropenia, lymphocytopenia and mild hypogammaglobulinemia. However, the clinical consequences are quite variable across patient cohorts and within families. Each complication is important as a cause for morbidity and a source for patient and family concerns.


Subject(s)
Agammaglobulinemia , Family , Mutation , Primary Immunodeficiency Diseases , Receptors, CXCR4/genetics , Registries , Warts , Agammaglobulinemia/diagnosis , Agammaglobulinemia/genetics , Agammaglobulinemia/pathology , Agammaglobulinemia/therapy , Female , Humans , Male , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/pathology , Primary Immunodeficiency Diseases/therapy , Risk Factors , Warts/diagnosis , Warts/genetics , Warts/pathology , Warts/therapy
13.
Hum Genet ; 139(6-7): 745-757, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32067110

ABSTRACT

Primary immunodeficiencies (PIDs) comprise a diverse group of over 400 genetic disorders that result in clinically apparent immune dysfunction. Although PIDs are classically considered as Mendelian disorders with complete penetrance, we now understand that absent or partial clinical disease is often noted in individuals harboring disease-causing genotypes. Despite the frequency of incomplete penetrance in PID, no conceptual framework exists to categorize and explain these occurrences. Here, by reviewing decades of reports on incomplete penetrance in PID we identify four recurrent themes of incomplete penetrance, namely genotype quality, (epi)genetic modification, environmental influence, and mosaicism. For each of these principles, we review what is known, underscore what remains unknown, and propose future experimental approaches to fill the gaps in our understanding. Although the content herein relates specifically to inborn errors of immunity, the concepts are generalizable across genetic diseases.


Subject(s)
Epigenesis, Genetic , Gene-Environment Interaction , Genetic Predisposition to Disease , Genetic Variation , Penetrance , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/pathology , Humans , Mosaicism , Primary Immunodeficiency Diseases/immunology
14.
Hum Genet ; 139(6-7): 783-794, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32419033

ABSTRACT

Anti-cytokine autoantibodies may cause immunodeficiency and have been recently recognized as 'autoimmune phenocopies of primary immunodeficiencies' and are found in particular, but not exclusively in adult patients. By blocking the cytokine's biological function, patients with anti-cytokine autoantibodies may present with a similar clinical phenotype as the related inborn genetic disorders. So far, autoantibodies to interferon (IFN)-γ, GM-CSF, to a group of TH-17 cytokines and to IL-6 have been found to be causative or closely associated with susceptibility to infection. This review compares infectious diseases associated with anti-cytokine autoantibodies with primary immunodeficiencies affecting similar cytokines or related pathways.


Subject(s)
Autoantibodies/immunology , Cytokines/immunology , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/pathology , Autoantibodies/blood , Humans , Phenotype , Primary Immunodeficiency Diseases/blood
15.
Clin Genet ; 98(3): 231-239, 2020 09.
Article in English | MEDLINE | ID: mdl-32441320

ABSTRACT

Primary immune deficiencies are usually attributed to genetic defects and, therefore, frequently referred to as inborn errors of immunity (IEI). We subjected the genomic DNA of 333 patients with clinical signs of IEI to next generation sequencing (NGS) analysis of 344 immunity-related genes and, in some instances, additional genetic techniques. Genetic causes of the disease were identified in 69/333 (21%) of subjects, including 11/18 (61%) of children with syndrome-associated IEIs, 45/202 (22%) of nonsyndromic patients with Jeffrey Modell Foundation (JMF) warning signs, 9/56 (16%) of subjects with periodic fever, 3/30 (10%) of cases of autoimmune cytopenia, 1/21 (5%) of patients with unusually severe infections and 0/6 (0%) of individuals with isolated elevation of IgE level. There were unusual clinical observations: twins with severe immunodeficiency carried a de novo CHARGE syndrome-associated SEMA3E c.2108C>T (p.S703L) allele; however, they lacked clinical features of CHARGE syndrome. Additionally, there were genetically proven instances of Netherton syndrome, Х-linked agammaglobulinemia, severe combined immune deficiency (SCID), IPEX and APECED syndromes, among others. Some patients carried recurrent pathogenic alleles, such as AIRE c.769C>T (p.R257*), NBN c.657del5, DCLRE1C c.103C>G (p.H35D), NLRP12 c.1054C>T (p.R352C) and c.910C>T (p.H304Y). NGS is a powerful tool for high-throughput examination of patients with malfunction of immunity.


Subject(s)
Agammaglobulinemia/genetics , CHARGE Syndrome/genetics , Genetic Diseases, X-Linked/genetics , Primary Immunodeficiency Diseases/genetics , Severe Combined Immunodeficiency/genetics , Adolescent , Agammaglobulinemia/immunology , Agammaglobulinemia/pathology , CHARGE Syndrome/immunology , CHARGE Syndrome/pathology , Child , Child, Preschool , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Endonucleases/deficiency , Endonucleases/genetics , Endonucleases/immunology , Female , Genetic Diseases, X-Linked/immunology , Genetic Diseases, X-Linked/pathology , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/pathology , Russia/epidemiology , Semaphorins/genetics , Semaphorins/immunology , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/pathology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/immunology , AIRE Protein
16.
Scand J Immunol ; 92(4): e12913, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32506568

ABSTRACT

Cartilage-hair hypoplasia (CHH) is an autosomal recessive syndromic immunodeficiency with skeletal dysplasia, short stature, hypotrichosis, variable degree of immune dysfunction and increased incidence of anaemia, Hirschsprung disease and malignancy. CHH is caused by variants in the RMRP gene, encoding the untranslated RNA molecule of the mitochondrial RNA-processing endoribonuclease, which participates in for example cell cycle regulation and telomere maintenance. Recent studies have expanded our understanding of the complex pathogenesis of CHH. Immune dysfunction has a major impact on clinical course and prognosis. Clinical features of immune dysfunction are highly variable, progressive and include infections, lung disease, immune dysregulation and malignancy. Mortality is increased compared with the general population, due to infections, malignancy and pulmonary disease. Several risk factors for early mortality have been reported in the Finnish CHH cohort and can be used to guide management. Newborn screening for severe combined immunodeficiency can possibly be of prognostic value in CHH. Regular follow-up by a multidisciplinary team should be implemented to address immune dysfunction in all patients with CHH, also in asymptomatic cases. Haematopoietic stem cell transplantation can cure immune dysfunction, but its benefits in mildly symptomatic patients with CHH remain debatable. Further research is needed to understand the mechanisms behind the variability of clinical features, to search for potential molecular treatment targets, to examine and validate risk factors for early mortality outside the Finnish CHH cohort and to develop management guidelines. This review focuses on the pathogenesis, clinical course and management of CHH.


Subject(s)
Hair/abnormalities , Hirschsprung Disease/immunology , Osteochondrodysplasias/congenital , Primary Immunodeficiency Diseases/immunology , Animals , Hair/immunology , Hair/pathology , Hair/physiopathology , Hirschsprung Disease/pathology , Hirschsprung Disease/physiopathology , Humans , Osteochondrodysplasias/immunology , Osteochondrodysplasias/pathology , Osteochondrodysplasias/physiopathology , Primary Immunodeficiency Diseases/pathology , Primary Immunodeficiency Diseases/physiopathology
17.
Am J Med Genet A ; 182(9): 2168-2174, 2020 09.
Article in English | MEDLINE | ID: mdl-32681719

ABSTRACT

A family with three affected males and a second family with a single affected male with intellectual disability, microcephaly, ophthalmoplegia, deafness, and Involuntary limb movements were reported by Schimke and Associates in 1984. The affected males with Schimke X-linked intellectual disability (XLID) syndrome (OMIM# 312840) had a similar facial appearance with deep-set eyes, downslanting palpebral fissures, hypotelorism, narrow nose and alae nasi, cupped ears and spacing of the teeth. Two mothers had mild hearing loss but no other manifestations of the disorder. The authors considered the disorder to be distinctive and likely X-linked. Whole genome sequencing in the single affected male available and the three carrier females from one of the families with Schimke XLID syndrome identified a 2 bp deletion in the BCAP31 gene. During the past decade, pathogenic alterations of the BCAP31 gene have been associated with deafness, dystonia, and central hypomyelination, an XLID condition given the eponym DDCH syndrome. A comparison of clinical findings in Schimke XLID syndrome and DDCH syndrome shows them to be the same clinical entity. The BCAP31 protein functions in endoplasmic reticulum-associated degradation to promote ubiquitination and destruction of misfolded proteins.


Subject(s)
Arteriosclerosis/pathology , Gene Deletion , Membrane Proteins/genetics , Mental Retardation, X-Linked/pathology , Mutation , Nephrotic Syndrome/pathology , Osteochondrodysplasias/pathology , Phenotype , Primary Immunodeficiency Diseases/pathology , Pulmonary Embolism/pathology , Adolescent , Adult , Arteriosclerosis/genetics , Child, Preschool , Female , Genetic Association Studies , Humans , Male , Mental Retardation, X-Linked/genetics , Nephrotic Syndrome/genetics , Osteochondrodysplasias/genetics , Pedigree , Primary Immunodeficiency Diseases/genetics , Pulmonary Embolism/genetics , Syndrome , Young Adult
18.
Pediatr Blood Cancer ; 67(2): e28091, 2020 02.
Article in English | MEDLINE | ID: mdl-31736244

ABSTRACT

BACKGROUND: Regarding the difficulties in recognition and management of the malignancies in primary immune deficiencies (PIDs), we aimed to present the types, risk factors, treatment options, and prognosis of the cancers in this specific group. METHODS: Seventeen patients with PID who developed malignancies or malignant-like diseases were evaluated for demographics, clinical features, treatment, toxicity, and prognosis. RESULTS: The median age of malignancy was 12.2 years (range, 2.2-26). Lymphoma was the most frequent malignancy (n = 7), followed by adenocarcinoma (n = 3), squamous cell carcinoma (n = 2), cholangiocarcinoma (n = 1), Wilms tumor (n = 1), and acute myeloid leukemia (n = 1). Nonneoplastic lymphoproliferation mimicking lymphoma was observed in five patients. The total overall survival (OS) was 62.5% ± 12.1%. The OS for lymphoma was 62.2% ± 17.1% and found to be inferior to non-PID patients with lymphoma (P = 0.001). CONCLUSION: In patients with PIDs, malignancy may occur and negatively affect the OS. The diagnosis can be challenging in the presence of nonneoplastic lymphoproliferative disease or bone marrow abnormalities. Awareness of susceptibility to malignant transformation and early diagnosis with multidisciplinary approach can save the patients' lives.


Subject(s)
Lymphoid Tissue/pathology , Lymphoma/classification , Lymphoma/diagnosis , Primary Immunodeficiency Diseases/complications , Adolescent , Adult , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Lymphoma/etiology , Lymphoma/therapy , Male , Primary Immunodeficiency Diseases/pathology , Prognosis , Survival Rate , Young Adult
19.
Exp Cell Res ; 384(1): 111588, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31473209

ABSTRACT

Given the critical role of the tumor microenvironment in PCa progression, we aimed to assess the prognostic effect of tumor infiltrating M2 macrophages (TIMMs) on biochemical recurrence (BCR) in patients with localized prostate cancer (PCa) after radical prostatectomy. A total of 127 localized PCa patients from GSE116918, 268 patients from TCGA database and 77 patients from GSE70770 were enrolled in our study. TIMMs were evaluated by the CIBERSORT method. Patients with high TIMMs had a significantly poorer recurrence free survival (RFS) (P = 0.017, P = 0.0063 and P = 0.001) in the three sets. In the multivariate analysis, the presence of high TIMMs (HR = 3.026, P = 0.023; HR = 2.679, P = 0.017; HR = 2.648, P = 0.005) was identified as an independent prognostic factor for RFS in the three sets. Harrell's Concordance index (C-index) increased in all three sets after combining TIMMs with traditional risk factors (PSA, clinical stage(T) and Gleason score). Gene Set Enrichment Analysis showed that T cell receptor signaling pathway, B cell receptor signaling pathway and primary immunodeficiency were significantly enriched in the low TIMMs group. TIMMs could serve as an independent prognostic factor for BCR in localized PCa patients after RP. Incorporation of TIMMs into traditional risk classification might further stratify patients with different prognosis.


Subject(s)
Macrophages/pathology , Neoplasm Recurrence, Local/pathology , Prostate/pathology , Prostatic Neoplasms/pathology , Aged , Disease Progression , Disease-Free Survival , Humans , Male , Neoplasm Grading/methods , Primary Immunodeficiency Diseases/pathology , Prognosis , Prostate/surgery , Prostatectomy/methods , Prostatic Neoplasms/surgery , Risk Factors , Signal Transduction/physiology , Tumor Microenvironment/physiology
20.
J Pediatr Hematol Oncol ; 42(2): 156-159, 2020 03.
Article in English | MEDLINE | ID: mdl-31033788

ABSTRACT

Gain of function mutations in the p110δ catalytic subunit of the phosphatidylinositol-3-OH kinase (PIK3CD) classified as activated phosphoinositide 3-kinase delta syndrome (APDS) are the cause of a primary immunodeficiency characterized by recurrent sinopulmonary infections, and lymphoproliferation. Previously, autoimmunity and Epstein-Barr virus-related B-cell lymphoma have been documented for patients with APDS; here, we present a case that extends the picture, as the patient shows the full diagnostic criteria of hemophagocytic lymphohistiocytosis at 6 months of age. He experienced Hodgkin lymphoma as a 2.5-year-old baby. Next-generation sequencing returned a de novo heterozygous missense variant in PIK3CD (LRG_191t1: c.3061G>A; p.Glu1021Lys), confirming the primary immunodeficiency. After 2 courses of ifosfamide, cisplatin, and etoposide combined with brentuximab, the patient successfully underwent allogeneic hematopoietic stem cell transplantation from his HLA full matched sister, and he has been well for 18 months after that. The hematologist treating Hodgkin lymphoma and/or hemophagocytic lymphohistiocytosis should be vigilant about the possible underlying immune deficiency, and they should consider APDS in their differential diagnosis.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Hodgkin Disease/pathology , Lymphohistiocytosis, Hemophagocytic/pathology , Mutation , Primary Immunodeficiency Diseases/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy , Hematopoietic Stem Cell Transplantation , Hodgkin Disease/complications , Hodgkin Disease/genetics , Hodgkin Disease/therapy , Humans , Infant , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/therapy , Male , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/therapy , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL