Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Nat Immunol ; 22(9): 1118-1126, 2021 09.
Article in English | MEDLINE | ID: mdl-34326534

ABSTRACT

Transcription factors specialized to limit the destructive potential of inflammatory immune cells remain ill-defined. We discovered loss-of-function variants in the X-linked ETS transcription factor gene ELF4 in multiple unrelated male patients with early onset mucosal autoinflammation and inflammatory bowel disease (IBD) characteristics, including fevers and ulcers that responded to interleukin-1 (IL-1), tumor necrosis factor or IL-12p40 blockade. Using cells from patients and newly generated mouse models, we uncovered ELF4-mutant macrophages having hyperinflammatory responses to a range of innate stimuli. In mouse macrophages, Elf4 both sustained the expression of anti-inflammatory genes, such as Il1rn, and limited the upregulation of inflammation amplifiers, including S100A8, Lcn2, Trem1 and neutrophil chemoattractants. Blockade of Trem1 reversed inflammation and intestine pathology after in vivo lipopolysaccharide challenge in mice carrying patient-derived variants in Elf4. Thus, ELF4 restrains inflammation and protects against mucosal disease, a discovery with broad translational relevance for human inflammatory disorders such as IBD.


Subject(s)
DNA-Binding Proteins/genetics , Hereditary Autoinflammatory Diseases/genetics , Inflammatory Bowel Diseases/genetics , Macrophages/immunology , Transcription Factors/genetics , Animals , Calgranulin A/metabolism , Female , Gene Expression Regulation/genetics , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/pathology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin 1 Receptor Antagonist Protein/immunology , Lipocalin-2/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th17 Cells/immunology , Transcription, Genetic/genetics , Triggering Receptor Expressed on Myeloid Cells-1/antagonists & inhibitors , Triggering Receptor Expressed on Myeloid Cells-1/metabolism
2.
Cell ; 159(7): 1563-77, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525875

ABSTRACT

The mechanism by which cells undergo death determines whether dying cells trigger inflammatory responses or remain immunologically silent. Mitochondria play a central role in the induction of cell death, as well as in immune signaling pathways. Here, we identify a mechanism by which mitochondria and downstream proapoptotic caspases regulate the activation of antiviral immunity. In the absence of active caspases, mitochondrial outer membrane permeabilization by Bax and Bak results in the expression of type I interferons (IFNs). This induction is mediated by mitochondrial DNA-dependent activation of the cGAS/STING pathway and results in the establishment of a potent state of viral resistance. Our results show that mitochondria have the capacity to simultaneously expose a cell-intrinsic inducer of the IFN response and to inactivate this response in a caspase-dependent manner. This mechanism provides a dual control, which determines whether mitochondria initiate an immunologically silent or a proinflammatory type of cell death.


Subject(s)
Apoptosis , Caspases/metabolism , Interferon Type I/metabolism , Signal Transduction , Animals , DNA, Mitochondrial/metabolism , Inflammation/immunology , Inflammation/metabolism , Interferon Type I/immunology , Mice , Mice, Knockout , Virus Diseases/immunology
3.
Neurobiol Dis ; 198: 106537, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38772452

ABSTRACT

Hereditary spastic paraplegia (HSP) comprises a large group of neurogenetic disorders characterized by progressive lower extremity spasticity. Neurological evaluation and genetic testing were completed in a Malian family with early-onset HSP. Three children with unaffected consanguineous parents presented with symptoms consistent with childhood-onset complicated HSP. Neurological evaluation found lower limb weakness, spasticity, dysarthria, seizures, and intellectual disability. Brain MRI showed corpus callosum thinning with cortical and spinal cord atrophy, and an EEG detected slow background in the index patient. Whole exome sequencing identified a homozygous missense variant in the adaptor protein (AP) complex 2 alpha-2 subunit (AP2A2) gene. Western blot analysis showed reduced levels of AP2A2 in patient-iPSC derived neuronal cells. Endocytosis of transferrin receptor (TfR) was decreased in patient-derived neurons. In addition, we observed increased axon initial segment length in patient-derived neurons. Xenopus tropicalis tadpoles with ap2a2 knockout showed cerebral edema and progressive seizures. Immunoprecipitation of the mutant human AP-2-appendage alpha-C construct showed defective binding to accessory proteins. We report AP2A2 as a novel genetic entity associated with HSP and provide functional data in patient-derived neuron cells and a frog model. These findings expand our understanding of the mechanism of HSP and improve the genetic diagnosis of this condition.


Subject(s)
Adaptor Protein Complex 2 , Endocytosis , Spastic Paraplegia, Hereditary , Animals , Child , Child, Preschool , Female , Humans , Male , Adaptor Protein Complex 2/genetics , Endocytosis/genetics , Endocytosis/physiology , Mutation/genetics , Mutation, Missense , Neurons/metabolism , Neurons/pathology , Pedigree , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/pathology , Xenopus
4.
J Clin Immunol ; 44(2): 44, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38231408

ABSTRACT

Defining monogenic drivers of autoinflammatory syndromes elucidates mechanisms of disease in patients with these inborn errors of immunity and can facilitate targeted therapeutic interventions. Here, we describe a cohort of patients with a Behçet's- and inflammatory bowel disease (IBD)-like disorder termed "deficiency in ELF4, X-linked" (DEX) affecting males with loss-of-function variants in the ELF4 transcription factor gene located on the X chromosome. An international cohort of fourteen DEX patients was assessed to identify unifying clinical manifestations and diagnostic criteria as well as collate findings informing therapeutic responses. DEX patients exhibit a heterogeneous clinical phenotype including weight loss, oral and gastrointestinal aphthous ulcers, fevers, skin inflammation, gastrointestinal symptoms, arthritis, arthralgia, and myalgia, with findings of increased inflammatory markers, anemia, neutrophilic leukocytosis, thrombocytosis, intermittently low natural killer and class-switched memory B cells, and increased inflammatory cytokines in the serum. Patients have been predominantly treated with anti-inflammatory agents, with the majority of DEX patients treated with biologics targeting TNFα.


Subject(s)
Arthritis , Behcet Syndrome , Biological Products , Inflammatory Bowel Diseases , Male , Humans , Behcet Syndrome/diagnosis , Behcet Syndrome/genetics , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/genetics , Arthralgia , DNA-Binding Proteins , Transcription Factors/genetics
5.
Genet Med ; 26(2): 101023, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37947183

ABSTRACT

PURPOSE: We sought to delineate a multisystem disorder caused by recessive cysteine-rich with epidermal growth factor-like domains 1 (CRELD1) gene variants. METHODS: The impact of CRELD1 variants was characterized through an international collaboration utilizing next-generation DNA sequencing, gene knockdown, and protein overexpression in Xenopus tropicalis, and in vitro analysis of patient immune cells. RESULTS: Biallelic variants in CRELD1 were found in 18 participants from 14 families. Affected individuals displayed an array of phenotypes involving developmental delay, early-onset epilepsy, and hypotonia, with about half demonstrating cardiac arrhythmias and some experiencing recurrent infections. Most harbored a frameshift in trans with a missense allele, with 1 recurrent variant, p.(Cys192Tyr), identified in 10 families. X tropicalis tadpoles with creld1 knockdown displayed developmental defects along with increased susceptibility to induced seizures compared with controls. Additionally, human CRELD1 harboring missense variants from affected individuals had reduced protein function, indicated by a diminished ability to induce craniofacial defects when overexpressed in X tropicalis. Finally, baseline analyses of peripheral blood mononuclear cells showed similar proportions of immune cell subtypes in patients compared with healthy donors. CONCLUSION: This patient cohort, combined with experimental data, provide evidence of a multisystem clinical syndrome mediated by recessive variants in CRELD1.


Subject(s)
Neurodevelopmental Disorders , Reinfection , Humans , Leukocytes, Mononuclear , Syndrome , Phenotype , Arrhythmias, Cardiac/genetics , Neurodevelopmental Disorders/genetics , Cell Adhesion Molecules/genetics , Extracellular Matrix Proteins/genetics
6.
Clin Genet ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38774940

ABSTRACT

Skeletal dysplasias are a heterogeneous group of disorders presenting mild to lethal defects. Several factors, such as genetic, prenatal, and postnatal environmental may contribute to reduced growth. Fourteen families of Pakistani origin, presenting the syndromic form of short stature either in the autosomal recessive or autosomal dominant manner were clinically and genetically investigated to uncover the underlying genetic etiology. Homozygosity mapping, whole exome sequencing, and Sanger sequencing were used to search for the disease-causing gene variants. In total, we have identified 13 sequence variants in 10 different genes. The variants in the HSPG2 and XRCC4 genes were not reported previously in the Pakistani population. This study will expand the mutation spectrum of the identified genes and will help in improved diagnosis of the syndromic form of short stature in the local population.

7.
Genet Med ; 25(8): 100856, 2023 08.
Article in English | MEDLINE | ID: mdl-37092537

ABSTRACT

PURPOSE: Dominant variants in the retinoic acid receptor beta (RARB) gene underlie a syndromic form of microphthalmia, known as MCOPS12, which is associated with other birth anomalies and global developmental delay with spasticity and/or dystonia. Here, we report 25 affected individuals with 17 novel pathogenic or likely pathogenic variants in RARB. This study aims to characterize the functional impact of these variants and describe the clinical spectrum of MCOPS12. METHODS: We used in vitro transcriptional assays and in silico structural analysis to assess the functional relevance of RARB variants in affecting the normal response to retinoids. RESULTS: We found that all RARB variants tested in our assays exhibited either a gain-of-function or a loss-of-function activity. Loss-of-function variants disrupted RARB function through a dominant-negative effect, possibly by disrupting ligand binding and/or coactivators' recruitment. By reviewing clinical data from 52 affected individuals, we found that disruption of RARB is associated with a more variable phenotype than initially suspected, with the absence in some individuals of cardinal features of MCOPS12, such as developmental eye anomaly or motor impairment. CONCLUSION: Our study indicates that pathogenic variants in RARB are functionally heterogeneous and associated with extensive clinical heterogeneity.


Subject(s)
Microphthalmos , Receptors, Retinoic Acid , Humans , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoids
8.
Hum Mol Genet ; 29(11): 1900-1921, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32196547

ABSTRACT

CTNND1 encodes the p120-catenin (p120) protein, which has a wide range of functions, including the maintenance of cell-cell junctions, regulation of the epithelial-mesenchymal transition and transcriptional signalling. Due to advances in next-generation sequencing, CTNND1 has been implicated in human diseases including cleft palate and blepharocheilodontic (BCD) syndrome albeit only recently. In this study, we identify eight novel protein-truncating variants, six de novo, in 13 participants from nine families presenting with craniofacial dysmorphisms including cleft palate and hypodontia, as well as congenital cardiac anomalies, limb dysmorphologies and neurodevelopmental disorders. Using conditional deletions in mice as well as CRISPR/Cas9 approaches to target CTNND1 in Xenopus, we identified a subset of phenotypes that can be linked to p120-catenin in epithelial integrity and turnover, and additional phenotypes that suggest mesenchymal roles of CTNND1. We propose that CTNND1 variants have a wider developmental role than previously described and that variations in this gene underlie not only cleft palate and BCD but may be expanded to a broader velocardiofacial-like syndrome.


Subject(s)
Catenins/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Craniofacial Abnormalities/genetics , Ectropion/genetics , Heart Defects, Congenital/genetics , Tooth Abnormalities/genetics , Adolescent , Adult , Animals , Anodontia/diagnostic imaging , Anodontia/genetics , Anodontia/physiopathology , Child , Child, Preschool , Cleft Lip/diagnostic imaging , Cleft Lip/physiopathology , Cleft Palate/diagnostic imaging , Cleft Palate/physiopathology , Craniofacial Abnormalities/diagnostic imaging , Craniofacial Abnormalities/physiopathology , Disease Models, Animal , Ectropion/diagnostic imaging , Ectropion/physiopathology , Female , Genetic Predisposition to Disease , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/physiopathology , Humans , Male , Mice , Tooth Abnormalities/diagnostic imaging , Tooth Abnormalities/physiopathology , Xenopus , Young Adult , Delta Catenin
9.
Am J Med Genet A ; 188(10): 2869-2878, 2022 10.
Article in English | MEDLINE | ID: mdl-35899841

ABSTRACT

The Pediatric Genomics Discovery Program (PGDP) at Yale uses next-generation sequencing (NGS) and translational research to evaluate complex patients with a wide range of phenotypes suspected to have rare genetic diseases. We conducted a retrospective cohort analysis of 356 PGDP probands evaluated between June 2015 and July 2020, querying our database for participant demographics, clinical characteristics, NGS results, and diagnostic and research findings. The three most common phenotypes among the entire studied cohort (n = 356) were immune system abnormalities (n = 105, 29%), syndromic or multisystem disease (n = 103, 29%), and cardiovascular system abnormalities (n = 62, 17%). Of 216 patients with final classifications, 77 (36%) received new diagnoses and 139 (64%) were undiagnosed; the remaining 140 patients were still actively being investigated. Monogenetic diagnoses were found in 67 (89%); the largest group had variants in known disease genes but with new contributions such as novel variants (n = 31, 40%) or expanded phenotypes (n = 14, 18%). Finally, five PGDP diagnoses (8%) were suggestive of novel gene-to-phenotype relationships. A broad range of patients can benefit from single subject studies combining NGS and functional molecular analyses. All pediatric providers should consider further genetics evaluations for patients lacking precise molecular diagnoses.


Subject(s)
Genomics , High-Throughput Nucleotide Sequencing , Cohort Studies , Genetic Testing , Humans , Phenotype , Retrospective Studies
10.
S D Med ; 75(6): 268-272, 2022 Jun.
Article in English | MEDLINE | ID: mdl-36206568

ABSTRACT

OBJECTIVE: To determine the yield of early endotracheal aspirate cultures in mechanically ventilated pediatric patients with acute respiratory failure due to acute respiratory tract infection and endeavor to guide antibiotic choice in acute respiratory failure with concern for infectious etiology. RESULTS: One-hundred ten admissions were included. Of those samples, 61 percent (67 out of 110) had bacterial growth in tracheal aspirate samples. Ninety percent (99 out of 110) patients have received antibiotics and in 47 percent (53 out of 110) antibiotics were optimized or discontinued according to the culture results. There were no difference in duration of mechanical ventilation or PICU stay in patients with positive versus negative cultures (p: 0.613, P: 0.337). CONCLUSIONS: Our study shows a high yield of positive tracheal aspirate cultures in infants, children and adolescents with acute respiratory failure. The cultures identify common organisms, helps to guide initial antibiotics choice, as well as later optimization or antibiotic discontinuation.


Subject(s)
Respiratory Distress Syndrome , Respiratory Insufficiency , Respiratory Tract Infections , Adolescent , Anti-Bacterial Agents/therapeutic use , Child , Humans , Infant , Respiration, Artificial/adverse effects , Respiratory Insufficiency/diagnosis , Respiratory Insufficiency/etiology , Respiratory Tract Infections/complications , Respiratory Tract Infections/diagnosis
11.
Mol Genet Genomics ; 296(4): 823-836, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33876311

ABSTRACT

Next-generation sequencing platforms are being increasingly applied in clinical genetic settings for evaluation of families with suspected heritable disease. These platforms potentially improve the diagnostic yield beyond that of disease-specific targeted gene panels, but also increase the number of rare or novel genetic variants that may confound precise diagnostics. Here, we describe a functional testing approach used to interpret the results of whole exome sequencing (WES) in a family presenting with syncope and sudden death. One individual had a prolonged QT interval on electrocardiogram (ECG) and carried a diagnosis of long QT syndrome (LQTS), but a second individual did not meet criteria for LQTS. Filtering WES results for uncommon variants with arrhythmia association identified four for further analyses. In silico analyses indicated that two of these variants, KCNH2 p.(Cys555Arg) and KCNQ1 p.(Arg293Cys), were likely to be causal in this family's LQTS. We subsequently performed functional characterization of these variants in a heterologous expression system. The expression of KCNQ1-Arg293Cys did not show a deleterious phenotype but KCNH2-Cys555Arg demonstrated a loss-of-function phenotype that was partially dominant. Our stepwise approach identified a precise genetic etiology in this family, which resulted in the establishment of a LQTS diagnosis in the second individual as well as an additional asymptomatic family member, enabling personalized clinical management. Given its ability to aid in the diagnosis, the application of functional characterization should be considered as a value adjunct to in silico analyses of WES.


Subject(s)
ERG1 Potassium Channel/genetics , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , AMP-Activated Protein Kinases/genetics , Amino Acid Substitution/genetics , DNA Mutational Analysis/methods , Electrocardiography , Family , Female , Genetic Testing/methods , HEK293 Cells , Heart Function Tests/methods , Humans , KCNQ1 Potassium Channel/genetics , Middle Aged , Mutation , Pedigree , Phenotype , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases/genetics , Exome Sequencing
12.
Am J Med Genet A ; 185(4): 1076-1080, 2021 04.
Article in English | MEDLINE | ID: mdl-33438828

ABSTRACT

De novo heterozygous variants in the brain-specific transcription factor Neuronal Differentiation Factor 2 (NEUROD2) have been recently associated with early-onset epileptic encephalopathy and developmental delay. Here, we report an adolescent with developmental delay without seizures who was found to have a novel de novo heterozygous NEUROD2 missense variant, p.(Leu163Pro). Functional testing using an in vivo assay of neuronal differentiation in Xenopus laevis tadpoles demonstrated that the patient variant of NEUROD2 displays minimal protein activity, strongly suggesting a loss of function effect. In contrast, a second rare NEUROD2 variant, p.(Ala235Thr), identified in an adolescent with developmental delay but lacking parental studies for inheritance, showed normal in vivo NEUROD2 activity. We thus provide clinical, genetic, and functional evidence that NEUROD2 variants can lead to developmental delay without accompanying early-onset seizures, and demonstrate how functional testing can complement genetic data when determining variant pathogenicity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/pathology , Developmental Disabilities/genetics , Neuropeptides/genetics , Adolescent , Animals , Brain/diagnostic imaging , Child , Developmental Disabilities/pathology , Disease Models, Animal , Female , Heterozygote , Humans , Larva/genetics , Male , Phenotype , Seizures/genetics , Seizures/pathology , Xenopus laevis/genetics
13.
Am J Med Genet A ; 182(10): 2291-2296, 2020 10.
Article in English | MEDLINE | ID: mdl-32812332

ABSTRACT

Recessive variants in the GLDN gene, which encodes the gliomedin protein and is involved in nervous system development, have recently been associated with Arthrogryposis Multiplex Congenita (AMC), a heterogenous condition characterized by congenital contractures of more than one joint. Two cohorts of patients with GLDN-associated AMC have previously been described, evolving the understanding of the condition from lethal to survivable with the provision of significant neonatal support. Here, we describe one additional patient currently living with the syndrome, having one novel variant, p.Leu365Phe, for which we provide functional data supporting its pathogenicity. We additionally provide experimental data for four other previously reported variants lacking functional evidence, including p.Arg393Lys, the second variant present in our patient. We discuss unique and defining clinical features, adding calcium-related findings which appear to be recurrent in the GLDN cohort. Finally, we compare all previously reported patients and draw new conclusions about scope of illness, with emphasis on the finding of pulmonary hypoplasia, suggesting that AMC secondary to GLDN variants may be best fitted under the umbrella of fetal akinesia deformation sequence (FADS).


Subject(s)
Arthrogryposis/genetics , Genetic Predisposition to Disease , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Arthrogryposis/pathology , Child, Preschool , Female , Humans , Mutation , Pedigree
14.
Am J Med Genet A ; 182(9): 2049-2057, 2020 09.
Article in English | MEDLINE | ID: mdl-32656949

ABSTRACT

Heterozygous variants in the DYNC1H1 gene have been associated chiefly with intellectual disability (ID), malformations in cortical development (MCD), spinal muscular atrophy (SMA), and Charcot-Marie-Tooth axonal type 20 (CMT), with fewer reports describing other intersecting phenotypes. To better characterize the variable syndromes associated with DYNC1H1, we undertook a detailed analysis of reported patients in the medical literature through June 30, 2019. In sum we identified 200 patients from 143 families harboring 103 different DYNC1H1 variants, and added reports for four unrelated patients identified at our center, three with novel variants. The most common features associated with DYNC1H1 were neuromuscular (NM) disease (largely associated with variants in the stem domain), ID with MCD (largely associated with variants in the motor domain), or a combination of these phenotypes. Despite these trends, exceptions are noted throughout. Overall, DYNC1H1 is associated with variable neurodevelopmental and/or neuromuscular phenotypes that overlap. To avoid confusion DYNC1H1 disorders may be best categorized at this time by more general descriptions rather than phenotype-specific nomenclature such as SMA or CMT. We therefore propose the terms: DYNC1H1-related NM disorder, DYNC1H1-related CNS disorder, and DYNC1H1-related combined disorder. Our single center's experience may be evidence that disease-causing variants in this gene are more prevalent than currently recognized.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Cytoplasmic Dyneins/genetics , Genetic Predisposition to Disease , Muscular Atrophy, Spinal/genetics , Adolescent , Charcot-Marie-Tooth Disease/pathology , Child , Child, Preschool , Female , Heterozygote , Humans , Infant , Male , Malformations of Cortical Development/genetics , Malformations of Cortical Development/pathology , Muscular Atrophy, Spinal/pathology , Mutation, Missense/genetics , Phenotype
15.
J Allergy Clin Immunol ; 143(1): 258-265, 2019 01.
Article in English | MEDLINE | ID: mdl-29935219

ABSTRACT

BACKGROUND: The lack of pathogen-protective, isotype-switched antibodies in patients with common variable immunodeficiency (CVID) suggests germinal center (GC) hypoplasia, yet a subset of patients with CVID is paradoxically affected by autoantibody-mediated autoimmune cytopenias (AICs) and lymphadenopathy. OBJECTIVE: We sought to compare the physical characteristics and immunologic output of GC responses in patients with CVID with AIC (CVID+AIC) and without AIC (CVID-AIC). METHODS: We analyzed GC size and shape in excisional lymph node biopsy specimens from 14 patients with CVID+AIC and 4 patients with CVID-AIC. Using paired peripheral blood samples, we determined how AICs specifically affected B-and T-cell compartments and antibody responses in patients with CVID. RESULTS: We found that patients with CVID+AIC displayed irregularly shaped hyperplastic GCs, whereas GCs were scarce and small in patients with CVID-AIC. GC hyperplasia was also evidenced by an increase in numbers of circulating follicular helper T cells, which correlated with decreased regulatory T-cell frequencies and function. In addition, patients with CVID+AIC had serum endotoxemia associated with a dearth of isotype-switched memory B cells that displayed significantly lower somatic hypermutation frequencies than their counterparts with CVID-AIC. Moreover, IgG+ B cells from patients with CVID+AIC expressed VH4-34-encoded antibodies with unmutated Ala-Val-Tyr and Asn-His-Ser motifs, which recognize both erythrocyte I/i self-antigens and commensal bacteria. CONCLUSIONS: Patients with CVID+AIC do not contain mucosal microbiota and exhibit hyperplastic yet inefficient GC responses that favor the production of untolerized IgG+ B-cell clones that recognize both commensal bacteria and hematopoietic I/i self-antigens.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/immunology , Common Variable Immunodeficiency/immunology , Germinal Center/immunology , Immunoglobulin G/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Aged , B-Lymphocytes/pathology , Biopsy , Child , Common Variable Immunodeficiency/pathology , Female , Germinal Center/pathology , Humans , Hyperplasia , Male , Middle Aged , T-Lymphocytes/pathology
17.
Drug Metab Dispos ; 47(1): 45-48, 2019 01.
Article in English | MEDLINE | ID: mdl-30385458

ABSTRACT

Uridine diphosphate glucuronosyltransferases (UGTs) are key enzymes responsible for the body's ability to process a variety of endogenous and exogenous compounds. Significant gains in understanding UGT function have come from the analysis of variants seen in patients. We cared for a Sudanese child who showed clinical features of type 1 Crigler-Najjar syndrome (CN-1), namely severe unconjugated hyperbilirubinemia leading to liver transplantation. CN-1 is an autosomal recessive disorder caused by damaging mutations in the gene for UGT1A1, the hepatic enzyme responsible for bilirubin conjugation in humans. Clinical genetic testing was unable to identify a known pathogenic UGT1A1 mutation in this child. Instead, a novel homozygous variant resulting in an in-frame deletion, p.Val275del, was noted. Sanger sequencing demonstrated that this variant segregated with the disease phenotype in this family. We further performed functional testing using recombinantly expressed UGT1A1 with and without the patient variant, demonstrating that p.Val275del results in a complete lack of glucuronidation activity, a hallmark of CN-1. Sequence analysis of this region shows a high degree of conservation across all known catalytically active human UGTs, further suggesting that it plays a key role in the enzymatic function of UGTs. Finally, we note that the patient's ethnicity likely played a role in his variant being previously undescribed and advocate for greater diversity and inclusion in genomic medicine.


Subject(s)
Crigler-Najjar Syndrome/genetics , Glucuronosyltransferase/genetics , Child, Preschool , Crigler-Najjar Syndrome/surgery , Genetic Testing , Homozygote , Humans , Liver Transplantation , Male , Sequence Deletion , Sudan
18.
Curr Opin Pediatr ; 31(3): 317-321, 2019 06.
Article in English | MEDLINE | ID: mdl-31090571

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to highlight the significant advances in the testing, interpretation, and diagnosis of genetic abnormalities in critically ill children and to emphasize that pediatric intensivists are uniquely positioned to search for genetic diagnoses in these patients. RECENT FINDINGS: Ten years following the first clinical diagnosis made through whole exome sequencing, we remain in the dark about the function of roughly 75% of our genes. However, steady advancements in molecular techniques, particularly next-generation sequencing, have spurred a rapid expansion of our understanding of the genetic underpinnings of severe congenital diseases. This has resulted in not only improved clinical diagnostics but also a greater availability of research programs actively investigating rare, undiagnosed diseases. In this background, the scarcity of clinical geneticists compels nongeneticists to familiarize themselves with the types of patients that could benefit from genetic testing, interpretations of test results as well as the available resources for these patients. SUMMARY: When caring for seriously ill children, critical care pediatricians should actively seek the possibility of an underlying genetic cause for their patients' conditions. This is true even in instances when a child has a descriptive diagnosis without a clear underlying molecular genetic mechanism. By promoting such diagnostics, in both clinical and research settings, pediatric intensivists can advance the care of their patients, improve the quality of information provided to families, and contribute to the knowledge of broad fields in medicine.


Subject(s)
Critical Illness , Genetic Testing , Genomics , Child , Diagnosis , Humans , Rare Diseases , Exome Sequencing
19.
BMC Nephrol ; 20(1): 271, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31315584

ABSTRACT

BACKGROUND: Monogenic mutations may be a significant cause of steroid-resistant nephrotic syndrome. NUP93 is a gene previously reported to cause isolated steroid-resistant nephrotic syndrome. CASE PRESENTATION: Here we describe a case of recessive, syndromic, steroid-resistant nephrotic syndrome caused by NUP93 mutation. CONCLUSIONS: NUP93 may convey a phenotype that has not only SRNS, but also other syndromic features.


Subject(s)
Mutation , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/genetics , Nuclear Pore Complex Proteins/genetics , Phenotype , Child, Preschool , Drug Resistance/genetics , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL