Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Front Immunol ; 14: 1271102, 2023.
Article in English | MEDLINE | ID: mdl-38022642

ABSTRACT

Autoimmune diseases arise from atypical immune responses that attack self-tissue epitopes, and their development is intricately connected to the disruption of the JAK-STAT signaling pathway, where SOCS proteins play crucial roles. Conditions such as autoimmune uveitis, psoriasis, lupus, and autoimmune encephalitis exhibit immune system dysfunctions associated with JAK-STAT signaling dysregulation. Emerging therapeutic strategies utilize JAK-STAT inhibitors and SOCS mimetics to modulate immune responses and alleviate autoimmune manifestations. Although more research and clinical studies are required to assess their effectiveness, safety profiles, and potential for personalized therapeutic approaches in autoimmune conditions, JAK-STAT inhibitors and SOCS mimetics show promise as potential treatment options. This review explores the action, effectiveness, safety profiles, and future prospects of JAK inhibitors and SOCS mimetics as therapeutic agents for psoriasis, autoimmune uveitis, systemic lupus erythematosus, and autoimmune encephalitis. The findings underscore the importance of investigating these targeted therapies to advance treatment options for individuals suffering from autoimmune diseases.


Subject(s)
Autoimmune Diseases of the Nervous System , Autoimmune Diseases , Janus Kinase Inhibitors , Psoriasis , Uveitis , Humans , Autoimmune Diseases/drug therapy , Uveitis/drug therapy
2.
Int J Mol Sci ; 24(9)2023 May 04.
Article in English | MEDLINE | ID: mdl-37175930

ABSTRACT

CLEC16A is emerging as an important genetic risk factor for several autoimmune disorders and for Parkinson disease (PD), opening new avenues for translational research and therapeutic development. While the exact role of CLEC16A in health and disease is still being elucidated, the gene plays a critical role in the regulation of autophagy, mitophagy, endocytosis, intracellular trafficking, immune function, and in biological processes such as insulin secretion and others that are important to cellular homeostasis. As shown in both human and animal modeling studies, CLEC16A hypofunction predisposes to both autoinflammatory phenotype and neurodegeneration. While the two are clearly related, further functional studies are needed to fully understand the mechanisms involved for optimized therapeutic interventions. Based on recent data, mitophagy-inducing drugs may be warranted, and such therapy should be tested in clinical trials as these drugs would tackle the underlying pathogenic mechanism (s) and could treat or prevent symptoms of autoimmunity and neurodegeneration in individuals with CLEC16A risk variants. Accordingly, interventions directed at reversing the dysregulated mitophagy and the consequences of loss of function of CLEC16A without activating other detrimental cellular pathways could present an effective therapy. This review presents the emerging role of CLEC16A in health and disease and provides an update on the disease processes that are attributed to variants located in the CLEC16A gene, which are responsible for autoimmune disorders and neurodegeneration with emphasis on how this information is being translated into practical and effective applications in the clinic.


Subject(s)
Autoimmune Diseases , Lectins, C-Type , Animals , Humans , Autoimmune Diseases/genetics , Autoimmunity/genetics , Autophagy/genetics , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Monosaccharide Transport Proteins/metabolism , Risk Factors
3.
Sci Rep ; 11(1): 7372, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795715

ABSTRACT

CLEC16A is implicated in multiple autoimmune diseases. We generated an inducible whole-body knockout (KO), Clec16aΔUBC mice to address the role of CLEC16A loss of function. KO mice exhibited loss of adipose tissue and severe weight loss in response to defective autophagic flux and exaggerated endoplasmic reticulum (ER) stress and robust cytokine storm. KO mice were glucose tolerant and displayed a state of systemic inflammation with elevated antibody levels, including IgM, IgA, Ig2b and IgG3, significantly reduced circulating insulin levels in the presence of normal food consumption. Metabolic analysis revealed disturbances in the lipid profile, white adipose decreasing concomitantly with enhanced inflammatory response, and energy wasting. Mechanistically, endoplasmic reticulum (ER) stress triggers excessive hormone sensitive lipases (HSL) mediated lipolysis which contributes to adipose inflammation via activation of JAK-STAT, stress kinases (ERK1/2, P38, JNK), and release of multiple proinflammatory mediators. Treatment with a JAK-STAT inhibitor (tofacitinib) partially rescued the inflammatory lipodystrophic phenotype and improved survival of Clec16aΔUBC mice by silencing cytokine release and modulating ER stress, lipolysis, mitophagy and autophagy. These results establish a mechanistic link between CLEC16A, lipid metabolism and the immune system perturbations. In summary, our Clec16aΔUBC mouse model highlights multifaceted roles of Clec16a in normal physiology, including a novel target for weight regulation and mutation-induced pathophysiology.


Subject(s)
Autoimmunity/genetics , Janus Kinase Inhibitors/pharmacology , Lectins, C-Type/genetics , Lipodystrophy/drug therapy , Lipodystrophy/genetics , Monosaccharide Transport Proteins/genetics , STAT1 Transcription Factor/antagonists & inhibitors , Adipose Tissue , Animals , Autophagy , Cytokines/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Female , Glucose Tolerance Test , Inflammation , Insulin/metabolism , Lipid Metabolism , Male , Mice , Mice, Knockout , Phenotype , Phosphorylation , Piperidines/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Sirolimus/pharmacology
4.
Sci Rep ; 11(1): 9319, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33927318

ABSTRACT

CLEC16A has been shown to play a role in autophagy/mitophagy processes. Additionally, genetic variants in CLEC16A have been implicated in multiple autoimmune diseases. We generated an inducible whole-body knockout, Clec16aΔUBC mice, to investigate the loss of function of CLEC16A. The mice exhibited a neuronal phenotype including tremors and impaired gait that rapidly progressed to dystonic postures. Nerve conduction studies and pathological analysis revealed loss of sensory axons that are associated with this phenotype. Activated microglia and astrocytes were found in regions of the CNS. Several mitochondrial-related proteins were up- or down-regulated. Upregulation of interferon stimulated gene 15 (IGS15) were observed in neuronal tissues. CLEC16A expression inversely related to IGS15 expression. ISG15 may be the link between CLEC16A and downstream autoimmune, inflammatory processes. Our results demonstrate that a whole-body, inducible knockout of Clec16a in mice results in an inflammatory neurodegenerative phenotype resembling spinocerebellar ataxia.


Subject(s)
Lectins, C-Type/physiology , Monosaccharide Transport Proteins/physiology , Nervous System Autoimmune Disease, Experimental , Spinocerebellar Ataxias , Animals , Cytokines/metabolism , Female , Gene Knockout Techniques , Male , Mice, Knockout , Neurons/ultrastructure , Ubiquitins/metabolism
5.
J Natl Cancer Inst ; 112(12): 1259-1266, 2020 12 14.
Article in English | MEDLINE | ID: mdl-32096864

ABSTRACT

BACKGROUND: Neuroblastoma is a childhood malignancy that arises from the developing sympathetic nervous system. Although mitochondrial dysfunctions have been implicated in the pathophysiology of neuroblastoma, the role of mitochondrial DNA (mtDNA) has not been extensively investigated. METHODS: A total of 2404 Caucasian children diagnosed with neuroblastoma and 9310 ancestry-matched controls were recruited at the Children's Hospital of Philadelphia. The mtDNA haplogroups were identified from SNP array data of two independent cohorts. We conducted a case-control study to explore potential associations of mtDNA haplogroups with the susceptibility of neuroblastoma. The genetic effect of neuroblastoma was measured by odds ratios (ORs) of mitochondrial haplogroups. All tests were two-sided. RESULTS: Haplogroup K was statistically significantly associated with reduced risk of neuroblastoma in the discovery cohort consisting of 1474 cases and 5699 controls (OR = 0.72, 95% confidence interval [CI] = 0.57 to 0.90; P = 4.8 × 10-3). The association was replicated in an independent cohort (OR = 0.69, 95% CI = 0.53 to 0.92; P = .01) of 930 cases and 3611 controls. Pooled analysis was performed by combining the two data sets. The association remained highly statistically significant after correction for multiple testing (OR = 0.71, 95% CI = 0.59 to 0.84, P = 1.96 × 10-4, Pcorrected = .002). Further analysis focusing on neuroblastoma subtypes indicated haplogroup K was more associated with high-risk neuroblastoma (OR = 0.57, 95% CI = 0.43 to 0.76; P = 1.46 × 10-4) than low-risk and intermediate-risk neuroblastoma. CONCLUSIONS: Haplogroup K is an independent genetic factor associated with reduced risk of developing neuroblastoma in European descents. These findings provide new insights into the genetic basis of neuroblastoma, implicating mitochondrial DNA encoded proteins in the etiology of neuroblastoma.


Subject(s)
DNA, Mitochondrial/genetics , Neuroblastoma/genetics , Polymorphism, Single Nucleotide , Age of Onset , Case-Control Studies , Cells, Cultured , Child , Cohort Studies , Datasets as Topic/statistics & numerical data , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Neuroblastoma/epidemiology
6.
Nat Commun ; 11(1): 255, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31937769

ABSTRACT

Copy number variants (CNVs) are suggested to have a widespread impact on the human genome and phenotypes. To understand the role of CNVs across human diseases, we examine the CNV genomic landscape of 100,028 unrelated individuals of European ancestry, using SNP and CGH array datasets. We observe an average CNV burden of ~650 kb, identifying a total of 11,314 deletion, 5625 duplication, and 2746 homozygous deletion CNV regions (CNVRs). In all, 13.7% are unreported, 58.6% overlap with at least one gene, and 32.8% interrupt coding exons. These CNVRs are significantly more likely to overlap OMIM genes (2.94-fold), GWAS loci (1.52-fold), and non-coding RNAs (1.44-fold), compared with random distribution (P < 1 × 10-3). We uncover CNV associations with four major disease categories, including autoimmune, cardio-metabolic, oncologic, and neurological/psychiatric diseases, and identify several drug-repurposing opportunities. Our results demonstrate robust frequency definition for large-scale rare variant association studies, identify CNVs associated with major disease categories, and illustrate the pleiotropic impact of CNVs in human disease.


Subject(s)
DNA Copy Number Variations , Genetic Predisposition to Disease/genetics , Genome, Human/genetics , White People/genetics , Comparative Genomic Hybridization , Databases, Genetic , Genetic Loci , Genetic Predisposition to Disease/ethnology , Genome-Wide Association Study , Humans , Molecular Sequence Annotation , Polymorphism, Single Nucleotide
7.
Curr Diab Rep ; 19(11): 116, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31686270

ABSTRACT

PURPOSE OF REVIEW: To provide an updated summary of discoveries made to date resulting from genome-wide association study (GWAS) and sequencing studies, and to discuss the latest loci added to the growing repertoire of genetic signals predisposing to type 1 diabetes (T1D). RECENT FINDINGS: Genetic studies have identified over 60 loci associated with T1D susceptibility. GWAS alone does not specifically inform on underlying mechanisms, but in combination with other sequencing and omics-data, advances are being made in our understanding of T1D genetic etiology and pathogenesis. Current knowledge indicates that genetic variation operating in both pancreatic ß cells and in immune cells is central in mediating T1D risk. One of the main challenges is to determine how these recently discovered GWAS-implicated variants affect the expression and function of gene products. Once we understand the mechanism of action for disease-causing variants, we will be well placed to apply targeted genomic approaches to impede the premature activation of the immune system in an effort to ultimately prevent the onset of T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Genome-Wide Association Study , Insulin-Secreting Cells , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Genomics , Humans , Polymorphism, Single Nucleotide , Prospective Studies
8.
Front Immunol ; 10: 68, 2019.
Article in English | MEDLINE | ID: mdl-30774629

ABSTRACT

CLEC16A locus polymorphisms have been associated with several autoimmune diseases. We overexpressed CLEC16A in YTS natural killer (NK) cells and observed reduced NK cell cytotoxicity and IFN-γ release, delayed dendritic cell (DC) maturation, decreased conjugate formation, cell-surface receptor downregulation and increased autophagy. In contrast, siRNA mediated knockdown resulted in increased NK cell cytotoxicity, reversal of receptor expression and disrupted mitophagy. Subcellular localization studies demonstrated that CLEC16A is a cytosolic protein that associates with Vps16A, a subunit of class C Vps-HOPS complex, and modulates receptor expression via autophagy. Clec16a knockout (KO) in mice resulted in altered immune cell populations, increased splenic NK cell cytotoxicity, imbalance of dendritic cell subsets, altered receptor expression, upregulated cytokine and chemokine secretion. Taken together, our findings indicate that CLEC16A restrains secretory functions including cytokine release and cytotoxicity and that a delicate balance of CLEC16A is needed for NK cell function and homeostasis.


Subject(s)
Killer Cells, Natural/immunology , Lectins, C-Type/immunology , Monosaccharide Transport Proteins/immunology , Animals , Autoimmune Diseases/genetics , Cell Line , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Genetic Predisposition to Disease , Humans , Interferon-gamma/immunology , Lectins, C-Type/genetics , Mice, Knockout , Monosaccharide Transport Proteins/genetics , Spleen/immunology
9.
PLoS One ; 13(9): e0203952, 2018.
Article in English | MEDLINE | ID: mdl-30226884

ABSTRACT

CLEC16A is implicated in multiple autoimmune diseases. We generated Clec16a inducible knockout (KO) mice to examine the functional link between CLEC16A auto-inflammation and autoimmunity. Clec16a KO mice exhibited weight loss and thymic and splenic atrophy. Mitochondrial potential was lowered in KO mice splenocytes resulting in aggregation of unhealthy mitochondria in B, T, and NK cells. In Clec16a KO mice we detected disrupted mitophagy in splenic B and T cells. NK cells from Clec16a KO mice exhibited increased cytotoxicity. Incomplete mitophagy was attenuated with PI3K and/or MEK inhibition in Clec16a KO mice. Our results demonstrate a functional link between CLEC16A and disrupted mitophagy in immune cells and show that incomplete mitophagy predisposes the KO mice to inflammation. Taken together, loss of function variants in CLEC16A that are associated with decreased CLEC16A expression levels may contribute to inflammation in autoimmunity through disrupted mitophagy. Drugs modulating mitophagy reverse the process and may be effective in treating and preventing autoimmunity in individuals with risk associated CLEC16A variants.


Subject(s)
Killer Cells, Natural/metabolism , Lectins, C-Type/physiology , MAP Kinase Signaling System , Monosaccharide Transport Proteins/physiology , Spleen/cytology , Animals , Blotting, Western , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Killer Cells, Natural/physiology , MAP Kinase Signaling System/physiology , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , Mitophagy , Spleen/metabolism , Spleen/physiology
10.
J Clin Endocrinol Metab ; 103(11): 4023-4032, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30137364

ABSTRACT

Context: Most cases of autosomal dominant isolated hypoparathyroidism are caused by gain-of-function mutations in CASR or GNA11 or dominant negative mutations in GCM2 or PTH. Objective: To identify the genetic etiology for dominantly transmitted isolated hypoparathyroidism in two multigenerational families with 14 affected family members. Methods: We performed whole exome sequencing of DNA from two families and examined the consequences of mutations by minigene splicing assay. Results: We discovered disease-causing mutations in both families. A splice-altering mutation in TBX1 (c.1009+1G>C) leading to skipping of exon 8 (101 bp) was identified in 10 affected family members and five unaffected subjects of family A, indicating reduced penetrance for this point mutation. In a second family from France (family B), we identified another splice-altering mutation (c.1009+2T>C) adjacent to the mutation identified in family A that results in skipping of the same exon; two subjects in family B had isolated hypoparathyroidism, whereas a third subject manifested the clinical triad of the 22q11.2 deletion syndrome, indicative of variable expressivity. Conclusions: We report evidence that heterozygous TBX1 mutations can cause isolated hypoparathyroidism. This study adds knowledge to the increasingly expanding list of causative and candidate genes in isolated hypoparathyroidism.


Subject(s)
DiGeorge Syndrome/genetics , Hypercalciuria/genetics , Hypocalcemia/genetics , Hypoparathyroidism/congenital , T-Box Domain Proteins/genetics , Aged , DiGeorge Syndrome/blood , DiGeorge Syndrome/diagnosis , Exons/genetics , Female , Heterozygote , Humans , Hypercalciuria/blood , Hypercalciuria/diagnosis , Hypocalcemia/blood , Hypocalcemia/diagnosis , Hypoparathyroidism/blood , Hypoparathyroidism/diagnosis , Hypoparathyroidism/genetics , Infant , Male , Mutation , Pedigree , Penetrance , RNA Splice Sites/genetics , Exome Sequencing
11.
Hum Mol Genet ; 27(18): 3233-3245, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29905864

ABSTRACT

Central conducting lymphatic anomaly (CCLA) is one of the complex lymphatic anomalies characterized by dilated lymphatic channels, lymphatic channel dysmotility and distal obstruction affecting lymphatic drainage. We performed whole exome sequencing (WES) of DNA from a four-generation pedigree and examined the consequences of the variant by transfection of mammalian cells and morpholino and rescue studies in zebrafish. WES revealed a heterozygous mutation in EPHB4 (RefSeq NM_004444.4; c.2334 + 1G>C) and RNA-Seq demonstrated that the EPHB4 mutation destroys the normal donor site, which leads to the use of a cryptic splice donor that results in retention of the intervening 12-bp intron sequence. Transient co-expression of the wild-type and mutant EPHB4 proteins showed reduced phosphorylation of tyrosine, consistent with a loss-of-function effect. Zebrafish ephb4a morpholino resulted in vessel misbranching and deformities in the lymphatic vessel development, indicative of possible differentiation defects in lymphatic vessels, mimicking the lymphatic presentations of the patients. Immunoblot analysis using zebrafish lysates demonstrated over-activation of mTORC1 as a consequence of reduced EPHB4 signaling. Strikingly, drugs that inhibit mTOR signaling or RAS-MAPK signaling effectively rescued the misbranching phenotype in a comparable manner. Moreover, knock-in of EPHB4 mutation in HEK293T cells also induced mTORC1 activity. Our data demonstrate the pathogenicity of the identified EPHB4 mutation as a novel cause of CCLA and suggesting that ERK inhibitors may have therapeutic benefits in such patients with complex lymphatic anomalies.


Subject(s)
Exome Sequencing , Lymphatic Abnormalities/genetics , Lymphatic Vessels/metabolism , Receptor, EphB4/genetics , Animals , Disease Models, Animal , HEK293 Cells , Heterozygote , Humans , Lymphatic Abnormalities/metabolism , Lymphatic Abnormalities/pathology , Lymphatic Vessels/pathology , Mechanistic Target of Rapamycin Complex 1/genetics , Pedigree , Phosphorylation , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Zebrafish/genetics
12.
Nat Commun ; 7: 12792, 2016 Sep 28.
Article in English | MEDLINE | ID: mdl-27677580

ABSTRACT

Acute otitis media (AOM) is among the most common pediatric diseases, and the most frequent reason for antibiotic treatment in children. Risk of AOM is dependent on environmental and host factors, as well as a significant genetic component. We identify genome-wide significance at a locus on 6q25.3 (rs2932989, Pmeta=2.15 × 10-09), and show that the associated variants are correlated with the methylation status of the FNDC1 gene (cg05678571, P=1.43 × 10-06), and further show it is an eQTL for FNDC1 (P=9.3 × 10-05). The mouse homologue, Fndc1, is expressed in middle ear tissue and its expression is upregulated upon lipopolysaccharide treatment. In this first GWAS of AOM and the largest OM genetic study to date, we identify the first genome-wide significant locus associated with AOM.

13.
Nat Commun ; 6: 8442, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26450413

ABSTRACT

Autoimmune diseases (AIDs) are polygenic diseases affecting 7-10% of the population in the Western Hemisphere with few effective therapies. Here, we quantify the heritability of paediatric AIDs (pAIDs), including JIA, SLE, CEL, T1D, UC, CD, PS, SPA and CVID, attributable to common genomic variations (SNP-h(2)). SNP-h(2) estimates are most significant for T1D (0.863±s.e. 0.07) and JIA (0.727±s.e. 0.037), more modest for UC (0.386±s.e. 0.04) and CD (0.454±0.025), largely consistent with population estimates and are generally greater than that previously reported by adult GWAS. On pairwise analysis, we observed that the diseases UC-CD (0.69±s.e. 0.07) and JIA-CVID (0.343±s.e. 0.13) are the most strongly correlated. Variations across the MHC strongly contribute to SNP-h(2) in T1D and JIA, but does not significantly contribute to the pairwise rG. Together, our results partition contributions of shared versus disease-specific genomic variations to pAID heritability, identifying pAIDs with unexpected risk sharing, while recapitulating known associations between autoimmune diseases previously reported in adult cohorts.


Subject(s)
Autoimmune Diseases/congenital , Autoimmune Diseases/genetics , Adolescent , Age of Onset , Case-Control Studies , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Polymorphism, Single Nucleotide , White People/genetics
14.
Nat Med ; 21(9): 1018-27, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26301688

ABSTRACT

Genome-wide association studies (GWASs) have identified hundreds of susceptibility genes, including shared associations across clinically distinct autoimmune diseases. We performed an inverse χ(2) meta-analysis across ten pediatric-age-of-onset autoimmune diseases (pAIDs) in a case-control study including more than 6,035 cases and 10,718 shared population-based controls. We identified 27 genome-wide significant loci associated with one or more pAIDs, mapping to in silico-replicated autoimmune-associated genes (including IL2RA) and new candidate loci with established immunoregulatory functions such as ADGRL2, TENM3, ANKRD30A, ADCY7 and CD40LG. The pAID-associated single-nucleotide polymorphisms (SNPs) were functionally enriched for deoxyribonuclease (DNase)-hypersensitivity sites, expression quantitative trait loci (eQTLs), microRNA (miRNA)-binding sites and coding variants. We also identified biologically correlated, pAID-associated candidate gene sets on the basis of immune cell expression profiling and found evidence of genetic sharing. Network and protein-interaction analyses demonstrated converging roles for the signaling pathways of type 1, 2 and 17 helper T cells (TH1, TH2 and TH17), JAK-STAT, interferon and interleukin in multiple autoimmune diseases.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/etiology , Child , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Risk Factors
15.
J Immunol ; 195(4): 1599-607, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26188062

ABSTRACT

Food allergy is a significant public health concern, especially among children. Previous candidate gene studies suggested a few susceptibility loci for food allergy, but no study investigated the contribution of copy number variations (CNVs) to food allergy on a genome-wide scale. To investigate the genetics of food allergy, we performed CNV assessment using high-resolution genome-wide single nucleotide polymorphism arrays. CNV calls from a total of 357 cases with confirmed food allergy and 3980 controls were analyzed within a discovery cohort, followed by a replication analysis composed of 167 cases and 1573 controls. We identified that CNVs in CTNNA3 were significantly associated with food allergy in both the discovery cohort and the replication cohort. Of particular interest, CTNNA3 CNVs hit exons or intron regions rich in histone marker H3K4Me1. CNVs in a second gene (RBFOX1) showed a significant association (p = 7.35 × 10(-5)) with food allergy at the genome-wide level in our meta-analysis of the European ancestry cohorts. The presence of these CNVs was confirmed by quantitative PCR. Furthermore, knockdown of CTNNA3 resulted in upregulation of CD63 and CD203c in mononuclear cells upon PMA stimulation, suggesting a role in sensitization to allergen. We uncovered at least two plausible genes harboring CNV loci that are enriched in pediatric patients with food allergies. The novel gene candidates discovered in this study by genome-wide CNV analysis are compelling drug and diagnostic targets for food allergy.


Subject(s)
DNA Copy Number Variations , Food Hypersensitivity/genetics , Food Hypersensitivity/immunology , Genetic Predisposition to Disease , RNA-Binding Proteins/genetics , alpha Catenin/genetics , Adolescent , Age Factors , Case-Control Studies , Child , Child, Preschool , Female , Gene Deletion , Genetic Association Studies , Humans , Male , Meta-Analysis as Topic , Middle Aged , Polymorphism, Single Nucleotide , RNA Splicing Factors , RNA, Small Interfering , Reproducibility of Results
16.
Nat Commun ; 6: 6804, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25891430

ABSTRACT

Common variable immunodeficiency disorder (CVID) is the most common symptomatic primary immunodeficiency in adults, characterized by B-cell abnormalities and inadequate antibody response. CVID patients have considerable autoimmune comorbidity and we therefore hypothesized that genetic susceptibility to CVID may overlap with autoimmune disorders. Here, in the largest genetic study performed in CVID to date, we compare 778 CVID cases with 10,999 controls across 123,127 single-nucleotide polymorphisms (SNPs) on the Immunochip. We identify the first non-HLA genome-wide significant risk locus at CLEC16A (rs17806056, P=2.0 × 10(-9)) and confirm the previously reported human leukocyte antigen (HLA) associations on chromosome 6p21 (rs1049225, P=4.8 × 10(-16)). Clec16a knockdown (KD) mice showed reduced number of B cells and elevated IgM levels compared with controls, suggesting that CLEC16A may be involved in immune regulatory pathways of relevance to CVID. In conclusion, the CLEC16A associations in CVID represent the first robust evidence of non-HLA associations in this immunodeficiency condition.


Subject(s)
B-Lymphocytes/metabolism , Common Variable Immunodeficiency/genetics , Lectins, C-Type/metabolism , Monosaccharide Transport Proteins/metabolism , Animals , Case-Control Studies , Cells, Cultured , Common Variable Immunodeficiency/epidemiology , Europe/epidemiology , Gene Expression Regulation/physiology , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Lectins, C-Type/genetics , Logistic Models , Mice , Monosaccharide Transport Proteins/genetics , Odds Ratio , Polymorphism, Single Nucleotide , Spleen/cytology , United States/epidemiology
17.
Cell ; 157(7): 1577-90, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949970

ABSTRACT

Clec16a has been identified as a disease susceptibility gene for type 1 diabetes, multiple sclerosis, and adrenal dysfunction, but its function is unknown. Here we report that Clec16a is a membrane-associated endosomal protein that interacts with E3 ubiquitin ligase Nrdp1. Loss of Clec16a leads to an increase in the Nrdp1 target Parkin, a master regulator of mitophagy. Islets from mice with pancreas-specific deletion of Clec16a have abnormal mitochondria with reduced oxygen consumption and ATP concentration, both of which are required for normal ß cell function. Indeed, pancreatic Clec16a is required for normal glucose-stimulated insulin release. Moreover, patients harboring a diabetogenic SNP in the Clec16a gene have reduced islet Clec16a expression and reduced insulin secretion. Thus, Clec16a controls ß cell function and prevents diabetes by controlling mitophagy. This pathway could be targeted for prevention and control of diabetes and may extend to the pathogenesis of other Clec16a- and Parkin-associated diseases.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Islets of Langerhans/pathology , Lectins, C-Type/metabolism , Mitophagy , Monosaccharide Transport Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Diabetes Mellitus, Type 1/pathology , Genetic Predisposition to Disease , Glucose/metabolism , Humans , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Lysosomes/chemistry , Lysosomes/metabolism , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , Polymorphism, Single Nucleotide , Ubiquitin-Protein Ligases
18.
Genes (Basel) ; 4(3): 499-521, 2013 Sep 16.
Article in English | MEDLINE | ID: mdl-24705215

ABSTRACT

Type 1 Diabetes (T1D) is a chronic multifactorial disease with a strong genetic component, which, through interactions with specific environmental factors, triggers disease onset. T1D typically manifests in early to mid childhood through the autoimmune destruction of pancreatic ß cells resulting in a lack of insulin production. Historically, prior to genome-wide association studies (GWAS), six loci in the genome were fully established to be associated with T1D. With the advent of high-throughput single nucleotide polymorphism (SNP) genotyping array technologies, enabling investigators to perform high-density GWAS, many additional T1D susceptibility genes have been discovered. Indeed, recent meta-analyses of multiple datasets from independent investigators have brought the tally of well-validated T1D disease genes to almost 60. In this mini-review, we address recent advances in the genetics of T1D and provide an update on the latest susceptibility loci added to the list of genes involved in the pathogenesis of T1D.

19.
PLoS Genet ; 7(9): e1002293, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21980299

ABSTRACT

Diabetes impacts approximately 200 million people worldwide, of whom approximately 10% are affected by type 1 diabetes (T1D). The application of genome-wide association studies (GWAS) has robustly revealed dozens of genetic contributors to the pathogenesis of T1D, with the most recent meta-analysis identifying in excess of 40 loci. To identify additional genetic loci for T1D susceptibility, we examined associations in the largest meta-analysis to date between the disease and ∼2.54 million SNPs in a combined cohort of 9,934 cases and 16,956 controls. Targeted follow-up of 53 SNPs in 1,120 affected trios uncovered three new loci associated with T1D that reached genome-wide significance. The most significantly associated SNP (rs539514, P = 5.66×10⁻¹¹) resides in an intronic region of the LMO7 (LIM domain only 7) gene on 13q22. The second most significantly associated SNP (rs478222, P = 3.50×10⁻9 resides in an intronic region of the EFR3B (protein EFR3 homolog B) gene on 2p23; however, the region of linkage disequilibrium is approximately 800 kb and harbors additional multiple genes, including NCOA1, C2orf79, CENPO, ADCY3, DNAJC27, POMC, and DNMT3A. The third most significantly associated SNP (rs924043, P = 8.06×10⁻9 lies in an intergenic region on 6q27, where the region of association is approximately 900 kb and harbors multiple genes including WDR27, C6orf120, PHF10, TCTE3, C6orf208, LOC154449, DLL1, FAM120B, PSMB1, TBP, and PCD2. These latest associated regions add to the growing repertoire of gene networks predisposing to T1D.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Genetic Loci , Genetic Predisposition to Disease , Cohort Studies , DNA, Intergenic , Female , Genome-Wide Association Study , Humans , LIM Domain Proteins/genetics , Linkage Disequilibrium , Male , Polymorphism, Single Nucleotide/genetics , Sequence Homology , Transcription Factors/genetics
20.
Ann Thorac Surg ; 92(3): 858-65, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21871270

ABSTRACT

BACKGROUND: One of the most important factors responsible for the calcific failure of bioprosthetic heart valves is glutaraldehyde crosslinking. Ethanol (EtOH) incubation after glutaraldehyde crosslinking has previously been reported to confer anticalcification efficacy for bioprostheses. The present studies investigated the anticalcification efficacy in vivo of the novel crosslinking agent, triglycidyl amine (TGA), with or without EtOH incubation, in comparison with glutaraldehyde. METHODS: The TGA crosslinking (±EtOH) was used to prepare porcine aortic valves for both rat subdermal implants and sheep mitral valve replacements, for comparisons with glutaraldehyde-fixed controls. Thermal denaturation temperature, an index of crosslinking, cholesterol extraction, and hydrodynamic properties were quantified. Explant endpoints included quantitative and morphologic assessment of calcification. RESULTS: Thermal denaturation temperatures after TGA were intermediate between unfixed and glutaraldehyde-fixed. EtOH incubation resulted in almost complete extraction of cholesterol from TGA or glutaraldehyde-fixed cusps. Rat subdermal explants (90 days) demonstrated that TGA-EtOH resulted in a significantly greater level of inhibition of calcification than other conditions. Thus, TGA-ethanol stent mounted porcine aortic valve bioprostheses were fabricated for comparisons with glutaraldehyde-pretreated controls. In hydrodynamic studies, TGA-EtOH bioprostheses had lower pressure gradients than glutaraldehyde-fixed. The TGA-ethanol bioprostheses used as mitral valve replacements in juvenile sheep (150 days) demonstrated significantly lower calcium levels in both explanted porcine aortic cusp and aortic wall samples compared with glutaraldehyde-fixed controls. However, TGA-EtOH sheep explants also demonstrated isolated calcific nodules and intracuspal hematomas. CONCLUSIONS: The TGA-EtOH pretreatment of porcine aortic valves confers significant calcification resistance in both rat subdermal and sheep circulatory implants, but with associated structural instability.


Subject(s)
Bioprosthesis , Calcinosis/prevention & control , Epoxy Compounds/pharmacology , Ethanol/pharmacology , Heart Valve Diseases/prevention & control , Heart Valve Prosthesis , Organ Preservation/methods , Animals , Calcinosis/pathology , Calorimetry , Disease Models, Animal , Drug Combinations , Heart Valve Diseases/pathology , Organ Preservation Solutions/pharmacology , Rats , Sheep , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...