Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 177(1): 32-37, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30901545

RESUMEN

The introduction of exome sequencing in the clinic has sparked tremendous optimism for the future of rare disease diagnosis, and there is exciting opportunity to further leverage these advances. To provide diagnostic clarity to all of these patients, however, there is a critical need for the field to develop and implement strategies to understand the mechanisms underlying all rare diseases and translate these to clinical care.


Asunto(s)
Secuenciación del Exoma/tendencias , Enfermedades Raras/diagnóstico , Investigación Biomédica Traslacional/métodos , Exoma , Pruebas Genéticas , Genoma Humano/genética , Secuenciación de Nucleótidos de Alto Rendimiento/tendencias , Humanos , Enfermedades Raras/genética , Análisis de Secuencia de ADN/métodos , Secuenciación del Exoma/métodos
2.
Cell ; 149(2): 295-306, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22484060

RESUMEN

Congenital scoliosis, a lateral curvature of the spine caused by vertebral defects, occurs in approximately 1 in 1,000 live births. Here we demonstrate that haploinsufficiency of Notch signaling pathway genes in humans can cause this congenital abnormality. We also show that in a mouse model, the combination of this genetic risk factor with an environmental condition (short-term gestational hypoxia) significantly increases the penetrance and severity of vertebral defects. We demonstrate that hypoxia disrupts FGF signaling, leading to a temporary failure of embryonic somitogenesis. Our results potentially provide a mechanism for the genesis of a host of common sporadic congenital abnormalities through gene-environment interaction.


Asunto(s)
Interacción Gen-Ambiente , Escoliosis/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Femenino , Haploinsuficiencia , Humanos , Hipoxia/metabolismo , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Linaje , Penetrancia , Receptores Notch/metabolismo , Escoliosis/congénito , Transducción de Señal , Columna Vertebral/embriología
3.
Am J Hum Genet ; 110(3): 419-426, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36868206

RESUMEN

Australian Genomics is a national collaborative partnership of more than 100 organizations piloting a whole-of-system approach to integrating genomics into healthcare, based on federation principles. In the first five years of operation, Australian Genomics has evaluated the outcomes of genomic testing in more than 5,200 individuals across 19 rare disease and cancer flagship studies. Comprehensive analyses of the health economic, policy, ethical, legal, implementation and workforce implications of incorporating genomics in the Australian context have informed evidence-based change in policy and practice, resulting in national government funding and equity of access for a range of genomic tests. Simultaneously, Australian Genomics has built national skills, infrastructure, policy, and data resources to enable effective data sharing to drive discovery research and support improvements in clinical genomic delivery.


Asunto(s)
Genómica , Política de Salud , Humanos , Australia , Enfermedades Raras , Atención a la Salud
4.
Differentiation ; 130: 28-31, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36543010

RESUMEN

The International Society of Differentiation was born from the First International Conference on Cell Differentiation conceived by D.V. and held in Nice, France in 1971. The conference also resulted in the creation of the journal of the Society named Differentiation. The Society advocates for the field of differentiation through the journal Differentiation, organizing and supporting international scientific conferences, honoring scientific achievements, and supporting trainees.


Asunto(s)
Diferenciación Celular , Sociedades Científicas , Sociedades Científicas/historia
5.
BMC Bioinformatics ; 24(1): 49, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36792982

RESUMEN

BACKGROUND: A wide range of tools are available for the detection of copy number variants (CNVs) from whole-genome sequencing (WGS) data. However, none of them focus on clinically-relevant CNVs, such as those that are associated with known genetic syndromes. Such variants are often large in size, typically 1-5 Mb, but currently available CNV callers have been developed and benchmarked for the discovery of smaller variants. Thus, the ability of these programs to detect tens of real syndromic CNVs remains largely unknown. RESULTS: Here we present ConanVarvar, a tool which implements a complete workflow for the targeted analysis of large germline CNVs from WGS data. ConanVarvar comes with an intuitive R Shiny graphical user interface and annotates identified variants with information about 56 associated syndromic conditions. We benchmarked ConanVarvar and four other programs on a dataset containing real and simulated syndromic CNVs larger than 1 Mb. In comparison to other tools, ConanVarvar reports 10-30 times less false-positive variants without compromising sensitivity and is quicker to run, especially on large batches of samples. CONCLUSIONS: ConanVarvar is a useful instrument for primary analysis in disease sequencing studies, where large CNVs could be the cause of disease.


Asunto(s)
Variaciones en el Número de Copia de ADN , Células Germinativas , Secuenciación Completa del Genoma , Flujo de Trabajo , Secuenciación de Nucleótidos de Alto Rendimiento
6.
Am J Hum Genet ; 106(1): 129-136, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31883644

RESUMEN

Birth defects occur in up to 3% of all live births and are the leading cause of infant death. Here we present five individuals from four unrelated families, individuals who share similar phenotypes with disease-causal bi-allelic variants in NADSYN1, encoding NAD synthetase 1, the final enzyme of the nicotinamide adenine dinucleotide (NAD) de novo synthesis pathway. Defects range from the isolated absence of both kidneys to multiple malformations of the vertebrae, heart, limbs, and kidney, and no affected individual survived for more than three months postnatally. NAD is an essential coenzyme for numerous cellular processes. Bi-allelic loss-of-function mutations in genes required for the de novo synthesis of NAD were previously identified in individuals with multiple congenital abnormalities affecting the heart, kidney, vertebrae, and limbs. Functional assessments of NADSYN1 missense variants, through a combination of yeast complementation and enzymatic assays, show impaired enzymatic activity and severely reduced NAD levels. Thus, NADSYN1 represents an additional gene required for NAD synthesis during embryogenesis, and NADSYN1 has bi-allelic missense variants that cause NAD deficiency-dependent malformations. Our findings expand the genotypic spectrum of congenital NAD deficiency disorders and further implicate mutation of additional genes involved in de novo NAD synthesis as potential causes of complex birth defects.


Asunto(s)
Ligasas de Carbono-Nitrógeno con Glutamina como Donante de Amida-N/genética , Anomalías Congénitas/etiología , Insuficiencia Multiorgánica/etiología , Mutación Missense , NAD/deficiencia , Alelos , Secuencia de Aminoácidos , Anomalías Congénitas/patología , Femenino , Genotipo , Edad Gestacional , Humanos , Lactante , Recién Nacido , Masculino , Insuficiencia Multiorgánica/patología , Linaje , Fenotipo , Embarazo , Homología de Secuencia
7.
Proc Natl Acad Sci U S A ; 117(7): 3738-3747, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32015132

RESUMEN

Causes for miscarriages and congenital malformations can be genetic, environmental, or a combination of both. Genetic variants, hypoxia, malnutrition, or other factors individually may not affect embryo development, however, they may do so collectively. Biallelic loss-of-function variants in HAAO or KYNU, two genes of the nicotinamide adenine dinucleotide (NAD) synthesis pathway, are causative of congenital malformation and miscarriage in humans and mice. The variants affect normal embryonic development by disrupting the synthesis of NAD, a key factor in multiple biological processes, from its dietary precursor tryptophan, resulting in NAD deficiency. This study demonstrates that congenital malformations caused by NAD deficiency can occur independent of genetic disruption of NAD biosynthesis. C57BL/6J wild-type mice had offspring exhibiting similar malformations when their supply of the NAD precursors tryptophan and vitamin B3 in the diet was restricted during pregnancy. When the dietary undersupply was combined with a maternal heterozygous variant in Haao, which alone does not cause NAD deficiency or malformations, the incidence of embryo loss and malformations was significantly higher, suggesting a gene-environment interaction. Maternal and embryonic NAD levels were deficient. Mild hypoxia as an additional factor exacerbated the embryo outcome. Our data show that NAD deficiency as a cause of embryo loss and congenital malformation is not restricted to the rare cases of biallelic mutations in NAD synthesis pathway genes. Instead, monoallelic genetic variants and environmental factors can result in similar outcomes. The results expand our understanding of the causes of congenital malformations and the importance of sufficient NAD precursor consumption during pregnancy.


Asunto(s)
Aborto Espontáneo/genética , Anomalías Congénitas/genética , Interacción Gen-Ambiente , NAD/deficiencia , Aborto Espontáneo/metabolismo , Animales , Anomalías Congénitas/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Humanos , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Ratones , Ratones Endogámicos C57BL , Embarazo
8.
Differentiation ; 128: 1-12, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36194927

RESUMEN

Myhre syndrome is a connective tissue disorder characterized by congenital cardiovascular, craniofacial, respiratory, skeletal, and cutaneous anomalies as well as intellectual disability and progressive fibrosis. It is caused by germline variants in the transcriptional co-regulator SMAD4 that localize at two positions within the SMAD4 protein, I500 and R496, with I500 V/T/M variants more commonly identified in individuals with Myhre syndrome. Here we assess the functional impact of SMAD4-I500V variant, identified in two previously unpublished individuals with Myhre syndrome, and provide novel insights into the molecular mechanism of SMAD4-I500V dysfunction. We show that SMAD4-I500V can dimerize, but its transcriptional activity is severely compromised. Our data show that SMAD4-I500V acts dominant-negatively on SMAD4 and on receptor-regulated SMADs, affecting transcription of target genes. Furthermore, SMAD4-I500V impacts the transcription and function of crucial developmental transcription regulator, NKX2-5. Overall, our data reveal a dominant-negative model of disease for SMAD4-I500V where the function of SMAD4 encoded on the remaining allele, and of co-factors, are perturbed by the continued heterodimerization of the variant, leading to dysregulation of TGF and BMP signaling. Our findings not only provide novel insights into the mechanism of Myhre syndrome pathogenesis but also extend the current knowledge of how pathogenic variants in SMAD proteins cause disease.


Asunto(s)
Deformidades Congénitas de la Mano , Discapacidad Intelectual , Humanos , Discapacidad Intelectual/genética , Proteína Smad4/genética , Mutación , Deformidades Congénitas de la Mano/genética , Factor de Crecimiento Transformador beta/genética
9.
Hum Mol Genet ; 29(7): 1068-1082, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31625560

RESUMEN

Pre-B cell leukemia factor 1 (PBX1) is an essential developmental transcription factor, mutations in which have recently been associated with CAKUTHED syndrome, characterized by multiple congenital defects including congenital heart disease (CHD). During analysis of a whole-exome-sequenced cohort of heterogeneous CHD patients, we identified a de novo missense variant, PBX1:c.551G>C p.R184P, in a patient with tetralogy of Fallot with absent pulmonary valve and extra-cardiac phenotypes. Functional analysis of this variant by creating a CRISPR-Cas9 gene-edited mouse model revealed multiple congenital anomalies. Congenital heart defects (persistent truncus arteriosus and ventricular septal defect), hypoplastic lungs, hypoplastic/ectopic kidneys, aplastic adrenal glands and spleen, as well as atretic trachea and palate defects were observed in the homozygous mutant embryos at multiple stages of development. We also observed developmental anomalies in a proportion of heterozygous embryos, suggestive of a dominant mode of inheritance. Analysis of gene expression and protein levels revealed that although Pbx1 transcripts are higher in homozygotes, amounts of PBX1 protein are significantly decreased. Here, we have presented the first functional model of a missense PBX1 variant and provided strong evidence that p.R184P is disease-causal. Our findings also expand the phenotypic spectrum associated with pathogenic PBX1 variants in both humans and mice.


Asunto(s)
Sistemas CRISPR-Cas/genética , Cardiopatías Congénitas/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Tronco Arterial Persistente/genética , Adulto , Animales , Modelos Animales de Enfermedad , Exoma/genética , Femenino , Cardiopatías Congénitas/patología , Heterocigoto , Humanos , Lactante , Masculino , Ratones , Mutación Missense/genética , Linaje , Fenotipo , Tronco Arterial Persistente/patología , Secuenciación del Exoma
10.
Hum Mol Genet ; 29(4): 566-579, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-31813956

RESUMEN

Congenital heart disease (CHD) is the most common birth defect and brings with it significant mortality and morbidity. The application of exome and genome sequencing has greatly improved the rate of genetic diagnosis for CHD but the cause in the majority of cases remains uncertain. It is clear that genetics, as well as environmental influences, play roles in the aetiology of CHD. Here we address both these aspects of causation with respect to the Notch signalling pathway. In our CHD cohort, variants in core Notch pathway genes account for 20% of those that cause disease, a rate that did not increase with the inclusion of genes of the broader Notch pathway and its regulators. This is reinforced by case-control burden analysis where variants in Notch pathway genes are enriched in CHD patients. This enrichment is due to variation in NOTCH1. Functional analysis of some novel missense NOTCH1 and DLL4 variants in cultured cells demonstrate reduced signalling activity, allowing variant reclassification. Although loss-of-function variants in DLL4 are known to cause Adams-Oliver syndrome, this is the first report of a hypomorphic DLL4 allele as a cause of isolated CHD. Finally, we demonstrate a gene-environment interaction in mouse embryos between Notch1 heterozygosity and low oxygen- or anti-arrhythmic drug-induced gestational hypoxia, resulting in an increased incidence of heart defects. This implies that exposure to environmental insults such as hypoxia could explain variable expressivity and penetrance of observed CHD in families carrying Notch pathway variants.


Asunto(s)
Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Genómica/métodos , Cardiopatías Congénitas/patología , Mutación , Receptor Notch1/genética , Animales , Estudios de Casos y Controles , Femenino , Cardiopatías Congénitas/etiología , Cardiopatías Congénitas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Secuenciación del Exoma
11.
Hum Mol Genet ; 29(22): 3662-3678, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33276377

RESUMEN

The genetic causes of multiple congenital anomalies are incompletely understood. Here, we report novel heterozygous predicted loss-of-function (LoF) and predicted damaging missense variants in the WW domain binding protein 11 (WBP11) gene in seven unrelated families with a variety of overlapping congenital malformations, including cardiac, vertebral, tracheo-esophageal, renal and limb defects. WBP11 encodes a component of the spliceosome with the ability to activate pre-messenger RNA splicing. We generated a Wbp11 null allele in mouse using CRISPR-Cas9 targeting. Wbp11 homozygous null embryos die prior to E8.5, indicating that Wbp11 is essential for development. Fewer Wbp11 heterozygous null mice are found than expected due to embryonic and postnatal death. Importantly, Wbp11 heterozygous null mice are small and exhibit defects in axial skeleton, kidneys and esophagus, similar to the affected individuals, supporting the role of WBP11 haploinsufficiency in the development of congenital malformations in humans. LoF WBP11 variants should be considered as a possible cause of VACTERL association as well as isolated Klippel-Feil syndrome, renal agenesis or esophageal atresia.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Unión al ADN/genética , Haploinsuficiencia/genética , Riñón/metabolismo , Factores de Empalme de ARN/genética , Anomalías Múltiples/patología , Canal Anal/anomalías , Canal Anal/patología , Animales , Esófago/anomalías , Esófago/metabolismo , Esófago/patología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Heterocigoto , Humanos , Riñón/anomalías , Riñón/patología , Deformidades Congénitas de las Extremidades/genética , Deformidades Congénitas de las Extremidades/patología , Mutación con Pérdida de Función/genética , Ratones , Empalme del ARN/genética , Columna Vertebral/anomalías , Columna Vertebral/patología , Tráquea/anomalías , Tráquea/patología
12.
Development ; 146(4)2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30787001

RESUMEN

Congenital heart disease (CHD) is the most common type of birth defect. In recent years, research has focussed on identifying the genetic causes of CHD. However, only a minority of CHD cases can be attributed to single gene mutations. In addition, studies have identified different environmental stressors that promote CHD, but the additive effect of genetic susceptibility and environmental factors is poorly understood. In this context, we have investigated the effects of short-term gestational hypoxia on mouse embryos genetically predisposed to heart defects. Exposure of mouse embryos heterozygous for Tbx1 or Fgfr1/Fgfr2 to hypoxia in utero increased the incidence and severity of heart defects while Nkx2-5+/- embryos died within 2 days of hypoxic exposure. We identified the molecular consequences of the interaction between Nkx2-5 and short-term gestational hypoxia, which suggest that reduced Nkx2-5 expression and a prolonged hypoxia-inducible factor 1α response together precipitate embryo death. Our study provides insight into the causes of embryo loss and variable penetrance of monogenic CHD, and raises the possibility that cases of foetal death and CHD in humans could be caused by similar gene-environment interactions.


Asunto(s)
Interacción Gen-Ambiente , Cardiopatías Congénitas/genética , Corazón/embriología , Proteína Homeótica Nkx-2.5/genética , Proteínas de Homeodominio/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Animales , Apoptosis , Proliferación Celular , Embrión de Mamíferos/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Corazón/diagnóstico por imagen , Heterocigoto , Proteína Homeótica Nkx-2.5/fisiología , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxígeno/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Proteínas de Dominio T Box/genética , Factores de Tiempo
13.
Am Heart J ; 254: 166-171, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36115390

RESUMEN

Congenital heart disease (CHD) has a multifactorial aetiology, raising the possibility of an underlying genetic burden, predisposing to disease but also variable expression, including variation in disease severity, and incomplete penetrance. Using whole genome sequencing (WGS), the findings of this study, indicate that complex, critical CHD is distinct from other types of disease due to increased genetic burden in common variation, specifically among established CHD genes. Additionally, these findings highlight associations with regulatory genes and environmental "stressors" in the final presentation of disease.


Asunto(s)
Cardiopatías Congénitas , Humanos , Cardiopatías Congénitas/genética
14.
Am Heart J ; 244: 1-13, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34670123

RESUMEN

BACKGROUND: The most common cyanotic congenital heart disease (CHD) requiring management as a neonate is transposition of great arteries (TGA). Clinically, up to 50% of TGA patients develop some form of neurodevelopmental disability (NDD), thought to have a significant genetic component. A "ciliopathy" and links with laterality disorders have been proposed. This first report of whole genome sequencing in TGA, sought to identify clinically relevant variants contributing to heart, brain and laterality defects. METHODS: Initial whole genome sequencing analyses on 100 TGA patients focussed on established disease genes related to CHD (n = 107), NDD (n = 659) and heterotaxy (n = 74). Single variant as well as copy number variant analyses were conducted. Variant pathogenicity was assessed using the American College of Medical Genetics and Genomics-Association for Molecular Pathology guidelines. RESULTS: Fifty-five putatively damaging variants were identified in established disease genes associated with CHD, NDD and heterotaxy; however, no clinically relevant variants could be attributed to disease. Notably, case-control analyses identified significantly more predicted-damaging, silent and total variants in TGA cases than healthy controls in established CHD genes (P < .001), NDD genes (P < .001) as well as across the three gene panels (P < .001). CONCLUSION: We present compelling evidence that the majority of TGA is not caused by monogenic rare variants and is most likely oligogenic and/or polygenic in nature, highlighting the complex genetic architecture and multifactorial influences on this CHD sub-type and its long-term sequelae. Assessment of variant burden in key heart, brain and/or laterality genes may be required to unravel the genetic contributions to TGA and related disabilities.


Asunto(s)
Cardiopatías Congénitas , Transposición de los Grandes Vasos , Arterias , Encéfalo/diagnóstico por imagen , Cardiopatías Congénitas/genética , Humanos , Recién Nacido , Transposición de los Grandes Vasos/genética , Secuenciación Completa del Genoma
15.
FASEB J ; 35(9): e21833, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34365659

RESUMEN

Macrophages are the principal component of the innate immune system. They play very crucial and multifaceted roles in the pathogenesis of inflammatory vascular diseases. There is an increasing recognition that transcriptionally dynamic macrophages are the key players in the pathogenesis of inflammatory vascular diseases. In this context, the accumulation and aberrant activation of macrophages in the subendothelial layers govern atherosclerotic plaque development. Macrophage-mediated inflammation is an explicitly robust biological response that involves broad alterations in inflammatory gene expression. Thus, cell-intrinsic negative regulatory mechanisms must exist which can restrain inflammatory response in a spatiotemporal manner. In this study, we identified CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxyl-terminal domain 2 (CITED2) as one such cell-intrinsic negative regulator of inflammation. Our in vivo studies show that myeloid-CITED2-deficient mice on the Apoe-/- background have larger atherosclerotic lesions on both control and high-fat/high-cholesterol diets. Our integrated transcriptomics and gene set enrichment analyses studies show that CITED2 deficiency elevates STAT1 and interferon regulatory factor 1 (IRF1) regulated pro-inflammatory gene expression in macrophages. At the molecular level, our studies identify that CITED2 deficiency elevates IFNγ-induced STAT1 transcriptional activity and STAT1 enrichment on IRF1 promoter in macrophages. More importantly, siRNA-mediated knockdown of IRF1 completely reversed elevated pro-inflammatory target gene expression in CITED2-deficient macrophages. Collectively, our study findings demonstrate that CITED2 restrains the STAT1-IRF1 signaling axis in macrophages and limits the development of atherosclerotic plaques.


Asunto(s)
Aterosclerosis/genética , Factor 1 Regulador del Interferón/genética , Proteínas Represoras/genética , Factor de Transcripción STAT1/genética , Transducción de Señal/genética , Transactivadores/genética , Animales , Femenino , Inflamación/genética , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Células RAW 264.7 , Transcripción Genética/genética
16.
Hum Mutat ; 42(7): 862-876, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33942433

RESUMEN

Nicotinamide adenine dinucleotide (NAD) is an essential coenzyme involved in over 400 cellular reactions. During embryogenesis, mammals synthesize NAD de novo from dietary l -tryptophan via the kynurenine pathway. Biallelic, inactivating variants in three genes encoding enzymes of this biosynthesis pathway (KYNU, HAAO, and NADSYN1) disrupt NAD synthesis and have been identified in patients with multiple malformations of the heart, kidney, vertebrae, and limbs; these patients have Congenital NAD Deficiency Disorder HAAO and four families with biallelic variants in KYNU. These patients present similarly with multiple malformations of the heart, kidney, vertebrae, and limbs, of variable severity. We show that each variant identified in these patients results in loss-of-function, revealed by a significant reduction in NAD levels via yeast genetic complementation assays. For the first time, missense mutations are identified as a cause of malformation and shown to disrupt enzyme function. These missense and frameshift variants cause moderate to severe NAD deficiency in yeast, analogous to insufficient synthesized NAD in patients. We hereby expand the genotypic and corresponding phenotypic spectrum of Congenital NAD Deficiency Disorder.


Asunto(s)
NAD , Columna Vertebral , Animales , Genotipo , Humanos , Mamíferos , Mutación Missense , Columna Vertebral/anomalías
17.
Anal Biochem ; 633: 114409, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34648806

RESUMEN

Nicotinamide adenine dinucleotide (NAD) is a key metabolic intermediate found in all cells and involved in numerous cellular functions. Perturbances in the NAD metabolome are linked to various diseases such as diabetes and schizophrenia, and to congenital malformations and recurrent miscarriage. Mouse models are central to the investigation of these and other NAD-related conditions because mice can be readily genetically modified and treated with diets with altered concentrations of NAD precursors. Simultaneous quantification of as many metabolites of the NAD metabolome as possible is required to understand which pathways are affected in these disease conditions and what are the functional consequences. Here, we report the development of a fit-for-purpose method to simultaneously quantify 26 NAD-related metabolites and creatinine in mouse plasma, whole blood, and liver tissue using ultra-high performance liquid chromatography - tandem mass spectrometry (UHPLC-MS/MS). The included metabolites represent dietary precursors, intermediates, enzymatic cofactors, and excretion products. Sample preparation was optimized for each matrix and included 21 isotope-labeled internal standards. The method reached adequate precision and accuracy for the intended context of use of exploratory pathway-related biomarker discovery in mouse models. The method was tested by determining metabolite concentrations in mice fed a special diet with defined precursor content.


Asunto(s)
Hígado/química , NAD/análisis , Animales , Cromatografía Líquida de Alta Presión , Femenino , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , NAD/metabolismo , Espectrometría de Masas en Tándem
18.
FASEB J ; 34(9): 12100-12113, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32697413

RESUMEN

Monocyte-derived macrophages are the major innate immune cells that provide the first line of cellular defense against infections or injuries. These recruited macrophages at the site of inflammation are exposed to a broad range of cytokines that categorically incite a robust pro-inflammatory response. However, macrophage pro-inflammatory activation must be under exquisite control to avert unbridled inflammation. Thus, endogenous mechanisms must exist that rigorously preserve macrophage quiescence and yet, allow nimble pro-inflammatory macrophage response with precise spatiotemporal control. Herein, we identify the CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxyl-terminal domain 2 (CITED2) as a critical intrinsic negative regulator of inflammation, which broadly attenuates pro-inflammatory gene programs in macrophages. Our in vivo studies revealed that myeloid-CITED2 deficiency significantly heightened macrophages and neutrophils recruitment to the site of inflammation. Our integrated transcriptomics and gene set enrichment analysis (GSEA) studies uncovered that CITED2 deficiency broadly enhances NFκB targets, IFNγ/IFNα responses, and inflammatory response gene expression in macrophages. Using complementary gain- and loss-of-function studies, we observed that CITED2 overexpression attenuate and CITED2 deficiency elevate LPS-induced NFκB transcriptional activity and NFκB-p65 recruitment to target gene promoter in macrophages. More importantly, blockade of NFκB signaling completely reversed elevated pro-inflammatory gene expression in macrophages. Collectively, our findings show that CITED2 restrains NFκB activation and curtails broad pro-inflammatory gene programs in myeloid cells.


Asunto(s)
Regulación de la Expresión Génica , Macrófagos/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Animales , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/toxicidad , Macrófagos/patología , Ratones , Neutrófilos/metabolismo , Neutrófilos/patología , Células RAW 264.7 , Proteínas Represoras/genética , Transactivadores/genética , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
19.
Circ Res ; 124(4): 553-563, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30582441

RESUMEN

RATIONALE: Familial recurrence studies provide strong evidence for a genetic component to the predisposition to sporadic, nonsyndromic Tetralogy of Fallot (TOF), the most common cyanotic congenital heart disease phenotype. Rare genetic variants have been identified as important contributors to the risk of congenital heart disease, but relatively small numbers of TOF cases have been studied to date. OBJECTIVE: We used whole exome sequencing to assess the prevalence of unique, deleterious variants in the largest cohort of nonsyndromic TOF patients reported to date. METHODS AND RESULTS: Eight hundred twenty-nine TOF patients underwent whole exome sequencing. The presence of unique, deleterious variants was determined; defined by their absence in the Genome Aggregation Database and a scaled combined annotation-dependent depletion score of ≥20. The clustering of variants in 2 genes, NOTCH1 and FLT4, surpassed thresholds for genome-wide significance (assigned as P<5×10-8) after correction for multiple comparisons. NOTCH1 was most frequently found to harbor unique, deleterious variants. Thirty-one changes were observed in 37 probands (4.5%; 95% CI, 3.2%-6.1%) and included 7 loss-of-function variants 22 missense variants and 2 in-frame indels. Sanger sequencing of the unaffected parents of 7 cases identified 5 de novo variants. Three NOTCH1 variants (p.G200R, p.C607Y, and p.N1875S) were subjected to functional evaluation, and 2 showed a reduction in Jagged1-induced NOTCH signaling. FLT4 variants were found in 2.4% (95% CI, 1.6%-3.8%) of TOF patients, with 21 patients harboring 22 unique, deleterious variants. The variants identified were distinct to those that cause the congenital lymphoedema syndrome Milroy disease. In addition to NOTCH1, FLT4 and the well-established TOF gene, TBX1, we identified potential association with variants in several other candidates, including RYR1, ZFPM1, CAMTA2, DLX6, and PCM1. CONCLUSIONS: The NOTCH1 locus is the most frequent site of genetic variants predisposing to nonsyndromic TOF, followed by FLT4. Together, variants in these genes are found in almost 7% of TOF patients.


Asunto(s)
Exoma , Tasa de Mutación , Tetralogía de Fallot/genética , Autoantígenos/genética , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Proteínas de Homeodominio/genética , Humanos , Mutación con Pérdida de Función , Mutación Missense , Proteínas Nucleares/genética , Receptor Notch1/genética , Transactivadores/genética , Factores de Transcripción/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
20.
J Biol Chem ; 294(21): 8617-8629, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30967472

RESUMEN

We previously reported that the cell cycle-related cyclin-dependent kinase 4-retinoblastoma (RB) transcriptional corepressor pathway is essential for stroke-induced cell death both in vitro and in vivo However, how this signaling pathway induces cell death is unclear. Previously, we found that the cyclin-dependent kinase 4 pathway activates the pro-apoptotic transcriptional co-regulator Cited2 in vitro after DNA damage. In the present study, we report that Cited2 protein expression is also dramatically increased following stroke/ischemic insult. Critically, utilizing conditional knockout mice, we show that Cited2 is required for neuronal cell death, both in culture and in mice after ischemic insult. Importantly, determining the mechanism by which Cited2 levels are regulated, we found that E2F transcription factor (E2F) family members participate in Cited2 regulation. First, E2F1 expression induced Cited2 transcription, and E2F1 deficiency reduced Cited2 expression. Moreover, determining the potential E2F-binding regions on the Cited2 gene regulatory sequence by ChIP analysis, we provide evidence that E2F1/4 proteins bind to this DNA region. A luciferase reporter assay to probe the functional outcomes of this interaction revealed that E2F1 activates and E2F4 inhibits Cited2 transcription. Moreover, we identified the functional binding motif for E2F1 in the Cited2 gene promoter by demonstrating that mutation of this site dramatically reduces E2F1-mediated Cited2 transcription. Finally, E2F1 and E2F4 regulated Cited2 expression in neurons after stroke-related insults. Taken together, these results indicate that the E2F-Cited2 regulatory pathway is critically involved in stroke injury.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F4/metabolismo , Regulación de la Expresión Génica , Neuronas/metabolismo , Proteínas Represoras/biosíntesis , Accidente Cerebrovascular/metabolismo , Transactivadores/biosíntesis , Secuencias de Aminoácidos , Animales , Muerte Celular , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F4/genética , Ratones , Ratones Transgénicos , Neuronas/patología , Proteínas Represoras/genética , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Transactivadores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA