Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Blood Adv ; 7(17): 4782-4793, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36399516

RESUMO

Sickle cell disease nephropathy (SCDN), a common SCD complication, is strongly associated with mortality. Polygenic risk scores calculated from recent transethnic meta-analyses of urinary albumin-to-creatinine ratio and estimated glomerular filtration rate (eGFR) trended toward association with proteinuria and eGFR in SCD but the model fit was poor (R2 < 0.01), suggesting that there are likely unique genetic risk factors for SCDN. Therefore, we performed genome-wide association studies (GWAS) for 2 critical manifestations of SCDN, proteinuria and decreased eGFR, in 2 well-characterized adult SCD cohorts, representing, to the best of our knowledge, the largest SCDN sample to date. Meta-analysis identified 6 genome-wide significant associations (false discovery rate, q ≤ 0.05): 3 for proteinuria (CRYL1, VWF, and ADAMTS7) and 3 for eGFR (LRP1B, linc02288, and FPGT-TNNI3K/TNNI3K). These associations are independent of APOL1 risk and represent novel SCDN loci, many with evidence for regulatory function. Moreover, GWAS SNPs in CRYL1, VWF, ADAMTS7, and linc02288 are associated with gene expression in kidney and pathways important to both renal function and SCD biology, supporting the hypothesis that SCDN pathophysiology is distinct from other forms of kidney disease. Together, these findings provide new targets for functional follow-up that could be tested prospectively and potentially used to identify patients with SCD who are at risk, before onset of kidney dysfunction.


Assuntos
Anemia Falciforme , Nefropatias , Doenças Vasculares , Adulto , Humanos , Estudo de Associação Genômica Ampla , Proteína ADAMTS7/genética , Fator de von Willebrand/genética , Nefropatias/genética , Anemia Falciforme/complicações , Anemia Falciforme/genética , Proteinúria/complicações , Proteínas Serina-Treonina Quinases/genética , Apolipoproteína L1/genética
2.
JCI Insight ; 7(11)2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35472001

RESUMO

COVID-19 infection causes collapse of glomerular capillaries and loss of podocytes, culminating in a severe kidney disease called COVID-19-associated nephropathy (COVAN). The underlying mechanism of COVAN is unknown. We hypothesized that cytokines induced by COVID-19 trigger expression of pathogenic APOL1 via JAK/STAT signaling, resulting in podocyte loss and COVAN phenotype. Here, based on 9 biopsy-proven COVAN cases, we demonstrated for the first time, to the best of our knowledge, that APOL1 protein was abundantly expressed in podocytes and glomerular endothelial cells (GECs) of COVAN kidneys but not in controls. Moreover, a majority of patients with COVAN carried 2 APOL1 risk alleles. We show that recombinant cytokines induced by SARS-CoV-2 acted synergistically to drive APOL1 expression through the JAK/STAT pathway in primary human podocytes, GECs, and kidney micro-organoids derived from a carrier of 2 APOL1 risk alleles, but expression was blocked by a JAK1/2 inhibitor, baricitinib. We demonstrate that cytokine-induced JAK/STAT/APOL1 signaling reduced the viability of kidney organoid podocytes but was rescued by baricitinib. Together, our results support the conclusion that COVID-19-induced cytokines are sufficient to drive COVAN-associated podocytopathy via JAK/STAT/APOL1 signaling and that JAK inhibitors could block this pathogenic process. These findings suggest JAK inhibitors may have therapeutic benefits for managing cytokine-induced, APOL1-mediated podocytopathy.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19 , Citocinas , Inibidores de Janus Quinases , Nefropatias , Apolipoproteína L1/genética , Azetidinas/farmacologia , COVID-19/metabolismo , Citocinas/metabolismo , Células Endoteliais/metabolismo , Humanos , Inibidores de Janus Quinases/farmacologia , Janus Quinases/metabolismo , Nefropatias/tratamento farmacológico , Nefropatias/metabolismo , Nefropatias/virologia , Organoides/metabolismo , Purinas/farmacologia , Pirazóis/farmacologia , SARS-CoV-2/isolamento & purificação , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
3.
Nat Commun ; 11(1): 5903, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33214552

RESUMO

The neuronal primary cilium and centriolar satellites have functions in neurogenesis, but little is known about their roles in the postnatal brain. We show that ablation of pericentriolar material 1 in the mouse leads to progressive ciliary, anatomical, psychomotor, and cognitive abnormalities. RNAseq reveals changes in amine- and G-protein coupled receptor pathways. The physiological relevance of this phenotype is supported by decreased available dopamine D2 receptor (D2R) levels and the failure of antipsychotic drugs to rescue adult behavioral defects. Immunoprecipitations show an association with Pcm1 and D2Rs. Finally, we sequence PCM1 in two human cohorts with severe schizophrenia. Systematic modeling of all discovered rare alleles by zebrafish in vivo complementation reveals an enrichment for pathogenic alleles. Our data emphasize a role for the pericentriolar material in the postnatal brain, with progressive degenerative ciliary and behavioral phenotypes; and they support a contributory role for PCM1 in some individuals diagnosed with schizophrenia.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Cílios/patologia , Predisposição Genética para Doença/genética , Esquizofrenia/genética , Adulto , Idoso , Alelos , Aminas/metabolismo , Animais , Antipsicóticos/uso terapêutico , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cílios/metabolismo , Resistência a Medicamentos/genética , Humanos , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Fenótipo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Esquizofrenia/tratamento farmacológico , Esquizofrenia/patologia , Esquizofrenia/fisiopatologia , Transdução de Sinais , Adulto Jovem , Peixe-Zebra
4.
PLoS One ; 14(6): e0217042, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31158233

RESUMO

Kidney failure occurs in 5-13% of individuals with sickle cell disease and is associated with early mortality. Two APOL1 alleles (G1 and G2) have been identified as risk factors for sickle cell disease nephropathy. Both risk alleles are prevalent in individuals with recent African ancestry and have been associated with nephropathic complications in other diseases. Despite the association of G1 and G2 with kidney dysfunction, the mechanisms by which these variants contribute to increased risk remain poorly understood. Previous work in zebrafish models suggest that the G2 risk allele functions as a dominant negative, whereas the G1 allele is a functional null. To understand better the cellular pathology attributed to APOL1 G2, we investigated the in vivo effects of the G2 risk variant on distinct cell types using RNA sequencing. We surveyed APOL1 G2 associated transcriptomic alterations in podocytes and vascular endothelial cells isolated from zebrafish larvae expressing cell-type specific reporters. Our analysis identified many transcripts (n = 7,523) showing differential expression between APOL1 G0 (human wild-type) and APOL1 G2 exposed podocytes. Conversely, relatively few transcripts (n = 107) were differentially expressed when comparing APOL1 G0 and APOL1 G2 exposed endothelial cells. Pathway analysis of differentially expressed transcripts in podocytes showed enrichment for autophagy associated terms such as "Lysosome" and "Phagosome", implicating these pathways in APOL1 G2 associated kidney dysfunction. This work provides insight into the molecular pathology of APOL1 G2 nephropathy which may offer new therapeutic strategies for multiple disease contexts such as sickle cell nephropathy.


Assuntos
Anemia Falciforme/patologia , Apolipoproteína L1/genética , Variação Genética , Nefropatias/patologia , Podócitos/patologia , Análise de Sequência de RNA , Peixe-Zebra , Animais , Expressão Gênica , Predisposição Genética para Doença/genética , Humanos , Larva/genética , Podócitos/metabolismo , RNA Mensageiro/genética , Risco , Transcrição Gênica
5.
Am J Hematol ; 93(12): 1451-1460, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30144150

RESUMO

Sickle cell disease (SCD) nephropathy and lower estimated glomerular filtration rate (eGFR) are risk factors for early mortality. Furthermore, rate of eGFR decline predicts progression to end-stage renal disease in many clinical settings. However, factors predicting renal function decline in SCD are poorly documented. Using clinical, laboratory, genetic, and metabolomic data, we evaluated predictors of renal function decline in a longitudinal cohort of 288 adults (mean age 33.0 years). In 193 subjects with 5-year follow-up data, mean rate of eGFR decline was 2.35 mL/min/1.73 m2 /year, nearly twice that of African American adults overall. Hyperfiltration was prevalent at baseline (61.1%), and 36.8% of subjects experienced rapid eGFR decline (≥3 mL/min/1.73 m2 /year). Severe Hb genotype; proteinuria; higher platelet and reticulocyte counts, and systolic BP; and lower Hb level and BMI were associated with rapid decline. A risk scoring system was created using these 7 variables and was highly predictive of rapid eGFR decline, with odds of rapid decline increasing 1.635-fold for every point increment (P < 0.0001). Rapid eGFR decline was also associated with higher organ system severity score and peak creatinine. Additionally, two metabolites (asymmetric dimethylarginine and quinolinic acid) were associated with rapid decline. Further investigation into longitudinal SCD nephropathy (SCDN) trajectory, early markers of SCDN, and tools for risk stratification should inform interventional studies targeted to slowing GFR decline and improving SCD outcomes.


Assuntos
Anemia Falciforme/complicações , Progressão da Doença , Taxa de Filtração Glomerular , Insuficiência Renal Crônica/etiologia , Adulto , Anemia Falciforme/fisiopatologia , Creatinina/sangue , Feminino , Humanos , Masculino , Medição de Risco , Fatores de Risco
6.
BMC Genomics ; 16: 11, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25609184

RESUMO

BACKGROUND: Expression quantitative trait loci (eQTL) play an important role in the regulation of gene expression. Gene expression levels and eQTLs are expected to vary from tissue to tissue, and therefore multi-tissue analyses are necessary to fully understand complex genetic conditions in humans. Dura mater tissue likely interacts with cranial bone growth and thus may play a role in the etiology of Chiari Type I Malformation (CMI) and related conditions, but it is often inaccessible and its gene expression has not been well studied. A genetic basis to CMI has been established; however, the specific genetic risk factors are not well characterized. RESULTS: We present an assessment of eQTLs for whole blood and dura mater tissue from individuals with CMI. A joint-tissue analysis identified 239 eQTLs in either dura or blood, with 79% of these eQTLs shared by both tissues. Several identified eQTLs were novel and these implicate genes involved in bone development (IPO8, XYLT1, and PRKAR1A), and ribosomal pathways related to marrow and bone dysfunction, as potential candidates in the development of CMI. CONCLUSIONS: Despite strong overall heterogeneity in expression levels between blood and dura, the majority of cis-eQTLs are shared by both tissues. The power to detect shared eQTLs was improved by using an integrative statistical approach. The identified tissue-specific and shared eQTLs provide new insight into the genetic basis for CMI and related conditions.


Assuntos
Malformação de Arnold-Chiari/genética , Locos de Características Quantitativas , Adolescente , Malformação de Arnold-Chiari/patologia , Desenvolvimento Ósseo/genética , Criança , Pré-Escolar , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/sangue , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Dura-Máter/metabolismo , Feminino , Redes Reguladoras de Genes , Genótipo , Humanos , Masculino , Pentosiltransferases/sangue , Pentosiltransferases/genética , Pentosiltransferases/metabolismo , Polimorfismo de Nucleotídeo Único , beta Carioferinas/sangue , beta Carioferinas/genética , beta Carioferinas/metabolismo , UDP Xilose-Proteína Xilosiltransferase
7.
Birth Defects Res A Clin Mol Teratol ; 100(8): 642-6, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25044326

RESUMO

BACKGROUND: Neural tube defects (NTD) have a strong genetic component, with up to 70% of variance in human prevalence determined by heritable factors. Although the identification of causal DNA variants by sequencing candidate genes from functionally relevant pathways and model organisms has provided some success, alternative approaches are demanded. METHODS: Next generation sequencing platforms are facilitating the production of massive amounts of sequencing data, primarily from the protein coding regions of the genome, at a faster rate and cheaper cost than has previously been possible. These platforms are permitting the identification of variants (de novo, rare, and common) that are drivers of NYTD etiology, and the cost of the approach allows for the screening of increased numbers of affected and unaffected individuals from NTD families and in simplex cases. CONCLUSION: The next generation sequencing platforms represent a powerful tool in the armory of the genetics researcher to identify the causal genetic basis of NTDs.


Assuntos
Exoma/genética , Predisposição Genética para Doença , Defeitos do Tubo Neural/genética , Sequência de Bases , Variação Genética , Estudo de Associação Genômica Ampla , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Tubo Neural/embriologia , Análise de Sequência de DNA
8.
Birth Defects Res B Dev Reprod Toxicol ; 98(5): 365-73, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24323870

RESUMO

Neural tube defects (NTDs) are caused by improper neural tube closure during the early stages of embryonic development. NTDs are hypothesized to have a complex genetic origin and numerous candidate genes have been proposed. The nitric oxide synthase 3 (NOS3) G594T polymorphism has been implicated in risk for spina bifida, and interactions between that single nucleotide polymorphism (SNP) and the methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism have also been observed. To evaluate other genetic variation in the NO pathway in the development of NTDs, we examined all three NOS genes: NOS1, NOS2, and NOS3. Using 3109 Caucasian samples in 745 families, we evaluated association in the overall dataset and within specific phenotypic subsets. Haplotype tagging SNPs in the NOS genes were tested for genetic association with NTD subtypes, both for main effects as well as for the presence of interactions with the MTHFR C677T polymorphism. Nominal main effect associations were found with all subtypes, across all three NOS genes, and interactions were observed between SNPs in all three NOS genes and MTHFR C677T. Unlike the previous report, the most significant associations in our dataset were with cranial subtypes and the AG genotype of rs4795067 in NOS2 (p = 0.0014) and the interaction between the rs9658490 G allele in NOS1 and MTHFR 677TT genotype (p = 0.0014). Our data extend the previous findings by implicating a role for all three NOS genes, independently and through interactions with MTHFR, in risk not only for spina bifida, but all NTD subtypes.


Assuntos
Defeitos do Tubo Neural/genética , Óxido Nítrico Sintase/genética , Polimorfismo de Nucleotídeo Único , Genótipo , Haplótipos , Humanos , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo III/genética , Fenótipo
9.
J Biol Chem ; 279(44): 46162-71, 2004 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-15292259

RESUMO

GRP94 is the endoplasmic reticulum paralog of cytoplasmic Hsp90. Models of Hsp90 action posit an ATP-dependent conformational switch in the N-terminal ligand regulatory domain of the chaperone. However, crystal structures of the isolated N-domain of Hsp90 in complex with a variety of ligands have yet to demonstrate such a conformational change. We have determined the structure of the N-domain of GRP94 in complex with ATP, ADP, and AMP. Compared with the N-ethylcarboxamidoadenosine and radicicol-bound forms, these structures reveal a large conformational rearrangement in the protein. The nucleotide-bound form exposes new surfaces that interact to form a biochemically plausible dimer that is reminiscent of those seen in structures of MutL and DNA gyrase. Weak ATP binding and a conformational change in response to ligand identity are distinctive mechanistic features of GRP94 and suggest a model for how GRP94 functions in the absence of co-chaperones and ATP hydrolysis.


Assuntos
Proteínas de Choque Térmico HSP70/química , Proteínas de Membrana/química , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Animais , Cristalização , DNA Girase/química , Cães , Proteínas de Choque Térmico HSP90/química , Ligantes , Conformação Proteica
10.
J Biol Chem ; 278(48): 48330-8, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-12970348

RESUMO

GRP94, the endoplasmic reticulum (ER) paralog of the chaperone Hsp90, plays an essential role in the structural maturation or secretion of a subset of proteins destined for transport to the cell surface, such as the Toll-like receptors 2 and 4, and IgG, respectively. GRP94 differs from cytoplasmic Hsp90 by exhibiting very weak ATP binding and hydrolysis activity. GRP94 also binds selectively to a series of substituted adenosine analogs. The high resolution crystal structures at 1.75-2.1 A of the N-terminal and adjacent charged domains of GRP94 in complex with N-ethylcarboxamidoadenosine, radicicol, and 2-chlorodideoxyadenosine reveals a structural mechanism for ligand discrimination among hsp90 family members. The structures also identify a putative subdomain that may act as a ligand-responsive switch. The residues of the charged region fold into a disordered loop whose termini are ordered and continue the twisted beta sheet that forms the structural core of the N-domain. This continuation of the beta sheet past the charged domain suggests a structural basis for the association of the N-terminal and middle domains of the full-length chaperone.


Assuntos
Didesoxiadenosina/análogos & derivados , Proteínas de Choque Térmico HSP70/química , Proteínas de Membrana/química , Adenosina/química , Adenosina-5'-(N-etilcarboxamida)/química , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Citoplasma/metabolismo , Didesoxiadenosina/química , Dimerização , Cães , Elétrons , Retículo Endoplasmático/metabolismo , Glutationa Transferase/metabolismo , Proteínas de Choque Térmico HSP90/química , Lactonas/química , Ligantes , Macrolídeos , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA