Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Am Soc Nephrol ; 32(10): 2517-2528, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34088853

RESUMO

BACKGROUND: AKI is a complication of coronavirus disease 2019 (COVID-19) that is associated with high mortality. Despite documented kidney tropism of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there are no consistent reports of viral detection in urine or correlation with AKI or COVID-19 severity. Here, we hypothesize that quantification of the viral load of SARS-CoV-2 in urine sediment from patients with COVID-19 correlates with occurrence of AKI and mortality. METHODS: The viral load of SARS-CoV-2 in urine sediments (U-viral load) was quantified by qRT-PCR in 52 patients with PCR-confirmed COVID-19 diagnosis, who were hospitalized between March 15 and June 8, 2020. Immunolabeling of SARS-CoV-2 proteins Spike and Nucleocapsid was performed in two COVID-19 kidney biopsy specimens and urine sediments. Viral infectivity assays were performed from 32 urine sediments. RESULTS: A total of 20 patients with COVID-19 (39%) had detectable SARS-CoV-2 U-viral load, of which 17 (85%) developed AKI with an average U-viral load four-times higher than patients with COVID-19 who did not have AKI. U-viral load was highest (7.7-fold) within 2 weeks after AKI diagnosis. A higher U-viral load correlated with mortality but not with albuminuria or AKI stage. SARS-CoV-2 proteins partially colocalized with the viral receptor ACE2 in kidney biopsy specimens in tubules and parietal cells, and in urine sediment cells. Infective SARS-CoV-2 was not detected in urine sediments. CONCLUSION: Our results further support SARS-CoV-2 kidney tropism. A higher SARS-CoV-2 viral load in urine sediments from patients with COVID-19 correlated with increased incidence of AKI and mortality. Urinary viral detection could inform the medical care of patients with COVID-19 and kidney injury to improve prognosis.


Assuntos
Injúria Renal Aguda/virologia , COVID-19/complicações , SARS-CoV-2/isolamento & purificação , Carga Viral , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/urina , Adulto , Idoso , Enzima de Conversão de Angiotensina 2/análise , COVID-19/urina , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Urina/virologia
2.
JCI Insight ; 3(21)2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30385718

RESUMO

Elevated blood pressure (BP) and renal dysfunction are complex traits representing major global health problems. Single nucleotide polymorphisms identified by genome-wide association studies have identified the Alström syndrome 1 (ALMS1) gene locus to render susceptibility for renal dysfunction, hypertension, and chronic kidney disease (CKD). Mutations in the ALMS1 gene in humans causes Alström syndrome, characterized by progressive metabolic alterations including hypertension and CKD. Despite compelling genetic evidence, the underlying biological mechanism by which mutations in the ALMS1 gene lead to the above-mentioned pathophysiology is not understood. We modeled this effect in a KO rat model and showed that ALMS1 genetic deletion leads to hypertension. We demonstrate that the link between ALMS1 and hypertension involves the activation of the renal Na+/K+/2Cl- cotransporter NKCC2, mediated by regulation of its endocytosis. Our findings establish a link between the genetic susceptibility to hypertension, CKD, and the expression of ALMS1 through its role in a salt-reabsorbing tubular segment of the kidney. These data point to ALMS1 as a potentially novel gene involved in BP and renal function regulation.


Assuntos
Síndrome de Alstrom/genética , Hipertensão/metabolismo , Proteínas/genética , Insuficiência Renal Crônica/metabolismo , Síndrome de Alstrom/diagnóstico , Síndrome de Alstrom/fisiopatologia , Animais , Proteínas de Ciclo Celular , Endocitose/fisiologia , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla/métodos , Hipertensão/fisiopatologia , Masculino , Modelos Animais , Mutação , Polimorfismo de Nucleotídeo Único/genética , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Insuficiência Renal Crônica/fisiopatologia , Membro 1 da Família 12 de Carreador de Soluto/metabolismo
3.
Nat Cell Biol ; 18(7): 752-764, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27214281

RESUMO

WNT ligands induce Ca(2+) signalling on target cells. PKD1 (polycystin 1) is considered an orphan, atypical G-protein-coupled receptor complexed with TRPP2 (polycystin 2 or PKD2), a Ca(2+)-permeable ion channel. Inactivating mutations in their genes cause autosomal dominant polycystic kidney disease (ADPKD), one of the most common genetic diseases. Here, we show that WNTs bind to the extracellular domain of PKD1 and induce whole-cell currents and Ca(2+) influx dependent on TRPP2. Pathogenic PKD1 or PKD2 mutations that abrogate complex formation, compromise cell surface expression of PKD1, or reduce TRPP2 channel activity suppress activation by WNTs. Pkd2(-/-) fibroblasts lack WNT-induced Ca(2+) currents and are unable to polarize during directed cell migration. In Xenopus embryos, pkd1, Dishevelled 2 (dvl2) and wnt9a act within the same pathway to preserve normal tubulogenesis. These data define PKD1 as a WNT (co)receptor and implicate defective WNT/Ca(2+) signalling as one of the causes of ADPKD.


Assuntos
Cálcio/metabolismo , Via de Sinalização Wnt , Animais , Membrana Celular/metabolismo , Proteínas Desgrenhadas/metabolismo , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Ligação Proteica , Canais de Cátion TRPP/metabolismo , Xenopus
4.
Cells ; 5(1)2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26840332

RESUMO

Cilia and flagella are evolutionarily conserved, membrane-bound, microtubule-based organelles on the surface of most eukaryotic cells. They play important roles in coordinating a variety of signaling pathways during growth, development, cell mobility, and tissue homeostasis. Defects in ciliary structure or function are associated with multiple human disorders called ciliopathies. These diseases affect diverse tissues, including, but not limited to the eyes, kidneys, brain, and lungs. Many processes must be coordinated simultaneously in order to initiate ciliogenesis. These include cell cycle, vesicular trafficking, and axonemal extension. Centrioles play a central role in both cell cycle progression and ciliogenesis, making the transition between basal bodies and mitotic spindle organizers integral to both processes. The maturation of centrioles involves a functional shift from cell division toward cilium nucleation which takes place concurrently with its migration and fusion to the plasma membrane. Several proteinaceous structures of the distal appendages in mother centrioles are required for this docking process. Ciliary assembly and maintenance requires a precise balance between two indispensable processes; so called assembly and disassembly. The interplay between them determines the length of the resulting cilia. These processes require a highly conserved transport system to provide the necessary substances at the tips of the cilia and to recycle ciliary turnover products to the base using a based microtubule intraflagellar transport (IFT) system. In this review; we discuss the stages of ciliogenesis as well as mechanisms controlling the lengths of assembled cilia.

5.
EMBO J ; 34(19): 2424-40, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26206584

RESUMO

Primary cilia start forming within the G1 phase of the cell cycle and continue to grow as cells exit the cell cycle (G0). They start resorbing when cells re-enter the cell cycle (S phase) and are practically invisible in mitosis. The mechanisms by which cilium biogenesis and disassembly are coupled to the cell cycle are complex and not well understood. We previously identified the centrosomal phosphoprotein NDE1 as a negative regulator of ciliary length and showed that its levels inversely correlate with ciliogenesis. Here, we identify the tumor suppressor FBW7 (also known as FBXW7, CDC4, AGO, or SEL-10) as the E3 ligase that mediates the destruction of NDE1 upon entry into G1. CDK5, a kinase active in G1/G0, primes NDE1 for FBW7-mediated recognition. Cells depleted of FBW7 or CDK5 show enhanced levels of NDE1 and a reduction in ciliary length, which is corrected in cells depleted of both FBW7 or CDK5 and NDE1. These data show that cell cycle-dependent mechanisms can control ciliary length through a CDK5-FBW7-NDE1 pathway.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Quinase 5 Dependente de Ciclina/metabolismo , Proteínas F-Box/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/fisiologia , Animais , Células 3T3 BALB , Proteínas de Ciclo Celular/genética , Cílios/genética , Cílios/metabolismo , Quinase 5 Dependente de Ciclina/genética , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Células HEK293 , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases/genética
6.
Nat Commun ; 4: 2130, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23945651

RESUMO

Anticancer drug therapy activates both molecular cell death and autophagy pathways. Here we show that even sublethal concentrations of DNA-damaging drugs, such as etoposide and cisplatin, induce the expression of autophagy-related protein 5 (ATG5), which is both necessary and sufficient for the subsequent induction of mitotic catastrophe. We demonstrate that ATG5 translocates to the nucleus, where it physically interacts with survivin in response to DNA-damaging agents both in vitro and in carcinoma tissues obtained from patients who had undergone radiotherapy and/or chemotherapy. As a consequence, elements of the chromosomal passenger complex are displaced during mitosis, resulting in chromosome misalignment and segregation defects. Pharmacological inhibition of autophagy does not prevent ATG5-dependent mitotic catastrophe, but shifts the balance to an early caspase-dependent cell death. Our data suggest a dual role for ATG5 in response to drug-induced DNA damage, where it acts in two signalling pathways in two distinct cellular compartments, the cytosol and the nucleus.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/efeitos dos fármacos , Neoplasias/metabolismo , Transporte Ativo do Núcleo Celular , Aurora Quinase B/metabolismo , Autofagia/genética , Proteína 5 Relacionada à Autofagia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Cisplatino/farmacologia , Dano ao DNA/genética , Etoposídeo/farmacologia , Proteínas de Choque Térmico HSP90/metabolismo , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Células Jurkat , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Mitose/genética , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos , Survivina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...