Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
PLoS One ; 18(8): e0289778, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37540694

RESUMO

PKD1 is the most commonly mutated gene causing autosomal dominant polycystic kidney disease (ADPKD). It encodes Polycystin-1 (PC1), a putative membrane protein that undergoes a set of incompletely characterized post-transcriptional cleavage steps and has been reported to localize in multiple subcellular locations, including the primary cilium and mitochondria. However, direct visualization of PC1 and detailed characterization of its binding partners remain challenging. We now report a new mouse model with HA epitopes and eGFP knocked-in frame into the endogenous mouse Pkd1 gene by CRISPR/Cas9. Using this model, we sought to visualize endogenous PC1-eGFP and performed affinity-purification mass spectrometry (AP-MS) and network analyses. We show that the modified Pkd1 allele is fully functional but the eGFP-tagged protein cannot be detected without signal amplification by secondary antibodies. Using nanobody-coupled beads and large quantities of tissue, AP-MS identified an in vivo PC1 interactome, which is enriched for mitochondrial proteins and components of metabolic pathways. These studies suggest this mouse model and interactome data will be useful to understand PC1 function, but that new methods and brighter tags will be required to track endogenous PC1.


Assuntos
Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Camundongos , Animais , Canais de Cátion TRPP/química , Rim Policístico Autossômico Dominante/genética , Modelos Animais de Doenças
3.
Adv Kidney Dis Health ; 30(3): 209-219, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37088523

RESUMO

Autosomal dominant polycystic kidney disease is the most common inherited cause of end-stage kidney disease worldwide. Most cases result from mutation of either of 2 genes, PKD1 and PKD2, which encode proteins that form a probable receptor/channel complex. Studies suggest that a loss of function of the complex below an indeterminate threshold triggers cyst initiation, which ultimately results in dysregulation of multiple metabolic processes and downstream pathways and subsequent cyst growth. Noncell autonomous factors may also promote cyst growth. In this report, we focus primarily on the process of early cyst formation and factors that contribute to its variability with brief consideration of how new studies suggest this process may be reversible.


Assuntos
Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Humanos , Canais de Cátion TRPP/genética , Doenças Renais Policísticas/genética , Mutação , Rim Policístico Autossômico Dominante/genética
4.
Int J Mater Form ; 15(5): 61, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35855077

RESUMO

This article details the ESAFORM Benchmark 2021. The deep drawing cup of a 1 mm thick, AA 6016-T4 sheet with a strong cube texture was simulated by 11 teams relying on phenomenological or crystal plasticity approaches, using commercial or self-developed Finite Element (FE) codes, with solid, continuum or classical shell elements and different contact models. The material characterization (tensile tests, biaxial tensile tests, monotonic and reverse shear tests, EBSD measurements) and the cup forming steps were performed with care (redundancy of measurements). The Benchmark organizers identified some constitutive laws but each team could perform its own identification. The methodology to reach material data is systematically described as well as the final data set. The ability of the constitutive law and of the FE model to predict Lankford and yield stress in different directions is verified. Then, the simulation results such as the earing (number and average height and amplitude), the punch force evolution and thickness in the cup wall are evaluated and analysed. The CPU time, the manpower for each step as well as the required tests versus the final prediction accuracy of more than 20 FE simulations are commented. The article aims to guide students and engineers in their choice of a constitutive law (yield locus, hardening law or plasticity approach) and data set used in the identification, without neglecting the other FE features, such as software, explicit or implicit strategy, element type and contact model.

5.
Kidney360 ; 2(10): 1576-1591, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-35372986

RESUMO

Background: Multiple studies of tissue and cell samples from patients and preclinical models of autosomal dominant polycystic kidney disease report abnormal mitochondrial function and morphology and suggest metabolic reprogramming is an intrinsic feature of this disease. Peroxisomes interact with mitochondria physically and functionally, and congenital peroxisome biogenesis disorders can cause various phenotypes, including mitochondrial defects, metabolic abnormalities, and renal cysts. We hypothesized that a peroxisomal defect might contribute to the metabolic and mitochondrial impairments observed in autosomal dominant polycystic kidney disease. Methods: Using control and Pkd1-/- kidney epithelial cells, we investigated peroxisome abundance, biogenesis, and morphology by immunoblotting, immunofluorescence, and live cell imaging of peroxisome-related proteins and assayed peroxisomal specific ß-oxidation. We further analyzed fatty acid composition by mass spectrometry in kidneys of Pkd1fl/fl;Ksp-Cre mice. We also evaluated peroxisome lipid metabolism in published metabolomics datasets of Pkd1 mutant cells and kidneys. Lastly, we investigated if the C terminus or full-length polycystin-1 colocalize with peroxisome markers by imaging studies. Results: Peroxisome abundance, morphology, and peroxisome-related protein expression in Pkd1-/- cells were normal, suggesting preserved peroxisome biogenesis. Peroxisomal ß-oxidation was not impaired in Pkd1-/- cells, and there was no obvious accumulation of very-long-chain fatty acids in kidneys of mutant mice. Reanalysis of published datasets provide little evidence of peroxisomal abnormalities in independent sets of Pkd1 mutant cells and cystic kidneys, and provide further evidence of mitochondrial fatty acid oxidation defects. Imaging studies with either full-length polycystin-1 or its C terminus, a fragment previously shown to go to the mitochondria, showed minimal colocalization with peroxisome markers restricted to putative mitochondrion-peroxisome contact sites. Conclusions: Our studies showed that loss of Pkd1 does not disrupt peroxisome biogenesis nor peroxisome-dependent fatty acid metabolism.


Assuntos
Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Proteína Quinase C/metabolismo , Animais , Humanos , Metabolismo dos Lipídeos/genética , Camundongos , Mutação , Peroxissomos/metabolismo , Doenças Renais Policísticas/genética , Rim Policístico Autossômico Dominante/genética
6.
Cell Signal ; 74: 109701, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32649993

RESUMO

Systems-based, agnostic approaches focusing on transcriptomics data have been employed to understand the pathogenesis of polycystic kidney diseases (PKD). While multiple signaling pathways, including Wnt, mTOR and G-protein-coupled receptors, have been implicated in late stages of disease, there were few insights into the transcriptional cascade immediately downstream of Pkd1 inactivation. One of the consistent findings has been transcriptional evidence of dysregulated metabolic and cytoskeleton remodeling pathways. Recent technical developments, including bulk and single-cell RNA sequencing technologies and spatial transcriptomics, offer new angles to investigate PKD. In this article, we review what has been learned based on transcriptional approaches and consider future opportunities.


Assuntos
Doenças Renais Policísticas/metabolismo , Transcriptoma , Animais , Perfilação da Expressão Gênica , Humanos , Canais de Cátion TRPP/metabolismo
7.
Glob Chang Biol ; 26(2): 509-522, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31486174

RESUMO

Tropical forests store large amounts of carbon and high biodiversity, but are being degraded at alarming rates. The emerging global Forest and Landscape Restoration (FLR) agenda seeks to limit global climate change by removing carbon dioxide from the atmosphere through the growth of trees. In doing so, it may also protect biodiversity as a free cobenefit, which is vital given the massive shortfall in funding for biodiversity conservation. We investigated whether natural forest regeneration on abandoned pastureland offers such cobenefits, focusing for the first time on the recovery of taxonomic diversity (TD), phylogenetic diversity (PD) and functional diversity (FD) of trees, including the recovery of threatened and endemic species richness, within isolated secondary forest (SF) fragments. We focused on the globally threatened Brazilian Atlantic Forest, where commitments have been made to restore 1 million hectares under FLR. Three decades after land abandonment, regenerating forests had recovered ~20% (72 Mg/ha) of the above-ground carbon stocks of a primary forest (PF), with cattle pasture containing just 3% of stocks relative to PFs. Over this period, SF recovered ~76% of TD, 84% of PD and 96% of FD found within PFs. In addition, SFs had on average recovered 65% of threatened and ~30% of endemic species richness of primary Atlantic forest. Finally, we find positive relationships between carbon stock and tree diversity recovery. Our results emphasize that SF fragments offer cobenefits under FLR and other carbon-based payments for ecosystem service schemes (e.g. carbon enhancements under REDD+). They also indicate that even isolated patches of SF could help to mitigate climate change and the biodiversity extinction crisis by recovering species of high conservation concern and improving landscape connectivity.


Assuntos
Ecossistema , Florestas , Animais , Biodiversidade , Brasil , Bovinos , Conservação dos Recursos Naturais , Filogenia , Clima Tropical
8.
Sci Rep ; 8(1): 2743, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29426897

RESUMO

Recent studies have reported intrinsic metabolic reprogramming in Pkd1 knock-out cells, implicating dysregulated cellular metabolism in the pathogenesis of polycystic kidney disease. However, the exact nature of the metabolic changes and their underlying cause remains controversial. We show herein that Pkd1 k o /ko renal epithelial cells have impaired fatty acid utilization, abnormal mitochondrial morphology and function, and that mitochondria in kidneys of ADPKD patients have morphological alterations. We further show that a C-terminal cleavage product of polycystin-1 (CTT) translocates to the mitochondria matrix and that expression of CTT in Pkd1 ko/ko cells rescues some of the mitochondrial phenotypes. Using Drosophila to model in vivo effects, we find that transgenic expression of mouse CTT results in decreased viability and exercise endurance but increased CO2 production, consistent with altered mitochondrial function. Our results suggest that PC1 may play a direct role in regulating mitochondrial function and cellular metabolism and provide a framework to understand how impaired mitochondrial function could be linked to the regulation of tubular diameter in both physiological and pathological conditions.


Assuntos
Rim , Mitocôndrias , Proteínas Mitocondriais/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Proteólise , Canais de Cátion TRPP/metabolismo , Idoso , Animais , Animais Geneticamente Modificados , Cães , Drosophila melanogaster , Embrião de Mamíferos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Ácidos Graxos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Rim/metabolismo , Rim/patologia , Células Madin Darby de Rim Canino , Masculino , Camundongos , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Canais de Cátion TRPP/genética
9.
An. acad. bras. ciênc ; 89(3): 1935-1942, July-Sept. 2017. tab
Artigo em Inglês | LILACS | ID: biblio-886759

RESUMO

ABSTRACT The use of animals from dairy farms is an alternative to meat production since it provides an increment of total income for farmers. This study aims to evaluate the performance of Holstein calves finished in two feeding systems (feedlot or pasture). Forty-three animals with 58 days old and 57 kg were divided in two treatments: 23 animals finished in feedlot with corn silage plus concentrate based on corn and soybean meal (40:60); 20 animals kept in cultivated pastures according to the period of the year: Italian ryegrass (Lolium multiflorum) and pearl millet (Pennisetum americanum) with supplementation with the same feedlot-concentrate at 1% body weight. Animals were slaughtered with 200 kg. Dry matter and nutrient intake were determined, with the use of chromium oxide for estimating pasture intake. Feedlot animals had greater total intake and total digestible nutrients, resulting in higher average daily gain (0.949 vs 0.694 kg day-1). Crude protein intake, neutral detergent fiber and feed conversion did not show significant differences. Holstein calves have improved performance when finished in feedlot.


Assuntos
Animais , Masculino , Silagem/análise , Ingestão de Energia/fisiologia , Ração Animal , Criação de Animais Domésticos , Estações do Ano , Lolium , Bovinos , Pennisetum , Fenômenos Fisiológicos da Nutrição Animal , Valor Nutritivo
10.
Sci Rep ; 7(1): 7733, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28798345

RESUMO

Autosomal recessive polycystic kidney disease (ARPKD) is an important childhood nephropathy, occurring 1 in 20,000 live births. The major clinical phenotypes are expressed in the kidney with dilatation of the collecting ducts, systemic hypertension, and progressive renal insufficiency, and in the liver with biliary dysgenesis, portal tract fibrosis, and portal hypertension. The systemic hypertension has been attributed to enhanced distal sodium reabsorption in the kidney, the structural defects have been ascribed to altered cellular morphology, and fibrosis to increased TGF-ß signaling in the kidney and biliary tract, respectively. The pathogenic mechanisms underlying these abnormalities have not been determined. In the current report, we find that disrupting PKHD1 results in altered sub-cellular localization and function of the C2-WWW-HECT domain E3 family of ligases regulating these processes. We also demonstrate altered activity of RhoA and increased TGF-ß signaling and ENaC activity. Linking these phenomena, we found that vesicles containing the PKHD1/Pkhd1 gene product, FPC, also contain the NEDD4 ubiquitin ligase interacting protein, NDFIP2, which interacts with multiple members of the C2-WWW-HECT domain E3 family of ligases. Our results provide a mechanistic explanation for both the cellular effects and in vivo phenotypic abnormalities in mice and humans that result from Pkhd1/PKHD1 mutation.


Assuntos
Ubiquitina-Proteína Ligases Nedd4/metabolismo , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/metabolismo , Receptores de Superfície Celular/deficiência , Animais , Biomarcadores , Linhagem Celular , Modelos Animais de Doenças , Ativação Enzimática , Expressão Gênica , Humanos , Espaço Intracelular/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Mutação , Rim Policístico Autossômico Recessivo/patologia , Transporte Proteico , Ratos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo
11.
An Acad Bras Cienc ; 89(3): 1935-1942, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28767892

RESUMO

The use of animals from dairy farms is an alternative to meat production since it provides an increment of total income for farmers. This study aims to evaluate the performance of Holstein calves finished in two feeding systems (feedlot or pasture). Forty-three animals with 58 days old and 57 kg were divided in two treatments: 23 animals finished in feedlot with corn silage plus concentrate based on corn and soybean meal (40:60); 20 animals kept in cultivated pastures according to the period of the year: Italian ryegrass (Lolium multiflorum) and pearl millet (Pennisetum americanum) with supplementation with the same feedlot-concentrate at 1% body weight. Animals were slaughtered with 200 kg. Dry matter and nutrient intake were determined, with the use of chromium oxide for estimating pasture intake. Feedlot animals had greater total intake and total digestible nutrients, resulting in higher average daily gain (0.949 vs 0.694 kg day-1). Crude protein intake, neutral detergent fiber and feed conversion did not show significant differences. Holstein calves have improved performance when finished in feedlot.


Assuntos
Ração Animal , Criação de Animais Domésticos , Ingestão de Energia/fisiologia , Silagem/análise , Fenômenos Fisiológicos da Nutrição Animal , Animais , Bovinos , Lolium , Masculino , Valor Nutritivo , Pennisetum , Estações do Ano
12.
EBioMedicine ; 5: 183-92, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27077126

RESUMO

BACKGROUND: The major gene mutated in autosomal dominant polycystic kidney disease was first identified over 20 years ago, yet its function remains poorly understood. We have used a systems-based approach to examine the effects of acquired loss of Pkd1 in adult mouse kidney as it transitions from normal to cystic state. METHODS: We performed transcriptional profiling of a large set of male and female kidneys, along with metabolomics and lipidomics analyses of a subset of male kidneys. We also assessed the effects of a modest diet change on cyst progression in young cystic mice. Fatty acid oxidation and glycolytic rates were measured in five control and mutant pairs of epithelial cells. RESULTS: We find that females have a significantly less severe kidney phenotype and correlate this protection with differences in lipid metabolism. We show that sex is a major determinant of the transcriptional profile of mouse kidneys and that some of this difference is due to genes involved in lipid metabolism. Pkd1 mutant mice have transcriptional profiles consistent with changes in lipid metabolism and distinct metabolite and complex lipid profiles in kidneys. We also show that cells lacking Pkd1 have an intrinsic fatty acid oxidation defect and that manipulation of lipid content of mouse chow modifies cystic disease. INTERPRETATION: Our results suggest PKD could be a disease of altered cellular metabolism.


Assuntos
Ácidos Graxos/metabolismo , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/genética , Animais , Modelos Animais de Doenças , Feminino , Humanos , Rim/metabolismo , Rim/patologia , Metabolismo dos Lipídeos/genética , Masculino , Camundongos , Camundongos Transgênicos , Oxirredução , Rim Policístico Autossômico Dominante/fisiopatologia , Canais de Cátion TRPP/metabolismo
13.
Mol Biol Cell ; 25(19): 2919-33, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25103236

RESUMO

Defects in centrosome and cilium function are associated with phenotypically related syndromes called ciliopathies. Cby1, the mammalian orthologue of the Drosophila Chibby protein, localizes to mature centrioles, is important for ciliogenesis in multiciliated airway epithelia in mice, and antagonizes canonical Wnt signaling via direct regulation of ß-catenin. We report that deletion of the mouse Cby1 gene results in cystic kidneys, a phenotype common to ciliopathies, and that Cby1 facilitates the formation of primary cilia and ciliary recruitment of the Joubert syndrome protein Arl13b. Localization of Cby1 to the distal end of mature centrioles depends on the centriole protein Ofd1. Superresolution microscopy using both three-dimensional SIM and STED reveals that Cby1 localizes to an ∼250-nm ring at the distal end of the mature centriole, in close proximity to Ofd1 and Ahi1, a component of the transition zone between centriole and cilium. The amount of centriole-localized Ahi1, but not Ofd1, is reduced in Cby1(-/-) cells. This suggests that Cby1 is required for efficient recruitment of Ahi1, providing a possible molecular mechanism for the ciliogenesis defect in Cby1(-/-) cells.


Assuntos
Proteínas de Transporte/genética , Centríolos/metabolismo , Cílios/genética , Doenças Renais Císticas/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Ribosilação do ADP/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Cílios/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/metabolismo , Estrutura Terciária de Proteína , Proteínas/metabolismo , Via de Sinalização Wnt , beta Catenina
14.
PLoS Genet ; 8(11): e1003053, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23209428

RESUMO

Autosomal Dominant Polycystic Kidney Disease (ADPKD; MIM ID's 173900, 601313, 613095) leads to end-stage kidney disease, caused by mutations in PKD1 or PKD2. Inactivation of Pkd1 before or after P13 in mice results in distinct early- or late-onset disease. Using a mouse model of ADPKD carrying floxed Pkd1 alleles and an inducible Cre recombinase, we intensively analyzed the relationship between renal maturation and cyst formation by applying transcriptomics and metabolomics to follow disease progression in a large number of animals induced before P10. Weighted gene co-expression network analysis suggests that Pkd1-cystogenesis does not cause developmental arrest and occurs in the context of gene networks similar to those that regulate/maintain normal kidney morphology/function. Knowledge-based Ingenuity Pathway Analysis (IPA) software identifies HNF4α as a likely network node. These results are further supported by a meta-analysis of 1,114 published gene expression arrays in Pkd1 wild-type tissues. These analyses also predict that metabolic pathways are key elements in postnatal kidney maturation and early steps of cyst formation. Consistent with these findings, urinary metabolomic studies show that Pkd1 cystic mutants have a distinct profile of excreted metabolites, with pathway analysis suggesting altered activity in several metabolic pathways. To evaluate their role in disease, metabolic networks were perturbed by inactivating Hnf4α and Pkd1. The Pkd1/Hnf4α double mutants have significantly more cystic kidneys, thus indicating that metabolic pathways could play a role in Pkd1-cystogenesis.


Assuntos
Redes Reguladoras de Genes , Fator 4 Nuclear de Hepatócito , Rim Policístico Autossômico Dominante , Proteína Quinase C , Alelos , Animais , Proliferação de Células , Modelos Animais de Doenças , Regulação da Expressão Gênica , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Camundongos , Mutação , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Proteína Quinase C/genética , Proteína Quinase C/metabolismo
15.
PLoS One ; 5(9)2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20862291

RESUMO

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of inherited renal failure that results from mutations in PKD1 and PKD2. The disorder is characterized by focal cyst formation that involves somatic mutation of the wild type allele in a large fraction of cysts. Consistent with a two-hit mechanism, mice that are homozygous for inactivating mutations of either Pkd1 or Pkd2 develop cystic kidneys, edema and hemorrhage and typically die in midgestation. Cystic kidney disease is unlikely to be the cause of fetal loss since renal function is not required to complete gestation. One hypothesis is that embryonic demise is due to leaky vessels or cardiac pathology. METHODOLOGY/PRINCIPAL FINDINGS: In these studies we used a series of genetically modified Pkd1 and Pkd2 murine models to investigate the cause of embryonic lethality in mutant embryos. Since placental defects are a frequent cause of fetal loss, we conducted histopathologic analyses of placentas from Pkd1 null mice and detected abnormalities of the labyrinth layer beginning at E12.5. We performed placental rescue experiments using tetraploid aggregation and conditional inactivation of Pkd1 with the Meox2 Cre recombinase. We found that both strategies improved the viability of Pkd1 null embryos. Selective inactivation of Pkd1 and Pkd2 in endothelial cells resulted in polyhydramnios and abnormalities similar to those observed in Pkd1(-/-) placentas. However, endothelial cell specific deletion of Pkd1 or Pkd2 did not yield the dramatic vascular phenotypes observed in null animals. CONCLUSIONS/SIGNIFICANCE: Placental abnormalities contribute to the fetal demise of Pkd(-/-) embryos. Endothelial cell specific deletion of Pkd1 or Pkd2 recapitulates a subset of findings seen in Pkd null animals. Our studies reveal a complex role for polycystins in maintaining vascular integrity.


Assuntos
Placenta/embriologia , Placenta/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Células Endoteliais/metabolismo , Feminino , Masculino , Camundongos , Camundongos Knockout , Gravidez , Canais de Cátion TRPP/genética
16.
J Am Soc Nephrol ; 20(11): 2389-402, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19833899

RESUMO

Mutations in PKD1 cause the majority of cases of autosomal dominant polycystic kidney disease (ADPKD). Because polycystin 1 modulates cell proliferation, cell differentiation, and apoptosis, its lower biologic activity observed in ADPKD might influence the degree of injury after renal ischemia/reperfusion. We induced renal ischemia/reperfusion in 10- to 12-wk-old male noncystic Pkd1(+/-) and wild-type mice. Compared with wild-type mice, heterozygous mice had higher fractional excretions of sodium and potassium and higher serum creatinine after 48 h. In addition, in heterozygous mice, also cortical damage, rates of apoptosis, and inflammatory infiltration into the interstitium at time points out to 14 d after injury all increased, as well as cell proliferation at 48 h and 7 d. The mRNA and protein expression of p21 was lower in heterozygous mice than wild-type mice at 48 h. After 6 wk, we observed dilated tubules, microcysts, and increased renal fibrosis in heterozygotes. The early mortality of heterozygotes was significantly higher than that of wild-type mice when we extended the duration of ischemia from 32 to 35 min. In conclusion, ischemia/reperfusion induces a more severe injury in kidneys of Pkd1-haploinsufficient mice, a process that apparently depends on a relative deficiency of p21 activity, tubular dilation, and microcyst formation. These data suggest the possibility that humans with ADPKD from PKD1 mutations may be at greater risk for damage from renal ischemia/reperfusion injury.


Assuntos
Doenças Renais Císticas/etiologia , Nefropatias/etiologia , Mutação , Traumatismo por Reperfusão/complicações , Canais de Cátion TRPP/genética , Animais , Masculino , Camundongos , Camundongos Mutantes
17.
J Cell Mol Med ; 13(7): 1279-90, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19292732

RESUMO

The longest open reading frame of PKHD1 (polycystic kidney and hepatic disease 1), the autosomal recessive polycystic kidney disease (ARPKD) gene, encodes a single-pass, integral membrane protein named polyductin or fibrocystin. A fusion protein comprising its intracellular C-terminus, FP2, was previously used to raise a polyclonal antiserum shown to detect polyductin in several human tissues, including liver. In the current study, we aimed to investigate by immunohistochemistry the detailed polyductin localization pattern in normal (ductal plate [DP], remodelling ductal plate [RDP], remodelled bile ducts) and abnormal development of the primitive intrahepatic biliary system, known as ductal plate malformation (DPM). This work also included the characterization of polyductin expression profile in various histological forms of neonatal and infantile cholestasis, and in cholangiocellular carcinoma (CCC) and hepatocellular carcinoma (HCC). We detected polyductin expression in the intrahepatic biliary system during the DP and the RDP stages as well as in DPM. No specific staining was found at the stage of remodelled bile ducts. Polyductin was also detected in liver biopsies with neonatal cholestasis, including mainly biliary atresia and neonatal hepatitis with ductular reaction as well as congenital hepatic fibrosis. In addition, polyductin was present in CCC, whereas it was absent in HCC. Polyductin was also co-localized in some DP cells together with oval stem cell markers. These results represent the first systematic study of polyductin expression in human pathologies associated with abnormal development of intrahepatic biliary tree, and support the following conclusions: (i) polyductin expression mirrors developmental properties of the primitive intrahepatic biliary system; (ii) polyductin is re-expressed in pathological conditions associated with DPM and (iii) polyductin might be a potential marker to distinguish CCC from HCC.


Assuntos
Ductos Biliares Intra-Hepáticos/anormalidades , Neoplasias do Sistema Biliar/patologia , Neoplasias Hepáticas/patologia , Fígado/patologia , Receptores de Superfície Celular/metabolismo , Células-Tronco/metabolismo , Células-Tronco/patologia , Adulto , Ductos Biliares Intra-Hepáticos/embriologia , Ductos Biliares Intra-Hepáticos/metabolismo , Neoplasias do Sistema Biliar/metabolismo , Biomarcadores/metabolismo , Feto/patologia , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo
18.
Methods Cell Biol ; 94: 273-97, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20362096

RESUMO

Cystic kidney diseases are characterized by dilated or cystic kidney tubular segments. Changes in planar cell polarity, flow sensing, and/or proliferation have been proposed to explain these disorders. Over the last few years, several groups have suggested that ciliary dysfunction is a central component of cyst formation. We review evidence for and against each of these models, stressing some of the inconsistencies that should be resolved if an accurate understanding of cyst formation is to be achieved. We also comment on data supporting a model in which ciliary function could play different roles at different developmental stages and on the relevance of dissecting potential differences between pathways required for tubule formation and/or maintenance.


Assuntos
Polaridade Celular , Cílios/patologia , Doenças Renais Policísticas/patologia , Animais , Proliferação de Células , Criança , Cílios/metabolismo , Regulação da Expressão Gênica , Humanos , Lactente , Rim/citologia , Rim/metabolismo , Rim/patologia , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/fisiopatologia , Transdução de Sinais/fisiologia , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Proteínas Wnt/metabolismo
19.
Am J Pathol ; 172(2): 417-29, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18202188

RESUMO

Autosomal recessive polycystic kidney disease is a hereditary fibrocystic disease that involves the kidneys and the biliary tract. Mutations in the PKHD1 gene are responsible for typical forms of autosomal recessive polycystic kidney disease. We have generated a mouse model with targeted mutation of Pkhd1 by disrupting exon 4, resulting in a mutant transcript with deletion of 66 codons and expression at approximately 30% of wild-type levels. Pkhd1(del4/del4) mice develop intrahepatic bile duct proliferation with progressive cyst formation and associated periportal fibrosis. In addition, these mice exhibit extrahepatic manifestations, including pancreatic cysts, splenomegaly, and common bile duct dilation. The kidneys are unaffected both histologically and functionally. Fibrocystin is expressed in the apical membranes and cilia of bile ducts and distal nephron segments but is absent from the proximal tubule. This pattern is unchanged in orthologous models of autosomal dominant polycystic kidney disease due to mutation in Pkd1 or Pkd2. Mutant fibrocystin in Pkhd1(del4/del4) mice also retains this expression pattern. The hypomorphic Pkhd1(del4/del4) mouse model provides evidence that reduced functional levels of fibrocystin are sufficient for cystogenesis and fibrosis in the liver and pancreas, but not the kidney, and supports the hypothesis of species-dependent differences in susceptibility of tissues to Pkhd1 mutations.


Assuntos
Sistema Biliar/patologia , Hepatopatias/genética , Pancreatopatias/genética , Rim Policístico Autossômico Recessivo/patologia , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Animais , Ductos Biliares/metabolismo , Ductos Biliares/patologia , Southern Blotting , Western Blotting , Cílios/metabolismo , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Hibridização In Situ , Túbulos Renais Proximais/metabolismo , Hepatopatias/patologia , Camundongos , Camundongos Mutantes , Mutação , Pancreatopatias/patologia , Rim Policístico Autossômico Recessivo/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Proc Natl Acad Sci U S A ; 104(47): 18688-93, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18003909

RESUMO

Polycystin-1 (PC1) has an essential function in renal tubular morphogenesis and disruption of its function causes cystogenesis in human autosomal dominant polycystic kidney disease. We have previously shown that recombinant human PC1 is cis-autoproteolytically cleaved at the G protein-coupled receptor proteolytic site domain. To investigate the role of cleavage in vivo, we generated by gene targeting a Pkd1 knockin mouse (Pkd1(V/V)) that expresses noncleavable PC1. The Pkd1(V/V) mice show a hypomorphic phenotype, characterized by a delayed onset and distal nephron segment involvement of cystogenesis at postnatal maturation stage. We show that PC1 is ubiquitously and incompletely cleaved in wild-type mice, so that uncleaved and cleaved PC1 molecules coexist. Our study establishes a critical but restricted role of cleavage for PC1 function and suggests a differential function of the two types of PC1 molecules in vivo.


Assuntos
Túbulos Renais/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Canais de Cátion TRPP/metabolismo , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Sequência de Bases , Células Cultivadas , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/patologia , Camundongos , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...