Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(4): e2317344121, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38241440

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic cause of chronic kidney disease and the fourth leading cause of end-stage kidney disease, accounting for over 50% of prevalent cases requiring renal replacement therapy. There is a pressing need for improved therapy for ADPKD. Recent insights into the pathophysiology of ADPKD revealed that cyst cells undergo metabolic changes that up-regulate aerobic glycolysis in lieu of mitochondrial respiration for energy production, a process that ostensibly fuels their increased proliferation. The present work leverages this metabolic disruption as a way to selectively target cyst cells for apoptosis. This small-molecule therapeutic strategy utilizes 11beta-dichloro, a repurposed DNA-damaging anti-tumor agent that induces apoptosis by exacerbating mitochondrial oxidative stress. Here, we demonstrate that 11beta-dichloro is effective in delaying cyst growth and its associated inflammatory and fibrotic events, thus preserving kidney function in perinatal and adult mouse models of ADPKD. In both models, the cyst cells with homozygous inactivation of Pkd1 show enhanced oxidative stress following treatment with 11beta-dichloro and undergo apoptosis. Co-administration of the antioxidant vitamin E negated the therapeutic benefit of 11beta-dichloro in vivo, supporting the conclusion that oxidative stress is a key component of the mechanism of action. As a preclinical development primer, we also synthesized and tested an 11beta-dichloro derivative that cannot directly alkylate DNA, while retaining pro-oxidant features. This derivative nonetheless maintains excellent anti-cystic properties in vivo and emerges as the lead candidate for development.


Assuntos
Cistos , Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Camundongos , Animais , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Proliferação de Células , Doenças Renais Policísticas/metabolismo , Apoptose , Estresse Oxidativo , Cistos/metabolismo , DNA/metabolismo , Rim/metabolismo , Canais de Cátion TRPP/genética
2.
J Am Soc Nephrol ; 34(1): 110-121, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36270750

RESUMO

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in Pkd1 and Pkd2. They encode the polytopic integral membrane proteins polycystin-1 (PC1) and polycystin-2 (PC2), respectively, which are expressed on primary cilia. Formation of kidney cysts in ADPKD starts when a somatic second hit mechanism inactivates the wild-type Pkd allele. Approximately one quarter of families with ADPDK due to Pkd1 have germline nonsynonymous amino acid substitution (missense) mutations. A subset of these mutations is hypomorphic, retaining some residual PC1 function. Previous studies have shown that the highly conserved Ire1 α -XBP1 pathway of the unfolded protein response can modulate levels of functional PC1 in the presence of mutations in genes required for post-translational maturation of integral membrane proteins. We examine how activity of the endoplasmic reticulum chaperone-inducing transcription factor XBP1 affects ADPKD in a murine model with missense Pkd1 . METHODS: We engineered a Pkd1 REJ domain missense murine model, Pkd1 R2216W , on the basis of the orthologous human hypomorphic allele Pkd1 R2220W , and examined the effects of transgenic activation of XBP1 on ADPKD progression. RESULTS: Expression of active XBP1 in cultured cells bearing PC1 R2216W mutations increased levels and ciliary trafficking of PC1 R2216W . Mice homozygous for Pkd1 R2216W or heterozygous for Pkd1 R2216Win trans with a conditional Pkd1 fl allele exhibit severe ADPKD following inactivation in neonates or adults. Transgenic expression of spliced XBP1 in tubule segments destined to form cysts reduced cell proliferation and improved Pkd progression, according to structural and functional parameters. CONCLUSIONS: Modulating ER chaperone function through XBP1 activity improved Pkd in a murine model of PC1, suggesting therapeutic targeting of hypomorphic mutations.


Assuntos
Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Adulto , Camundongos , Humanos , Animais , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Modelos Animais de Doenças , Doenças Renais Policísticas/metabolismo , Mutação , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo
4.
Kidney360 ; 1(10): 1068-1076, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33554127

RESUMO

BACKGROUND: A major difference between autosomal recessive polycystic kidney disease (ARPKD) and autosomal dominant polycystic kidney disease (ADPKD) lies in the pattern of inheritance, and the resultant timing and focality of cyst formation. In both diseases, cysts form in the kidney and liver as a consequence of the cellular recessive genotype of the respective disease gene, but this occurs by germline inheritance in ARPKD and somatic second hit mutations to the one normal allele in ADPKD. The fibrocystic liver phenotype in ARPKD is attributed to abnormal ductal plate formation because of the absence of PKHD1 expression during embryogenesis and organ development. The finding of polycystic liver disease in a subset of adult PKHD1 heterozygous carriers raises the question of whether somatic second hit mutations in PKHD1 in adults may also result in bile duct-derived cyst formation. METHODS: We used an adult-inducible Pkhd1 mouse model to examine whether Pkhd1 has a functional role in maintaining bile duct homeostasis after normal liver development. RESULTS: Inactivation of Pkhd1 beginning at 4 weeks of age resulted in a polycystic liver phenotype with minimal fibrosis at 17 weeks. Increased biliary epithelium, which lines these liver cysts, was most pronounced in female mice. We assessed genetic interaction of this phenotype with either reduced or increased copies of Pkd1, and found no significant effects on the Pkhd1 phenotype in the liver or kidney from altered Pkd1 expression. CONCLUSIONS: Somatic adult inactivation of Pkhd1 results in a polycystic liver phenotype. Pkhd1 is a required gene in adulthood for biliary structural homeostasis independent of Pkd1. This suggests that PKHD1 heterozygous carrier patients can develop liver cysts after somatic mutations in their normal copy of PKHD1.


Assuntos
Cistos , Hepatopatias , Rim Policístico Autossômico Recessivo , Animais , Cistos/genética , Feminino , Hepatopatias/genética , Camundongos , Rim Policístico Autossômico Recessivo/genética , Receptores de Superfície Celular/genética
5.
J Am Soc Nephrol ; 30(3): 443-459, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30745418

RESUMO

BACKGROUND: SEC63 encodes a resident protein in the endoplasmic reticulum membrane that, when mutated, causes human autosomal dominant polycystic liver disease. Selective inactivation of Sec63 in all distal nephron segments in embryonic mouse kidney results in polycystin-1-mediated polycystic kidney disease (PKD). It also activates the Ire1α-Xbp1 branch of the unfolded protein response, producing Xbp1s, the active transcription factor promoting expression of specific genes to alleviate endoplasmic reticulum stress. Simultaneous inactivation of Xbp1 and Sec63 worsens PKD in this model. METHODS: We explored the renal effects of postnatal inactivation of Sec63 alone or with concomitant inactivation of Xbp1 or Ire1α, specifically in the collecting ducts of neonatal mice. RESULTS: The later onset of inactivation of Sec63 restricted to the collecting duct does not result in overt activation of the Ire1α-Xbp1 pathway or cause polycystin-1-dependent PKD. Inactivating Sec63 along with either Xbp1 or Ire1α in this model causes interstitial inflammation and associated fibrosis with decline in kidney function over several months. Re-expression of XBP1s in vivo completely rescues the chronic kidney injury observed after inactivation of Sec63 with either Xbp1 or Ire1α. CONCLUSIONS: In the absence of Sec63, basal levels of Xbp1s activity in collecting ducts is both necessary and sufficient to maintain proteostasis (protein homeostasis) and protect against inflammation, myofibroblast activation, and kidney functional decline. The Sec63-Xbp1 double knockout mouse offers a novel genetic model of chronic tubulointerstitial kidney injury, using collecting duct proteostasis defects as a platform for discovery of signals that may underlie CKD of disparate etiologies.

6.
Curr Biol ; 27(20): 3120-3131.e4, 2017 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-29033332

RESUMO

Oriented cell division (OCD) and convergent extension (CE) shape developing renal tubules, and their disruption has been associated with polycystic kidney disease (PKD) genes, the majority of which encode proteins that localize to primary cilia. Core planar cell polarity (PCP) signaling controls OCD and CE in other contexts, leading to the hypothesis that disruption of PCP signaling interferes with CE and/or OCD to produce PKD. Nonetheless, the contribution of PCP to tubulogenesis and cystogenesis is uncertain, and two major questions remain unanswered. Specifically, the inference that mutation of PKD genes interferes with PCP signaling is untested, and the importance of PCP signaling for cystogenic PKD phenotypes has not been examined. We show that, during proliferative stages, PCP signaling polarizes renal tubules to control OCD. However, we find that, contrary to the prevailing model, PKD mutations do not disrupt PCP signaling but instead act independently and in parallel with PCP signaling to affect OCD. Indeed, PCP signaling that is normally downregulated once development is completed is retained in cystic adult kidneys. Disrupting PCP signaling results in inaccurate control of tubule diameter, a tightly regulated parameter with important physiological ramifications. However, we show that disruption of PCP signaling is not cystogenic. Our results suggest that regulating tubule diameter is a key function of PCP signaling but that loss of this control does not induce cysts.


Assuntos
Polaridade Celular/fisiologia , Túbulos Renais/fisiologia , Morfogênese , Doenças Renais Policísticas/fisiopatologia , Transdução de Sinais , Animais , Feminino , Túbulos Renais/fisiopatologia , Masculino , Camundongos
7.
J Clin Invest ; 127(9): 3558, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28862642

RESUMO

Dominantly inherited isolated polycystic liver disease (PCLD) consists of liver cysts that are radiologically and pathologically identical to those seen in autosomal dominant polycystic kidney disease, but without clinically relevant kidney cysts. The causative genes are known for fewer than 40% of PCLD index cases. Here, we have used whole exome sequencing in a discovery cohort of 102 unrelated patients who were excluded for mutations in the 2 most common PCLD genes, PRKCSH and SEC63, to identify heterozygous loss-of-function mutations in 3 additional genes, ALG8, GANAB, and SEC61B. Similarly to PRKCSH and SEC63, these genes encode proteins that are integral to the protein biogenesis pathway in the endoplasmic reticulum. We inactivated these candidate genes in cell line models to show that loss of function of each results in defective maturation and trafficking of polycystin-1, the central determinant of cyst pathogenesis. Despite acting in a common pathway, each PCLD gene product demonstrated distinct effects on polycystin-1 biogenesis. We also found enrichment on a genome-wide basis of heterozygous mutations in the autosomal recessive polycystic kidney disease gene PKHD1, indicating that adult PKHD1 carriers can present with clinical PCLD. These findings define genetic and biochemical modulators of polycystin-1 function and provide a more complete definition of the spectrum of dominant human polycystic diseases.

8.
J Clin Invest ; 127(5): 1772-1785, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28375157

RESUMO

Dominantly inherited isolated polycystic liver disease (PCLD) consists of liver cysts that are radiologically and pathologically identical to those seen in autosomal dominant polycystic kidney disease, but without clinically relevant kidney cysts. The causative genes are known for fewer than 40% of PCLD index cases. Here, we have used whole exome sequencing in a discovery cohort of 102 unrelated patients who were excluded for mutations in the 2 most common PCLD genes, PRKCSH and SEC63, to identify heterozygous loss-of-function mutations in 3 additional genes, ALG8, GANAB, and SEC61B. Similarly to PRKCSH and SEC63, these genes encode proteins that are integral to the protein biogenesis pathway in the endoplasmic reticulum. We inactivated these candidate genes in cell line models to show that loss of function of each results in defective maturation and trafficking of polycystin-1, the central determinant of cyst pathogenesis. Despite acting in a common pathway, each PCLD gene product demonstrated distinct effects on polycystin-1 biogenesis. We also found enrichment on a genome-wide basis of heterozygous mutations in the autosomal recessive polycystic kidney disease gene PKHD1, indicating that adult PKHD1 carriers can present with clinical PCLD. These findings define genetic and biochemical modulators of polycystin-1 function and provide a more complete definition of the spectrum of dominant human polycystic diseases.


Assuntos
Cistos , Glucosiltransferases , Heterozigoto , Hepatopatias , Mutação , Canais de Translocação SEC , Canais de Cátion TRPP , Adulto , Animais , Proteínas de Ligação ao Cálcio , Linhagem Celular Transformada , Cistos/genética , Cistos/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Feminino , Estudo de Associação Genômica Ampla , Glucosidases/genética , Glucosidases/metabolismo , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Hepatopatias/genética , Hepatopatias/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Chaperonas Moleculares , Proteínas de Ligação a RNA , Canais de Translocação SEC/genética , Canais de Translocação SEC/metabolismo , Canais de Cátion TRPP/biossíntese , Canais de Cátion TRPP/genética
9.
Artigo em Inglês | MEDLINE | ID: mdl-28320755

RESUMO

Autosomal-dominant polycystic kidney disease (ADPKD) is a disease of defective tissue homeostasis resulting in active remodeling of nephrons and bile ducts to form fluid-filled sacs called cysts. The causal genes PKD1 and PKD2 encode transmembrane proteins polycystin 1 (PC1) and polycystin 2 (PC2), respectively. Together, the polycystins localize to the solitary primary cilium that protrudes from the apical surface of most kidney tubule cells and is thought to function as a privileged compartment that the cell uses for signal integration of sensory inputs. It has been proposed that PC1 and PC2 form a receptor-channel complex that detects external stimuli and transmit a local calcium-mediated signal, which may control a multitude of cellular processes by an as-yet unknown mechanism. Genetic studies using mouse models of cilia and polycystin dysfunction have shown that polycystins regulate an unknown cilia-dependent signal that is normally part of the homeostatic maintenance of nephron structure. ADPKD ensues when this pathway is dysregulated by absence of polycystins from intact cilia, but disruption of cilia also disrupts this signaling mechanism and ameliorates ADPKD even in the absence of polycystins. Understanding the role of cilia and ciliary signaling in ADPKD is challenging, but success will provide saltatory advances in our understanding of how tubule structure is maintained in healthy kidneys and how disruption of polycystin or cilia function leads to the pathological tissue remodeling process underlying ADPKD.


Assuntos
Cílios/fisiologia , Cistos/patologia , Rim Policístico Autossômico Dominante/patologia , Animais , Proteínas Hedgehog/metabolismo , Homeostase , Humanos , Transdução de Sinais
10.
Trends Mol Med ; 22(12): 997-999, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27793600

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 and PKD2, encoding polycystin-1 and polycystin-2, respectively. Optimizing the folding environment for polycystin-1 missense mutations may have a critical effect on the progression of ADPKD in animal models and could potentially lead to tangible therapeutic options for subgroups of ADPKD patients.


Assuntos
Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Animais , Descoberta de Drogas , Humanos , Mutação de Sentido Incorreto/efeitos dos fármacos , Rim Policístico Autossômico Dominante/tratamento farmacológico , Dobramento de Proteína/efeitos dos fármacos , Canais de Cátion TRPP/química
11.
J Clin Invest ; 125(5): 1955-67, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25844898

RESUMO

The HSP40 cochaperone SEC63 is associated with the SEC61 translocon complex in the ER. Mutations in the gene encoding SEC63 cause polycystic liver disease in humans; however, it is not clear how altered SEC63 influences disease manifestations. In mice, loss of SEC63 induces cyst formation both in liver and kidney as the result of reduced polycystin-1 (PC1). Here we report that inactivation of SEC63 induces an unfolded protein response (UPR) pathway that is protective against cyst formation. Specifically, using murine genetic models, we determined that SEC63 deficiency selectively activates the IRE1α-XBP1 branch of UPR and that SEC63 exists in a complex with PC1. Concomitant inactivation of both SEC63 and XBP1 exacerbated the polycystic kidney phenotype in mice by markedly suppressing cleavage at the G protein-coupled receptor proteolysis site (GPS) in PC1. Enforced expression of spliced XBP1 (XBP1s) enhanced GPS cleavage of PC1 in SEC63-deficient cells, and XBP1 overexpression in vivo ameliorated cystic disease in a murine model with reduced PC1 function that is unrelated to SEC63 inactivation. Collectively, the findings show that SEC63 function regulates IRE1α/XBP1 activation, SEC63 and XBP1 are required for GPS cleavage and maturation of PC1, and activation of XBP1 can protect against polycystic disease in the setting of impaired biogenesis of PC1.


Assuntos
DNA Helicases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Endorribonucleases/metabolismo , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Recessivo/genética , Proteínas Serina-Treonina Quinases/metabolismo , Canais de Cátion TRPP/deficiência , Fatores de Transcrição/fisiologia , Resposta a Proteínas não Dobradas/fisiologia , Animais , Linhagem Celular , DNA Helicases/deficiência , DNA Helicases/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Feminino , Glucosidases/deficiência , Glucosidases/genética , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Chaperonas Moleculares , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Recessivo/metabolismo , Estrutura Terciária de Proteína , Splicing de RNA , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição de Fator Regulador X , Canais de Cátion TRPP/biossíntese , Canais de Cátion TRPP/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transfecção , Proteína 1 de Ligação a X-Box
12.
J Clin Invest ; 124(12): 5129-44, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25365220

RESUMO

The most severe form of autosomal dominant polycystic kidney disease occurs in patients with mutations in the gene (PKD1) encoding polycystin-1 (PC1). PC1 is a complex polytopic membrane protein expressed in cilia that undergoes autoproteolytic cleavage at a G protein-coupled receptor proteolytic site (GPS). A quarter of PKD1 mutations are missense variants, though it is not clear how these mutations promote disease. Here, we established a cell-based system to evaluate these mutations and determined that GPS cleavage is required for PC1 trafficking to cilia. A common feature among a subset of pathogenic missense mutations is a resulting failure of PC1 to traffic to cilia regardless of GPS cleavage. The application of our system also identified a missense mutation in the gene encoding polycystin-2 (PC2) that prevented this protein from properly trafficking to cilia. Using a Pkd1-BAC recombineering approach, we developed murine models to study the effects of these mutations and confirmed that only the cleaved form of PC1 exits the ER and can rescue the embryonically lethal Pkd1-null mutation. Additionally, steady-state expression levels of the intramembranous COOH-terminal fragment of cleaved PC1 required an intact interaction with PC2. The results of this study demonstrate that PC1 trafficking and expression require GPS cleavage and PC2 interaction, respectively, and provide a framework for functional assays to categorize the effects of missense mutations in polycystins.


Assuntos
Doenças Renais Policísticas/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Linhagem Celular , Cílios/genética , Cílios/metabolismo , Cílios/patologia , Humanos , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/prevenção & controle , Estabilidade Proteica , Estrutura Terciária de Proteína , Transporte Proteico/genética , Canais de Cátion TRPP/genética
13.
Biochem Pharmacol ; 88(3): 412-21, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24518257

RESUMO

Cyst enlargement in autosomal dominant polycystic kidney disease (ADPKD) is associated with cAMP-activated proliferation of cyst-lining epithelial cells and transepithelial fluid secretion into the cyst lumen via cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel leading to renal failure for which no effective treatment is currently available. We previously reported that steviol retards Madin-Darby canine kidney (MDCK) cyst enlargement by inhibiting CFTR channel activity and promoting proteasomal-mediated CFTR degradation. It is imperative to examine the effect of steviol in animal models of ADPKD. Therefore, we examined the effect of steviol on renal cyst growth in an orthologous mouse model of human ADPKD (Pkd1(flox/flox):Pkhd1-Cre). The results showed that daily treatment with both 200mg/kg BW of steviol and 1000mg/kg BW of stevioside for 14 days markedly decreased kidney weight and cystic index in these mice. However, only steviol markedly reduced blood urea nitrogen and creatinine values. Steviol also reduced cell proliferation but had no effect on cell apoptosis. In addition, steviol suppressed CFTR and mTOR/S6K expression in renal cyst-lining epithelial cells. Interestingly, steviol was found to stimulate AMP-activated protein kinase (AMPK). Our findings indicate that steviol slows cyst progression in ADPKD mouse model, in part, through the activation of AMPK which subsequently inhibits CFTR chloride channel expression and inhibits renal epithelial cell proliferation via mTOR/S6K pathway. Most importantly, steviol could markedly improve kidney function in a mouse model of ADPKD. Steviol thus has potential application for further development as a therapeutic compound for the treatment of polycystic kidney disease.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Diterpenos do Tipo Caurano/farmacologia , Células Epiteliais/efeitos dos fármacos , Doenças Renais Policísticas/tratamento farmacológico , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Apoptose , Proliferação de Células , Diterpenos do Tipo Caurano/uso terapêutico , Ativação Enzimática , Células Epiteliais/patologia , Glucosídeos/farmacologia , Glucosídeos/uso terapêutico , Humanos , Camundongos , Camundongos Mutantes , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia
14.
Trends Mol Med ; 20(5): 251-60, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24491980

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is the most common potentially lethal monogenic disorder, with more than 12 million cases worldwide. The two causative genes for ADPKD, PKD1 and PKD2, encode protein products polycystin-1 (PC1) and polycystin-2 (PC2 or TRPP2), respectively. Recent data have shed light on the role of PC1 in regulating the severity of the cystic phenotypes in ADPKD, autosomal recessive polycystic kidney disease (ARPKD), and isolated autosomal dominant polycystic liver disease (ADPLD). These studies showed that the rate for cyst growth was a regulated trait, a process that can be either sped up or slowed down by alterations in functional PC1. These findings redefine the previous understanding that cyst formation occurs as an 'on-off' process. Here, we review these and other related studies with an emphasis on their translational implications for polycystic diseases.


Assuntos
Cistos/metabolismo , Doenças Renais Policísticas/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Cistos/genética , Humanos , Doenças Renais Policísticas/genética , Canais de Cátion TRPP/genética
15.
Pediatr Nephrol ; 28(8): 1161-72, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23161205

RESUMO

Epithelial cell polarity is essential for organ development; aberrations in this process have been implicated in various diseases, including polycystic kidney disease. Establishment and maintenance of cell polarity is governed by a number of molecular processes and how these processes operate remains an interesting question. Conserved protein complexes guide both apical-basolateral polarity and planar cell polarity. In this review we discuss the recent findings that provide insights into polarity mechanisms and the intriguing crosstalk between apical-basolateral polarity and planar cell polarity, and their relationship to cystic kidney disease.


Assuntos
Polaridade Celular , Células Epiteliais/patologia , Doenças Renais Císticas/patologia , Rim/patologia , Animais , Cílios/metabolismo , Cílios/patologia , Células Epiteliais/metabolismo , Humanos , Junções Intercelulares/metabolismo , Junções Intercelulares/patologia , Rim/metabolismo , Doenças Renais Císticas/metabolismo , Transdução de Sinais
16.
Nat Genet ; 43(7): 639-47, 2011 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-21685914

RESUMO

Autosomal dominant polycystic liver disease results from mutations in PRKCSH or SEC63. The respective gene products, glucosidase IIß and SEC63p, function in protein translocation and quality control pathways in the endoplasmic reticulum. Here we show that glucosidase IIß and Sec63p are required in mice for adequate expression of a functional complex of the polycystic kidney disease gene products, polycystin-1 and polycystin-2. We find that polycystin-1 is the rate-limiting component of this complex and that there is a dose-response relationship between cystic dilation and levels of functional polycystin-1 following mutation of Prkcsh or Sec63. Reduced expression of polycystin-1 also serves to sensitize the kidney to cyst formation resulting from mutations in Pkhd1, the recessive polycystic kidney disease gene. Finally, we show that proteasome inhibition increases steady-state levels of polycystin-1 in cells lacking glucosidase IIß and that treatment with a proteasome inhibitor reduces cystic disease in orthologous gene models of human autosomal dominant polycystic liver disease.


Assuntos
Cistos/patologia , Glucosidases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Hepatopatias/metabolismo , Doenças Renais Policísticas/metabolismo , Receptores de Superfície Celular/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Apoptose , Western Blotting , Proliferação de Células , Cistos/genética , Cistos/metabolismo , Feminino , Glucosidases/genética , Técnicas Imunoenzimáticas , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/genética , Hepatopatias/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Doenças Renais Policísticas/genética , Receptores de Superfície Celular/genética , Canais de Cátion TRPP/genética
17.
Biochem J ; 433(2): 285-94, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21044049

RESUMO

PKD2 is one of the two genes mutated in ADPKD (autosomal-dominant polycystic kidney disease). The protein product of PKD2, polycystin-2, functions as a non-selective cation channel in the endoplasmic reticulum and possibly at the plasma membrane. Hydrophobicity plots and its assignment to the TRP (transient receptor potential) family of cation channels suggest that polycystin-2 contains six transmembrane domains and that both the N- and C-termini extend into the cytoplasm. However, no experimental evidence for this model has so far been provided. To determine the orientation of the different loops of polycystin-2, we truncated polycystin-2 within the predicted loops 1-5 and tagged the constructs at the C-terminus with an HA (haemagglutinin) epitope. After transient expression and selective membrane permeabilization, immunofluorescence staining for the HA epitope revealed that loops 1, 3 and 5 extend into the lumen of the endoplasmic reticulum or the extracellular space, whereas loops 2 and 4 extend into the cytoplasm. This approach also confirmed the cytoplasmic orientation of the N- and C-termini of polycystin-2. In accordance with the immunofluorescence data, protease protection assays from microsomal preparations yielded protected fragments when polycystin-2 was truncated in loops 1, 3 and 5, whereas no protected fragments could be detected when polycystin-2 was truncated in loops 2 and 4. The results of the present study therefore provide the first experimental evidence for the topological orientation of polycystin-2.


Assuntos
Membrana Celular/química , Espaço Intracelular/química , Canais de Cátion TRPP/química , Sequência de Aminoácidos , Animais , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Humanos , Espaço Intracelular/metabolismo , Dados de Sequência Molecular , Canais de Cátion TRPP/metabolismo
18.
Adv Chronic Kidney Dis ; 17(2): 118-30, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20219615

RESUMO

Autosomal dominant polycystic disease (ADPKD) is the most common form of inherited kidney disease that results in renal failure. The understanding of the pathogenesis of ADPKD has advanced significantly since the discovery of the 2 causative genes, PKD1 and PKD2. Dominantly inherited gene mutations followed by somatic second-hit mutations inactivating the normal copy of the respective gene result in renal tubular cyst formation that deforms the kidney and eventually impairs its function. The respective gene products, polycystin-1 and polycystin-2, work together in a common cellular pathway. Polycystin-1, a large receptor molecule, forms a receptor-channel complex with polycystin-2, which is a cation channel belonging to the TRP family. Both polycystin proteins have been localized to the primary cilium, a nonmotile microtubule-based structure that extends from the apical membrane of tubular cells into the lumen. Here we discuss recent insights in the pathogenesis of ADPKD including the genetics of ADPKD, the properties of the respective polycystin proteins, the role of cilia, and some cell-signaling pathways that have been implicated in the pathways related to PKD1 and PKD2.


Assuntos
Rim Policístico Autossômico Dominante , Canais de Cátion TRPP/genética , Animais , Cílios/patologia , Humanos , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Rim Policístico Autossômico Dominante/fisiopatologia , Estrutura Terciária de Proteína , Transdução de Sinais/fisiologia , Canais de Cátion TRPP/química , Canais de Cátion TRPP/metabolismo
19.
J Am Soc Nephrol ; 21(2): 295-302, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19959710

RESUMO

Polycystic kidney disease (PKD) can arise from either developmental or postdevelopmental processes. Recessive PKD, caused by mutations in PKHD1, is a developmental defect, whereas dominant PKD, caused by mutations in PKD1 or PKD2, occurs by a cellular recessive mechanism in mature kidneys. Oriented cell division is a feature of planar cell polarity that describes the orientation of the mitotic axes of dividing cells during development with respect to the luminal vector of the elongating nephron. In polycystic mutant mice, the loss of oriented cell division may also contribute to the pathogenesis of PKD. Here, we examined the role of oriented cell division in mouse models based on mutations in Pkd1, Pkd2, and Pkhd1. Precystic tubules after kidney-selective inactivation of either Pkd1 or Pkd2 did not lose oriented division before cystic dilation but lost oriented division after tubular dilation began. In contrast, Pkhd1(del4/del4) mice lost oriented cell division but did not develop kidney cysts. Increased intercalation of cells into the plane of the tubular epithelium maintained the normal tubular morphology in Pkhd1(del4/del4) mice, which had more cells present in transverse tubular profiles. In conclusion, loss of oriented cell division is a feature of Pkhd1 mutation and cyst formation, but it is neither sufficient to produce kidney cysts nor required to initiate cyst formation after mutation in Pkd1 or Pkd2.


Assuntos
Divisão Celular/fisiologia , Polaridade Celular/fisiologia , Túbulos Renais/patologia , Doenças Renais Policísticas/fisiopatologia , Animais , Modelos Animais de Doenças , Túbulos Renais/metabolismo , Camundongos , Camundongos Transgênicos , Mutação/genética , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo
20.
Histochem Cell Biol ; 132(2): 199-210, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19381676

RESUMO

Nephronophthisis belongs to a family of recessive cystic kidney diseases and may arise from mutations in multiple genes. In this report we have used a spontaneous mouse mutant of type 3 nephronophthisis to examine whether the doxycycline-inducible synthesis of Timp-2, a natural inhibitor of matrix metalloproteinases, can influence renal cyst growth in transgenic mice. Metalloproteinases may exert either a negative or a positive effect on the progression of cystic kidney disease, and we reasoned that this may be most effectively examined by using a natural inhibitor. Surprisingly, already the application of doxycycline, which also inhibits matrix metalloproteinases, accelerated renal cyst growth and led to increased renal fibrosis, an additional effect of Timp-2 was not detected. The positive effect of doxycycline on kidney size was not due to a non-specific "anabolic effect" but was specific for cystic kidneys because it was not observed in non-cystic kidneys. When looking for potential metabolic changes we noticed that the urine of control animals led to an increase in the calcium response of LLC-PK(1) cells, whereas the urine of doxycycline-treated mice showed the opposite effect and even antagonized the urine of control animals. Further experiments demonstrated that the urine of control animals contained a heat-labile, proteinase K-resistant substance which appears to be responsible for the induction of a calcium response in LLC-PK(1) cells. We conclude that doxycycline accelerates cyst growth possibly by the induction of a substance which lowers the intracellular calcium concentration. Our data also add a note of caution when interpreting phenotypes of animal models based upon the tet system.


Assuntos
Cistos/enzimologia , Rim/enzimologia , Rim/patologia , Doenças Renais Policísticas/enzimologia , Inibidor Tecidual de Metaloproteinase-2/biossíntese , Animais , Cistos/induzido quimicamente , Cistos/genética , Modelos Animais de Doenças , Doxiciclina/farmacologia , Fibrose , Células HeLa , Humanos , Rim/efeitos dos fármacos , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Doenças Renais Policísticas/induzido quimicamente , Doenças Renais Policísticas/genética , Receptores de Superfície Celular/genética , Inibidor Tecidual de Metaloproteinase-2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...