Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 150(3): 533-48, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22863007

RESUMO

Nephronophthisis-related ciliopathies (NPHP-RC) are degenerative recessive diseases that affect kidney, retina, and brain. Genetic defects in NPHP gene products that localize to cilia and centrosomes defined them as "ciliopathies." However, disease mechanisms remain poorly understood. Here, we identify by whole-exome resequencing, mutations of MRE11, ZNF423, and CEP164 as causing NPHP-RC. All three genes function within the DNA damage response (DDR) pathway. We demonstrate that, upon induced DNA damage, the NPHP-RC proteins ZNF423, CEP164, and NPHP10 colocalize to nuclear foci positive for TIP60, known to activate ATM at sites of DNA damage. We show that knockdown of CEP164 or ZNF423 causes sensitivity to DNA damaging agents and that cep164 knockdown in zebrafish results in dysregulated DDR and an NPHP-RC phenotype. Our findings link degenerative diseases of the kidney and retina, disorders of increasing prevalence, to mechanisms of DDR.


Assuntos
Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Exoma , Doenças Renais Císticas/genética , Proteínas dos Microtúbulos/metabolismo , Animais , Cílios/metabolismo , Técnicas de Silenciamento de Genes , Genes Recessivos , Humanos , Proteína Homóloga a MRE11 , Camundongos , Proteínas , Transdução de Sinais , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
2.
Nucleic Acids Res ; 50(20): 11401-11414, 2022 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-35944903

RESUMO

Current therapies for Duchenne muscular dystrophy (DMD) use phosphorodiamidate morpholino oligomers (PMO) to induce exon skipping in the dystrophin pre-mRNA, enabling the translation of a shortened but functional dystrophin protein. This strategy has been hampered by insufficient delivery of PMO to cardiac and skeletal muscle. To overcome these limitations, we developed the FORCETM platform consisting of an antigen-binding fragment, which binds the transferrin receptor 1, conjugated to an oligonucleotide. We demonstrate that a single dose of the mouse-specific FORCE-M23D conjugate enhances muscle delivery of exon skipping PMO (M23D) in mdx mice, achieving dose-dependent and robust exon skipping and durable dystrophin restoration. FORCE-M23D-induced dystrophin expression reached peaks of 51%, 72%, 62%, 90% and 77%, of wild-type levels in quadriceps, tibialis anterior, gastrocnemius, diaphragm, and heart, respectively, with a single 30 mg/kg PMO-equivalent dose. The shortened dystrophin localized to the sarcolemma, indicating expression of a functional protein. Conversely, a single 30 mg/kg dose of unconjugated M23D displayed poor muscle delivery resulting in marginal levels of exon skipping and dystrophin expression. Importantly, FORCE-M23D treatment resulted in improved functional outcomes compared with administration of unconjugated M23D. Our results suggest that FORCE conjugates are a potentially effective approach for the treatment of DMD.


The biggest problem confronting oligonucleotide therapeutics is a lack of compounds capable of targeting compounds to diseased tissues. This paper reports a major advance targeting the transferrin receptor to increase the delivery of morpholine oligomers to muscle cells in vivo. This work suggests the possibility for improved treatments of muscular dystrophy and other diseases.


Assuntos
Distrofina , Éxons , Morfolinos , Distrofia Muscular de Duchenne , Oligonucleotídeos Antissenso , Animais , Camundongos , Distrofina/genética , Éxons/genética , Camundongos Endogâmicos mdx , Morfolinos/farmacologia , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/farmacologia , Receptores da Transferrina/genética
3.
Hum Mol Genet ; 29(15): 2508-2522, 2020 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-32620959

RESUMO

Bardet-Biedl syndrome (BBS) is a pleiotropic autosomal recessive ciliopathy affecting multiple organs. The development of potential disease-modifying therapy for BBS will require concurrent targeting of multi-systemic manifestations. Here, we show for the first time that monosialodihexosylganglioside accumulates in Bbs2-/- cilia, indicating impairment of glycosphingolipid (GSL) metabolism in BBS. Consequently, we tested whether BBS pathology in Bbs2-/- mice can be reversed by targeting the underlying ciliary defect via reduction of GSL metabolism. Inhibition of GSL synthesis with the glucosylceramide synthase inhibitor Genz-667161 decreases the obesity, liver disease, retinal degeneration and olfaction defect in Bbs2-/- mice. These effects are secondary to preservation of ciliary structure and signaling, and stimulation of cellular differentiation. In conclusion, reduction of GSL metabolism resolves the multi-organ pathology of Bbs2-/- mice by directly preserving ciliary structure and function towards a normal phenotype. Since this approach does not rely on the correction of the underlying genetic mutation, it might translate successfully as a treatment for other ciliopathies.


Assuntos
Síndrome de Bardet-Biedl/genética , Cílios/genética , Ciliopatias/genética , Proteínas/genética , Animais , Síndrome de Bardet-Biedl/tratamento farmacológico , Síndrome de Bardet-Biedl/patologia , Diferenciação Celular/efeitos dos fármacos , Cílios/patologia , Ciliopatias/tratamento farmacológico , Ciliopatias/patologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Gangliosídeos/biossíntese , Gangliosídeos/genética , Glucosiltransferases/antagonistas & inibidores , Glucosiltransferases/genética , Glicoesfingolipídeos/biossíntese , Glicoesfingolipídeos/genética , Camundongos Knockout
4.
Am J Physiol Renal Physiol ; 315(1): F57-F73, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29537311

RESUMO

Following the discovery of (R)-roscovitine's beneficial effects in three polycystic kidney disease (PKD) mouse models, cyclin-dependent kinases (CDKs) inhibitors have been investigated as potential treatments. We have used various affinity chromatography approaches to identify the molecular targets of roscovitine and its more potent analog (S)-CR8 in human and murine polycystic kidneys. These methods revealed casein kinases 1 (CK1) as additional targets of the two drugs. CK1ε expression at the mRNA and protein levels is enhanced in polycystic kidneys of 11 different PKD mouse models as well as in human polycystic kidneys. A shift in the pattern of CK1α isoforms is observed in all PKD mouse models. Furthermore, the catalytic activities of both CK1ε and CK1α are increased in mouse polycystic kidneys. Inhibition of CK1ε and CK1α may thus contribute to the long-lasting attenuating effects of roscovitine and (S)-CR8 on cyst development. CDKs and CK1s may constitute a dual therapeutic target to develop kinase inhibitory PKD drug candidates.


Assuntos
Caseína Quinase 1 épsilon/antagonistas & inibidores , Caseína Quinase Ialfa/antagonistas & inibidores , Rim/efeitos dos fármacos , Doenças Renais Policísticas/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Piridinas/farmacologia , Roscovitina/farmacologia , Animais , Caseína Quinase 1 épsilon/genética , Caseína Quinase 1 épsilon/metabolismo , Caseína Quinase Ialfa/genética , Caseína Quinase Ialfa/metabolismo , Catálise , Cromatografia de Afinidade/métodos , Modelos Animais de Doenças , Humanos , Rim/enzimologia , Rim/patologia , Camundongos Transgênicos , Doenças Renais Policísticas/enzimologia , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Ligação Proteica , Inibidores de Proteínas Quinases/metabolismo , Purinas/metabolismo , Piridinas/metabolismo , Roscovitina/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Hum Mol Genet ; 25(11): 2245-2255, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27053712

RESUMO

Polycystic kidney diseases (PKDs) comprise a subgroup of ciliopathies characterized by the formation of fluid-filled kidney cysts and progression to end-stage renal disease. A mechanistic understanding of cystogenesis is crucial for the development of viable therapeutic options. Here, we identify CDK5, a kinase active in post mitotic cells, as a new and important mediator of PKD progression. We show that long-lasting attenuation of PKD in the juvenile cystic kidneys (jck) mouse model of nephronophthisis by pharmacological inhibition of CDK5 using either R-roscovitine or S-CR8 is accompanied by sustained shortening of cilia and a more normal epithelial phenotype, suggesting this treatment results in a reprogramming of cellular differentiation. Also, a knock down of Cdk5 in jck cells using small interfering RNA results in significant shortening of ciliary length, similar to what we observed with R-roscovitine. Finally, conditional inactivation of Cdk5 in the jck mice significantly attenuates cystic disease progression and is associated with shortening of ciliary length as well as restoration of cellular differentiation. Our results suggest that CDK5 may regulate ciliary length by affecting tubulin dynamics via its substrate collapsin response mediator protein 2. Taken together, our data support therapeutic approaches aimed at restoration of ciliogenesis and cellular differentiation as a promising strategy for the treatment of renal cystic diseases.


Assuntos
Cílios/efeitos dos fármacos , Quinase 5 Dependente de Ciclina/genética , Falência Renal Crônica/tratamento farmacológico , Doenças Renais Policísticas/tratamento farmacológico , Animais , Diferenciação Celular/efeitos dos fármacos , Cílios/patologia , Quinase 5 Dependente de Ciclina/antagonistas & inibidores , Modelos Animais de Doenças , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Falência Renal Crônica/genética , Falência Renal Crônica/patologia , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Purinas/administração & dosagem , Roscovitina , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
6.
Am J Physiol Renal Physiol ; 310(11): F1414-22, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27076647

RESUMO

Polycystic kidney diseases (PKDs) are genetic diseases characterized by renal cyst formation with increased cell proliferation, apoptosis, and transition to a secretory phenotype at the expense of terminal differentiation. Despite recent progress in understanding PKD pathogenesis and the emergence of potential therapies, the key molecular mechanisms promoting cystogenesis are not well understood. Here, we demonstrate that mechanisms including endoplasmic reticulum stress, oxidative damage, and compromised mitochondrial function all contribute to nephronophthisis-associated PKD. Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is emerging as a critical mediator of these cellular processes. Therefore, we reasoned that pharmacological targeting of CaMKII may translate into effective inhibition of PKD in jck mice. Our data demonstrate that CaMKII is activated within cystic kidney epithelia in jck mice. Blockade of CaMKII with a selective inhibitor results in effective inhibition of PKD in jck mice. Mechanistic experiments in vitro and in vivo demonstrated that CaMKII inhibition relieves endoplasmic reticulum stress and oxidative damage and improves mitochondrial integrity and membrane potential. Taken together, our data support CaMKII inhibition as a new and effective therapeutic avenue for the treatment of cystic diseases.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Rim/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo/fisiologia , Doenças Renais Policísticas/metabolismo , Animais , Camundongos
7.
Hum Mol Genet ; 21(15): 3397-407, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22563011

RESUMO

Genetic forms of polycystic kidney diseases (PKDs), including nephronophthisis, are characterized by formation of fluid-filled cysts in the kidneys and progression to end-stage renal disease. No therapies are currently available to treat cystic diseases, making it imperative to dissect molecular mechanisms in search of therapeutic targets. Accumulating evidence suggests a pathogenic role for glucosylceramide (GlcCer) in multiple forms of PKD. It is not known, however, whether other structural glycosphingolipids (GSLs) or bioactive signaling sphingolipids (SLs) modulate cystogenesis. Therefore, we set out to address the role of a specific GSL (ganglioside GM3) and signaling SL (sphingosine-1-phosphate, S1P) in PKD progression, using the jck mouse model of nephronopthisis. To define the role of GM3 accumulation in cystogenesis, we crossed jck mice with mice carrying a targeted mutation in the GM3 synthase (St3gal5) gene. GM3-deficient jck mice displayed milder PKD, revealing a pivotal role for ganglioside GM3. Mechanistic changes in regulation of the cell-cycle machinery and Akt-mTOR signaling were consistent with reduced cystogenesis. Dramatic overexpression of sphingosine kinase 1 (Sphk1) mRNA in jck kidneys suggested a pathogenic role for S1P. Surprisingly, genetic loss of Sphk1 exacerbated cystogenesis and was associated with increased levels of GlcCer and GM3. On the other hand, increasing S1P accumulation through pharmacologic inhibition of S1P lyase had no effect on the progression of cystogenesis or kidney GSL levels. Together, these data suggest that genes involved in the SL metabolism may be modifiers of cystogenesis, and suggest GM3 synthase as a new anti-cystic therapeutic target.


Assuntos
Fosfotransferases (Aceptor do Grupo Álcool)/genética , Doenças Renais Policísticas/genética , Sialiltransferases/genética , Animais , Modelos Animais de Doenças , Glucosilceramidas/metabolismo , Glicoesfingolipídeos/metabolismo , Camundongos , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Doenças Renais Policísticas/enzimologia , Sialiltransferases/metabolismo , Esfingosina/metabolismo , Serina-Treonina Quinases TOR/metabolismo
8.
Nature ; 444(7121): 949-52, 2006 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-17122773

RESUMO

Polycystic kidney diseases (PKDs) are primarily characterized by the growth of fluid-filled cysts in renal tubules leading to end-stage renal disease. Mutations in the PKD1 or PKD2 genes lead to autosomal dominant PKD (ADPKD), a slowly developing adult form. Autosomal recessive polycystic kidney disease results from mutations in the PKHD1 gene, affects newborn infants and progresses very rapidly. No effective treatment is currently available for PKD. A previously unrecognized site of subcellular localization was recently discovered for all proteins known to be disrupted in PKD: primary cilia. Because ciliary functions seem to be involved in cell cycle regulation, disruption of proteins associated with cilia or centrioles may directly affect the cell cycle and proliferation, resulting in cystic disease. We therefore reasoned that the dysregulated cell cycle may be the most proximal cause of cystogenesis, and that intervention targeted at this point could provide significant therapeutic benefit for PKD. Here we show that treatment with the cyclin-dependent kinase (CDK) inhibitor (R)-roscovitine does indeed yield effective arrest of cystic disease in jck and cpk mouse models of PKD. Continuous daily administration of the drug is not required to achieve efficacy; pulse treatment provides a robust, long-lasting effect, indicating potential clinical benefits for a lifelong therapy. Molecular studies of the mechanism of action reveal effective cell-cycle arrest, transcriptional inhibition and attenuation of apoptosis. We found that roscovitine is active against cysts originating from different parts of the nephron, a desirable feature for the treatment of ADPKD, in which cysts form in multiple nephron segments. Our results indicate that inhibition of CDK is a new and effective approach to the treatment of PKD.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Modelos Animais de Doenças , Doenças Renais Policísticas/tratamento farmacológico , Purinas/farmacologia , Purinas/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Progressão da Doença , Rim/efeitos dos fármacos , Rim/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Doenças Renais Policísticas/patologia , Purinas/administração & dosagem , Roscovitina , Fatores de Tempo
9.
Sci Rep ; 12(1): 9340, 2022 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-35660779

RESUMO

A significant population of patients with chronic kidney disease (CKD) develops cardiac hypertrophy, which can lead to heart failure and sudden cardiac death. Soluble klotho (sKL), the shed ectodomain of the transmembrane protein klotho, protects the heart against hypertrophic growth. We have shown that sKL protects the heart by regulating the formation and function of lipid rafts by targeting the sialic acid moiety of gangliosides, GM1/GM3. Reduction in circulating sKL contributes to an increased risk of cardiac hypertrophy in mice. sKL replacement therapy has been considered but its use is limited by the inability to mass produce the protein. Therefore, alternative methods to protect the heart are proposed. Glucosylation of ceramide catalyzed by glucosylceramide synthase is the entry step for the formation of gangliosides. Here we show that oral administration of a glucosylceramide synthase inhibitor (GCSi) reduces plasma and heart tissue glycosphingolipids, including gangliosides. Administration of GCSi is protective in two mouse models of cardiac stress-induction, one with isoproterenol overstimulation and the other with 5/6 nephrectomy-induced CKD. Treatment with GCSi does not alter the severity of renal dysfunction and hypertension in CKD. These results provide proof of principle for targeting glucosylceramide synthase to decrease gangliosides as a treatment for cardiac hypertrophy. They also support the hypothesis that sKL protects the heart by targeting gangliosides.


Assuntos
Cardiomegalia , Insuficiência Renal Crônica , Animais , Cardiomegalia/tratamento farmacológico , Cardiomegalia/prevenção & controle , Gangliosídeos/metabolismo , Glucosiltransferases , Humanos , Camundongos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/tratamento farmacológico
10.
Cells ; 11(4)2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-35203245

RESUMO

Col4a3-/- Alport mice serve as an animal model for renal fibrosis. MicroRNA-21 (miR-21) expression has been shown to be increased in the kidneys of Alport syndrome patients. Here, we investigated the nephroprotective effects of Lademirsen anti-miR-21 therapy. We used a fast-progressing Col4a3-/- mouse model with a 129/SvJ background and an intermediate-progressing F1 hybrid mouse model with a mixed genetic background, with angiotensin-converting enzyme inhibitor (ACEi) monotherapy in combination with anti-miR-21 therapy. In the fast-progressing model, the anti miR-21 and ACEi therapies showed an additive effect in the reduction in fibrosis, the decline of proteinuria, the preservation of kidney function and increased survival. In the intermediate-progressing F1 model, the anti-miR-21 and ACEi therapies individually improved kidney pathology. Both also improved kidney function and survival; however, the combination showed a significant additive effect, particularly for survival. RNA sequencing (RNA-seq) gene expression profiling revealed that the anti-miR-21 and ACEi therapies modulate several common pathways. However, anti-miR-21 was particularly effective at normalizing the expression profiles of the genes involved in renal tubulointerstitial injury pathways. In conclusion, significant additive effects were detected for the combination of anti-miR-21 and ACEi therapies on kidney function, pathology and survival in Alport mouse models, as well as a strong differential effect of anti-miR-21 on the renal expression of fibrotic factors. These results support the addition of anti-miR-21 to the current standard of care (ACEi) in ongoing clinical trials in patients with Alport syndrome.


Assuntos
Inibidores da Enzima Conversora de Angiotensina , MicroRNAs , Nefrite Hereditária , Insuficiência Renal , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Antagomirs , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Modelos Animais de Doenças , Fibrose , Humanos , Camundongos , Camundongos Knockout , MicroRNAs/antagonistas & inibidores , Nefrite Hereditária/tratamento farmacológico , Nefrite Hereditária/genética , Insuficiência Renal/tratamento farmacológico
11.
JBMR Plus ; 5(9): e10530, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34532615

RESUMO

Osteogenesis imperfecta (OI), is a genetic disorder of bone fragility caused by mutations in collagen I or proteins involved in collagen processing. Previous studies in mice and human OI bones have shown that excessive activation of TGF-ß signaling plays an important role in dominant and recessive OI disease progression. Inhibition of TGF-ß signaling with a murine pan-specific TGF-ß neutralizing antibody (1D11) was shown to significantly increase trabecular bone volume and long bone strength in mouse models of OI. To investigate the frequency of dosing and dose options of TGF-ß neutralizing antibody therapy, we assessed the effect of 1D11 on disease progression in a dominant OI mouse model (col1a2 gene mutation at G610C). In comparison with OI mice treated with a control antibody, we attempted to define mechanistic effects of 1D11 measured via µCT, biomechanical, dynamic histomorphometry, and serum biomarkers of bone turnover. In addition, osteoblast and osteoclast numbers in histological bone sections were assessed to better understand the mechanism of action of the 1D11 antibody in OI. Here we show that 1D11 treatment resulted in both dose and frequency dependency, increases in trabecular bone volume fraction and ultimate force in lumbar bone, and ultimate force, bending strength, yield force, and yield strength in the femur (p ≤ 0.05). Suppression of serum biomarkers of osteoblast differentiation, osteocalcin, resorption, CTx-1, and bone formation were observed after 1D11 treatment of OI mice. Immunohistochemical analysis showed dose and frequency dependent decreases in runt-related transcription factor, and increase in alkaline phosphatase in lumbar bone sections. In addition, a significant decrease in TRACP and the number of osteoclasts to bone surface area was observed with 1D11 treatment. Our results show that inhibition of the TGF-ß pathway corrects the high-turnover aspects of bone disease and improves biomechanical properties of OI mice. These results highlight the potential for a novel treatment for osteogenesis imperfecta. © 2021 Sanofi-Genzyme. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

12.
Cell Signal ; 69: 109526, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31911181

RESUMO

Sphingolipids and glycosphingolipids are classes of structurally and functionally important lipids that regulate multiple cellular processes, including membrane organization, proliferation, cell cycle regulation, apoptosis, transport, migration, and inflammatory signalling pathways. Imbalances in sphingolipid levels or subcellular localization result in dysregulated cellular processes and lead to the development and progression of multiple disorders, including polycystic kidney disease. This review will describe metabolic pathways of glycosphingolipids with a focus on the evidence linking glycosphingolipid mediated regulation of cell signalling, lipid microdomains, cilia, and polycystic kidney disease. We will discuss molecular mechanisms of glycosphingolipid dysregulation and their impact on cystogenesis. We will further highlight how modulation of sphingolipid metabolism can be translated into new approaches for the treatment of polycystic kidney disease and describe current clinical studies with glucosylceramide synthase inhibitors in Autosomal Dominant Polycystic Kidney Disease.


Assuntos
Cílios , Glicoesfingolipídeos/metabolismo , Rim Policístico Autossômico Dominante , Animais , Cílios/metabolismo , Cílios/patologia , Cistos/metabolismo , Cistos/patologia , Humanos , Rim/metabolismo , Rim/patologia , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Transdução de Sinais
13.
Hum Gene Ther ; 30(7): 865-881, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30808234

RESUMO

Alport syndrome is a genetic disease caused by mutations in type IV collagen and is characterized by progressive kidney disease. The Col4α3-/- mouse model recapitulates the main features of human Alport syndrome. Previously, it was reported that kidney microRNA-21 (miR-21) expression is significantly increased in Col4α3-/- mice, and administration of anti-miR-21 oligonucleotides (anti-miR-21) attenuates kidney disease progression in Col4α3-/- mice, indicating that miR-21 is a viable therapeutic target for Alport syndrome. However, the expression pattern of miR-21 in the kidneys of patients with human Alport syndrome has not been evaluated. Paraffin-embedded kidney specimens were obtained from 27 patients with Alport syndrome and from 10 normal controls. They were evaluated for miR-21 expression and for in situ hybridization and mRNA expression by quantitative polymerase chain reaction. In addition, anti-miR-21 was administrated to Col4α3-/- mice at different stages of disease, and changes in proteinuria, kidney function, and survival were monitored. Transcriptomic analysis of mouse kidney was conducted using RNA sequencing. miR-21 expression was significantly elevated in kidney specimens from patients with Alport syndrome compared to normal controls. Elevated renal miR-21 expression positively correlated with 24 h urine protein, serum blood urea nitrogen, serum creatinine, and severity of kidney pathology. On histological evaluation, high levels of miR-21 were localized to damaged tubular epithelial cells and glomeruli. Kidney specimens from both humans and mice with Alport syndrome exhibited abnormal expression of genes involved in kidney injury, fibrosis, inflammation, mitochondrial function, and lipid metabolism. Administration of anti-miR-21 to Alport mice resulted in slowing of kidney function decline, partial reversal of abnormal gene expression associated with disease pathology, and improved survival. Increased levels of miR-21 in human Alport kidney samples showed a correlation with kidney disease severity measured by proteinuria, biomarkers of kidney function, and kidney histopathology scores. These human data, combined with the finding that a reduction of miR-21 in Col4α3-/- mice improves kidney phenotype and survival, support miR-21 as a viable therapeutic target for the treatment of Alport syndrome.


Assuntos
Regulação da Expressão Gênica , Predisposição Genética para Doença , MicroRNAs/genética , Nefrite Hereditária/genética , Adolescente , Animais , Autoantígenos , Biomarcadores , Biópsia , Criança , Colágeno Tipo IV/deficiência , Modelos Animais de Doenças , Feminino , Fibrose , Perfilação da Expressão Gênica , Estudos de Associação Genética , Humanos , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Knockout , Nefrite Hereditária/diagnóstico , Nefrite Hereditária/metabolismo , Índice de Gravidade de Doença
14.
J Clin Invest ; 114(10): 1433-43, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15545994

RESUMO

Polycystin-1, which is encoded by a gene that is mutated in autosomal dominant polycystic kidney disease (ADPKD), is involved in cell-matrix interactions as well as in ciliary signaling. The precise mechanisms by which it functions, however, remain unclear. Here we find that polycystin-1 undergoes a proteolytic cleavage that releases its C-terminal tail (CTT), which enters the nucleus and initiates signaling processes. The cleavage occurs in vivo in association with alterations in mechanical stimuli. Polycystin-2, the product of the second gene mutated in ADPKD, modulates the signaling properties of the polycystin-1 CTT. These data reveal a novel pathway by which polycystin-1 transmits messages directly to the nucleus.


Assuntos
Núcleo Celular/metabolismo , Proteínas/química , Proteínas/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Células CHO , Células COS , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Cães , Embrião de Mamíferos , Células Epiteliais/citologia , Túbulos Renais/citologia , Túbulos Renais/embriologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Proteínas/genética , Deleção de Sequência , Estresse Mecânico , Canais de Cátion TRPP , Fator de Transcrição AP-1/metabolismo
15.
Nephrol Ther ; 2 Suppl 2: S109-14, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17373209

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) cystogenesis has been extensively studied and major characteristic abnormalities were identified including increased proliferation, apoptosis, changes in cellular polarity, abnormal matrix composition and fluid secretion. We set out to understand how mutated polycystin-1 leads to these abnormalities and how well mechanisms of cystogenesis in vivo can be understood using in vitro models. Specifically, we addressed the dynamic role of polycystin-1 in the context of cellular rearrangements accompanying cystogenesis and tubulogenesis in vitro. We demonstrated that polycystin-1 plays an important role in cell-cell adhesion through homophilic interactions of its Ig-like domains. To define the role of polycystin-1 in formation of intercellular contacts and cell polarity during epithelial morphogenesis, we have utilized a 3D MDCK in vitro model of tubulogenesis and cystogenesis. We demonstrate that polycystin-1 is a component of desmosomal junctions of epithelial cells. A striking down-regulation of polycystin-1 mRNA was detected in cysts as compared to tubules, leading to altered protein expression and localization. While polycystin-1 is localized to basolateral membranes of MDCK tubules, it is only detected in cytoplasmic pools in cystic cells. To address the impact of mutated PC-1 on intercellular adhesion, we have analyzed the structure/function of desmosomal junctions in primary cells derived from ADPKD cysts. We demonstrated that, in the absence of functional polycystin-1, desmosomal junctions can not be properly assembled and remain sequestered in cytoplasmic compartments. Our data show that the MDCK in vitro model of cystogenesis and tubulogenesis adequately reflects several aspects of cystogenesis in vivo. We have used in vitro cystogenesis assay for a high throughput screen of small molecule drugs and identified drugs specifically inhibiting cystogenesis but not tubulogenesis in vitro.


Assuntos
Doenças Renais Policísticas/prevenção & controle , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/metabolismo , Apoptose , Adesão Celular , Divisão Celular , Polaridade Celular , Células Epiteliais/fisiologia , Regulação da Expressão Gênica , Humanos , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Doenças Renais Policísticas/fisiopatologia
16.
Physiol Rep ; 4(12)2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27356569

RESUMO

Development of a disease-modifying therapy to treat autosomal dominant polycystic kidney disease (ADPKD) requires well-characterized preclinical models that accurately reflect the pathology and biochemical changes associated with the disease. Using a Pkd1 conditional knockout mouse, we demonstrate that subtly altering the timing and extent of Pkd1 deletion can have a significant impact on the origin and severity of kidney cyst formation. Pkd1 deletion on postnatal day 1 or 2 results in cysts arising from both the cortical and medullary regions, whereas deletion on postnatal days 3-8 results in primarily medullary cyst formation. Altering the extent of Pkd1 deletion by modulating the tamoxifen dose produces dose-dependent changes in the severity, but not origin, of cystogenesis. Limited Pkd1 deletion produces progressive kidney cystogenesis, accompanied by interstitial fibrosis and loss of kidney function. Cyst growth occurs in two phases: an early, rapid growth phase, followed by a later, slow growth period. Analysis of biochemical pathway changes in cystic kidneys reveals dysregulation of the cell cycle, increased proliferation and apoptosis, activation of Mek-Erk, Akt-mTOR, and Wnt-ß-catenin signaling pathways, and altered glycosphingolipid metabolism that resemble the biochemical changes occurring in human ADPKD kidneys. These pathways are normally active in neonatal mouse kidneys until repressed around 3 weeks of age; however, they remain active following Pkd1 deletion. Together, this work describes the key parameters to accurately model the pathological and biochemical changes associated with ADPKD in a conditional mouse model.


Assuntos
Deleção de Genes , Doenças Renais Policísticas/genética , Canais de Cátion TRPP/metabolismo , Animais , Modelos Animais de Doenças , Fibrose , Rim/metabolismo , Rim/patologia , Sistema de Sinalização das MAP Quinases , Camundongos , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia , Canais de Cátion TRPP/genética , Via de Sinalização Wnt
17.
Physiol Genomics ; 8(2): 87-98, 2002 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-11875186

RESUMO

Polycystin-1 (PKD1) mutations account for approximately 85% of autosomal dominant polycystic kidney disease (ADPKD). We have shown previously that oocyte surface expression of a transmembrane fusion protein encoding part of the cytoplasmic COOH terminus of PKD1 increases activity of a Ca2+-permeable cation channel. We show here that human ADPKD mutations incorporated into this fusion protein attenuated or abolished encoded cation currents. Point mutations and truncations showed that cation current expression requires integrity of a region encompassing the putative coiled coil domain of the PKD1 cytoplasmic tail. Whereas these loss-of-function mutants did not exhibit dominant negative phenotypes, coexpression of a fusion protein expressing the interacting COOH-terminal cytoplasmic tail of PKD2 did suppress cation current. Liganding of the ectodomain of the PKD1 fusion protein moderately activated cation current. The divalent cation permeability and pharmacological profile of the current has been extended. Inducible expression of the PKD1 fusion in EcR-293 cells was also associated with activation of cation channels and increased Ca2+ entry.


Assuntos
Canais de Cálcio/fisiologia , Mutação de Sentido Incorreto , Fragmentos de Peptídeos/fisiologia , Proteínas/fisiologia , Animais , Cálcio/antagonistas & inibidores , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Cátions Bivalentes/antagonistas & inibidores , Cátions Bivalentes/metabolismo , Linhagem Celular , Citoplasma/genética , Citoplasma/fisiologia , Análise Mutacional de DNA , Humanos , Ligantes , Oócitos/química , Oócitos/citologia , Oócitos/metabolismo , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/fisiopatologia , Biossíntese de Proteínas , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Proteínas/química , Proteínas/genética , Receptores de IgG/biossíntese , Receptores de IgG/química , Receptores de IgG/genética , Receptores de IgG/fisiologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Canais de Cátion TRPP , Regulação para Cima/genética , Xenopus laevis/embriologia
18.
Cell Cycle ; 11(21): 4040-6, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23032260

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) and other forms of PKD are associated with dysregulated cell cycle and proliferation. Although no effective therapy for the treatment of PKD is currently available, possible mechanism-based approaches are beginning to emerge. A therapeutic intervention targeting aberrant cilia-cell cycle connection using CDK-inhibitor R-roscovitine showed effective arrest of PKD in jck and cpk models that are not orthologous to human ADPKD. To evaluate whether CDK inhibition approach will translate into efficacy in an orthologous model of ADPKD, we tested R-roscovitine and its derivative S-CR8 in a model with a conditionally inactivated Pkd1 gene (Pkd1 cKO). Similar to ADPKD, Pkd1 cKO mice developed renal and hepatic cysts. Treatment of Pkd1 cKO mice with R-roscovitine and its more potent and selective analog S-CR8 significantly reduced renal and hepatic cystogenesis and attenuated kidney function decline. Mechanism of action studies demonstrated effective blockade of cell cycle and proliferation and reduction of apoptosis. Together, these data validate CDK inhibition as a novel and effective approach for the treatment of ADPKD.


Assuntos
Adenina/análogos & derivados , Quinases Ciclina-Dependentes/antagonistas & inibidores , Doenças Renais Císticas/tratamento farmacológico , Hepatopatias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Purinas/uso terapêutico , Adenina/química , Adenina/farmacologia , Adenina/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Modelos Animais de Doenças , Humanos , Doenças Renais Císticas/enzimologia , Doenças Renais Císticas/patologia , Hepatopatias/enzimologia , Hepatopatias/patologia , Camundongos , Camundongos Knockout , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/enzimologia , Rim Policístico Autossômico Dominante/patologia , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Purinas/química , Purinas/farmacologia , Roscovitina
19.
Trends Mol Med ; 17(11): 625-33, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21775207

RESUMO

Polycystic kidney diseases (PKDs) comprise a large group of genetic disorders characterized by formation of cysts in the kidneys and other organs, ultimately leading to end-stage renal disease. Although PKDs can be caused by mutations in different genes, they converge on a set of common molecular mechanisms involved in cystogenesis and ciliary dysfunction, and can be qualified as ciliopathies. Recent advances in understanding the mechanisms regulating disease progression have led to the development of new therapies that are being tested in both preclinical and clinical trials. In this article, we briefly review a network of molecular pathways of cystogenesis that are regulated by ciliary functions. We discuss the mTOR pathway in depth, highlighting recent progress in understanding its role in PKD and the current results of clinical trials.


Assuntos
Doenças Renais Policísticas/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Cílios/metabolismo , Cílios/patologia , Ensaios Clínicos como Assunto , Humanos , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Mutação , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/genética , Proteína Quinase D2 , Proteínas Quinases/genética , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Canais de Cátion TRPP/genética
20.
Nat Med ; 16(7): 788-92, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20562878

RESUMO

Polycystic kidney disease (PKD) represents a family of genetic disorders characterized by renal cystic growth and progression to kidney failure. No treatment is currently available for people with PKD, although possible therapeutic interventions are emerging. Despite genetic and clinical heterogeneity, PKDs have in common defects of cystic epithelia, including increased proliferation, apoptosis and activation of growth regulatory pathways. Sphingolipids and glycosphingolipids are emerging as major regulators of these cellular processes. We sought to evaluate the therapeutic potential for glycosphingolipid modulation as a new approach to treat PKD. Here we demonstrate that kidney glucosylceramide (GlcCer) and ganglioside GM3 levels are higher in human and mouse PKD tissue as compared to normal tissue, regardless of the causative mutation. Blockade of GlcCer accumulation with the GlcCer synthase inhibitor Genz-123346 effectively inhibits cystogenesis in mouse models orthologous to human autosomal dominant PKD (Pkd1 conditional knockout mice) and nephronophthisis (jck and pcy mice). Molecular analysis in vitro and in vivo indicates that Genz-123346 acts through inhibition of the two key pathways dysregulated in PKD: Akt protein kinase-mammalian target of rapamycin signaling and cell cycle machinery. Taken together, our data suggest that inhibition of GlcCer synthesis represents a new and effective treatment option for PKD.


Assuntos
Dioxanos/farmacologia , Glucosilceramidas/biossíntese , Doenças Renais Policísticas/metabolismo , Pirrolidinas/farmacologia , Animais , Ciclo Celular , Modelos Animais de Doenças , Gangliosídeo G(M3)/metabolismo , Glucosiltransferases/antagonistas & inibidores , Glicoesfingolipídeos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Doenças Renais Policísticas/tratamento farmacológico , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA