Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Am J Physiol Renal Physiol ; 324(4): F404-F422, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36794754

RESUMO

Polycystic kidney disease is a disorder of renal epithelial growth and differentiation. Transcription factor EB (TFEB), a master regulator of lysosome biogenesis and function, was studied for a potential role in this disorder. Nuclear translocation and functional responses to TFEB activation were studied in three murine models of renal cystic disease, including knockouts of folliculin, folliculin interacting proteins 1 and 2, and polycystin-1 (Pkd1) as well as in mouse embryonic fibroblasts lacking Pkd1 and three-dimensional cultures of Madin-Darby canine kidney cells. Nuclear translocation of Tfeb characterized cystic but not noncystic renal tubular epithelia in all three murine models as both an early and sustained response to cyst formation. Epithelia expressed elevated levels of Tfeb-dependent gene products, including cathepsin B and glycoprotein nonmetastatic melanoma protein B. Nuclear Tfeb translocation was observed in mouse embryonic fibroblasts lacking Pkd1 but not wild-type fibroblasts. Pkd1 knockout fibroblasts were characterized by increased Tfeb-dependent transcripts, lysosomal biogenesis and repositioning, and increased autophagy. The growth of Madin-Darby canine kidney cell cysts was markedly increased following exposure to the TFEB agonist compound C1, and nuclear Tfeb translocation was observed in response to both forskolin and compound C1 treatment. Nuclear TFEB also characterized cystic epithelia but not noncystic tubular epithelia in human patients with autosomal dominant polycystic kidney disease. Noncanonical activation of TFEB is characteristic of cystic epithelia in multiple models of renal cystic disease including those associated with loss of Pkd1. Nuclear TFEB translocation is functionally active in these models and may be a component of a general pathway contributing to cystogenesis and growth.NEW & NOTEWORTHY Changes in epithelial cell metabolism are important in renal cyst development. The role of TFEB, a transcriptional regulator of lysosomal function, was explored in several models of renal cystic disease and human ADPKD tissue sections. Nuclear TFEB translocation was uniformly observed in cystic epithelia in each model of renal cystic disease examined. TFEB translocation was functionally active and associated with lysosomal biogenesis and perinuclear repositioning, increased TFEB-associated protein expression, and activation of autophagic flux. Compound C1, a TFEB agonist, promoted cyst growth in 3-D cultures of MDCK cells. Nuclear TFEB translocation is an underappreciated signaling pathway for cystogenesis that may represent a new paradigm for cystic kidney disease.


Assuntos
Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Animais , Cães , Humanos , Camundongos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Cistos , Fibroblastos/metabolismo , Células Madin Darby de Rim Canino , Doenças Renais Policísticas/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/genética
2.
J Lipid Res ; 62: 100089, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34087196

RESUMO

Phospholipidosis, the excessive accumulation of phospholipids within lysosomes, is a pathological response observed following exposure to many drugs across multiple therapeutic groups. A clear mechanistic understanding of the causes and implications of this form of drug toxicity has remained elusive. We previously reported the discovery and characterization of a lysosome-specific phospholipase A2 (PLA2G15) and later reported that amiodarone, a known cause of drug-induced phospholipidosis, inhibits this enzyme. Here, we assayed a library of 163 drugs for inhibition of PLA2G15 to determine whether this phospholipase was the cellular target for therapeutics other than amiodarone that cause phospholipidosis. We observed that 144 compounds inhibited PLA2G15 activity. Thirty-six compounds not previously reported to cause phospholipidosis inhibited PLA2G15 with IC50 values less than 1 mM and were confirmed to cause phospholipidosis in an in vitro assay. Within this group, fosinopril was the most potent inhibitor (IC50 0.18 µM). Additional characterization of the inhibition of PLA2G15 by fosinopril was consistent with interference of PLA2G15 binding to liposomes. PLA2G15 inhibition was more accurate in predicting phospholipidosis compared with in silico models based on pKa and ClogP, measures of protonation, and transport-independent distribution in the lysosome, respectively. In summary, PLA2G15 is a primary target for cationic amphiphilic drugs that cause phospholipidosis, and PLA2G15 inhibition by cationic amphiphilic compounds provides a potentially robust screening platform for potential toxicity during drug development.


Assuntos
Inibidores Enzimáticos/farmacologia , Fosfolipases A2/metabolismo , Fosfolipídeos/metabolismo , Animais , Inibidores Enzimáticos/química , Humanos , Lisossomos/enzimologia , Fosfolipases A2/genética
3.
Mol Pharm ; 16(9): 3985-3995, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31356752

RESUMO

Folate-based small molecule drug conjugates (SMDCs) are currently under development and have shown promising preclinical and clinical results against various cancers and polycystic kidney disease. Two requisites for response to a folate-based SMDC are (i) folate receptor alpha (FRα) protein is expressed in the diseased tissues, and (ii) FRα in those tissues is accessible and functionally competent to bind systemically administered SMDCs. Here we report on the development of a small molecule reporter conjugate (SMRC), called EC2220, which is composed of a folate ligand for FRα binding, a multilysine containing linker that can cross-link to FRα in the presence of formaldehyde fixation, and a small hapten (fluorescein) used for immunohistochemical detection. Data show that EC2220 produces a far greater IHC signal in FRα-positive tissues over that produced with EC17, a folate-fluorescein SMRC that is released from the formaldehyde-denatured FRα protein. Furthermore, the extent of the EC2220 IHC signal was proportional to the level of FRα expression. This EC2220-based assay was qualified both in vitro and in vivo using normal tissue, cancer tissue, and polycystic kidneys. Overall, EC2220 is a sensitive and effective reagent for evaluating functional and accessible receptor expression in vitro and in vivo.


Assuntos
Receptor 1 de Folato/metabolismo , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Doenças Renais Policísticas/tratamento farmacológico , Células A549 , Animais , Doxiciclina/farmacologia , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/metabolismo , Receptor 1 de Folato/análise , Ácido Fólico/análogos & derivados , Ácido Fólico/química , Ácido Fólico/metabolismo , Células HeLa , Humanos , Lisina/análogos & derivados , Lisina/química , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos Transgênicos , Neoplasias/metabolismo , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Doenças Renais Policísticas/induzido quimicamente , Doenças Renais Policísticas/metabolismo , Proteína Quinase C/genética , Distribuição Tecidual , Compostos de Tritil/química , Compostos de Tritil/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Am J Physiol Renal Physiol ; 315(2): F395-F405, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717938

RESUMO

Autosomal-dominant polycystic kidney disease (ADPKD) is a very common genetic disease leading to renal failure. Numerous aberrantly regulated signaling pathways have been identified as promising molecular drug targets for ADPKD therapy. In rodent models, many small-molecule drugs against such targets have proven effective in reducing renal cyst growth. For example, mammalian target of rapamycin (mTOR) inhibition with rapamycin greatly ameliorates renal cystic disease in several rodent models. However, clinical trials with mTOR inhibitors were disappointing largely due to the intolerable extrarenal side effects during long-term treatment with these drugs. Most other potential drug targets in ADPKD are also widely expressed in extrarenal tissues, which makes it likely that untargeted therapies with small-molecule inhibitors against such targets will lead to systemic adverse effects during the necessary long-term treatment of years and decades in ADPKD patients. To overcome this problem, we previously demonstrated that folate-conjugated rapamycin (FC-rapa) targets polycystic kidneys due to the high expression of the folate receptor (FRα) and that treatment of a nonortholgous PKD mouse model leads to inhibition of renal cyst growth. Here we show, in a head-to-head comparison with unconjugated rapamycin, that FCrapa inhibits renal cyst growth, mTOR activation, cell cycling, and fibrosis in an orthologous Pkd1 mouse model. Both unconjugated rapamycin and FC-rapa are similarly effective on polycystic kidneys in this model. However, FC-rapa lacks the extrarenal effects of unconjugated rapamycin, in particular immunosuppressive effects. We conclude that folate-conjugation is a promising avenue for increasing the tissue specificity of small-molecule compounds to facilitate very long-term treatment in ADPKD.


Assuntos
Ácido Fólico/farmacologia , Rim/efeitos dos fármacos , Rim Policístico Autossômico Dominante/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Células A549 , Animais , Modelos Animais de Doenças , Composição de Medicamentos , Receptor 1 de Folato/metabolismo , Ácido Fólico/análogos & derivados , Ácido Fólico/metabolismo , Humanos , Integrases/genética , Rim/enzimologia , Camundongos Knockout , Rim Policístico Autossômico Dominante/enzimologia , Rim Policístico Autossômico Dominante/genética , Inibidores de Proteínas Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/análogos & derivados , Sirolimo/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Canais de Cátion TRPP/deficiência , Canais de Cátion TRPP/genética , Distribuição Tecidual
5.
Proc Natl Acad Sci U S A ; 108(19): 7985-90, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21518865

RESUMO

Mutations in polycystin-1 (PC1) lead to autosomal-dominant polycystic kidney disease (ADPKD), a leading cause of renal failure for which no treatment is available. PC1 is an integral membrane protein, which has been implicated in the regulation of multiple signaling pathways including the JAK/STAT pathway. Here we show that membrane-anchored PC1 activates STAT3 in a JAK2-dependent manner, leading to tyrosine phosphorylation and transcriptional activity. The C-terminal cytoplasmic tail of PC1 can undergo proteolytic cleavage and nuclear translocation. Tail-cleavage abolishes the ability of PC1 to directly activate STAT3 but the cleaved PC1 tail now coactivates STAT3 in a mechanism requiring STAT phosphorylation by cytokines or growth factors. This leads to an exaggerated cytokine response. Hence, PC1 can regulate STAT activity by a dual mechanism. In ADPKD kidneys PC1 tail fragments are overexpressed, including a unique ∼15-kDa fragment (P15). STAT3 is strongly activated in cyst-lining epithelial cells in human ADPKD, and orthologous and nonorthologous polycystic mouse models. STAT3 is also activated in developing, postnatal kidneys but inactivated in adult kidneys. These results indicate that STAT3 signaling is regulated by PC1 and is a driving factor for renal epithelial proliferation during normal renal development and during cyst growth.


Assuntos
Fatores de Transcrição STAT/metabolismo , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Animais , Morte Celular , Linhagem Celular , Proliferação de Células , Modelos Animais de Doenças , Cães , Humanos , Interferon gama/metabolismo , Interferon gama/farmacologia , Rim/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Mutação , Fosforilação , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Transcrição STAT/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Canais de Cátion TRPP/química , Transfecção
6.
Proc Natl Acad Sci U S A ; 108(44): 18067-72, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22025716

RESUMO

Autosomal-dominant (AD) polycystic kidney disease (PKD) is a leading cause of renal failure in the United States, and currently lacks available treatment options to slow disease progression. Mutations in the gene coding for polycystin-1 (PC1) underlie the majority of cases but the function of PC1 has remained poorly understood. We have previously shown that PC1 regulates the transcriptional activity of signal transducer and activator of transcription-6 (STAT6). Here we show that STAT6 is aberrantly activated in cyst-lining cells in PKD mouse models. Activation of the STAT6 pathway leads to a positive feedback loop involving auto/paracrine signaling by IL13 and the IL4/13 receptor. The presence of IL13 in cyst fluid and the overexpression of IL4/13 receptor chains suggests a mechanism of sustained STAT6 activation in cysts. Genetic inactivation of STAT6 in a PKD mouse model leads to significant inhibition of proliferation and cyst growth and preservation of renal function. We show that the active metabolite of leflunomide, a drug approved for treatment of arthritis, inhibits STAT6 in renal epithelial cells. Treatment of PKD mice with this drug leads to amelioration of the renal cystic disease similar to genetic STAT6 inactivation. These results suggest STAT6 as a promising drug target for treatment of ADPKD.


Assuntos
Doenças Renais Policísticas/patologia , Fator de Transcrição STAT6/antagonistas & inibidores , Animais , Linhagem Celular , Crotonatos/uso terapêutico , Modelos Animais de Doenças , Cães , Hidroxibutiratos , Camundongos , Nitrilas , Doenças Renais Policísticas/tratamento farmacológico , Toluidinas/uso terapêutico
7.
J Am Soc Nephrol ; 23(10): 1674-81, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22859856

RESUMO

Activation of the mammalian target of rapamycin (mTOR) signaling pathway is aberrant in autosomal-dominant polycystic kidney disease (ADPKD). The mTOR inhibitors, such as rapamycin, ameliorate PKD in rodent models, but clinical trials have not shown benefit, possibly as a result of low tissue concentrations of rapamycin at clinically tolerable doses. To overcome this limitation, we synthesized a folate-conjugated form of rapamycin (FC-rapa) that is taken up by folate receptor-mediated endocytosis and cleaved intracellularly to reconstitute the active drug. We found that renal cyst-lining cells highly express the folate receptor in ADPKD and mouse models. In vitro, FC-rapa inhibited mTOR activity in a dose- and folate receptor-dependent manner. Treatment of a PKD mouse model with FC-rapa inhibited mTOR in the target tissue, strongly attenuated proliferation and growth of renal cysts and preserved renal function. Furthermore, FC-rapa inhibited mTOR activity in the kidney but not in other organs. In summary, these results suggest that targeting the kidney using FC-rapa may overcome the significant side effects and lack of renal efficacy observed in clinical trials with mTOR inhibitors in ADPKD.


Assuntos
Rim Policístico Autossômico Dominante/tratamento farmacológico , Sirolimo/uso terapêutico , Animais , Linhagem Celular , Modelos Animais de Doenças , Endocitose , Receptores de Folato com Âncoras de GPI/metabolismo , Ácido Fólico/análogos & derivados , Ácido Fólico/uso terapêutico , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Camundongos , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Transdução de Sinais/efeitos dos fármacos , Sirolimo/análogos & derivados , Serina-Treonina Quinases TOR/metabolismo
8.
Nephrology (Carlton) ; 17(8): 739-47, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22725947

RESUMO

AIM: Polycystic kidney disease (PKD) in humans involves kidney cyst expansion beginning in utero. Recessive PKD can result in end-stage renal disease (ESRD) within the first decade, whereas autosomal dominant PKD (ADPKD), caused by mutations in the PKD1 or PKD2 gene, typically leads to ESRD by the fifth decade of life. Inhibition of mTOR signalling was recently found to halt cyst formation in adult ADPKD mice. In contrast, no studies have investigated potential treatments to prevent cyst formation in utero in recessive PKD. Given that homozygous Pkd1 mutant mice exhibit cyst formation in utero, we decided to investigate whether mTOR inhibition in utero ameliorates kidney cyst formation in foetal Pkd1 homozygous mutant mice. METHODS: Pregnant Pkd1(+/-) female mice (mated with Pkd1(+/-) male mice) were treated with rapamycin from E14.5 to E17.5. Foetal kidneys were dissected, genotyped and evaluated by cyst size as well as expression of the developmental marker, Pax2. RESULTS: Numerous cysts were present in Pkd1(-/-) kidneys, which were twice the weight of wild-type kidneys. Cyst size was reduced by a third in rapamycin-treated Pkd1(-/-) kidney sections and kidney mass was reduced to near wild-type levels. However, total cyst number was not reduced compared with control embryos. Pax2 expression and kidney development were unaltered in rapamycin-treated mice but some lethality was observed in Pkd1(-/-) null embryos. CONCLUSION: Rapamycin treatment reduces cyst formation in Pkd1(-/-) mutant mice; therefore, the prevention of kidney cyst expansion in utero by mTOR inhibition is feasible. However, selective rapamycin-associated lethality limits its usefulness as a treatment in utero.


Assuntos
Embrião de Mamíferos/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim Policístico Autossômico Dominante/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Canais de Cátion TRPP/deficiência , Animais , Embrião de Mamíferos/enzimologia , Embrião de Mamíferos/patologia , Estudos de Viabilidade , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Idade Gestacional , Homozigoto , Rim/embriologia , Rim/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Fator de Transcrição PAX2/metabolismo , Fenótipo , Rim Policístico Autossômico Dominante/embriologia , Rim Policístico Autossômico Dominante/enzimologia , Rim Policístico Autossômico Dominante/genética , Inibidores de Proteínas Quinases/toxicidade , Deleção de Sequência , Transdução de Sinais/efeitos dos fármacos , Sirolimo/toxicidade , Serina-Treonina Quinases TOR/metabolismo , Canais de Cátion TRPP/genética
9.
J Am Soc Nephrol ; 21(3): 489-97, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20075061

RESUMO

Aberrant activation of the mammalian target of rapamycin (mTOR) pathway occurs in polycystic kidney disease (PKD). mTOR inhibitors, such as rapamycin, are highly effective in several rodent models of PKD, but these models result from mutations in genes other than Pkd1 and Pkd2, which are the primary genes responsible for human autosomal dominant PKD. To address this limitation, we tested the efficacy of rapamycin in a mouse model that results from conditional inactivation of Pkd1. Mosaic deletion of Pkd1 resulted in PKD and replicated characteristic features of human PKD including aberrant mTOR activation, epithelial proliferation and apoptosis, and progressive fibrosis. Treatment with rapamycin was highly effective: It reduced cyst growth, preserved renal function, inhibited epithelial cell proliferation, increased apoptosis of cyst-lining cells, and inhibited fibrosis. These data provide in vivo evidence that rapamycin is effective in a human-orthologous mouse model of PKD.


Assuntos
Doenças Renais Policísticas/genética , Doenças Renais Policísticas/fisiopatologia , Sirolimo/farmacologia , Canais de Cátion TRPP/genética , Animais , Apoptose/fisiologia , Nitrogênio da Ureia Sanguínea , Divisão Celular/fisiologia , Modelos Animais de Doenças , Fibrose , Expressão Gênica/efeitos dos fármacos , Humanos , Imunossupressores/farmacologia , Proteínas de Filamentos Intermediários/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Túbulos Renais Coletores/patologia , Túbulos Renais Coletores/fisiopatologia , Túbulos Renais Distais/patologia , Túbulos Renais Distais/fisiopatologia , Camundongos , Mosaicismo , Proteínas do Tecido Nervoso/genética , Nestina , Fenótipo , Doenças Renais Policísticas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinases TOR , Canais de Cátion TRPP/metabolismo
10.
Mol Cell Biol ; 26(15): 5809-26, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16847333

RESUMO

Transcription of immediate-early genes--as well as multiple genes affecting muscle function, cytoskeletal integrity, apoptosis control, and wound healing/angiogenesis--is regulated by serum response factor (Srf). Extracellular signals regulate Srf in part via a pathway involving megakaryoblastic leukemia 1 (Mkl1, also known as myocardin-related transcription factor A [Mrtf-a]), which coactivates Srf-responsive genes downstream of Rho GTPases. Here we investigate Mkl1 function using gene targeting and show the protein to be essential for the physiologic preparation of the mammary gland during pregnancy and the maintenance of lactation. Lack of Mkl1 causes premature involution and impairs expression of Srf-dependent genes in the mammary myoepithelial cells, which control milk ejection following oxytocin-induced contraction. Despite the importance of Srf in multiple transcriptional pathways and widespread Mkl1 expression, the spectrum of abnormalities associated with Mkl1 absence appears surprisingly restricted.


Assuntos
Lactação/fisiologia , Glândulas Mamárias Animais/anatomia & histologia , Glândulas Mamárias Animais/fisiologia , Transativadores/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Criança , Insuficiência de Crescimento , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Marcação de Genes , Coração/anatomia & histologia , Coração/embriologia , Humanos , Lactente , Leucemia Megacarioblástica Aguda , Masculino , Glândulas Mamárias Animais/anormalidades , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Leite , Miócitos Cardíacos/patologia , Miócitos Cardíacos/ultraestrutura , Análise de Sequência com Séries de Oligonucleotídeos , Ocitocina/metabolismo , Gravidez , Prolactina/metabolismo , Fator de Transcrição STAT3 , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transativadores/genética
11.
Clin Kidney J ; 11(Suppl 1): i27-i38, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30581563

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is a widespread genetic disease that leads to renal failure in the majority of patients. The very first pharmacological treatment, tolvaptan, received Food and Drug Administration approval in 2018 after previous approval in Europe and other countries. However, tolvaptan is moderately effective and may negatively impact a patient's quality of life due to potentially significant side effects. Additional and improved therapies are still urgently needed, and several clinical trials are underway, which are discussed in the companion paper Müller and Benzing (Management of autosomal-dominant polycystic kidney disease-state-of-the-art) Clin Kidney J 2018; 11: i2-i13. Here, we discuss new therapeutic avenues that are currently being investigated at the preclinical stage. We focus on mammalian target of rapamycin and dual kinase inhibitors, compounds that target inflammation and histone deacetylases, RNA-targeted therapeutic strategies, glucosylceramide synthase inhibitors, compounds that affect the metabolism of renal cysts and dietary restriction. We discuss tissue targeting to renal cysts of small molecules via the folate receptor, and of monoclonal antibodies via the polymeric immunoglobulin receptor. A general problem with potential pharmacological approaches is that the many molecular targets that have been implicated in ADPKD are all widely expressed and carry out important functions in many organs and tissues. Because ADPKD is a slowly progressing, chronic disease, it is likely that any therapy will have to continue over years and decades. Therefore, systemically distributed drugs are likely to lead to potentially prohibitive extra-renal side effects during extended treatment. Tissue targeting to renal cysts of such drugs is one potential way around this problem. The use of dietary, instead of pharmacological, interventions is another.

12.
Oncogene ; 22(25): 3875-87, 2003 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-12813461

RESUMO

The Wnt/beta-catenin signaling pathway is critical for normal mammalian development, the specification of epidermal cells and neoplastic transformation of intestinal epithelium. However, precise molecular information regarding cell-specific responses to beta-catenin signaling has been limited. This question was addressed using a mouse model in which exon 3 of the beta-catenin gene was deleted in several cell types with loxP-mediated recombination utilizing a Cre transgene under control of the mouse mammary tumor virus-long terminal repeat (MMTV-LTR). The stabilization of beta-catenin in prostate epithelium resulted in hyperplasias and extensive transdifferentiation into epidermal-like structures, which expressed keratins 1 and 6, filaggrin, loricrin and involucrin. The cell-specific loss of NKCC1 protein and reduced nuclear Stat5a is further suggestive of a loss of prostate epithelial characteristics. In addition to the prostate, hyperplasias and squamous metaplasias were detected in epithelia of the epididymis, vas deferens, coagulating gland, preputial gland and salivary gland. However, and in contrast to a recent study, no lesions reminiscent of high-grade prostate intraepithelial neoplasia were detected. Since beta-catenin was activated in several cell types and impinged upon the viability of these mice, it was not possible to evaluate the cumulative effect over more than 3 months. To assess long-term consequences of beta-catenin activation, mutant and control prostate tissues were transplanted into the mammary fat pads of wild-type males. Notably, squamous metaplasias, intra-acinous hyperplasia and possible neoplastic transformation were observed after a total of 18 weeks of beta-catenin stimulation. This suggests that the transdifferentiation into squamous metaplasias is an early response of endoderm-derived cells to beta-catenin, and that the development of intra-acinous hyperplasias or neoplastic foci is a later event.


Assuntos
Proteínas do Citoesqueleto/fisiologia , Próstata/patologia , Transativadores/fisiologia , Tecido Adiposo , Animais , Antígenos de Diferenciação/análise , Biomarcadores , Diferenciação Celular , Linhagem da Célula , Transformação Celular Neoplásica/genética , Proteínas do Citoesqueleto/genética , Endoderma/citologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/transplante , Éxons/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hiperplasia , Masculino , Glândulas Mamárias Animais , Vírus do Tumor Mamário do Camundongo/genética , Metaplasia , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Próstata/transplante , Proteínas Recombinantes de Fusão/fisiologia , Deleção de Sequência , Sequências Repetidas Terminais , Transativadores/genética , Transgenes , Transplante Heterotópico , beta Catenina
13.
Mol Endocrinol ; 16(6): 1309-21, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12040017

RESUMO

Despite the fact that physiological evidence points to the existence of a functional Na-K-Cl cotransporter in the mammary gland, the molecular identity of this transport process remains unknown. We now show that the Na-K-Cl cotransporter isoform, NKCC1, is expressed in mammary tissue. Developmental profiling revealed that the level of NKCC1 protein was significantly influenced by the stage of mammary gland development, and immunolocalization studies demonstrated that NKCC1 was present on the basolateral membrane of mammary epithelial cells. To examine whether functional NKCC1 is required for mammary epithelial cell development, we used NKCC1 -/- mice. We demonstrate that NKCC1 -/- mammary epithelium exhibited a significant delay in ductal outgrowth and an increase in branching morphogenesis during virgin development. These effects were autonomous to the epithelium as assessed by mammary gland transplantation. Although the absence of NKCC1 had no apparent effect on gross mammary epithelial cell morphology during lactation, pups born to NKCC1 -/- mice failed to thrive. Finally, analysis of NKCC1 protein in mouse models that exhibit defects in mammary gland development demonstrate that high levels of NKCC1 protein are indicative of ductal epithelial cells, and the presence of NKCC1 protein is characteristic of mammary epithelial cell identity.


Assuntos
Mama/crescimento & desenvolvimento , Mama/metabolismo , Células Epiteliais/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Animais , Diferenciação Celular , Divisão Celular , Células Epiteliais/citologia , Feminino , Camundongos , Camundongos Nus , Morfogênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Simportadores de Cloreto de Sódio-Potássio/genética , Membro 2 da Família 12 de Carreador de Soluto , Transplante de Tecidos
14.
Mol Endocrinol ; 16(3): 563-70, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11875116

RESUMO

The PRL receptor (PrlR) and the signal transducer and activator of transcription 5a (Stat5a) are essential for the proliferation and differentiation of mammary epithelium during pregnancy. Based on tissue culture cell experiments, Jak2 is the tyrosine kinase responsible for the phosphorylation of both the PrlR and Stat5. We have now used a genetic approach to test the role of Jak2 in the mammary gland, a PrlR-responsive tissue. Because Jak2-null embryos die at E12.5, we transplanted Jak2-null mammary anlagen into cleared fat pads of wild-type mice and investigated epithelial development during pregnancy. In the absence of Jak2, no secretory alveoli were present at parturition, and epithelial cell proliferation was reduced by 95% after an acute hormone treatment. Furthermore, the Na-K-Cl cotransporter, a ductal marker, was maintained in Jak2-null epithelium and the sodium-phosphate cotransporter type IIb, a secretory cell marker, was absent. Nuclear Stat5a was only observed in a few epithelial cells in Jak2-null glands at pregnancy and parturition compared with most epithelial cells in wild-type glands. Taken together, our results demonstrate that Jak2 is a critical tyrosine kinase that conveys intracellular signals necessary for proliferation and differentiation of mammary epithelium during pregnancy.


Assuntos
Glândulas Mamárias Animais/fisiologia , Proteínas do Leite , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas , Animais , Transporte Biológico , Diferenciação Celular , Divisão Celular , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/citologia , Epitélio/química , Epitélio/fisiologia , Feminino , Janus Quinase 2 , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Gravidez , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Fator de Transcrição STAT5 , Simportadores de Cloreto de Sódio-Potássio/análise , Transativadores/análise , Transativadores/metabolismo
15.
J Histochem Cytochem ; 51(5): 555-65, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12704203

RESUMO

Through the use of transgenic and gene knockout mice, several studies have identified specific genes required for the functional development of mammary epithelium. Although histological and milk protein gene analyses can provide useful information regarding functional differentiation, they are limited in their ability to precisely define the molecular lesions. For example, mice that carry a mutation in one of the subunits of the IkappaB kinase, IKKalpha, cannot lactate despite the presence of histologically normal alveolar compartment and the expression of milk protein genes. To further define and understand such lesions on a molecular level, we sought evidence for proteins that are differentially expressed during mammary gland development with a view to generating a tissue proteotype. Using database screens and immunohistochemical analyses, we have identified three proteins that exhibit distinct profiles. Here, using mouse models as test biological systems, we demonstrate the development and application of mammary tissue proteotyping and its use in the elucidation of specific developmental lesions. We propose that the technique of proteotyping will have wide applications in the analyses of defects in other mouse models.


Assuntos
Aquaporinas/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteínas de Membrana , Proteínas do Leite , Proteínas Proto-Oncogênicas , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Simportadores/metabolismo , Animais , Aquaporina 5 , Biomarcadores/análise , Proteínas de Ligação a DNA/genética , Feminino , Quinase I-kappa B , Imuno-Histoquímica , Subunidades beta de Inibinas/genética , Janus Quinase 2 , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , Camundongos Nus , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/fisiologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Receptores da Prolactina/genética , Fator de Transcrição STAT3 , Fator de Transcrição STAT5 , Transdução de Sinais , Proteínas Cotransportadoras de Sódio-Fosfato , Membro 2 da Família 12 de Carreador de Soluto , Transativadores/genética
16.
Proc Natl Acad Sci U S A ; 103(14): 5466-71, 2006 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-16567633

RESUMO

Autosomal-dominant polycystic kidney disease (ADPKD) is a common genetic disorder that frequently leads to renal failure. Mutations in polycystin-1 (PC1) underlie most cases of ADPKD, but the function of PC1 has remained poorly understood. No preventive treatment for this disease is available. Here, we show that the cytoplasmic tail of PC1 interacts with tuberin, and the mTOR pathway is inappropriately activated in cyst-lining epithelial cells in human ADPKD patients and mouse models. Rapamycin, an inhibitor of mTOR, is highly effective in reducing renal cystogenesis in two independent mouse models of PKD. Treatment of human ADPKD transplant-recipient patients with rapamycin results in a significant reduction in native polycystic kidney size. These results indicate that PC1 has an important function in the regulation of the mTOR pathway and that this pathway provides a target for medical therapy of ADPKD.


Assuntos
Rim/patologia , Doenças Renais Policísticas/prevenção & controle , Proteínas Quinases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Humanos , Camundongos , Doenças Renais Policísticas/patologia , Proteínas Quinases/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Canais de Cátion TRPP
17.
Development ; 131(13): 3047-55, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15163627

RESUMO

Mammary gland development is a complex process that is dependent on interactions between the developing mammary epithelium and the surrounding stromal tissues. We show that mice lacking the triglyceride synthesis enzyme acyl CoA:diacylglycerol transferase 1 (DGAT1) have impaired mammary gland development, characterized by decreased epithelial proliferation and alveolar development, and reduced expression of markers of functional differentiation. Transplantation studies demonstrate that the impaired development results from a deficiency of DGAT1 in both the stromal and epithelial tissues. Our findings are the first to link defects in stromal lipid metabolism to impaired mammary gland development.


Assuntos
Aciltransferases/biossíntese , Aciltransferases/fisiologia , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/embriologia , Células Estromais/metabolismo , Animais , Apoptose , Diferenciação Celular , Diacilglicerol O-Aciltransferase , Processamento de Imagem Assistida por Computador , Immunoblotting , Imuno-Histoquímica , Lactação , Metabolismo dos Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Testes de Precipitina , RNA/metabolismo , Fatores de Tempo , Transplante de Tecidos
18.
Development ; 130(21): 5257-68, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12954715

RESUMO

The ERBB family of type 1 receptor tyrosine kinases and their ligands have crucial functions during mammopoiesis, but the signaling networks that ultimately regulate ERBB activity in the breast have remained elusive. Here, we show that mice with Cre-lox mediated deletions of both Erbb4 alleles within the developing mammary gland (Erbb4(Flox/Flox)Wap-Cre) fail to accumulate lobuloalveoli or successfully engage lactation at parturition owing, in part, to impaired epithelial proliferation. Analysis of the mammary differentiation factor STAT5 by immunohistochemistry and western blot revealed a complete ablation of STAT5 activation in Erbb4(Flox/Flox)Wap-Cre mammary epithelium at parturition. Consistent with disrupted STAT5 function, Erbb4(Flox/Flox)Wap-Cre mammary glands at parturition failed to express the mammary epithelial differentiation marker NPT2B. Defects in epithelial functional differentiation at parturition were accompanied by a profound reduction in expression of the STAT5-regulated milk genes casein beta and whey acidic protein. We propose that ERBB4 functions as an essential mediator of STAT5 signaling, and that loss of STAT5 activity contributes to the impaired functional differentiation of mammary glands observed in mice containing conditional Erbb4 deletions.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Receptores ErbB/metabolismo , Lactação/fisiologia , Glândulas Mamárias Animais/fisiologia , Transativadores/metabolismo , Animais , Caseínas/genética , Clonagem Molecular , Proteínas de Ligação a DNA/genética , Ativação Enzimática , Epitélio/anatomia & histologia , Epitélio/fisiologia , Receptores ErbB/genética , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos , Proteínas do Leite/genética , Gravidez , Receptor ErbB-4 , Fator de Transcrição STAT5 , Transdução de Sinais/fisiologia , Transativadores/genética
19.
Proc Natl Acad Sci U S A ; 99(1): 219-24, 2002 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-11773619

RESUMO

Mammary anlagen are formed in the embryo as a derivative of the epidermis, a process that is controlled by Lef-1 and therefore possibly by beta-catenin. To investigate the role of beta-catenin signaling in mammary alveolar epithelium, we have stabilized endogenous beta-catenin in differentiating alveolar epithelium through the deletion of exon 3 (amino acids 5-80) of the beta-catenin gene. This task was accomplished in mice carrying a floxed beta-catenin gene and a Cre transgene under control of the mammary-specific whey acidic protein (WAP) gene promoter or the mouse mammary tumor virus-long terminal repeat (MMTV-LTR). Stabilized beta-catenin was obtained during the first pregnancy, and its presence resulted in the dedifferentiation of alveolar epithelium followed by a transdifferentiation into epidermal and pilar structures. Extensive squamous metaplasia, but no adenocarcinomas, developed upon beta-catenin activation during pregnancy and persisted throughout involution. These data demonstrate that the activation of beta-catenin signaling induces a program that results in loss of mammary epithelial cell differentiation and induction of epidermal structures.


Assuntos
Adenocarcinoma/metabolismo , Carcinoma de Células Escamosas/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Células Epidérmicas , Metaplasia/metabolismo , Transdução de Sinais , Transativadores , Animais , Western Blotting , Diferenciação Celular , Citoplasma/metabolismo , Proteínas do Citoesqueleto/biossíntese , Epiderme/embriologia , Éxons , Deleção de Genes , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Proteínas do Leite/genética , Modelos Biológicos , Regiões Promotoras Genéticas , Fatores de Tempo , beta Catenina
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa