Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Blood ; 131(3): 323-327, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29167177

RESUMO

The stromal signals that promote B lymphopoiesis remain poorly understood. Hedgehog (Hh) signaling promotes B lymphopoiesis in a non-cell-autonomous fashion in vitro, and depletion of the Hh effector Smoothened (Smo) from stromal cells is associated with the loss of osteoblastoid markers. These observations suggested that Hh signaling in the osteoblastoid lineage promotes B lymphopoiesis in vivo. To test this, we employed a mouse model for conditional ablation of Smo in the osteoblastoid lineage. Depletion of Smo from osteoblastoid cells is associated with profound and selective reductions in the number and proportion of bone marrow B-lymphoid progenitors. Upon partial bone marrow ablation, mutant animals exhibit delayed repopulation of the B-lymphoid compartment after the early lymphoid progenitor stage. Primary osteoblasts from mutant mice are defective in supporting B lymphopoiesis in vitro, whereas hematopoietic progenitors from mutant mice exhibit normal differentiation. We conclude that efficient B lymphopoiesis in vivo is dependent on the maintenance of Hh signaling in the osteoblastoid lineage.


Assuntos
Linfócitos B/metabolismo , Linhagem da Célula , Linfopoese , Osteoblastos/citologia , Transdução de Sinais , Receptor Smoothened/metabolismo , Animais , Medula Óssea/metabolismo , Proteínas Hedgehog/metabolismo , Camundongos Transgênicos
2.
Proc Natl Acad Sci U S A ; 114(1): 125-130, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-27999180

RESUMO

Kabuki syndrome is a Mendelian intellectual disability syndrome caused by mutations in either of two genes (KMT2D and KDM6A) involved in chromatin accessibility. We previously showed that an agent that promotes chromatin opening, the histone deacetylase inhibitor (HDACi) AR-42, ameliorates the deficiency of adult neurogenesis in the granule cell layer of the dentate gyrus and rescues hippocampal memory defects in a mouse model of Kabuki syndrome (Kmt2d+/ßGeo). Unlike a drug, a dietary intervention could be quickly transitioned to the clinic. Therefore, we have explored whether treatment with a ketogenic diet could lead to a similar rescue through increased amounts of beta-hydroxybutyrate, an endogenous HDACi. Here, we report that a ketogenic diet in Kmt2d+/ßGeo mice modulates H3ac and H3K4me3 in the granule cell layer, with concomitant rescue of both the neurogenesis defect and hippocampal memory abnormalities seen in Kmt2d+/ßGeo mice; similar effects on neurogenesis were observed on exogenous administration of beta-hydroxybutyrate. These data suggest that dietary modulation of epigenetic modifications through elevation of beta-hydroxybutyrate may provide a feasible strategy to treat the intellectual disability seen in Kabuki syndrome and related disorders.


Assuntos
Anormalidades Múltiplas/dietoterapia , Dieta Cetogênica/métodos , Face/anormalidades , Doenças Hematológicas/dietoterapia , Hipocampo/metabolismo , Histonas/biossíntese , Deficiência Intelectual/dietoterapia , Neurogênese/fisiologia , Doenças Vestibulares/dietoterapia , Ácido 3-Hidroxibutírico/metabolismo , Anormalidades Múltiplas/genética , Animais , Modelos Animais de Doenças , Doenças Hematológicas/genética , Hipocampo/citologia , Histona Desmetilases/genética , Histona-Lisina N-Metiltransferase/genética , Deficiência Intelectual/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína de Leucina Linfoide-Mieloide/genética , Neurogênese/genética , Doenças Vestibulares/genética
3.
Nature ; 503(7474): 126-30, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24107997

RESUMO

In systemic sclerosis (SSc), a common and aetiologically mysterious form of scleroderma (defined as pathological fibrosis of the skin), previously healthy adults acquire fibrosis of the skin and viscera in association with autoantibodies. Familial recurrence is extremely rare and causal genes have not been identified. Although the onset of fibrosis in SSc typically correlates with the production of autoantibodies, whether they contribute to disease pathogenesis or simply serve as a marker of disease remains controversial and the mechanism for their induction is largely unknown. The study of SSc is hindered by a lack of animal models that recapitulate the aetiology of this complex disease. To gain a foothold in the pathogenesis of pathological skin fibrosis, we studied stiff skin syndrome (SSS), a rare but tractable Mendelian disorder leading to childhood onset of diffuse skin fibrosis with autosomal dominant inheritance and complete penetrance. We showed previously that SSS is caused by heterozygous missense mutations in the gene (FBN1) encoding fibrillin-1, the main constituent of extracellular microfibrils. SSS mutations all localize to the only domain in fibrillin-1 that harbours an Arg-Gly-Asp (RGD) motif needed to mediate cell-matrix interactions by binding to cell-surface integrins. Here we show that mouse lines harbouring analogous amino acid substitutions in fibrillin-1 recapitulate aggressive skin fibrosis that is prevented by integrin-modulating therapies and reversed by antagonism of the pro-fibrotic cytokine transforming growth factor ß (TGF-ß). Mutant mice show skin infiltration of pro-inflammatory immune cells including plasmacytoid dendritic cells, T helper cells and plasma cells, and also autoantibody production; these findings are normalized by integrin-modulating therapies or TGF-ß antagonism. These results show that alterations in cell-matrix interactions are sufficient to initiate and sustain inflammatory and pro-fibrotic programmes and highlight new therapeutic strategies.


Assuntos
Autoimunidade/efeitos dos fármacos , Contratura/tratamento farmacológico , Contratura/patologia , Integrinas/efeitos dos fármacos , Integrinas/metabolismo , Escleroderma Sistêmico/tratamento farmacológico , Escleroderma Sistêmico/patologia , Dermatopatias Genéticas/tratamento farmacológico , Dermatopatias Genéticas/patologia , Motivos de Aminoácidos/genética , Substituição de Aminoácidos/genética , Animais , Anticorpos Antinucleares/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Neutralizantes/uso terapêutico , Autoimunidade/imunologia , Contratura/imunologia , Contratura/prevenção & controle , Células Dendríticas/efeitos dos fármacos , Feminino , Fibrilina-1 , Fibrilinas , Fibrose/tratamento farmacológico , Fibrose/patologia , Fibrose/prevenção & controle , Masculino , Camundongos , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Mutação de Sentido Incorreto/genética , Plasmócitos/efeitos dos fármacos , Escleroderma Sistêmico/imunologia , Escleroderma Sistêmico/prevenção & controle , Dermatopatias Genéticas/imunologia , Dermatopatias Genéticas/prevenção & controle , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/imunologia
4.
Hepatology ; 63(6): 1828-41, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26679751

RESUMO

UNLABELLED: Wilson disease (WD) is a hepatoneurological disorder caused by mutations in the copper-transporter, ATP7B. Copper accumulation in the liver is a hallmark of WD. Current therapy is based on copper chelation, which decreases the manifestations of liver disease, but often worsens neurological symptoms. We demonstrate that in Atp7b(-/-) mice, an animal model of WD, liver function can be significantly improved without copper chelation. Analysis of transcriptional and metabolic changes in samples from WD patients and Atp7b(-/-) mice identified dysregulation of nuclear receptors (NRs), especially the liver X receptor (LXR)/retinoid X receptor heterodimer, as an important event in WD pathogenesis. Treating Atp7b(-/-) mice with the LXR agonist, T0901317, ameliorated disease manifestations despite significant copper overload. Genetic markers of liver fibrosis and inflammatory cytokines were significantly decreased, lipid profiles normalized, and liver function and histology were improved. CONCLUSIONS: The results demonstrate the major role of an altered NR function in the pathogenesis of WD and suggest that modulation of NR activity should be explored as a supplementary approach to improving liver function in WD. (Hepatology 2016;63:1828-1841).


Assuntos
Degeneração Hepatolenticular/tratamento farmacológico , Hidrocarbonetos Fluorados/uso terapêutico , Metabolismo dos Lipídeos/efeitos dos fármacos , Receptores X do Fígado/agonistas , Fígado/efeitos dos fármacos , Sulfonamidas/uso terapêutico , Adenosina Trifosfatases/genética , Animais , Proteínas de Transporte de Cátions/genética , Cobre/metabolismo , ATPases Transportadoras de Cobre , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Degeneração Hepatolenticular/genética , Humanos , Hidrocarbonetos Fluorados/farmacologia , Fígado/metabolismo , Testes de Função Hepática , Receptores X do Fígado/metabolismo , Camundongos Knockout , Receptores X de Retinoides/metabolismo , Sulfonamidas/farmacologia
5.
J Infect Dis ; 214(1): 122-9, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26908749

RESUMO

BACKGROUND: Chronic inflammation and composition of the colon microbiota have been associated with colorectal cancer in humans. The human commensal enterotoxigenic Bacteroides fragilis (ETBF) is linked to both inflammatory bowel disease and colorectal cancer and, in our murine model, causes interleukin 17A (IL-17A)-dependent colon tumors. In these studies, we hypothesized that persistent colonization by ETBF is required for tumorigenesis. METHODS: We established a method for clearing ETBF in mice, using the antibiotic cefoxitin. Multiple intestinal neoplasia mice were colonized with ETBF for the experiment duration or were cleared of infection after 5 or 14 days. Gross tumors and/or microadenomas were then evaluated. In parallel, IL-17A expression was evaluated in wild-type littermates. RESULTS: Cefoxitin treatment resulted in complete and durable clearance of ETBF colonization. We observed a stepwise increase in median colon tumor numbers as the duration of ETBF colonization increased before cefoxitin treatment. ETBF eradication also significantly decreased mucosal IL-17A expression. CONCLUSIONS: The timing of ETBF clearance profoundly influences colon adenoma formation, defining a period during which the colon is susceptible to IL-17A-dependent tumorigenesis in this murine model. This model system can be used to study the microbiota-dependent and molecular mechanisms contributing to IL-17A-dependent colon tumor initiation.


Assuntos
Carcinogênese/efeitos dos fármacos , Cefoxitina/efeitos adversos , Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias do Colo/complicações , Neoplasias do Colo/tratamento farmacológico , Enterotoxinas/efeitos adversos , Enterotoxinas/uso terapêutico , Animais , Bacteroides fragilis/química , Colo/microbiologia , Neoplasias do Colo/microbiologia , Humanos , Camundongos
6.
J Proteome Res ; 15(12): 4176-4187, 2016 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-27696867

RESUMO

Because colorectal cancer (CRC) remains a leading cause of cancer mortality worldwide, more accessible screening tests are urgently needed to identify early stage lesions. We hypothesized that highly sensitive, metabolic profile analysis of stool samples will identify metabolites associated with early stage lesions and could serve as a noninvasive screening test. We therefore applied traveling wave ion mobility mass spectrometry (TWIMMS) coupled with ultraperformance liquid chromatography (UPLC) to investigate metabolic aberrations in stool samples in a transgenic model of premalignant polyposis aberrantly expressing the gene encoding the high mobility group A (Hmga1) chromatin remodeling protein. Here, we report for the first time that the fecal metabolome of Hmga1 mice is distinct from that of control mice and includes metabolites previously identified in human CRC. Significant alterations were observed in fatty acid metabolites and metabolites associated with bile acids (hypoxanthine xanthine, taurine) in Hmga1 mice compared to controls. Surprisingly, a marked increase in the levels of distinctive short, arginine-enriched, tetra-peptide fragments was observed in the transgenic mice. Together these findings suggest that specific metabolites are associated with Hmga1-induced polyposis and abnormal proliferation in intestinal epithelium. Although further studies are needed, these data provide a compelling rationale to develop fecal metabolomic analysis as a noninvasive screening tool to detect early precursor lesions to CRC in humans.


Assuntos
Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/metabolismo , Detecção Precoce de Câncer/métodos , Fezes/química , Proteínas HMGA/genética , Metaboloma , Polipose Adenomatosa do Colo/genética , Animais , Ácidos e Sais Biliares/metabolismo , Cromatografia Líquida de Alta Pressão , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo
7.
Kidney Int ; 90(1): 90-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27165822

RESUMO

Abnormal proliferation of cyst-lining epithelium and increased intracystic fluid secretion via the cystic fibrosis transmembrane conductance regulator (CFTR) are thought to contribute to cyst growth in autosomal dominant polycystic kidney disease (ADPKD). Histone deacetylase 6 (HDAC6) expression and activity are increased in certain cancers, neurodegenerative diseases, and in Pkd1-mutant renal epithelial cells. Inhibition of HDAC6 activity with specific inhibitors slows cancer growth. Here we studied the effect of tubacin, a specific HDAC6 inhibitor, on cyst growth in polycystic kidney disease. Treatment with tubacin prevented cyst formation in MDCK cells, an in vitro model of cystogenesis. Cyclic AMP stimulates cell proliferation and activates intracystic CFTR-mediated chloride secretion in ADPKD. Treatment with tubacin downregulated cyclic AMP levels, inhibited cell proliferation, and inhibited cyclic AMP-activated CFTR chloride currents in MDCK cells. We also found that tubacin reduced cyst growth by inhibiting proliferation of cyst-lining epithelial cells, downregulated cyclic AMP levels, and improved renal function in a Pkd1-conditional mouse model of ADPKD. Thus, HDAC6 could play a role in cyst formation and could serve as a potential therapeutic target in ADPKD.


Assuntos
Anilidas/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Ácidos Hidroxâmicos/farmacologia , Rim/efeitos dos fármacos , Rim Policístico Autossômico Dominante/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Cloretos/sangue , Cloretos/metabolismo , AMP Cíclico/sangue , Modelos Animais de Doenças , Cães , Regulação para Baixo , Células Epiteliais/metabolismo , Feminino , Desacetilase 6 de Histona , Histona Desacetilases/genética , Humanos , Rim/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética
8.
Gynecol Oncol ; 141(3): 580-587, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27001612

RESUMO

OBJECTIVES: Although uterine cancer is the fourth most common cause for cancer death in women worldwide, the molecular underpinnings of tumor progression remain poorly understood. The High Mobility Group A1 (HMGA1) gene is overexpressed in aggressive cancers and high levels portend adverse outcomes in diverse tumors. We previously reported that Hmga1a transgenic mice develop uterine tumors with complete penetrance. Because HMGA1 drives tumor progression by inducing MatrixMetalloproteinase (MMP) and other genes involved in invasion, we explored the HMGA1-MMP-2 pathway in uterine cancer. METHODS: To investigate MMP-2 in uterine tumors driven by HMGA1, we used a genetic approach with mouse models. Next, we assessed HMGA1 and MMP-2 expression in primary human uterine tumors, including low-grade carcinomas (endometrial endometrioid) and more aggressive tumors (endometrial serous carcinomas, uterine carcinosarcomas/malignant mesodermal mixed tumors). RESULTS: Here, we report for the first time that uterine tumor growth is impaired in Hmga1a transgenic mice crossed on to an Mmp-2 deficient background. In human tumors, we discovered that HMGA1 is highest in aggressive carcinosarcomas and serous carcinomas, with lower levels in the more indolent endometrioid carcinomas. Moreover, HMGA1 and MMP-2 were positively correlated, but only in a subset of carcinosarcomas. HMGA1 also occupies the MMP-2 promoter in human carcinosarcoma cells. CONCLUSIONS: Together, our studies define a novel HMGA1-MMP-2 pathway involved in a subset of human carcinosarcomas and tumor progression in murine models. Our work also suggests that targeting HMGA1 could be effective adjuvant therapy for more aggressive uterine cancers and provides compelling data for further preclinical studies.


Assuntos
Carcinossarcoma/genética , Cistadenocarcinoma Seroso/genética , Proteína HMGA1a/genética , Metaloproteinase 2 da Matriz/genética , Neoplasias Uterinas/genética , Animais , Carcinossarcoma/metabolismo , Imunoprecipitação da Cromatina , Cistadenocarcinoma Seroso/metabolismo , Feminino , Inativação Gênica , Proteína HMGA1a/biossíntese , Humanos , Masculino , Metaloproteinase 2 da Matriz/biossíntese , Camundongos Transgênicos , Regiões Promotoras Genéticas , Regulação para Cima , Neoplasias Uterinas/metabolismo
9.
Proc Natl Acad Sci U S A ; 110(52): 21113-8, 2013 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-24255108

RESUMO

FMS-like tyrosine kinase 3 (FLT3) is mutated in approximately one third of acute myeloid leukemia cases. The most common FLT3 mutations in acute myeloid leukemia are internal tandem duplication (ITD) mutations in the juxtamembrane domain (23%) and point mutations in the tyrosine kinase domain (10%). The mutation substituting the aspartic acid at position 838 (equivalent to the human aspartic acid residue at position 835) with a tyrosine (referred to as FLT3/D835Y hereafter) is the most frequent kinase domain mutation, converting aspartic acid to tyrosine. Although both of these mutations constitutively activate FLT3, patients with an ITD mutation have a significantly poorer prognosis. To elucidate the mechanisms behind this prognostic difference, we have generated a knock-in mouse model with a D838Y point mutation in FLT3 that corresponds to the FLT3/D835Y mutation described in humans. Compared with FLT3/ITD knock-in mice, the FLT3/D835Y knock-in mice survive significantly longer. The majority of these mice develop myeloproliferative neoplasms with a less-aggressive phenotype. In addition, FLT3/D835Y mice have distinct hematopoietic development patterns. Unlike the tremendous depletion of the hematopoietic stem cell compartment we have observed in FLT3/ITD mice, FLT3/D835Y mutant mice are not depleted in hematopoietic stem cells. Further comparisons of these FLT3/D835Y knock-in mice with FLT3/ITD mice should provide an ideal platform for dissecting the molecular mechanisms that underlie the prognostic differences between the two different types of FLT3 mutations.


Assuntos
Técnicas de Introdução de Genes/métodos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Modelos Animais , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Imuno-Histoquímica , Camundongos , Mutação de Sentido Incorreto/genética , Prognóstico , Duplicações Segmentares Genômicas/genética
10.
Proc Natl Acad Sci U S A ; 110(36): 14717-22, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-23959886

RESUMO

Nanoparticle gene therapy holds great promise for the treatment of malignant disease in light of the large number of potent, tumor-specific therapeutic payloads potentially available for delivery. To be effective, gene therapy vehicles must be able to deliver their therapeutic payloads to metastatic lesions after systemic administration. Here we describe nanoparticles comprised of a core of high molecular weight linear polyethylenimine (LPEI) complexed with DNA and surrounded by a shell of polyethyleneglycol-modified (PEGylated) low molecular weight LPEI. Compared with a state-of-the-art commercially available in vivo gene delivery formulation, i.v. delivery of the core/PEGylated shell (CPS) nanoparticles provided more than a 16,000-fold increase in the ratio of tumor to nontumor transfection. The vast majority of examined liver and lung metastases derived from a colorectal cancer cell line showed transgene expression after i.v. CPS injection in an animal model of metastasis. Histological examination of tissues from transfected mice revealed that the CPS nanoparticles selectively transfected neoplastic cells rather than stromal cells within primary and metastatic tumors. However, only a small fraction of neoplastic cells (<1%) expressed the transgene, and the extent of delivery varied with the tumor cell line, tumor site, and host mouse strain used. Our results demonstrate that these CPS nanoparticles offer substantial advantages over previously described formulations for in vivo nanoparticle gene therapeutics. At the same time, they illustrate that major increases in the effectiveness of such approaches are needed for utility in patients with metastatic cancer.


Assuntos
Terapia Genética/métodos , Proteínas Luminescentes/genética , Nanopartículas/administração & dosagem , Neoplasias/terapia , Transfecção/métodos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células HCT116 , Células HT29 , Células Hep G2 , Humanos , Proteínas Luminescentes/química , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Nanopartículas/química , Nanopartículas/ultraestrutura , Metástase Neoplásica , Neoplasias/genética , Neoplasias/patologia , Polietilenoglicóis/química , Polietilenoimina/química , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Am Soc Nephrol ; 25(1): 81-91, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24071006

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of renal failure that is due to mutations in two genes, PKD1 and PKD2. Vascular complications, including aneurysms, are a well recognized feature of ADPKD, and a subgroup of families exhibits traits reminiscent of Marfan syndrome (MFS). MFS is caused by mutations in fibrillin-1 (FBN1), which encodes an extracellular matrix protein with homology to latent TGF-ß binding proteins. It was recently demonstrated that fibrillin-1 deficiency is associated with upregulation of TGF-ß signaling. We investigated the overlap between ADPKD and MFS by breeding mice with targeted mutations in Pkd1 and Fbn1. Double heterozygotes displayed an exacerbation of the typical Fbn1 heterozygous aortic phenotype. We show that the basis of this genetic interaction results from further upregulation of TGF-ß signaling caused by Pkd1 haploinsufficiency. In addition, we demonstrate that loss of PKD1 alone is sufficient to induce a heightened responsiveness to TGF-ß. Our data link the interaction of two important diseases to a fundamental signaling pathway.


Assuntos
Proteínas dos Microfilamentos/genética , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/genética , Fator de Crescimento Transformador beta/metabolismo , Doenças Vasculares/genética , Doenças Vasculares/metabolismo , Animais , Modelos Animais de Doenças , Epistasia Genética , Feminino , Fibrilina-1 , Fibrilinas , Estudos de Associação Genética , Haploinsuficiência , Heterozigoto , Humanos , Masculino , Síndrome de Marfan/etiologia , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Proteínas dos Microfilamentos/deficiência , Mutação , Rim Policístico Autossômico Dominante/complicações , Transdução de Sinais , Canais de Cátion TRPP/deficiência , Doenças Vasculares/etiologia
12.
Am J Physiol Renal Physiol ; 307(11): F1179-86, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25234309

RESUMO

Autosomal-dominant polycystic kidney disease (ADPKD) is a progressive, proliferative renal disease. Kidneys from ADPKD patients are characterized by the presence of cysts that are marked by enhanced proliferation and apoptosis of renal tubular epithelial cells. Current treatment of this disease is supportive, as there are few if any clinically validated targeted therapeutics. Given the parallels between cystic disease and cancer, and in light of our findings of the efficacy of the nuclear transport inhibitors in kidney cancer, which has similarities to ADPKD, we asked whether such inhibitors show utility in ADPKD. In this study, we tested selective inhibitors of nuclear export (SINE) in two human ADPKD cell lines and in an in vivo mouse model of ADPKD. After effective downregulation of a nuclear exporter, exportin 1 (XPO1), with KPT-330, both cell lines showed dose-dependent inhibition of cell proliferation through G0/G1 arrest associated with downregulation of CDK4, with minimal apoptosis. To analyze mechanisms of CDK4 decrease by XPO1 inhibition, localization of various XPO1 target proteins was examined, and C/EBPß was found to be localized in the nucleus by XPO1 inhibition, resulting in an increase of C/EBPα, which activates degradation of CDK4. Furthermore, inhibition of XPO1 with the parallel inhibitor KPT-335 attenuated cyst growth in vivo in the PKD1 mutant mouse model Pkd1(v/v). Thus, inhibition of nuclear export by KPT-330, which has shown no adverse effects in renal serum chemistries and urinalyses in animal models, and which is already in phase 1 trials for cancers, will be rapidly translatable to human ADPKD.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/biossíntese , Cistos/patologia , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/patologia , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Humanos , Hidrazinas/farmacologia , Carioferinas/antagonistas & inibidores , Camundongos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Triazóis/farmacologia , Proteína Exportina 1
13.
Blood ; 119(12): 2883-94, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-22323452

RESUMO

Constitutive activation of FLT3 by internal tandem duplication (ITD) is one of the most common molecular alterations in acute myeloid leukemia (AML). FLT3/ITD mutations have also been observed in myelodysplastic syndrome patients both before and during progression to AML. Previous work has shown that insertion of an FLT3/ITD mutation into the murine Flt3 gene induces a myeloproliferative neoplasm, but not progression to acute leukemia, suggesting that additional cooperating events are required. We therefore combined the FLT3/ITD mutation with a model of myelodysplastic syndrome involving transgenic expression of the Nup98-HoxD13 (NHD13) fusion gene. Mice expressing both the FLT3/ITD and NHD13 transgene developed AML with 100% penetrance and short latency. These leukemias were driven by mutant FLT3 expression and were susceptible to treatment with FLT3 tyrosine kinase inhibitors. We also observed a spontaneous loss of the wild-type Flt3 allele in these AMLs, further modeling the loss of the heterozygosity phenomenon that is seen in human AML with FLT3-activating mutations. Because resistance to FLT3 inhibitors remains an important clinical issue, this model may help identify new molecular targets in collaborative signaling pathways.


Assuntos
Modelos Animais de Doenças , Proteínas de Homeodomínio/genética , Leucemia Mieloide Aguda/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Proteínas de Fusão Oncogênica/genética , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Western Blotting , Citometria de Fluxo , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Blood ; 120(7): 1516-27, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22740442

RESUMO

Blood vessel networks form in a 2-step process of sprouting angiogenesis followed by selective branch regression and stabilization of remaining vessels. Pericytes are known to function in stabilizing blood vessels, but their role in vascular sprouting and selective vessel regression is poorly understood. The endosialin (CD248) receptor is expressed by pericytes associated with newly forming but not stable quiescent vessels. In the present study, we used the Endosialin(-/-) mouse as a means to uncover novel roles for pericytes during the process of vascular network formation. We demonstrate in a postnatal retina model that Endosialin(-/-) mice have normal vascular sprouting but are defective in selective vessel regression, leading to increased vessel density. Examination of the Endosialin(-/-) mouse tumor vasculature revealed an equivalent phenotype, indicating that pericytes perform a hitherto unidentified function to promote vessel destabilization and regression in vivo in both physiologic and pathologic angiogenesis. Mechanistically, Endosialin(-/-) mice have no defect in pericyte recruitment. Rather, endosialin binding to an endothelial associated, but not a pericyte associated, basement membrane component induces endothelial cell apoptosis and detachment. The results of the present study advance our understanding of pericyte biology and pericyte/endothelial cell cooperation during vascular patterning and have implications for the design of both pro- and antiangiogenic therapies.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/patologia , Padronização Corporal , Neovascularização Fisiológica , Pericitos/patologia , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Aorta/crescimento & desenvolvimento , Aorta/patologia , Apoptose , Membrana Basal/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/metabolismo , Pericitos/metabolismo , Ratos , Retina/metabolismo , Retina/patologia , Vasos Retinianos/crescimento & desenvolvimento , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Proc Natl Acad Sci U S A ; 108(37): 15354-9, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21876161

RESUMO

It is estimated that the etiology of 20-30% of epithelial cancers is directly associated with inflammation, although the direct molecular events linking inflammation and carcinogenesis are poorly defined. In the context of gastrointestinal disease, the bacterium enterotoxigenic Bacteroides fragilis (ETBF) is a significant source of chronic inflammation and has been implicated as a risk factor for colorectal cancer. Spermine oxidase (SMO) is a polyamine catabolic enzyme that is highly inducible by inflammatory stimuli resulting in increased reactive oxygen species (ROS) and DNA damage. We now demonstrate that purified B. fragilis toxin (BFT) up-regulates SMO in HT29/c1 and T84 colonic epithelial cells, resulting in SMO-dependent generation of ROS and induction of γ-H2A.x, a marker of DNA damage. Further, ETBF-induced colitis in C57BL/6 mice is associated with increased SMO expression and treatment of mice with an inhibitor of polyamine catabolism, N(1),N(4)-bis(2,3-butandienyl)-1,4-butanediamine (MDL 72527), significantly reduces ETBF-induced chronic inflammation and proliferation. Most importantly, in the multiple intestinal neoplasia (Min) mouse model, treatment with MDL 72527 reduces ETBF-induced colon tumorigenesis by 69% (P < 0.001). The results of these studies indicate that SMO is a source of bacteria-induced ROS directly associated with tumorigenesis and could serve as a unique target for chemoprevention.


Assuntos
Bacteroides fragilis/fisiologia , Neoplasias do Colo/microbiologia , Poliaminas/metabolismo , Lesões Pré-Cancerosas/microbiologia , Acetiltransferases/metabolismo , Animais , Toxinas Bacterianas/toxicidade , Bacteroides fragilis/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Colite/patologia , Neoplasias do Colo/complicações , Neoplasias do Colo/patologia , Dano ao DNA , Modelos Animais de Doenças , Indução Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Humanos , Inflamação/complicações , Inflamação/patologia , Intestinos/efeitos dos fármacos , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/biossíntese , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Lesões Pré-Cancerosas/patologia , Putrescina/análogos & derivados , Putrescina/farmacologia , Proteínas Recombinantes/toxicidade , Poliamina Oxidase
16.
Blood ; 118(18): 4935-45, 2011 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21908433

RESUMO

Clinical evidence has shown that FLT3 internal tandem duplication (ITD) mutation confers poor prognosis in acute myeloid leukemia. Loss of the FLT3 wild-type (WT) allele is associated with even worse prognosis. We have previously reported that heterozygous FLT3(wt/ITD) "knockin" mice develop a slowly fatal myeloproliferative neoplasm (MPN). To study the roles of the WT FLT3 and ITD alleles in the development of MPNs, we generated FLT3/ITD homozygous (FLT3(ITD/ITD)) and hemizygous (FLT3(-/ITD)) mice. FLT3(-/ITD) mice, with the loss of WT allele, display a more severe MPN, as evidenced by even larger spleen, higher white blood cell counts, and shorter survival, compared with FLT3(wt/ITD) mice. Reintroduction of the WT FLT3 allele into FLT3(-/ITD) BM slowed the progression of MPN in recipient mice. FLT3(ITD/ITD) mice had an even severe MPN compared with the FLT3(-/ITD) and FLT3(wt/ITD) mice. Spontaneous leukemia developed in a small fraction of the FLT3(ITD/ITD) mice but was never observed in the FLT3(-/ITD) and FLT3(wt/ITD) mice. Our results suggest that loss of the WT allele contributes to the development of a more severe phenotype. Thus, the WT FLT3 allele seemingly functions as a tumor suppressor, attenuating the function of the FLT3/ITD allele in leukemia harboring FLT3/ITD mutations.


Assuntos
Proliferação de Células , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Células Mieloides/fisiologia , Tirosina Quinase 3 Semelhante a fms/genética , Alelos , Animais , Progressão da Doença , Técnicas de Introdução de Genes , Genes Supressores de Tumor/fisiologia , Perda de Heterozigosidade/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Invasividade Neoplásica , Sequências de Repetição em Tandem/genética , Sequências de Repetição em Tandem/fisiologia , Tirosina Quinase 3 Semelhante a fms/química , Tirosina Quinase 3 Semelhante a fms/metabolismo , Tirosina Quinase 3 Semelhante a fms/fisiologia
17.
BMC Cancer ; 13: 157, 2013 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-23530816

RESUMO

BACKGROUND: Colorectal cancer (CRC) is the second leading cause of death from cancer in the United States. Colorectal cancers have a prolonged latency following initiation that may span decades providing ample time for implementing a chemoprevention strategy that could block or reverse the progression to CRC. Cdk4 pathway alterations have been linked to a number of cancers including CRC. In these experiments we focused on the Cdk4 pathway and its role in intestinal tumorigenesis as a possible target in chemoprevention strategies. METHODS: We evaluated the effect of Cdk4 blockade on the prevention of intestinal tumor formation by crossing Cdk4(-/-) mice to Apc(-/+) mice. In addition, we tested the effect of the dietary compound silibinin on the Cdk4 pathway in Apc(-/+) mice and HT-29 colon cancer cells in culture. RESULTS: Cdk4(-/-) mice backcrossed to Apc(-/+) mice reduced intestinal adenoma formation compared to Apc(-/+) controls. Silibinin effectively targeted the Cdk4 pathway causing hypophosphorylation of the retinoblastoma protein, inhibited cell growth, and induced apoptosis. As a result silibinin blocked the development of intestinal adenomas by 52% in this genetic model (Apc(-/+) mice) of early events in colorectal cancer formation. No toxic abnormalities were detected in mice which received silibinin. CONCLUSIONS: Modification of the Cdk4 pathway using a natural plant-derived compound such as silibinin may be a useful chemopreventive strategy for colorectal carcinomas.


Assuntos
Quinase 4 Dependente de Ciclina/metabolismo , Genes APC , Transdução de Sinais/efeitos dos fármacos , Silimarina/farmacologia , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Adenoma/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimioprevenção , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Colorretais/prevenção & controle , Ciclina D1/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Knockout , Proteína do Retinoblastoma/metabolismo , Silibina , Silimarina/uso terapêutico
18.
FASEB J ; 26(6): 2648-56, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22415308

RESUMO

After separating from a primary tumor, metastasizing cells enter the circulatory system and interact with host cells before lodging in secondary organs. Previous studies have implicated the surface glycoproteins CD44 and carcinoembryonic antigen (CEA) in adhesion, migration, and invasion, suggesting that they may influence metastatic progression. To elucidate the role of these multifunctional molecules while avoiding the potential drawbacks of ectopic expression or monoclonal antibody treatments, we silenced the expression of CD44 and/or CEA in LS174T colon carcinoma cells and analyzed their ability to metastasize in 2 independent mouse models. Quantitative PCR revealed that CD44 knockdown increased lung and liver metastasis >10-fold, while metastasis was decreased by >50% following CEA knockdown. These findings were corroborated by in vitro experiments assessing the metastatic potential of LS174T cells. Cell migration was decreased as a result of silencing CEA but was enhanced in CD44-knockdown cells. In addition, CD44 silencing promoted homotypic aggregation of LS147T cells, a phenotype that was reversed by additional CEA knockdown. Finally, CD44-knockdown cells exhibited greater mechanical compliance than control cells, a property that correlates with increased metastatic potential. Collectively, these data indicate that CEA, but not CD44, is a viable target for therapeutics aimed at curbing colon carcinoma metastasis.


Assuntos
Antígeno Carcinoembrionário/fisiologia , Neoplasias do Colo/patologia , Receptores de Hialuronatos/fisiologia , Metástase Neoplásica/fisiopatologia , Animais , Movimento Celular/fisiologia , Neoplasias do Colo/fisiopatologia , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Camundongos , Células Tumorais Cultivadas
19.
Arthritis Rheum ; 64(10): 3334-43, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22674221

RESUMO

OBJECTIVE: CD248 (tumor endothelial marker 1/endosialin) is found on stromal cells and is highly expressed during malignancy and inflammation. Studies have shown a reduction in inflammatory arthritis in CD248-knockout (CD248(-/-) ) mice. The aim of the present study was to investigate the functional effect of genetic deletion of CD248 on bone mass. METHODS: Western blotting, polymerase chain reaction, and immunofluorescence were used to investigate the expression of CD248 in humans and mice. Micro-computed tomography and the 3-point bending test were used to measure bone parameters and mechanical properties of the tibiae of 10-week-old wild-type (WT) or CD248(-/-) mice. Human and mouse primary osteoblasts were cultured in medium containing 10 mM ß-glycerophosphate and 50 µg/ml ascorbic acid to induce mineralization, and then treated with platelet-derived growth factor BB (PDGF-BB). The mineral apposition rate in vivo was calculated by identifying newly formed bone via calcein labeling. RESULTS: Expression of CD248 was seen in human and mouse osteoblasts, but not osteoclasts. CD248(-/-) mouse tibiae had higher bone mass and superior mechanical properties (increased load required to cause fracture) compared to WT mice. Primary osteoblasts from CD248(-/-) mice induced increased mineralization in vitro and produced increased bone over 7 days in vivo. There was no decrease in bone mineralization and no increase in proliferation of osteoblasts in response to stimulation with PDGF-BB, which could be attributed to a defect in PDGF signal transduction in the CD248(-/-) mice. CONCLUSION: There is an unmet clinical need to address rheumatoid arthritis-associated bone loss. Genetic deletion of CD248 in mice results in high bone mass due to increased osteoblast-mediated bone formation, suggesting that targeting CD248 in rheumatoid arthritis may have the effect of increasing bone mass in addition to the previously reported effect of reducing inflammation.


Assuntos
Antígenos CD/metabolismo , Osso e Ossos/metabolismo , Proteínas de Neoplasias/metabolismo , Osteoblastos/metabolismo , Osteogênese/fisiologia , Animais , Antígenos CD/genética , Becaplermina , Osso e Ossos/efeitos dos fármacos , Calcificação Fisiológica/efeitos dos fármacos , Calcificação Fisiológica/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis/farmacologia
20.
Cancer Cell ; 7(6): 561-73, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15950905

RESUMO

PIK3CA is mutated in diverse human cancers, but the functional effects of these mutations have not been defined. To evaluate the consequences of PIK3CA alterations, the two most common mutations were inactivated by gene targeting in colorectal cancer (CRC) cells. Biochemical analyses of these cells showed that mutant PIK3CA selectively regulated the phosphorylation of AKT and the forkhead transcription factors FKHR and FKHRL1. PIK3CA mutations had little effect on growth under standard conditions, but reduced cellular dependence on growth factors. PIK3CA mutations resulted in attenuation of apoptosis and facilitated tumor invasion. Treatment with the PI3K inhibitor LY294002 abrogated PIK3CA signaling and preferentially inhibited growth of PIK3CA mutant cells. These data have important implications for therapy of cancers harboring PIK3CA alterations.


Assuntos
Proliferação de Células , Invasividade Neoplásica/patologia , Fosfatidilinositol 3-Quinases/genética , Substituição de Aminoácidos , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose , Sequência de Bases , Linhagem Celular Tumoral , Movimento Celular/genética , Cromonas/farmacologia , Classe I de Fosfatidilinositol 3-Quinases , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead , Marcação de Genes , Substâncias de Crescimento/deficiência , Humanos , Insulina/deficiência , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Nus , Morfolinas/farmacologia , Mutação , Invasividade Neoplásica/genética , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa