Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 1201, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36882420

RESUMO

SNAIL is a key transcriptional regulator in embryonic development and cancer. Its effects in physiology and disease are believed to be linked to its role as a master regulator of epithelial-to-mesenchymal transition (EMT). Here, we report EMT-independent oncogenic SNAIL functions in cancer. Using genetic models, we systematically interrogated SNAIL effects in various oncogenic backgrounds and tissue types. SNAIL-related phenotypes displayed remarkable tissue- and genetic context-dependencies, ranging from protective effects as observed in KRAS- or WNT-driven intestinal cancers, to dramatic acceleration of tumorigenesis, as shown in KRAS-induced pancreatic cancer. Unexpectedly, SNAIL-driven oncogenesis was not associated with E-cadherin downregulation or induction of an overt EMT program. Instead, we show that SNAIL induces bypass of senescence and cell cycle progression through p16INK4A-independent inactivation of the Retinoblastoma (RB)-restriction checkpoint. Collectively, our work identifies non-canonical EMT-independent functions of SNAIL and unravel its complex context-dependent role in cancer.


Assuntos
Neoplasias Pancreáticas , Fatores de Transcrição da Família Snail , Carcinogênese , Transformação Celular Neoplásica , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas p21(ras) , Animais , Fatores de Transcrição da Família Snail/genética
2.
Gut ; 71(4): 766-777, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-33846140

RESUMO

OBJECTIVE: Molecular taxonomy of tumours is the foundation of personalised medicine and is becoming of paramount importance for therapeutic purposes. Four transcriptomics-based classification systems of pancreatic ductal adenocarcinoma (PDAC) exist, which consistently identified a subtype of highly aggressive PDACs with basal-like features, including ΔNp63 expression and loss of the epithelial master regulator GATA6. We investigated the precise molecular events driving PDAC progression and the emergence of the basal programme. DESIGN: We combined the analysis of patient-derived transcriptomics datasets and tissue samples with mechanistic experiments using a novel dual-recombinase mouse model for Gata6 deletion at late stages of KRasG12D-driven pancreatic tumorigenesis (Gata6LateKO). RESULTS: This comprehensive human-to-mouse approach showed that GATA6 loss is necessary, but not sufficient, for the expression of ΔNp63 and the basal programme in patients and in mice. The concomitant loss of HNF1A and HNF4A, likely through epigenetic silencing, is required for the full phenotype switch. Moreover, Gata6 deletion in mice dramatically increased the metastatic rate, with a propensity for lung metastases. Through RNA-Seq analysis of primary cells isolated from mouse tumours, we show that Gata6 inhibits tumour cell plasticity and immune evasion, consistent with patient-derived data, suggesting that GATA6 works as a barrier for acquiring the fully developed basal and metastatic phenotype. CONCLUSIONS: Our work provides both a mechanistic molecular link between the basal phenotype and metastasis and a valuable preclinical tool to investigate the most aggressive subtype of PDAC. These data, therefore, are important for understanding the pathobiological features underlying the heterogeneity of pancreatic cancer in both mice and human.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animais , Carcinoma Ductal Pancreático/patologia , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Camundongos , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas
3.
Cancer Discov ; 11(12): 3158-3177, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34282029

RESUMO

Biliary tract cancer ranks among the most lethal human malignancies, representing an unmet clinical need. Its abysmal prognosis is tied to an increasing incidence and a fundamental lack of mechanistic knowledge regarding the molecular basis of the disease. Here, we show that the Pdx1-positive extrahepatic biliary epithelium is highly susceptible toward transformation by activated PIK3CAH1047R but refractory to oncogenic KrasG12D. Using genome-wide transposon screens and genetic loss-of-function experiments, we discover context-dependent genetic interactions that drive extrahepatic cholangiocarcinoma (ECC) and show that PI3K signaling output strength and repression of the tumor suppressor p27Kip1 are critical context-specific determinants of tumor formation. This contrasts with the pancreas, where oncogenic Kras in concert with p53 loss is a key cancer driver. Notably, inactivation of p27Kip1 permits KrasG12D-driven ECC development. These studies provide a mechanistic link between PI3K signaling, tissue-specific tumor suppressor barriers, and ECC pathogenesis, and present a novel genetic model of autochthonous ECC and genes driving this highly lethal tumor subtype. SIGNIFICANCE: We used the first genetically engineered mouse model for extrahepatic bile duct carcinoma to identify cancer genes by genome-wide transposon-based mutagenesis screening. Thereby, we show that PI3K signaling output strength and p27Kip1 function are critical determinants for context-specific ECC formation. This article is highlighted in the In This Issue feature, p. 2945.


Assuntos
Neoplasias dos Ductos Biliares , Neoplasias do Sistema Biliar , Animais , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos/patologia , Neoplasias do Sistema Biliar/genética , Genes Supressores de Tumor , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/genética
4.
BMC Dev Biol ; 21(1): 4, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33517884

RESUMO

BACKGROUND: Our previous study have shown that the PSMD11 protein was an important survival factor for cancer cells except for its key role in regulation of assembly and activity of the 26S proteasome. To further investigate the role of PSMD11 in carcinogenesis, we constructed a conditional exon 5 floxed allele of PSMD11 (PSMD11flx) in mice. RESULTS: It was found that homozygous PSMD11 flx/flx mice showed normal and exhibited a normal life span and fertility, and showed roughly equivalent expression of PSMD11 in various tissues, suggesting that the floxed allele maintained the wild-type function. Cre recombinase could induce efficient knockout of the floxed PSMD11 allele both in vitro and in vivo. Mice with constitutive single allele deletion of PSMD11 derived from intercrossing between PSMD11flx/flx and CMV-Cre mice were all viable and fertile, and showed apparent growth retardation, suggesting that PSMD11 played a significant role in the development of mice pre- or postnatally. No whole-body PSMD11 deficient embryos (PSMD11-/-) were identified in E7.5-8.5 embryos in uteros, indicating that double allele knockout of PSMD11 leads to early embryonic lethality. To avoid embryonic lethality produced by whole-body PSMD11 deletion, we further developed conditional PSMD11 global knockout mice with genotype Flp;FSF-R26CAG - CreERT2/+; PSMD11 flx/flx, and demonstrated that PSMD11 could be depleted in a temporal and tissue-specific manner. Meanwhile, it was found that depletion of PSMD11 could induce massive apoptosis in MEFs. CONCLUSIONS: In summary, our data demonstrated that we have successfully generated a conditional knockout allele of PSMD11 in mice, and found that PSMD11 played a key role in early and postnatal development in mice, the PSMD11 flx/flx mice will be an invaluable tool to explore the functions of PSMD11 in development and diseases.


Assuntos
Alelos , Complexo de Endopeptidases do Proteassoma/genética , Animais , Homozigoto , Camundongos , Camundongos Knockout
5.
J Cell Biochem ; 119(8): 6644-6656, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29665121

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most devastating disease with the 5-year survival rate less than 6%. In this study, we investigated if inhibiting protein synthesis directly with homoharringtonine (HHT) could induce acute apoptosis in pancreatic cancer cells through quick depletion of multiple short-lived critical members of the central proteome, example, PSMD11(26S proteasome non-ATPase regulatory subunit 11). It was shown that although HHT could inhibit proliferation and growth of MiaPaCa-2 and PANC-1 cells in a time- and dose-dependent manner, only part of pancreatic cancer cells could be induced to die through acute apoptosis. Mechanistic studies showed that HHT could induce quick protein synthesis of PSMD11 through activating MEK1/ERK1/2 signaling pathway in pancreatic cancer cells. Inhibiting MEK1/ERK1/2 pathway with sorafenib could improve the cytotoxity of HHT in vitro and in a genetically engineered mouse model of pancreatic cancer. These results suggest that quick induction of PSMD11 or other acute apoptosis inhibitors through activation of the MEK1/ERK1/2 signaling pathway may be one of the important surviving mechanism which can help pancreatic cancer cells avoid acute apoptosis, it may have significant implications for the targeted therapy of pancreatic ductal adenocarcinoma.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Mepesuccinato de Omacetaxina/farmacologia , MAP Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Complexo de Endopeptidases do Proteassoma/biossíntese , Biossíntese de Proteínas/efeitos dos fármacos , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Humanos , MAP Quinase Quinase 1/genética , Sistema de Sinalização das MAP Quinases/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Complexo de Endopeptidases do Proteassoma/genética
6.
Stem Cells Dev ; 27(12): 819-830, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29336212

RESUMO

Microgravity-induced alterations in the autonomic nervous system (ANS) contribute to derangements in both the mechanical and electrophysiological function of the cardiovascular system, leading to severe symptoms in humans following space travel. Because the ANS forms embryonically from neural crest (NC) progenitors, we hypothesized that microgravity can impair NC-derived cardiac structures. Accordingly, we conducted in vitro simulated microgravity experiments employing NC genetic lineage tracing in mice with cKitCreERT2/+, Isl1nLacZ, and Wnt1-Cre reporter alleles. Inducible fate mapping in adult mouse hearts and pluripotent stem cells (iPSCs) demonstrated reduced cKitCreERT2/+-mediated labeling of both NC-derived cardiomyocytes and autonomic neurons (P < 0.0005 vs. controls). Whole transcriptome analysis, suggested that this effect was associated with repressed cardiac NC- and upregulated mesoderm-related gene expression profiles, coupled with abnormal bone morphogenetic protein (BMP)/transforming growth factor beta (TGF-ß) and Wnt/ß-catenin signaling. To separate the manifestations of simulated microgravity on NC versus mesodermal-cardiac derivatives, we conducted Isl1nLacZ lineage analyses, which indicated an approximately 3-fold expansion (P < 0.05) in mesoderm-derived Isl-1+ pacemaker sinoatrial nodal cells; and an approximately 3-fold reduction (P < 0.05) in cardiac NC-derived ANS cells, including sympathetic nerves and Isl-1+ cardiac ganglia. Finally, NC-specific fate mapping with a Wnt1-Cre reporter iPSC model of murine NC development confirmed that simulated microgravity directly impacted the in vitro development of cardiac NC progenitors and their contribution to the sympathetic and parasympathetic innervation of the iPSC-derived myocardium. Altogether, these findings reveal an important role for gravity in the development of NCs and their postnatal derivatives, and have important therapeutic implications for human space exploration, providing insights into cellular and molecular mechanisms of microgravity-induced cardiomyopathies/channelopathies.


Assuntos
Sistema Nervoso Autônomo/metabolismo , Diferenciação Celular , Sistema de Condução Cardíaco/metabolismo , Crista Neural/metabolismo , Neurogênese , Ausência de Peso/efeitos adversos , Via de Sinalização Wnt , Animais , Sistema Nervoso Autônomo/patologia , Sistema de Condução Cardíaco/patologia , Humanos , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Crista Neural/patologia , Simulação de Ausência de Peso
7.
Nature ; 554(7690): 62-68, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29364867

RESUMO

The poor correlation of mutational landscapes with phenotypes limits our understanding of the pathogenesis and metastasis of pancreatic ductal adenocarcinoma (PDAC). Here we show that oncogenic dosage-variation has a critical role in PDAC biology and phenotypic diversification. We find an increase in gene dosage of mutant KRAS in human PDAC precursors, which drives both early tumorigenesis and metastasis and thus rationalizes early PDAC dissemination. To overcome the limitations posed to gene dosage studies by the stromal richness of PDAC, we have developed large cell culture resources of metastatic mouse PDAC. Integration of cell culture genomes, transcriptomes and tumour phenotypes with functional studies and human data reveals additional widespread effects of oncogenic dosage variation on cell morphology and plasticity, histopathology and clinical outcome, with the highest KrasMUT levels underlying aggressive undifferentiated phenotypes. We also identify alternative oncogenic gains (Myc, Yap1 or Nfkb2), which collaborate with heterozygous KrasMUT in driving tumorigenesis, but have lower metastatic potential. Mechanistically, different oncogenic gains and dosages evolve along distinct evolutionary routes, licensed by defined allelic states and/or combinations of hallmark tumour suppressor alterations (Cdkn2a, Trp53, Tgfß-pathway). Thus, evolutionary constraints and contingencies direct oncogenic dosage gain and variation along defined routes to drive the early progression of PDAC and shape its downstream biology. Our study uncovers universal principles of Ras-driven oncogenesis that have potential relevance beyond pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Evolução Molecular , Dosagem de Genes , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Alelos , Animais , Carcinogênese/genética , Proteínas de Ciclo Celular , Inibidor p16 de Quinase Dependente de Ciclina/genética , Progressão da Doença , Feminino , Genes myc , Genes p53 , Humanos , Masculino , Camundongos , Mutação , Subunidade p52 de NF-kappa B/genética , Metástase Neoplásica/genética , Proteínas Nucleares/genética , Fenótipo , Fosfoproteínas/genética , Fatores de Transcrição/genética , Transcriptoma/genética , Fator de Crescimento Transformador beta1/genética , Proteínas de Sinalização YAP
8.
Neuron ; 96(6): 1419-1431.e5, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-29224725

RESUMO

Animals depend on sensory feedback from mechanosensory afferents for the dynamic control of movement. This sensory feedback needs to be selectively modulated in a task- and context-dependent manner. Here, we show that inhibitory interneurons (INs) expressing the RORß orphan nuclear receptor gate sensory feedback to the spinal motor system during walking and are required for the production of a fluid locomotor rhythm. Genetic manipulations that abrogate inhibitory RORß IN function result in an ataxic gait characterized by exaggerated flexion movements and marked alterations to the step cycle. Inactivation of RORß in inhibitory neurons leads to reduced presynaptic inhibition and changes to sensory-evoked reflexes, arguing that the RORß inhibitory INs function to suppress the sensory transmission pathways that activate flexor motor reflexes and interfere with the ongoing locomotor program. VIDEO ABSTRACT.


Assuntos
Interneurônios/fisiologia , Locomoção/fisiologia , Membro 2 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Medula Espinal/citologia , Caminhada/fisiologia , Vias Aferentes , Animais , Animais Recém-Nascidos , Estimulação Elétrica , Retroalimentação Sensorial , GABAérgicos/farmacologia , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Proteínas da Membrana Plasmática de Transporte de Glicina/genética , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Articulação do Quadril/inervação , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/fisiologia , Inibição Neural/genética , Inibição Neural/fisiologia , Membro 2 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX2/metabolismo , Reflexo/genética , Reflexo/fisiologia , Limiar Sensorial/fisiologia
9.
Stem Cell Res ; 23: 20-32, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28672156

RESUMO

Progress in cell therapy for retinal disorders has been challenging. Recognized retinal progenitors are a heterogeneous population of cells that lack surface markers for the isolation of live cells for clinical implementation. In the present application, our objective was to use the stem cell factor receptor c-Kit (CD117), a surface marker, to isolate and evaluate a distinct progenitor cell population from retinas of postnatal and adult mice. Here we report that, by combining traditional methods with fate mapping, we have identified a c-Kit-positive (c-Kit+) retinal progenitor cell (RPC) that is self-renewing and clonogenic in vitro, and capable of generating many cell types in vitro and in vivo. Based on cell lineage tracing, significant subpopulations of photoreceptors in the outer nuclear layer and bipolar, horizontal, amacrine and Müller cells in the inner nuclear layer are the progeny of c-Kit+ cells in vivo. The RPC progeny contributes to retinal neurons and glial cells, which are responsible for the conversion of light into visual signals. The ability to isolate and expand in vitro live c-Kit+ RPCs makes them a future therapeutic option for retinal diseases.


Assuntos
Envelhecimento/fisiologia , Retina/citologia , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Linhagem da Célula/efeitos dos fármacos , Separação Celular , Células Ependimogliais/citologia , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Tamoxifeno/farmacologia , Fatores de Tempo
10.
Development ; 143(23): 4394-4404, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27789621

RESUMO

Olfactory epithelium (OE) has a lifelong capacity for neurogenesis due to the presence of basal stem cells. Despite the ability to generate short-term cultures, the successful in vitro expansion of purified stem cells from adult OE has not been reported. We sought to establish expansion-competent OE stem cell cultures to facilitate further study of the mechanisms and cell populations important in OE renewal. Successful cultures were prepared using adult mouse basal cells selected for expression of c-KIT. We show that c-KIT signaling regulates self-renewal capacity and prevents neurodifferentiation in culture. Inhibition of TGFß family signaling, a known negative regulator of embryonic basal cells, is also necessary for maintenance of the proliferative, undifferentiated state in vitro Characterizing successful cultures, we identified expression of BMI1 and other Polycomb proteins not previously identified in olfactory basal cells but known to be essential for self-renewal in other stem cell populations. Inducible fate mapping demonstrates that BMI1 is expressed in vivo by multipotent OE progenitors, validating our culture model. These findings provide mechanistic insights into the renewal and potency of olfactory stem cells.


Assuntos
Autorrenovação Celular/fisiologia , Neurogênese/fisiologia , Mucosa Olfatória/citologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/citologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Proliferação de Células/fisiologia , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais
11.
Circ Res ; 119(8): 921-30, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27481956

RESUMO

RATIONALE: Culture-expanded cells originating from cardiac tissue that express the cell surface receptor cKit are undergoing clinical testing as a cell source for heart failure and congenital heart disease. Although accumulating data support that mesenchymal stem cells (MSCs) enhance the efficacy of cardiac cKit(+) cells (CSCs), the underlying mechanism for this synergistic effect remains incompletely understood. OBJECTIVE: To test the hypothesis that MSCs stimulate endogenous CSCs to proliferate, migrate, and differentiate via the SDF1/CXCR4 and stem cell factor/cKit pathways. METHODS AND RESULTS: Using genetic lineage-tracing approaches, we show that in the postnatal murine heart, cKit(+) cells proliferate, migrate, and form cardiomyocytes, but not endothelial cells. CSCs exhibit marked chemotactic and proliferative responses when cocultured with MSCs but not with cardiac stromal cells. Antagonism of the CXCR4 pathway with AMD3100 (an SDF1/CXCR4 antagonist) inhibited MSC-induced CSC chemotaxis but stimulated CSC cardiomyogenesis (P<0.0001). Furthermore, MSCs enhanced CSC proliferation via the stem cell factor/cKit and SDF1/CXCR4 pathways (P<0.0001). CONCLUSIONS: Together these findings show that MSCs exhibit profound, yet differential, effects on CSC migration, proliferation, and differentiation and suggest a mechanism underlying the improved cardiac regeneration associated with combination therapy using CSCs and MSCs. These findings have important therapeutic implications for cell-based therapy strategies that use mixtures of CSCs and MSCs.


Assuntos
Quimiocina CXCL12/biossíntese , Células-Tronco Mesenquimais/fisiologia , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-kit/biossíntese , Receptores CXCR4/biossíntese , Fator de Células-Tronco/biossíntese , Animais , Animais Recém-Nascidos , Movimento Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Humanos , Camundongos , Camundongos Transgênicos , Projetos Piloto , Transdução de Sinais/fisiologia , Suínos
12.
Dev Neurobiol ; 76(3): 241-51, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26016700

RESUMO

Olfactory tissue undergoes lifelong renewal, due to the presence of basal neural stem cells. Multiple categories of globose basal stem cells have been identified, expressing markers such as Lgr5, Ascl1, GBC-2, and c-Kit. The differentiation potential of individual globose cells has remained unclear. Here, we utilized Cre/loxP lineage tracing with a multicolor reporter system to define c-Kit+ cell contributions at clonal resolution. We determined that reporter expression permitted identification of c-Kit derived progeny with fine cellular detail, and that clones were found to be comprised by neurons only, microvillar cells only, microvillar cells and neurons, or gland/duct cells. Quantification of reporter-labeled cells indicated that c-Kit+ cells behave as transit amplifying or immediate precursors, although we also found evidence for longer-term c-Kit+ cell contributions. Our results from the application of multicolor fate mapping delineate the clonal contributions of c-Kit+ cells to olfactory epithelial renewal, and provide novel insight into tissue maintenance of an adult neuroepithelium.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula , Células-Tronco Neurais/citologia , Mucosa Olfatória/citologia , Neurônios Receptores Olfatórios/citologia , Animais , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Proteínas Proto-Oncogênicas c-kit/biossíntese
13.
Proc Natl Acad Sci U S A ; 112(42): 13051-6, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26438843

RESUMO

The degree to which cKit-expressing progenitors generate cardiomyocytes in the heart is controversial. Genetic fate-mapping studies suggest minimal contribution; however, whether or not minimal contribution reflects minimal cardiomyogenic capacity is unclear because the embryonic origin and role in cardiogenesis of these progenitors remain elusive. Using high-resolution genetic fate-mapping approaches with cKit(CreERT2/+) and Wnt1::Flpe mouse lines, we show that cKit delineates cardiac neural crest progenitors (CNC(kit)). CNC(kit) possess full cardiomyogenic capacity and contribute to all CNC derivatives, including cardiac conduction system cells. Furthermore, by modeling cardiogenesis in cKit(CreERT2)-induced pluripotent stem cells, we show that, paradoxically, the cardiogenic fate of CNC(kit) is regulated by bone morphogenetic protein antagonism, a signaling pathway activated transiently during establishment of the cardiac crescent, and extinguished from the heart before CNC invasion. Together, these findings elucidate the origin of cKit(+) cardiac progenitors and suggest that a nonpermissive cardiac milieu, rather than minimal cardiomyogenic capacity, controls the degree of CNC(kit) contribution to myocardium.


Assuntos
Miócitos Cardíacos/metabolismo , Crista Neural/citologia , Proteínas Proto-Oncogênicas c-kit/genética , Células-Tronco/citologia , Animais , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Crista Neural/metabolismo
14.
J Physiol ; 593(2): 403-14, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25630261

RESUMO

Oesophageal achalasia is a disease known to result from reduced relaxation of the lower oesophageal sphincter (LES). Nitric oxide (NO) is one of the main inhibitory transmitters. NO-sensitive guanylyl cyclase (NO-GC) acts as the key target of NO and, by the generation of cGMP, mediates nitrergic relaxation in the LES. To date, the exact mechanism of nitrergic LES relaxation is still insufficiently elucidated. To clarify the role of NO-GC in LES relaxation, we used cell-specific knockout (KO) mouse lines for NO-GC. These include mice lacking NO-GC in smooth muscle cells (SMC-GCKO), in interstitial cells of Cajal (ICC-GCKO) and in both SMC/ICC (SMC/ICC-GCKO). We applied oesophageal manometry to study the functionality of LES in vivo. Isometric force studies were performed to monitor LES responsiveness to exogenous NO and electric field stimulation of intrinsic nerves in vitro. Cell-specific expression/deletion of NO-GC was monitored by immunohistochemistry. Swallowing-induced LES relaxation is strongly reduced by deletion of NO-GC in ICC. Basal LES tone is affected by NO-GC deletion in either SMC or ICC. Lack of NO-GC in both cells leads to a complete interruption of NO-induced relaxation and, therefore, to an achalasia-like phenotype similar to that seen in global GCKO mice. Our data indicate that regulation of basal LES tone is based on a dual mechanism mediated by NO-GC in SMC and ICC whereas swallow-induced LES relaxation is mainly regulated by nitrergic mechanisms in ICC.


Assuntos
Esfíncter Esofágico Inferior/metabolismo , Guanilato Ciclase/metabolismo , Células Intersticiais de Cajal/metabolismo , Relaxamento Muscular , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Esfíncter Esofágico Inferior/citologia , Esfíncter Esofágico Inferior/fisiologia , Guanilato Ciclase/genética , Células Intersticiais de Cajal/fisiologia , Contração Isométrica , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Receptores Citoplasmáticos e Nucleares/genética , Guanilil Ciclase Solúvel
15.
J Comp Neurol ; 523(1): 15-31, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25044230

RESUMO

The olfactory epithelium houses chemosensory neurons, which transmit odor information from the nose to the brain. In adult mammals, the olfactory epithelium is a uniquely robust neuroproliferative zone, with the ability to replenish its neuronal and non-neuronal populations due to the presence of germinal basal cells. The stem and progenitor cells of these germinal layers, and their regulatory mechanisms, remain incompletely defined. Here we show that progenitor cells expressing c-Kit, a receptor tyrosine kinase marking stem cells in a variety of embryonic tissues, are required for maintenance of the adult neuroepithelium. Mouse genetic fate-mapping analyses show that embryonically, a c-Kit(+) population contributes to olfactory neurogenesis. In adults under conditions of normal turnover, there is relatively sparse c-Kit(+) progenitor cell (ckPC) activity. However, after experimentally induced neuroepithelial injury, ckPCs are activated such that they reconstitute the neuronal population. There are also occasional non-neuronal cells found to arise from ckPCs. Moreover, the selective depletion of the ckPC population, utilizing temporally controlled targeted diphtheria toxin A expression, results in failure of neurogenesis after experimental injury. Analysis of this model indicates that most ckPCs reside among the globose basal cell populations and act downstream of horizontal basal cells, which can serve as stem cells. Identification of the requirement for olfactory c-Kit-expressing progenitors in olfactory maintenance provides new insight into the mechanisms involved in adult olfactory neurogenesis. Additionally, we define an important and previously unrecognized site of adult c-Kit activity.


Assuntos
Regeneração Nervosa/fisiologia , Células-Tronco Neurais/fisiologia , Mucosa Olfatória/fisiologia , Neurônios Receptores Olfatórios/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Western Blotting , Contagem de Células , Dermoscopia , Imuno-Histoquímica , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Neurogênese/fisiologia , Mucosa Olfatória/crescimento & desenvolvimento , Mucosa Olfatória/lesões , Proteínas Proto-Oncogênicas c-kit/genética
16.
Nat Genet ; 47(1): 47-56, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25485836

RESUMO

Here we describe a conditional piggyBac transposition system in mice and report the discovery of large sets of new cancer genes through a pancreatic insertional mutagenesis screen. We identify Foxp1 as an oncogenic transcription factor that drives pancreatic cancer invasion and spread in a mouse model and correlates with lymph node metastasis in human patients with pancreatic cancer. The propensity of piggyBac for open chromatin also enabled genome-wide screening for cancer-relevant noncoding DNA, which pinpointed a Cdkn2a cis-regulatory region. Histologically, we observed different tumor subentities and discovered associated genetic events, including Fign insertions in hepatoid pancreatic cancer. Our studies demonstrate the power of genetic screening to discover cancer drivers that are difficult to identify by other approaches to cancer genome analysis, such as downstream targets of commonly mutated human cancer genes. These piggyBac resources are universally applicable in any tissue context and provide unique experimental access to the genetic complexity of cancer.


Assuntos
Transformação Celular Neoplásica/genética , Elementos de DNA Transponíveis/genética , Redes Reguladoras de Genes , Mutagênese Insercional , Neoplasias Pancreáticas/genética , Sequência de Aminoácidos , Animais , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/antagonistas & inibidores , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Introdução de Genes , Genes Sintéticos , Genes p16 , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mariposas/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/patologia , ATPases Translocadoras de Prótons/genética , RNA Interferente Pequeno/farmacologia , Proteínas Repressoras/análise , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Transgenes , Transposases/genética , Transposases/fisiologia
17.
Nat Med ; 20(11): 1340-1347, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25326799

RESUMO

Genetically engineered mouse models (GEMMs) have dramatically improved our understanding of tumor evolution and therapeutic resistance. However, sequential genetic manipulation of gene expression and targeting of the host is almost impossible using conventional Cre-loxP-based models. We have developed an inducible dual-recombinase system by combining flippase-FRT (Flp-FRT) and Cre-loxP recombination technologies to improve GEMMs of pancreatic cancer. This enables investigation of multistep carcinogenesis, genetic manipulation of tumor subpopulations (such as cancer stem cells), selective targeting of the tumor microenvironment and genetic validation of therapeutic targets in autochthonous tumors on a genome-wide scale. As a proof of concept, we performed tumor cell-autonomous and nonautonomous targeting, recapitulated hallmarks of human multistep carcinogenesis, validated genetic therapy by 3-phosphoinositide-dependent protein kinase inactivation as well as cancer cell depletion and show that mast cells in the tumor microenvironment, which had been thought to be key oncogenic players, are dispensable for tumor formation.


Assuntos
Carcinoma Ductal Pancreático/patologia , Engenharia Genética/métodos , Terapia de Alvo Molecular , Medicina de Precisão/métodos , Recombinases/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Carcinoma Ductal Pancreático/metabolismo , Linhagem da Célula , Feminino , Proteínas de Fluorescência Verde/metabolismo , Masculino , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , Modelos Biológicos , Metástase Neoplásica , Oncogenes , Pâncreas/patologia , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Reprodutibilidade dos Testes , Especificidade da Espécie , Células Estromais/metabolismo , Células Estromais/patologia , Tamoxifeno , Fatores de Tempo
18.
Am J Physiol Gastrointest Liver Physiol ; 307(1): G98-106, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24833707

RESUMO

Nitric oxide (NO) is a major inhibitory neurotransmitter in the gastrointestinal (GI) tract. Its main effector, NO-sensitive guanylyl cyclase (NO-GC), is expressed in several GI cell types, including smooth muscle cells (SMC), interstitial cells of Cajal (ICC), and fibroblast-like cells. Up to date, the interplay between neurons and these cells to initiate a nitrergic inhibitory junction potential (IJP) is unclear. Here, we investigate the origin of the nitrergic IJP in murine fundus and colon. IJPs were determined in fundus and colon SMC of mice lacking NO-GC globally (GCKO) and specifically in SMC (SM-GCKO), ICC (ICC-GCKO), and both SMC/ICC (SM/ICC-GCKO). Nitrergic IJP was abolished in ICC-GCKO fundus and reduced in SM-GCKO fundus. In the colon, the amplitude of nitrergic IJP was reduced in ICC-GCKO, whereas nitrergic IJP in SM-GCKO was reduced in duration. These results were corroborated by loss of the nitrergic IJP in global GCKO. In conclusion, our results prove the obligatory role of NO-GC in ICC for the initiation of an IJP. NO-GC in SMC appears to enhance the nitrergic IJP, resulting in a stronger and prolonged hyperpolarization in fundus and colon SMC, respectively. Thus NO-GC in both cell types is mandatory to induce a full nitrergic IJP. Our data from the colon clearly reveal the nitrergic IJP to be biphasic, resulting from individual inputs of ICC and SMC.


Assuntos
Colo/inervação , Fundo Gástrico/inervação , Células Intersticiais de Cajal/metabolismo , Inibição Neural , Neurônios Nitrérgicos/metabolismo , Óxido Nítrico/metabolismo , Transmissão Sináptica , Animais , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Potenciais Pós-Sinápticos Inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/metabolismo , Fatores de Tempo
19.
Eur J Immunol ; 44(1): 296-306, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24127407

RESUMO

Mast cells are abundantly situated at contact sites between the body and its environment, such as the skin and, especially during certain immune responses, at mucosal surfaces. They mediate allergic reactions and degrade toxins as well as venoms. However, their roles during innate and adaptive immune responses remain controversial and it is likely that major functions remain to be discovered. Recent developments in mast cell-specific conditional gene targeting in the mouse promise to enhance our understanding of these fascinating cells. To complete the genetic toolbox to study mast cell development, homeostasis and function, it is imperative to inducibly manipulate their gene expression. Here, we report the generation of a novel knock-in mouse line expressing a tamoxifen-inducible version of the Cre recombinase from within the endogenous c-Kit locus. We demonstrate highly efficient and specific inducible expression of a fluorescent reporter protein in mast cells both in vivo and in vitro. Furthermore, induction of diphtheria toxin A expression allowed selective and efficient ablation of mast cells at various anatomical locations, while other hematopoietic cells remain unaffected. This novel mouse strain will hence be very valuable to study mast cell homeostasis and how specific genes influence their functions in physiology and pathology.


Assuntos
Toxina Diftérica/metabolismo , Marcação de Genes/métodos , Integrases/metabolismo , Mastócitos/imunologia , Camundongos Transgênicos/imunologia , Fragmentos de Peptídeos/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Toxina Diftérica/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Introdução de Genes , Loci Gênicos/genética , Integrases/genética , Mastócitos/efeitos dos fármacos , Mastócitos/patologia , Camundongos , Especificidade de Órgãos , Fragmentos de Peptídeos/genética , Proteínas Proto-Oncogênicas c-kit/genética , Tamoxifeno/administração & dosagem , Transgenes/genética
20.
Cancer Cell ; 24(1): 15-29, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23845441

RESUMO

We show that BRAF(V600E) initiates an alternative pathway to colorectal cancer (CRC), which progresses through a hyperplasia/adenoma/carcinoma sequence. This pathway underlies significant subsets of CRCs with distinctive pathomorphologic/genetic/epidemiologic/clinical characteristics. Genetic and functional analyses in mice revealed a series of stage-specific molecular alterations driving different phases of tumor evolution and uncovered mechanisms underlying this stage specificity. We further demonstrate dose-dependent effects of oncogenic signaling, with physiologic Braf(V600E) expression being sufficient for hyperplasia induction, but later stage intensified Mapk-signaling driving both tumor progression and activation of intrinsic tumor suppression. Such phenomena explain, for example, the inability of p53 to restrain tumor initiation as well as its importance in invasiveness control, and the late stage specificity of its somatic mutation. Finally, systematic drug screening revealed sensitivity of this CRC subtype to targeted therapeutics, including Mek or combinatorial PI3K/Braf inhibition.


Assuntos
Neoplasias Colorretais/etiologia , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Animais , Transformação Celular Neoplásica , Neoplasias Colorretais/tratamento farmacológico , Inibidor p16 de Quinase Dependente de Ciclina , Ensaios de Seleção de Medicamentos Antitumorais , Sistema de Sinalização das MAP Quinases , Camundongos , Instabilidade de Microssatélites , Invasividade Neoplásica , Proteínas de Neoplasias/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteína Supressora de Tumor p53/fisiologia , Via de Sinalização Wnt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...