Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(51): E10947-E10955, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29203670

RESUMO

KRAS mutant tumors are largely recalcitrant to targeted therapies. Genetically engineered mouse models (GEMMs) of Kras mutant cancer recapitulate critical aspects of this disease and are widely used for preclinical validation of targets and therapies. Through comprehensive profiling of exomes and matched transcriptomes of >200 KrasG12D-initiated GEMM tumors from one lung and two pancreatic cancer models, we discover that significant intratumoral and intertumoral genomic heterogeneity evolves during tumorigenesis. Known oncogenes and tumor suppressor genes, beyond those engineered, are mutated, amplified, and deleted. Unlike human tumors, the GEMM genomic landscapes are dominated by copy number alterations, while protein-altering mutations are rare. However, interspecies comparative analyses of the genomic landscapes demonstrate fidelity between genes altered in KRAS mutant human and murine tumors. Genes that are spontaneously altered during murine tumorigenesis are also among the most prevalent found in human indications. Using targeted therapies, we also demonstrate that this inherent tumor heterogeneity can be exploited preclinically to discover cancer-specific and genotype-specific therapeutic vulnerabilities. Focusing on Kras allelic imbalance, a feature shared by all three models, we discover that MAPK pathway inhibition impinges uniquely on this event, indicating distinct susceptibility and fitness advantage of Kras-mutant cells. These data reveal previously unknown genomic diversity among KrasG12D-initiated GEMM tumors, places them in context of human patients, and demonstrates how to exploit this inherent tumor heterogeneity to discover therapeutic vulnerabilities.


Assuntos
Genes ras , Heterogeneidade Genética , Neoplasias/genética , Neoplasias/patologia , Alelos , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Análise Mutacional de DNA , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Genômica/métodos , Humanos , Neoplasias Pulmonares/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Mutação , Neoplasias/metabolismo , Neoplasias/mortalidade , Prognóstico , Seleção Genética , Transcriptoma
2.
J Pathol ; 227(4): 417-30, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22611036

RESUMO

Resistance to anti-angiogenic therapy can occur via several potential mechanisms. Unexpectedly, recent studies showed that short-term inhibition of either VEGF or VEGFR enhanced tumour invasiveness and metastatic spread in preclinical models. In an effort to evaluate the translational relevance of these findings, we examined the consequences of long-term anti-VEGF monoclonal antibody therapy in several well-validated genetically engineered mouse tumour models of either neuroendocrine or epithelial origin. Anti-VEGF therapy decreased tumour burden and increased overall survival, either as a single agent or in combination with chemotherapy, in all four models examined. Importantly, neither short- nor long-term exposure to anti-VEGF therapy altered the incidence of metastasis in any of these autochthonous models, consistent with retrospective analyses of clinical trials. In contrast, we observed that sunitinib treatment recapitulated previously reported effects on tumour invasiveness and metastasis in a pancreatic neuroendocrine tumour (PNET) model. Consistent with these results, sunitinib treatment resulted in an up-regulation of the hypoxia marker GLUT1 in PNETs, whereas anti-VEGF did not. These results indicate that anti-VEGF mediates anti-tumour effects and therapeutic benefits without a paradoxical increase in metastasis. Moreover, these data underscore the concept that drugs targeting VEGF ligands and receptors may affect tumour metastasis in a context-dependent manner and are mechanistically distinct from one another.


Assuntos
Adenocarcinoma/tratamento farmacológico , Anticorpos Anti-Idiotípicos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Metástase Neoplásica/tratamento farmacológico , Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/imunologia , Adenocarcinoma/genética , Inibidores da Angiogênese/uso terapêutico , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Engenharia Genética , Indóis/uso terapêutico , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Camundongos , Tumores Neuroendócrinos/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Pirróis/uso terapêutico , Carcinoma de Pequenas Células do Pulmão/genética , Sunitinibe , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
3.
Nat Neurosci ; 11(4): 429-39, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18344991

RESUMO

Prevailing notions of cerebral vascularization imply that blood vessels sprout passively into the brain parenchyma from pial vascular plexuses to meet metabolic needs of growing neuronal populations. Endothelial cells, building blocks of blood vessels, are thought to be homogeneous in the brain with respect to their origins, gene expression patterns and developmental mechanisms. These current notions that cerebral angiogenesis is regulated by local environmental signals contrast with current models of cell-autonomous regulation of neuronal development. Here we demonstrate that telencephalic angiogenesis in mice progresses in an orderly, ventral-to-dorsal gradient regulated by compartment-specific homeobox transcription factors. Our data offer new perspectives on intrinsic regulation of angiogenesis in the embryonic telencephalon, call for a revision of the current models of telencephalic angiogenesis and support novel roles for endothelial cells in brain development.


Assuntos
Endotélio Vascular/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Telencéfalo/irrigação sanguínea , Telencéfalo/embriologia , Animais , Padronização Corporal/fisiologia , Circulação Cerebrovascular/fisiologia , Embrião de Mamíferos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Técnicas In Vitro , Camundongos , Camundongos Knockout , Camundongos Mutantes , Neovascularização Fisiológica/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Organogênese/fisiologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Gravidez , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
iScience ; 25(12): 105712, 2022 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-36582483

RESUMO

Here, we have developed an automated image processing algorithm for segmenting lungs and individual lung tumors in in vivo micro-computed tomography (micro-CT) scans of mouse models of non-small cell lung cancer and lung fibrosis. Over 3000 scans acquired across multiple studies were used to train/validate a 3D U-net lung segmentation model and a Support Vector Machine (SVM) classifier to segment individual lung tumors. The U-net lung segmentation algorithm can be used to estimate changes in soft tissue volume within lungs (primarily tumors and blood vessels), whereas the trained SVM is able to discriminate between tumors and blood vessels and identify individual tumors. The trained segmentation algorithms (1) significantly reduce time required for lung and tumor segmentation, (2) reduce bias and error associated with manual image segmentation, and (3) facilitate identification of individual lung tumors and objective assessment of changes in lung and individual tumor volumes under different experimental conditions.

5.
Cell Rep ; 38(6): 110351, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35139374

RESUMO

KRAS, which is mutated in ∼30% of all cancers, activates the RAF-MEK-ERK signaling cascade. CRAF is required for growth of KRAS mutant lung tumors, but the requirement for CRAF kinase activity is unknown. Here, we show that subsets of KRAS mutant tumors are dependent on CRAF for growth. Kinase-dead but not dimer-defective CRAF rescues growth inhibition, suggesting that dimerization but not kinase activity is required. Quantitative proteomics demonstrates increased levels of CRAF:ARAF dimers in KRAS mutant cells, and depletion of both CRAF and ARAF rescues the CRAF-loss phenotype. Mechanistically, CRAF depletion causes sustained ERK activation and induction of cell-cycle arrest, while treatment with low-dose MEK or ERK inhibitor rescues the CRAF-loss phenotype. Our studies highlight the role of CRAF in regulating MAPK signal intensity to promote tumorigenesis downstream of mutant KRAS and suggest that disrupting CRAF dimerization or degrading CRAF may have therapeutic benefit.


Assuntos
Carcinogênese/metabolismo , Dimerização , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Fosforilação/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas ras/genética
6.
J Neurosci ; 30(15): 5334-45, 2010 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-20392955

RESUMO

Dlx5 and Dlx6 homeobox genes are expressed in developing and mature cortical interneurons. Simultaneous deletion of Dlx5 and 6 results in exencephaly of the anterior brain; despite this defect, prenatal basal ganglia differentiation appeared largely intact, while tangential migration of Lhx6(+) and Mafb(+) interneurons to the cortex was reduced and disordered. The migration deficits were associated with reduced CXCR4 expression. Transplantation of mutant immature interneurons into a wild-type brain demonstrated that loss of either Dlx5 or Dlx5&6 preferentially reduced the number of mature parvalbumin(+) interneurons; those parvalbumin(+) interneurons that were present had increased dendritic branching. Dlx5/6(+/-) mice, which appear normal histologically, show spontaneous electrographic seizures and reduced power of gamma oscillations. Thus, Dlx5&6 appeared to be required for development and function of somal innervating (parvalbumin(+)) neocortical interneurons. This contrasts with Dlx1, whose function is required for dendrite innervating (calretinin(+), somatostatin(+), and neuropeptide Y(+)) interneurons (Cobos et al., 2005).


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiologia , Proteínas de Homeodomínio/metabolismo , Interneurônios/fisiologia , Parvalbuminas/metabolismo , Animais , Gânglios da Base/crescimento & desenvolvimento , Gânglios da Base/fisiologia , Gânglios da Base/fisiopatologia , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Córtex Cerebral/fisiopatologia , Dendritos/patologia , Dendritos/fisiologia , Proteínas de Homeodomínio/genética , Interneurônios/citologia , Interneurônios/patologia , Proteínas com Homeodomínio LIM , Fator de Transcrição MafB/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Periodicidade , Receptores CXCR4/metabolismo , Convulsões/patologia , Convulsões/fisiopatologia , Telencéfalo/crescimento & desenvolvimento , Telencéfalo/fisiologia , Telencéfalo/fisiopatologia , Fatores de Transcrição
7.
Dev Biol ; 340(1): 41-53, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20096683

RESUMO

Precise control of neuronal migration is essential for proper function of the brain. Taking a forward genetic screen, we isolated a mutant mouse with defects in interneuron migration. By genetic mapping, we identified a frame shift mutation in the pericentrin (Pcnt) gene. The Pcnt gene encodes a large centrosomal coiled-coil protein that has been implicated in schizophrenia. Recently, frame shift and premature termination mutations in the pericentrin (PCNT) gene were identified in individuals with Seckel syndrome and microcephalic osteodysplastic primordial dwarfism (MOPD II), both of which are characterized by greatly reduced body and brain sizes. The mouse Pcnt mutant shares features with the human syndromes in its overall growth retardation and reduced brain size. We found that dorsal lateral ganglionic eminence (dLGE)-derived olfactory bulb interneurons are severely affected and distributed abnormally in the rostral forebrain in the mutant. Furthermore, mutant interneurons exhibit abnormal migration behavior and RNA interference knockdown of Pcnt impairs cell migration along the rostal migratory stream (RMS) into the olfactory bulb. These findings indicate that pericentrin is required for proper migration of olfactory bulb interneurons and provide a developmental basis for association of pericentrin function with interneuron defects in human schizophrenia.


Assuntos
Antígenos/genética , Movimento Celular/fisiologia , Interneurônios/citologia , Mutação , Bulbo Olfatório/metabolismo , Animais , Centrossomo/metabolismo , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
Future Sci OA ; 7(8): FSO737, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34295539

RESUMO

The utilization of suitable mouse models is a critical step in the drug discovery oncology workflow as their generation and use are important for target identification and validation as well as toxicity and efficacy assessments. Current murine models have been instrumental in furthering insights into the mode of action of drugs before transitioning into the clinic. Recent advancements in genome editing with the development of the CRISPR/Cas9 system and the possibility of applying such technology directly in vivo have expanded the toolkit of preclinical models available. In this review, a brief presentation of the current models used in drug discovery will be provided with a particular emphasis on the novel CRISPR/Cas9 models.

9.
Clin Cancer Res ; 27(4): 1162-1173, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33023953

RESUMO

PURPOSE: Lung adenocarcinomas comprise the largest fraction of non-small cell lung cancer, which is the leading cause of cancer-related deaths. Seventy-five percent of adenocarcinomas lack targeted therapies because of scarcity of druggable drivers. Here, we classified tumors on the basis of signaling similarities and discovered subgroups within this unmet patient population. EXPERIMENTAL DESIGN: We leveraged transcriptional data from >800 early- and advanced-stage patients. RESULTS: We identified three robust subtypes dubbed mucinous, proliferative, and mesenchymal with respective pathway phenotypes. These transcriptional states lack discrete and causative mutational etiology as evidenced by similarly distributed oncogenic drivers, including KRAS and EGFR. The subtypes capture heterogeneity even among tumors lacking known oncogenic drivers. Paired multi-regional intratumoral biopsies demonstrated unified subtypes despite divergently evolved prooncogenic mutations, indicating subtype stability during selective pressure. Heterogeneity among in vitro and in vivo preclinical models is expounded by the human lung adenocarcinoma subtypes and can be leveraged to discover subtype-specific vulnerabilities. As proof of concept, we identified differential subtype response to MEK pathway inhibition in a chemical library screen of 89 lung cancer cell lines, which reproduces across model systems and a clinical trial. CONCLUSIONS: Our findings support forward translational relevance of transcriptional subtypes, where further exploration therein may improve lung adenocarcinoma treatment.See related commentary by Skoulidis, p. 913.


Assuntos
Adenocarcinoma de Pulmão/tratamento farmacológico , Biomarcadores Tumorais/genética , Neoplasias Pulmonares/tratamento farmacológico , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Adenocarcinoma de Pulmão/diagnóstico , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Animais , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Conjuntos de Dados como Assunto , Feminino , Heterogeneidade Genética , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Estadiamento de Neoplasias , Inibidores de Proteínas Quinases/farmacologia , RNA-Seq , Transcriptoma/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cereb Cortex ; 19 Suppl 1: i96-106, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19386638

RESUMO

Here we define the expression of approximately 100 transcription factors (TFs) in progenitors and neurons of the developing mouse medial and caudal ganglionic eminences, anlage of the basal ganglia and pallial interneurons. We have begun to elucidate the transcriptional hierarchy of these genes with respect to the Dlx homeodomain genes, which are essential for differentiation of most gamma-aminobutyric acidergic projection neurons of the basal ganglia. This analysis identified Dlx-dependent and Dlx-independent pathways. The Dlx-independent pathway depends in part on the function of the Mash1 basic helix-loop-helix (b-HLH) TF. These analyses define core transcriptional components that differentially specify the identity and differentiation of the globus pallidus, basal telencephalon, and pallial interneurons.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/fisiologia , Encéfalo/citologia , Encéfalo/metabolismo , Proteínas de Homeodomínio/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Animais , Encéfalo/embriologia , Diferenciação Celular , Camundongos , Camundongos Endogâmicos C57BL
11.
Cell Death Differ ; 26(11): 2416-2429, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30824837

RESUMO

Emerging research suggests that multiple tumor compartments can influence treatment responsiveness and relapse, yet the search for therapeutic resistance mechanisms remains largely focused on acquired genomic alterations in cancer cells. Here we show how treatment-induced changes occur in multiple tumor compartments during tumor relapse and can reduce benefit of follow-on therapies. By using serial biopsies, next-generation sequencing, and single-cell transcriptomics, we tracked the evolution of multiple cellular compartments within individual lesions during first-line treatment response, relapse, and second-line therapeutic interventions in an autochthonous model of melanoma. We discovered that although treatment-relapsed tumors remained genetically stable, they converged on a shared resistance phenotype characterized by dramatic changes in tumor cell differentiation state, immune infiltration, and extracellular matrix (ECM) composition. Similar alterations in tumor cell differentiation were also observed in more than half of our treatment-relapsed patient tumors. Tumor cell-state changes were coincident with ECM remodeling and increased tumor stiffness, which alone was sufficient to alter tumor cell fate and reduce treatment responses in melanoma cell lines in vitro. Despite the absence of acquired mutations in the targeted pathway, resistant tumors showed significantly decreased responsiveness to second-line therapy intervention within the same pathway. The ability to preclinically model relapse and refractory settings-while capturing dynamics within and crosstalk between all relevant tumor compartments-provides a unique opportunity to better design and sequence appropriate clinical interventions.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Matriz Extracelular/patologia , Melanoma/tratamento farmacológico , Melanoma/patologia , Animais , Azetidinas/farmacologia , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Variações do Número de Cópias de DNA/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Melanoma/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Piperidinas/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Vemurafenib/farmacologia , Sequenciamento do Exoma
12.
Neuron ; 44(2): 251-61, 2004 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-15473965

RESUMO

Most cortical interneurons arise from the subcortical telencephalon, but the molecules that control their migration remain largely unidentified. Here, we show that different isoforms of Neuregulin-1 are expressed in the developing cortex and in the route that migrating interneurons follow toward the cortex, whereas a population of the migrating interneurons express ErbB4, a receptor for Neuregulin-1. The different isoforms of Neuregulin-1 act as short- and long-range attractants for migrating interneurons, and perturbing ErbB4 function in vitro decreases the number of interneurons that tangentially migrate to the cortex. In vivo, loss of Neuregulin-1/ErbB4 signaling causes an alteration in the tangential migration of cortical interneurons and a reduction in the number of GABAergic interneurons in the postnatal cortex. These observations provide evidence that Neuregulin-1 and its ErbB4 receptor directly control neuronal migration in the nervous system.


Assuntos
Movimento Celular/fisiologia , Córtex Cerebral/embriologia , Interneurônios/fisiologia , Neuregulina-1/fisiologia , Animais , Células COS , Chlorocebus aethiops , Receptores ErbB/metabolismo , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Isoformas de Proteínas/fisiologia , Receptor ErbB-4 , Transdução de Sinais
13.
J Neurosci ; 27(12): 3230-43, 2007 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-17376983

RESUMO

Olfactory bulb interneuron development is a complex multistep process that involves cell specification in the ventral telencephalon, tangential migration into the olfactory bulb, and local neuronal maturation. Although several transcription factors have been implicated in this process, how or when they act remains to be elucidated. Here we explore the mechanisms that result in olfactory bulb interneuron defects in Dlx1&2-/- (distal-less homeobox 1 and 2) and Mash1-/- (mammalian achaete-schute homolog 1) mutants. We provide evidence that Dlx1&2 and Mash1 regulate parallel molecular pathways that are required for the generation of these cells, thereby providing new insights into the mechanisms underlying olfactory bulb development. The analysis also defined distinct anatomical zones related to olfactory bulb development. Finally we show that Dlx1&2 are required for promoting tangential migration to the olfactory bulb, potentially via regulating the expression of ErbB4 (v-erb-a erythroblastic leukemia viral oncogene homolog 4), Robo2 (roundabout homolog 2), Slit1 (slit homolog 1), and PK2 (prokineticin 2), which have all been shown to play essential roles in this migration.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Homeodomínio/fisiologia , Interneurônios/citologia , Interneurônios/fisiologia , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiologia , Fatores de Transcrição/fisiologia , Animais , Movimento Celular/fisiologia , Interneurônios/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Bulbo Olfatório/metabolismo , Transdução de Sinais/fisiologia
14.
J Neurosci ; 27(26): 6878-91, 2007 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-17596436

RESUMO

The subventricular zone (SVZ) of the postnatal brain continuously generates olfactory bulb (OB) interneurons. We show that calretinin+, calbindin+, and dopaminergic (TH+) periglomerular OB interneurons correspond to distinct subtypes of GABAergic cells; all were produced in the postnatal mouse brain, but they matured and were eliminated at different rates. The embryonic lateral ganglionic eminence (LGE) is thought to be the site of origin of postnatal SVZ neural progenitors. Consistently, grafts of the embryonic LGE into the adult brain SVZ generated many OB interneurons, including TH+ and calbindin+ periglomerular interneurons. However, calretinin+ cells were not produced from these LGE grafts. Surprisingly, pallial and septal embryonic progenitors transplanted into the adult brain SVZ also resulted in the generation of OB interneurons, including calretinin+ cells. A subset of Dlx2+ OB interneurons was derived from cells expressing Emx1, a transcription factor largely restricted to the pallium during development. Emx1 lineage-derived cells contributed a substantial portion of GABAergic cells in the OB, including calretinin+ interneurons. This is in contrast to cortex, in which Emx1 lineage-derived cells do not differentiate into GABAergic neurons. Our results suggest that some OB interneurons are derived from progenitors outside the LGE and that precursors expressing what has classically been considered a pallial transcription factor generate GABAergic interneurons.


Assuntos
Proteínas de Homeodomínio/metabolismo , Interneurônios/metabolismo , Bulbo Olfatório/embriologia , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Transplante de Tecido Encefálico , Calbindina 2 , Calbindinas , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Dopamina/metabolismo , Sobrevivência de Enxerto/fisiologia , Interneurônios/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Bulbo Olfatório/citologia , Bulbo Olfatório/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo , Células-Tronco/citologia , Telencéfalo/citologia , Telencéfalo/embriologia , Telencéfalo/metabolismo
15.
Clin Cancer Res ; 24(18): 4455-4467, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29798909

RESUMO

Purpose: The tumor microenvironment presents with altered extracellular matrix (ECM) and stroma composition, which may affect treatment efficacy and contribute to tissue stiffness. Ultrasound (US) elastography can visualize and quantify tissue stiffness noninvasively. However, the contributions of ECM and stromal components to stiffness are poorly understood. We therefore set out to quantify ECM and stroma density and their relation to tumor stiffness.Experimental Design: A modified clinical ultrasound system was used to measure tumor stiffness and perfusion during tumor growth in preclinical tumor models. In vivo measurements were compared with collagen mass spectroscopy and automatic analysis of matrix and stromal markers derived from immunofluorescence images.Results: US elastography estimates of tumor stiffness were positively correlated with tumor volume in collagen and myofibroblast-rich tumors, while no correlations were found for tumors with low collagen and myofibroblast content. US elastography measurements were strongly correlated with ex vivo mechanical testing and mass spectroscopy-based measurements of total collagen and immature collagen crosslinks. Registration of ultrasound and confocal microscopy data showed strong correlations between blood vessel density and T-cell density in syngeneic tumors, while no correlations were found for genetic tumor models. In contrast to collagen density, which was positively correlated with stiffness, no significant correlations were observed for hyaluronic acid density. Finally, localized delivery of collagenase led to a significant reduction in tumor stiffness without changes in perfusion 24 hours after treatment.Conclusions: US elastography can be used as a potential biomarker to assess changes in the tumor microenvironment, particularly changes affecting the ECM. Clin Cancer Res; 24(18); 4455-67. ©2018 AACR.


Assuntos
Contagem de Células , Técnicas de Imagem por Elasticidade , Matriz Extracelular/patologia , Melanoma Experimental/diagnóstico por imagem , Animais , Linhagem Celular Tumoral , Colágeno/metabolismo , Colágeno/ultraestrutura , Modelos Animais de Doenças , Matriz Extracelular/genética , Humanos , Ácido Hialurônico/ultraestrutura , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Microambiente Tumoral/genética
16.
J Neurosci ; 23(2): 568-78, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12533617

RESUMO

Induction, neurogenesis, and synaptogenesis of the olfactory bulb are thought to require interactions with the olfactory epithelium. The Dlx family of homeobox genes is expressed in both the olfactory bulb and olfactory epithelium. In particular, Dlx5 is expressed in the olfactory placode, olfactory epithelium, and local circuit neurons of the olfactory bulb. Here we analyzed mice lacking DLX5 function. The Dlx5-/- mutation reduces the size of the olfactory epithelium. Although some olfactory neurons are formed, they fail to generate olfactory axons that innervate the olfactory bulb. Despite the lack of innervation, the olfactory bulb forms, and neurogenesis of projection and local circuit neurons proceeds. However, the mutation has a cell-autonomous effect on the ability of neural progenitors to produce olfactory bulb local circuit neurons, with granule cells more severely affected than periglomerular cells. In addition, the mutation has a noncell-autonomous effect on the morphogenesis of mitral cells.


Assuntos
Proteínas de Homeodomínio/metabolismo , Bulbo Olfatório/metabolismo , Mucosa Olfatória/metabolismo , Condutos Olfatórios/metabolismo , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Bromodesoxiuridina , Morte Celular , Divisão Celular/genética , Movimento Celular/genética , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Bulbo Olfatório/citologia , Bulbo Olfatório/embriologia , Mucosa Olfatória/citologia , Mucosa Olfatória/embriologia , Nervo Olfatório/citologia , Nervo Olfatório/embriologia , Condutos Olfatórios/citologia , Condutos Olfatórios/embriologia , RNA Mensageiro/biossíntese , Células-Tronco/citologia , Células-Tronco/metabolismo , Ácido gama-Aminobutírico/biossíntese
17.
Stem Cell Reports ; 4(5): 768-79, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25937372

RESUMO

The adult prostate possesses a significant regenerative capacity that is of great interest for understanding adult stem cell biology. We demonstrate that leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5) is expressed in a rare population of prostate epithelial progenitor cells, and a castration-resistant Lgr5(+) population exists in regressed prostate tissue. Genetic lineage tracing revealed that Lgr5(+) cells and their progeny are primarily luminal. Lgr5(+) castration-resistant cells are long lived and upon regeneration, both luminal Lgr5(+) cells and basal Lgr5(+) cells expand. Moreover, single Lgr5(+) cells can generate multilineage prostatic structures in renal transplantation assays. Additionally, Lgr5(+) cell depletion revealed that the regenerative potential of the castrated adult prostate depends on Lgr5(+) cells. Together, these data reveal insights into the cellular hierarchy of castration-resistant Lgr5+ cells, indicate a requirement for Lgr5(+) cells during prostatic regeneration, and identify an Lgr5(+) adult stem cell population in the prostate.


Assuntos
Próstata/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/metabolismo , Androgênios/farmacologia , Animais , Linhagem da Célula , Células Cultivadas , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Orquiectomia , Próstata/citologia , Próstata/patologia , Ratos , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos , Células-Tronco/citologia , Testosterona/farmacologia
18.
Transl Oncol ; 8(2): 126-35, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25926079

RESUMO

Genetically engineered mouse models (GEMMs) of lung cancer closely recapitulate the human disease but suffer from the difficulty of evaluating tumor growth by conventional methods. Herein, a novel automated image analysis method for estimating the lung tumor burden from in vivo micro-computed tomography (micro-CT) data is described. The proposed tumor burden metric is the segmented soft tissue volume contained within a chest space region of interest, excluding an estimate of the heart volume. The method was validated by comparison with previously published manual analysis methods and applied in two therapeutic studies in a mutant K-ras GEMM of non-small cell lung carcinoma. Mice were imaged by micro-CT pre-treatment and stratified into four treatment groups: an antibody inhibiting vascular endothelial growth factor (anti-VEGF), chemotherapy, combination of anti-VEGF and chemotherapy, or control antibody. In the first study, post-treatment imaging was performed 4 weeks later. In the second study, mice were scanned serially on a high-throughput scanner every 2 weeks for 8 weeks during treatment. In both studies, the automated tumor burden estimates were well correlated with manual metrics (r value range: 0.83-0.93, P < .0001) and showed a similar, significant reduction in tumor growth in mice treated with anti-VEGF alone or in combination with chemotherapy. Given the fully automated nature of this technique, the proposed analysis method can provide a valuable tool in preclinical drug research for screening and randomizing animals into treatment groups and evaluating treatment efficacy in mouse models of lung cancer in a highly robust and efficient manner.

19.
J Comp Neurol ; 521(7): 1561-84, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23042297

RESUMO

Mice lacking the Dlx1 and Dlx2 homeobox genes (Dlx1/2 mutants) have severe deficits in subpallial differentiation, including overexpression of the Gsx1 and Gsx2 homeobox genes. To investigate whether Gsx overexpression contributes to the Dlx1/2 mutant phenotypes, we made compound loss-of-function mutants. Eliminating Gsx2 function from the Dlx1/2 mutants rescued the increased expression of Ascl1 and Hes5 (Notch signaling mediators) and Olig2 (oligodendrogenesis mediator). In addition, Dlx1/2;Gsx2 mutants, like Dlx1/2;Ascl1 mutants, exacerbated the Gsx2 and Dlx1/2 patterning and differentiation phenotypes, particularly in the lateral ganglionic eminence (LGE) caudal ganglionic eminence (CGE), and septum, including loss of GAD1 expression. On the other hand, eliminating Gsx1 function from the Dlx1/2 mutants (Dlx1/2;Gsx1 mutants) did not severely exacerbate their phenotype; on the contrary, it resulted in a partial rescue of medial ganglionic eminence (MGE) properties, including interneuron migration to the cortex. Thus, despite their redundant properties, Gsx1 and -2 have distinct interactions with Dlx1 and -2. Gsx2 interaction is strongest in the LGE, CGE, and septum, whereas the Gsx1 interaction is strongest in the MGE. From these studies, and earlier studies, we present a model of the transcriptional network that regulates early steps of subcortical development.


Assuntos
Encéfalo/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Neurogênese/fisiologia , Fatores de Transcrição/genética , Animais , Encéfalo/embriologia , Embrião de Mamíferos , Imunofluorescência , Hibridização In Situ , Camundongos , Camundongos Mutantes , Fenótipo
20.
Neuron ; 69(1): 61-76, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-21220099

RESUMO

CXCL12/CXCR4 signaling is critical for cortical interneuron migration and their final laminar distribution. No information is yet available on CXCR7, a newly defined CXCL12 receptor. Here we demonstrated that CXCR7 regulated interneuron migration autonomously, as well as nonautonomously through its expression in immature projection neurons. Migrating cortical interneurons coexpressed Cxcr4 and Cxcr7, and Cxcr7(-/-) and Cxcr4(-/-) mutants had similar defects in interneuron positioning. Ectopic CXCL12 expression and pharmacological blockade of CXCR4 in Cxcr7(-/-) mutants showed that both receptors were essential for responding to CXCL12 during interneuron migration. Furthermore, live imaging revealed that Cxcr4(-/-) and Cxcr7(-/-) mutants had opposite defects in interneuron motility and leading process morphology. In vivo inhibition of Gα(i/o) signaling in migrating interneurons phenocopied the interneuron lamination defects of Cxcr4(-/-) mutants. On the other hand, CXCL12 stimulation of CXCR7, but not CXCR4, promoted MAP kinase signaling. Thus, we suggest that CXCR4 and CXCR7 have distinct roles and signal transduction in regulating interneuron movement and laminar positioning.


Assuntos
Movimento Celular/fisiologia , Interneurônios/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Córtex Cerebral/citologia , Quimiocina CXCL12/metabolismo , Imuno-Histoquímica , Interneurônios/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Knockout , Receptores CXCR/deficiência , Receptores CXCR/genética , Receptores CXCR4/deficiência , Receptores CXCR4/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa