Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Sci Adv ; 10(12): eadn4649, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38517960

RESUMO

Genomic rearrangements are a hallmark of most childhood tumors, including medulloblastoma, one of the most common brain tumors in children, but their causes remain largely unknown. Here, we show that PiggyBac transposable element derived 5 (Pgbd5) promotes tumor development in multiple developmentally accurate mouse models of Sonic Hedgehog (SHH) medulloblastoma. Most Pgbd5-deficient mice do not develop tumors, while maintaining normal cerebellar development. Ectopic activation of SHH signaling is sufficient to enforce cerebellar granule cell progenitor-like cell states, which exhibit Pgbd5-dependent expression of distinct DNA repair and neurodevelopmental factors. Mouse medulloblastomas expressing Pgbd5 have increased numbers of somatic structural DNA rearrangements, some of which carry PGBD5-specific sequences at their breakpoints. Similar sequence breakpoints recurrently affect somatic DNA rearrangements of known tumor suppressors and oncogenes in medulloblastomas in 329 children. This identifies PGBD5 as a medulloblastoma mutator and provides a genetic mechanism for the generation of oncogenic DNA rearrangements in childhood cancer.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Humanos , Criança , Animais , Camundongos , Meduloblastoma/genética , Transposases/genética , Transposases/metabolismo , Proteínas Hedgehog/metabolismo , Fatores de Transcrição/genética , Mutagênese , Neoplasias Cerebelares/genética
2.
iScience ; 26(10): 107831, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37822508

RESUMO

The major cause of treatment failure and mortality among medulloblastoma patients is metastasis intracranially or along the spinal cord. The molecular mechanisms driving tumor metastasis in Sonic hedgehog-driven medulloblastoma (SHH-MB) patients, however, remain largely unknown. In this study we define a tumor suppressive role of KMT2D (MLL2), a gene frequently mutated in the most metastatic ß-subtype. Strikingly, genetic mouse models of SHH-MB demonstrate that heterozygous loss of Kmt2d in conjunction with activation of the SHH pathway causes highly penetrant disease with decreased survival, increased hindbrain invasion and spinal cord metastasis. Loss of Kmt2d attenuates neural differentiation and shifts the transcriptional/chromatin landscape of primary and metastatic tumors toward a decrease in differentiation genes and tumor suppressors and an increase in genes/pathways implicated in advanced stage cancer and metastasis (TGFß, Notch, Atoh1, Sox2, and Myc). Thus, secondary heterozygous KMT2D mutations likely have prognostic value for identifying SHH-MB patients prone to develop metastasis.

3.
Oncogene ; 40(2): 396-407, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33159168

RESUMO

The immune microenvironment of tumors can play a critical role in promoting or inhibiting tumor progression depending on the context. We present evidence that tumor-associated macrophages/microglia (TAMs) can promote tumor progression in the sonic hedgehog subgroup of medulloblastoma (SHH-MB). By combining longitudinal manganese-enhanced magnetic resonance imaging (MEMRI) and immune profiling of a sporadic mouse model of SHH-MB, we found the density of TAMs is higher in the ~50% of tumors that progress to lethal disease. Furthermore, reducing regulatory T cells or eliminating B and T cells in Rag1 mutants does not alter SHH-MB tumor progression. As TAMs are a dominant immune component in tumors and are normally dependent on colony-stimulating factor 1 receptor (CSF1R), we treated mice with a CSF1R inhibitor, PLX5622. Significantly, PLX5622 reduces a subset of TAMs, prolongs mouse survival, and reduces the volume of most tumors within 4 weeks of treatment. Moreover, concomitant with a reduction in TAMs the percentage of infiltrating cytotoxic T cells is increased, indicating a change in the tumor environment. Our studies in an immunocompetent preclinical mouse model demonstrate TAMs can have a functional role in promoting SHH-MB progression. Thus, CSF1R inhibition could have therapeutic potential for a subset of SHH-MB patients.


Assuntos
Neoplasias Cerebelares/prevenção & controle , Modelos Animais de Doenças , Proteínas Hedgehog/fisiologia , Meduloblastoma/prevenção & controle , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Macrófagos Associados a Tumor/imunologia , Animais , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proliferação de Células , Neoplasias Cerebelares/etiologia , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Feminino , Humanos , Masculino , Meduloblastoma/etiologia , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prognóstico , Células Tumorais Cultivadas , Microambiente Tumoral
4.
J Exp Med ; 217(6)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32196081

RESUMO

The activity and survival of retinal photoreceptors depend on support functions performed by the retinal pigment epithelium (RPE) and on oxygen and nutrients delivered by blood vessels in the underlying choroid. By combining single-cell and bulk RNA sequencing, we categorized mouse RPE/choroid cell types and characterized the tissue-specific transcriptomic features of choroidal endothelial cells. We found that choroidal endothelium adjacent to the RPE expresses high levels of Indian Hedgehog and identified its downstream target as stromal GLI1+ mesenchymal stem cell-like cells. In vivo genetic impairment of Hedgehog signaling induced significant loss of choroidal mast cells, as well as an altered inflammatory response and exacerbated visual function defects after retinal damage. Our studies reveal the cellular and molecular landscape of adult RPE/choroid and uncover a Hedgehog-regulated choroidal immunomodulatory signaling circuit. These results open new avenues for the study and treatment of retinal vascular diseases and choroid-related inflammatory blinding disorders.


Assuntos
Corioide/imunologia , Corioide/patologia , Endotélio/imunologia , Imunomodulação , Análise de Célula Única , Animais , Proliferação de Células , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Proteínas Hedgehog/metabolismo , Inflamação/genética , Mastócitos/metabolismo , Melanócitos/metabolismo , Melanócitos/patologia , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Epitélio Pigmentado da Retina/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteína GLI1 em Dedos de Zinco/metabolismo
5.
Magn Reson Med ; 83(1): 214-227, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31403226

RESUMO

PURPOSE: Genetically engineered mouse models of sporadic cancers are critical for studying tumor biology and for preclinical testing of therapeutics. We present an MRI-based pipeline designed to produce high resolution, quantitative information about tumor progression and response to novel therapies in mouse models of medulloblastoma (MB). METHODS: Sporadic MB was modeled in mice by inducing expression of an activated form of the Smoothened gene (aSmo) in a small number of cerebellar granule cell precursors. aSmo mice were imaged and analyzed at defined time-points using a 3D manganese-enhanced MRI-based pipeline optimized for high-throughput. RESULTS: A semi-automated segmentation protocol was established that estimates tumor volume in a time-frame compatible with a high-throughput pipeline. Both an empirical, volume-based classifier and a linear discriminant analysis-based classifier were tested to distinguish progressing from nonprogressing lesions at early stages of tumorigenesis. Tumor centroids measured at early stages revealed that there is a very specific location of the probable origin of the aSmo MB tumors. The efficacy of the manganese-enhanced MRI pipeline was demonstrated with a small-scale experimental drug trial designed to reduce the number of tumor associated macrophages and microglia. CONCLUSION: Our results revealed a high level of heterogeneity between tumors within and between aSmo MB models, indicating that meaningful studies of sporadic tumor progression and response to therapy could not be conducted without an imaging-based pipeline approach.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética , Meduloblastoma/diagnóstico por imagem , Algoritmos , Animais , Cerebelo/metabolismo , Análise Discriminante , Modelos Animais de Doenças , Progressão da Doença , Imageamento Tridimensional , Modelos Lineares , Camundongos , Reconhecimento Automatizado de Padrão , Transdução de Sinais , Receptor Smoothened/genética
6.
J Exp Med ; 216(10): 2265-2281, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31350310

RESUMO

Microglia, the brain resident macrophages, critically shape forebrain neuronal circuits. However, their precise function in the cerebellum is unknown. Here we show that human and mouse cerebellar microglia express a unique molecular program distinct from forebrain microglia. Cerebellar microglial identity was driven by the CSF-1R ligand CSF-1, independently of the alternate CSF-1R ligand, IL-34. Accordingly, CSF-1 depletion from Nestin+ cells led to severe depletion and transcriptional alterations of cerebellar microglia, while microglia in the forebrain remained intact. Strikingly, CSF-1 deficiency and alteration of cerebellar microglia were associated with reduced Purkinje cells, altered neuronal function, and defects in motor learning and social novelty interactions. These findings reveal a novel CSF-1-CSF-1R signaling-mediated mechanism that contributes to motor function and social behavior.


Assuntos
Comportamento Animal/fisiologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Microglia/metabolismo , Atividade Motora/fisiologia , Células de Purkinje/metabolismo , Transdução de Sinais/fisiologia , Comportamento Social , Animais , Humanos , Fator Estimulador de Colônias de Macrófagos/genética , Camundongos , Camundongos Transgênicos , Células de Purkinje/citologia , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo
7.
Nature ; 572(7767): 67-73, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31043743

RESUMO

Study of the origin and development of cerebellar tumours has been hampered by the complexity and heterogeneity of cerebellar cells that change over the course of development. Here we use single-cell transcriptomics to study more than 60,000 cells from the developing mouse cerebellum and show that different molecular subgroups of childhood cerebellar tumours mirror the transcription of cells from distinct, temporally restricted cerebellar lineages. The Sonic Hedgehog medulloblastoma subgroup transcriptionally mirrors the granule cell hierarchy as expected, while group 3 medulloblastoma resembles Nestin+ stem cells, group 4 medulloblastoma resembles unipolar brush cells, and PFA/PFB ependymoma and cerebellar pilocytic astrocytoma resemble the prenatal gliogenic progenitor cells. Furthermore, single-cell transcriptomics of human childhood cerebellar tumours demonstrates that many bulk tumours contain a mixed population of cells with divergent differentiation. Our data highlight cerebellar tumours as a disorder of early brain development and provide a proximate explanation for the peak incidence of cerebellar tumours in early childhood.


Assuntos
Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Evolução Molecular , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Transcrição Gênica , Animais , Neoplasias Cerebelares/classificação , Cerebelo/citologia , Cerebelo/embriologia , Cerebelo/metabolismo , Criança , Feminino , Feto/citologia , Glioma/classificação , Glioma/genética , Glioma/patologia , Humanos , Meduloblastoma/classificação , Meduloblastoma/genética , Meduloblastoma/patologia , Camundongos , Análise de Sequência de RNA , Análise de Célula Única , Fatores de Tempo , Transcriptoma/genética
8.
Proc Natl Acad Sci U S A ; 115(13): 3392-3397, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29531057

RESUMO

The main cell of origin of the Sonic hedgehog (SHH) subgroup of medulloblastoma (MB) is granule cell precursors (GCPs), a SHH-dependent transient amplifying population in the developing cerebellum. SHH-MBs can be further subdivided based on molecular and clinical parameters, as well as location because SHH-MBs occur preferentially in the lateral cerebellum (hemispheres). Our analysis of adult patient data suggests that tumors with Smoothened (SMO) mutations form more specifically in the hemispheres than those with Patched 1 (PTCH1) mutations. Using sporadic mouse models of SHH-MB with the two mutations commonly seen in adult MB, constitutive activation of Smo (SmoM2) or loss-of-Ptch1, we found that regardless of timing of induction or type of mutation, tumors developed primarily in the hemispheres, with SmoM2-mutants indeed showing a stronger specificity. We further uncovered that GCPs in the hemispheres are more susceptible to high-level SHH signaling compared with GCPs in the medial cerebellum (vermis), as more SmoM2 or Ptch1-mutant hemisphere cells remain undifferentiated and show increased tumorigenicity when transplanted. Finally, we identified location-specific GCP gene-expression profiles, and found that deletion of the genes most highly expressed in the hemispheres (Nr2f2) or vermis (Engrailed1) showed opposing effects on GCP differentiation. Our studies thus provide insights into intrinsic differences within GCPs that impact on SHH-MB progression.


Assuntos
Neoplasias Cerebelares/patologia , Cerebelo/patologia , Proteínas Hedgehog/metabolismo , Meduloblastoma/patologia , Receptor Patched-1/metabolismo , Receptor Smoothened/metabolismo , Adulto , Animais , Diferenciação Celular , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/metabolismo , Cerebelo/metabolismo , Proteínas Hedgehog/genética , Humanos , Lactente , Meduloblastoma/genética , Meduloblastoma/metabolismo , Camundongos , Receptor Patched-1/genética , Transdução de Sinais , Receptor Smoothened/genética , Transcriptoma
9.
Dis Model Mech ; 10(1): 39-52, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27935821

RESUMO

It is widely appreciated that reactive stroma or carcinoma-associated fibroblasts can influence epithelial tumor progression. In prostate cancer (PCa), the second most common male malignancy worldwide, the amount of reactive stroma is variable and has predictive value for tumor recurrence. By analyzing human PCa protein and RNA expression databases, we found smooth muscle cells (SMCs) are decreased in advanced tumors, whereas fibroblasts are maintained. In three mouse models of PCa, PB-MYC, ERG/PTEN and TRAMP, we found the composition of the stroma is distinct. SMCs are greatly depleted in advanced PB-MYC tumors and locally reduced in ERG/PTEN prostates, whereas in TRAMP tumors the SMC layers are increased. In addition, interductal fibroblast-like cells expand in PB-MYC and ERG/PTEN tumors, whereas in TRAMP PCa they expand little and stromal cells invade into intraductal adenomas. Fate mapping of SMCs showed that in PB-MYC tumors the cells are depleted, whereas they expand in TRAMP tumors and interestingly contribute to the stromal cells in intraductal adenomas. Hedgehog (HH) ligands secreted by epithelial cells are known to regulate prostate mesenchyme expansion differentially during development and regeneration. Any possible role of HH signaling in stromal cells during PCa progression is poorly understood. We found that HH signaling is high in SMCs and fibroblasts near tumor cells in all models, and epithelial Shh expression is decreased whereas Ihh and Dhh are increased. In human primary PCa, expression of IHH is the highest of the three HH genes, and elevated HH signaling correlates with high stromal gene expression. Moreover, increasing HH signaling in the stroma of PB-MYC PCa resulted in more intact SMC layers and decreased tumor progression (micro-invasive carcinoma). Thus, we propose HH signaling restrains tumor progression by maintaining the smooth muscle and preventing invasion by tumor cells. Our studies highlight the importance of understanding how HH signaling and stromal composition impact on PCa to optimize drug treatments.


Assuntos
Proteínas Hedgehog/metabolismo , Músculo Liso/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais , Actinas/metabolismo , Animais , Contagem de Células , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Invasividade Neoplásica , Neoplasias da Próstata/genética , Células Estromais/metabolismo , Células Estromais/patologia , Proteína GLI1 em Dedos de Zinco/metabolismo
10.
Bull Math Biol ; 78(5): 859-78, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27125657

RESUMO

Determining the cellular basis of brain growth is an important problem in developmental neurobiology. In the mammalian brain, the cerebellum is particularly amenable to studies of growth because it contains only a few cell types, including the granule cells, which are the most numerous neuronal subtype. Furthermore, in the mouse cerebellum granule cells are generated from granule cell precursors (gcps) in the external granule layer (EGL), from 1 day before birth until about 2 weeks of age. The complexity of the underlying cellular processes (multiple cell behaviors, three spatial dimensions, time-dependent changes) requires a quantitative framework to be fully understood. In this paper, a differential equation-based model is presented, which can be used to estimate temporal changes in granule cell numbers in the EGL. The model includes the proliferation of gcps and their differentiation into granule cells, as well as the process by which granule cells leave the EGL. Parameters describing these biological processes were derived from fitting the model to histological data. This mathematical model should be useful for understanding altered gcp and granule cell behaviors in mouse mutants with abnormal cerebellar development and cerebellar cancers.


Assuntos
Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Neurônios/citologia , Algoritmos , Animais , Animais Recém-Nascidos , Diferenciação Celular , Cerebelo/embriologia , Simulação por Computador , Conceitos Matemáticos , Camundongos , Camundongos Mutantes Neurológicos , Modelos Neurológicos , Células-Tronco Neurais/citologia , Neurônios/classificação
11.
Dev Biol ; 400(1): 94-104, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25641695

RESUMO

The prostate gland plays an important role in male reproduction, and is also an organ prone to diseases such as benign prostatic hyperplasia (BPH) and prostate cancer. The prostate consists of ducts with an inner layer of epithelium surrounded by stroma. Reciprocal signaling between these two cell compartments is instrumental to normal prostatic development, homeostasis, regeneration, as well as tumor formation. Hedgehog (HH) signaling is a master regulator in numerous developmental processes. In many organs, HH plays a key role in epithelial-mesenchymal signaling that regulates organ growth and tissue differentiation, and abnormal HH signaling has been implicated in the progression of various epithelial carcinomas. In this review, we focus on recent studies exploring the multipotency of endogenous postnatal and adult epithelial and stromal stem cells and studies addressing the role of HH in prostate development and cancer. We discuss the implications of the results for a new understanding of prostate development and disease. Insight into the cellular and molecular mechanisms underlying epithelial-mesenchymal growth regulation should provide a basis for devising innovative therapies to combat diseases of the prostate.


Assuntos
Células-Tronco Adultas/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Hedgehog/metabolismo , Próstata/embriologia , Próstata/crescimento & desenvolvimento , Neoplasias da Próstata/fisiopatologia , Transdução de Sinais/fisiologia , Animais , Humanos , Masculino , Camundongos , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Especificidade da Espécie
12.
Am J Respir Cell Mol Biol ; 52(1): 1-13, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25068457

RESUMO

Over the past two decades, the secreted protein sonic hedgehog (SHH) has emerged as a critical morphogen during embryonic lung development, regulating the interaction between epithelial and mesenchymal cell populations in the airway and alveolar compartments. There is increasing evidence that the SHH pathway is active in adult lung diseases such as pulmonary fibrosis, asthma, chronic obstructive pulmonary disease, and lung cancer, which raises two questions: (1) What role does SHH signaling play in these diseases? and (2) Is it a primary driver of the disease or a response (perhaps beneficial) to the primary disturbance? In this review we aim to fill the gap between the well-studied period of embryonic lung development and the adult diseased lung by reviewing the hedgehog (HH) pathway during the postnatal period and in adult uninjured and injured lungs. We elucidate the similarities and differences in the epithelial-mesenchymal interplay during the fibrosis response to injury in lung compared with other organs and present a critical appraisal of tools and agents available to evaluate HH signaling.


Assuntos
Proteínas Hedgehog/metabolismo , Pneumopatias/embriologia , Pneumopatias/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Transdução de Sinais , Adulto , Animais , Proteínas Hedgehog/genética , Humanos , Pulmão/patologia , Pneumopatias/genética , Pneumopatias/patologia , Mesoderma/embriologia , Mesoderma/metabolismo , Mesoderma/patologia , Mucosa Respiratória/embriologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
13.
Neoplasia ; 16(12): 993-1006, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25499213

RESUMO

Mouse models have increased our understanding of the pathogenesis of medulloblastoma (MB), the most common malignant pediatric brain tumor that often forms in the cerebellum. A major goal of ongoing research is to better understand the early stages of tumorigenesis and to establish the genetic and environmental changes that underlie MB initiation and growth. However, studies of MB progression in mouse models are difficult due to the heterogeneity of tumor onset times and growth patterns and the lack of clinical symptoms at early stages. Magnetic resonance imaging (MRI) is critical for noninvasive, longitudinal, three-dimensional (3D) brain tumor imaging in the clinic but is limited in resolution and sensitivity for imaging early MBs in mice. In this study, high-resolution (100 µm in 2 hours) and high-throughput (150 µm in 15 minutes) manganese-enhanced MRI (MEMRI) protocols were optimized for early detection and monitoring of MBs in a Patched-1 (Ptch1) conditional knockout (CKO) model. The high tissue contrast obtained with MEMRI revealed detailed cerebellar morphology and enabled detection of MBs over a wide range of stages including pretumoral lesions as early as 2 to 3 weeks postnatal with volumes close to 0.1 mm(3). Furthermore, longitudinal MEMRI allowed noninvasive monitoring of tumors and demonstrated that lesions within and between individuals have different tumorigenic potentials. 3D volumetric studies allowed quantitative analysis of MB tumor morphology and growth rates in individual Ptch1-CKO mice. These results show that MEMRI provides a powerful method for early in vivo detection and longitudinal imaging of MB progression in the mouse brain.


Assuntos
Neoplasias Cerebelares/diagnóstico , Cloretos , Meios de Contraste , Modelos Animais de Doenças , Imageamento por Ressonância Magnética/métodos , Compostos de Manganês , Meduloblastoma/diagnóstico , Animais , Progressão da Doença , Imageamento Tridimensional , Camundongos , Camundongos Knockout , Receptores Patched , Receptor Patched-1 , Receptores de Superfície Celular/genética
14.
Proc Natl Acad Sci U S A ; 110(51): 20611-6, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24218555

RESUMO

The adult mouse prostate has a seemingly endless capacity for regeneration, and sonic hedgehog (SHH) signaling has been implicated in this stem cell-driven process. However, it is not clear whether SHH acts on the epithelium or stromal cells that secrete factors required for epithelial expansion. Because little is known about stromal stem cells compared with their epithelial counterparts, we used in vivo mouse genetics tools to characterize four prostate stromal subtypes and their stem cells. Using knockin reporter alleles, we uncovered that SHH signals from prostate basal epithelial cells to adjacent stromal cells. Furthermore, the SHH target gene Gli1 is preferentially expressed in subepithelial fibroblast-like cells, one of four prostate stromal subtypes and the subtype closest to the epithelial source of SHH. Using Genetic Inducible Fate Mapping to mark adult Gli1- or Smooth muscle actin-expressing cells and follow their fate during regeneration, we uncovered that Gli1-expressing cells exhibit long-term self-renewal capacity during multiple rounds of androgen-mediated regeneration after castration-induced involution, and depleted smooth muscle cells are mainly replenished by preexisting smooth muscle cells. Based on our Genetic Inducible Fate Mapping studies, we propose a model where SHH signals to multiple stromal stem cells, which are largely unipotent in vivo.


Assuntos
Proteínas Hedgehog/metabolismo , Modelos Biológicos , Próstata/metabolismo , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Animais , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteínas Hedgehog/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Próstata/citologia , Células-Tronco/citologia , Células Estromais/citologia , Células Estromais/metabolismo , Proteína GLI1 em Dedos de Zinco
15.
J Neurosci ; 33(44): 17490-505, 2013 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-24174682

RESUMO

Sonic hedgehog (SHH), a key regulator of embryonic neurogenesis, signals directly to neural stem cells (NSCs) in the subventricular zone (SVZ) and to astrocytes in the adult mouse forebrain. The specific mechanism by which the GLI2 and GLI3 transcriptional activators (GLI2(A) and GLI3(A)) and repressors (GLI2(R) and GLI3(R)) carry out SHH signaling has not been addressed. We found that the majority of slow-cycling NSCs express Gli2 and Gli3, whereas Gli1 is restricted ventrally and all three genes are downregulated when NSCs transition into proliferating progenitors. Surprisingly, whereas conditional ablation of Smo in postnatal glial fibrillary acidic protein-expressing cells results in cell-autonomous loss of NSCs and a progressive reduction in SVZ proliferation, without an increase in glial cell production, removal of Gli2 or Gli3 does not alter adult SVZ neurogenesis. Significantly, removing Gli3 in Smo conditional mutants largely rescues neurogenesis and, conversely, expression of a constitutive GLI3(R) in the absence of normal Gli2 and Gli3 abrogates neurogenesis. Thus unattenuated GLI3(R) is a primary inhibitor of adult SVZ NSC function. Ablation of Gli2 and Gli3 revealed a minor role for GLI2(R) and little requirement for GLI(A) function in stimulating SVZ neurogenesis. Moreover, we found that similar rules of GLI activity apply to SHH signaling in regulating SVZ-derived olfactory bulb interneurons and maintaining cortical astrocyte function. Namely, fewer superficial olfactory bulb interneurons are generated in the absence of Gli2 and Gli3, whereas astrocyte partial gliosis results from an increase in GLI3(R). Thus precise titration of GLI(R) levels by SHH is critical to multiple functions of adult NSCs and astrocytes.


Assuntos
Astrócitos/fisiologia , Proteínas Hedgehog/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/fisiologia , Transdução de Sinais/genética , Animais , Astrócitos/patologia , Feminino , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Bulbo Olfatório/citologia , Bulbo Olfatório/patologia , Bulbo Olfatório/fisiologia , Proteína Gli3 com Dedos de Zinco
16.
Am J Respir Cell Mol Biol ; 48(6): 703-10, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23371063

RESUMO

Sonic Hedgehog (Shh) signaling is essential during embryonic lung development, but its role in postnatal lung development and adult lung are not known. Using Gli1(nlacZ) reporter mice to identify cells with active Hh signaling, we found that Gli1(nlacZ)-positive mesenchymal cells are densely and diffusely present up to 2 weeks after birth and decline in number thereafter. In adult mice, Gli1(nlacZ)-positive cells are present around large airways and vessels and are sparse in alveolar septa. Hh-stimulated cells are mostly fibroblasts; only 10% of Gli1(nlacZ)-positive cells are smooth muscle cells, and most smooth muscle cells do not have activation of Hh signaling. To assess its functional relevance, we influenced Hh signaling in the developing postnatal lung and adult injured lung. Inhibition of Hh signaling during early postnatal lung development causes airspace enlargement without diminished alveolar septation. After bleomycin injury in the adult lung, there are abundant Gli1(nlacZ)-positive mesenchymal cells in fibrotic lesions and increased numbers of Gli1(nlacZ)-positive cells in preserved alveolar septa. Inhibition of Hh signaling with an antibody against all Hedgehog isoforms does not reduce bleomycin-induced fibrosis, but adenovirus-mediated overexpression of Shh increases collagen production in this model. Our data provide strong evidence that Hh signaling can regulate lung stromal cell function in two critical scenarios: normal development in postnatal lung and lung fibrosis in adult lung.


Assuntos
Bleomicina/efeitos adversos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Pulmão/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Fatores Etários , Alelos , Animais , Animais Recém-Nascidos , Contagem de Células , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Proteínas Hedgehog/genética , Imuno-Histoquímica , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Transdução de Sinais , Proteína GLI1 em Dedos de Zinco
17.
Cell Rep ; 2(2): 386-96, 2012 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-22884371

RESUMO

Mosaic mutant analysis, the study of cellular defects in scattered mutant cells in a wild-type environment, is a powerful approach for identifying critical functions of genes and has been applied extensively to invertebrate model organisms. A highly versatile technique has been developed in mouse: MASTR (mosaic mutant analysis with spatial and temporal control of recombination), which utilizes the increasing number of floxed alleles and simultaneously combines conditional gene mutagenesis and cell marking for fate analysis. A targeted allele (R26(MASTR)) was engineered; the allele expresses a GFPcre fusion protein following FLP-mediated recombination, which serves the dual function of deleting floxed alleles and marking mutant cells with GFP. Within 24 hr of tamoxifen administration to R26(MASTR) mice carrying an inducible FlpoER transgene and a floxed allele, nearly all GFP-expressing cells have a mutant allele. The fate of single cells lacking FGF8 or SHH signaling in the developing hindbrain was analyzed using MASTR, and it was revealed that there is only a short time window when neural progenitors require FGFR1 for viability and that granule cell precursors differentiate rapidly when SMO is lost. MASTR is a powerful tool that provides cell-type-specific (spatial) and temporal marking of mosaic mutant cells and is broadly applicable to developmental, cancer, and adult stem cell studies.


Assuntos
Alelos , Transtornos Cromossômicos/genética , Engenharia Genética/métodos , Mutação , Recombinação Genética , Animais , Análise Mutacional de DNA/métodos , Camundongos , Camundongos Transgênicos , Mosaicismo
18.
Dev Biol ; 370(1): 110-24, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22841643

RESUMO

Anterior-posterior (AP) limb patterning is directed by sonic hedgehog (SHH) signaling from the posteriorly located zone of polarizing activity (ZPA). GLI3 and GLI2 are the transcriptional mediators generally utilized in SHH signaling, and each can function as an activator (A) and repressor (R). Although GLI3R has been suggested to be the primary effector of SHH signaling during limb AP patterning, a role for GLI3A or GLI2 has not been fully ruled out, nor has it been determined whether Gli3 plays distinct roles in limb development at different stages. By conditionally removing Gli3 in the limb at multiple different time points, we uncovered four Gli3-mediated functions in limb development that occur at distinct but partially over-lapping time windows: AP patterning of the proximal limb, AP patterning of the distal limb, regulation of digit number and bone differentiation. Furthermore, by removing Gli2 in Gli3 temporal conditional knock-outs, we uncovered an essential role for Gli2 in providing the remaining posterior limb patterning seen in Gli3 single mutants. To test whether GLIAs or GLIRs regulate different aspects of AP limb patterning and/or digit number, we utilized a knock-in allele in which GLI1, which functions solely as an activator, is expressed in place of the bifunctional GLI2 protein. Interestingly, we found that GLIAs contribute to AP patterning specifically in the posterior limb, whereas GLIRs predominantly regulate anterior patterning and digit number. Since GLI3 is a more effective repressor, our results explain why GLI3 is required only for anterior limb patterning and why GLI2 can compensate for GLI3A in posterior limb patterning. Taken together, our data suggest that establishment of a complete range of AP positional identities in the limb requires integration of the spatial distribution, timing, and dosage of GLI2 and GLI3 activators and repressors.


Assuntos
Padronização Corporal/fisiologia , Extremidades/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Primers do DNA/genética , Extremidades/anatomia & histologia , Proteínas Hedgehog/metabolismo , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Transdução de Sinais/genética , Tamoxifeno , Proteína Gli2 com Dedos de Zinco , Proteína Gli3 com Dedos de Zinco , beta-Galactosidase/metabolismo
19.
Neural Dev ; 6: 29, 2011 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-21689430

RESUMO

BACKGROUND: The ventral midbrain contains a diverse array of neurons, including dopaminergic neurons of the ventral tegmental area (VTA) and substantia nigra (SN) and neurons of the red nucleus (RN). Dopaminergic and RN neurons have been shown to arise from ventral mesencephalic precursors that express Sonic Hedgehog (Shh). However, Shh expression, which is initially confined to the mesencephalic ventral midline, expands laterally and is then downregulated in the ventral midline. In contrast, expression of the Hedgehog target gene Gli1 initiates in the ventral midline prior to Shh expression, but after the onset of Shh expression it is expressed in precursors lateral to Shh-positive cells. Given these dynamic gene expression patterns, Shh and Gli1 expression could delineate different progenitor populations at distinct embryonic time points. RESULTS: We employed genetic inducible fate mapping (GIFM) to investigate whether precursors that express Shh (Shh-GIFM) or transduce Shh signaling (Gli1-GIFM) at different time points give rise to different ventral midbrain cell types. We find that precursors restricted to the ventral midline are labeled at embryonic day (E)7.5 with Gli1-GIFM, and with Shh-GIFM at E8.5. These precursors give rise to all subtypes of midbrain dopaminergic neurons and the anterior RN. A broader domain of progenitors that includes the ventral midline is marked with Gli1-GIFM at E8.5 and with Shh-GIFM at E9.5; these fate-mapped cells also contribute to all midbrain dopaminergic subtypes and to the entire RN. In contrast, a lateral progenitor domain that is labeled with Gli1-GIFM at E9.5 and with Shh-GIFM at E11.5 has a markedly reduced potential to give rise to the RN and to SN dopaminergic neurons, and preferentially gives rise to the ventral-medial VTA. In addition, cells derived from Shh- and Gli1-expressing progenitors located outside of the ventral midline give rise to astrocytes. CONCLUSIONS: We define a ventral midbrain precursor map based on the timing of Gli1 and Shh expression, and suggest that the diversity of midbrain dopaminergic neurons is at least partially determined during their precursor stage when their medial-lateral position, differential gene expression and the time when they leave the ventricular zone influence their fate decisions.


Assuntos
Proteínas Hedgehog/genética , Proteínas Hedgehog/fisiologia , Mesencéfalo/fisiologia , Células-Tronco Neurais/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Animais , Astrócitos/fisiologia , Mapeamento Encefálico , Diferenciação Celular/genética , Dopamina/fisiologia , Feminino , Imunofluorescência , Hibridização In Situ , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/fisiologia , Mesencéfalo/citologia , Mesencéfalo/embriologia , Camundongos , Neurônios/fisiologia , Nervo Oculomotor/embriologia , Nervo Oculomotor/crescimento & desenvolvimento , Gravidez , RNA/biossíntese , RNA/genética , Núcleo Rubro/citologia , Núcleo Rubro/embriologia , Núcleo Rubro/fisiologia , Substância Negra/embriologia , Substância Negra/crescimento & desenvolvimento , Substância Negra/fisiologia , Proteína GLI1 em Dedos de Zinco
20.
Cell Stem Cell ; 8(5): 552-65, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21549329

RESUMO

In adult skin, stem cells in the hair follicle bulge cyclically regenerate the follicle, whereas a distinct stem cell population maintains the epidermis. The degree to which all bulge cells have equal regenerative potential is not known. We found that Sonic hedgehog (Shh) from neurons signals to a population of cells in the telogen bulge marked by the Hedgehog response gene Gli1. Gli1-expressing bulge cells function as multipotent stem cells in their native environment and repeatedly regenerate the anagen follicle. Shh-responding perineural bulge cells incorporate into healing skin wounds where, notably, they can change their lineage into epidermal stem cells. The perineural niche (including Shh) is dispensable for follicle contributions to acute wound healing and skin homeostasis, but is necessary to maintain bulge cells capable of becoming epidermal stem cells. Thus, nerves cultivate a microenvironment where Shh creates a molecularly and phenotypically distinct population of hair follicle stem cells.


Assuntos
Células-Tronco Adultas/metabolismo , Folículo Piloso/fisiologia , Proteínas Hedgehog/metabolismo , Células-Tronco Multipotentes/metabolismo , Neurônios/fisiologia , Células-Tronco Adultas/citologia , Animais , Animais Geneticamente Modificados , Linhagem da Célula , Sobrevivência Celular , Transdiferenciação Celular , Células Cultivadas , Epiderme/lesões , Epiderme/fisiologia , Regulação da Expressão Gênica , Técnicas de Introdução de Genes , Folículo Piloso/citologia , Camundongos , Células-Tronco Multipotentes/citologia , Neurônios/citologia , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Regeneração , Nicho de Células-Tronco , Transativadores/genética , Transativadores/metabolismo , Transgenes/genética , Cicatrização , Proteína GLI1 em Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA