Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Nat Commun ; 14(1): 8097, 2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-38062045

RESUMO

Innervation of the hypothalamic median eminence by Gonadotropin-Releasing Hormone (GnRH) neurons is vital to ensure puberty onset and successful reproduction. However, the molecular and cellular mechanisms underlying median eminence development and pubertal timing are incompletely understood. Here we show that Semaphorin-6A is strongly expressed by median eminence-resident oligodendrocytes positioned adjacent to GnRH neuron projections and fenestrated capillaries, and that Semaphorin-6A is required for GnRH neuron innervation and puberty onset. In vitro and in vivo experiments reveal an unexpected function for Semaphorin-6A, via its receptor Plexin-A2, in the control of median eminence vascular permeability to maintain neuroendocrine homeostasis. To support the significance of these findings in humans, we identify patients with delayed puberty carrying a novel pathogenic variant of SEMA6A. In all, our data reveal a role for Semaphorin-6A in regulating GnRH neuron patterning by tuning the median eminence vascular barrier and thereby controlling puberty onset.


Assuntos
Hormônio Liberador de Gonadotropina , Semaforinas , Humanos , Hormônio Liberador de Gonadotropina/metabolismo , Eminência Mediana/metabolismo , Permeabilidade Capilar , Neurônios/metabolismo , Puberdade , Semaforinas/genética , Semaforinas/metabolismo
3.
Sci Rep ; 12(1): 2341, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149717

RESUMO

The growth of glioblastoma (GBM), one of the deadliest adult cancers, is fuelled by a subpopulation of stem/progenitor cells, which are thought to be the source of resistance and relapse after treatment. Re-engagement of a latent capacity of these cells to re-enter a trajectory resulting in cell differentiation is a potential new therapeutic approach for this devastating disease. ASCL1, a proneural transcription factor, plays a key role in normal brain development and is also expressed in a subset of GBM cells, but fails to engage a full differentiation programme in this context. Here, we investigated the barriers to ASCL1-driven differentiation in GBM stem cells. We see that ASCL1 is highly phosphorylated in GBM stem cells where its expression is compatible with cell proliferation. However, overexpression of a form of ASCL1 that cannot be phosphorylated on Serine-Proline sites drives GBM cells down a neuronal lineage and out of cell cycle more efficiently than its wild-type counterpart, an effect further enhanced by deletion of the inhibitor of differentiation ID2, indicating mechanisms to reverse the block to GBM cell differentiation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatologia , Glioblastoma/metabolismo , Glioblastoma/fisiopatologia , Proteína 2 Inibidora de Diferenciação/genética , Células-Tronco Neoplásicas/metabolismo , Motivos de Aminoácidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Neoplasias Encefálicas/genética , Ciclo Celular , Diferenciação Celular , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Humanos , Proteína 2 Inibidora de Diferenciação/metabolismo , Células-Tronco Neoplásicas/citologia , Fosforilação
4.
Nature ; 594(7863): 442-447, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34079126

RESUMO

Interactions between tumour cells and the surrounding microenvironment contribute to tumour progression, metastasis and recurrence1-3. Although mosaic analyses in Drosophila have advanced our understanding of such interactions4,5, it has been difficult to engineer parallel approaches in vertebrates. Here we present an oncogene-associated, multicolour reporter mouse model-the Red2Onco system-that allows differential tracing of mutant and wild-type cells in the same tissue. By applying this system to the small intestine, we show that oncogene-expressing mutant crypts alter the cellular organization of neighbouring wild-type crypts, thereby driving accelerated clonal drift. Crypts that express oncogenic KRAS or PI3K secrete BMP ligands that suppress local stem cell activity, while changes in PDGFRloCD81+ stromal cells induced by crypts with oncogenic PI3K alter the WNT signalling environment. Together, these results show how oncogene-driven paracrine remodelling creates a niche environment that is detrimental to the maintenance of wild-type tissue, promoting field transformation dominated by oncogenic clones.


Assuntos
Neoplasias Colorretais/patologia , Intestino Delgado/patologia , Células-Tronco Neoplásicas/patologia , Oncogenes , Nicho de Células-Tronco , Animais , Células Clonais/patologia , Neoplasias Colorretais/genética , Feminino , Intestino Delgado/metabolismo , Masculino , Camundongos , Mutação , Células-Tronco Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Reprodutibilidade dos Testes , Análise de Célula Única , Nicho de Células-Tronco/genética , Microambiente Tumoral , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt
5.
Biol Open ; 10(2)2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33619017

RESUMO

Emerging three-dimensional (3D) cultures of glioblastoma are becoming powerful models to study glioblastoma stem cell behavior and the impact of cell-cell and cell-microenvironment interactions on tumor growth and invasion. Here we describe a method for culturing human glioblastoma stem cells (GSCs) in 3D by co-culturing them with pluripotent stem cell-derived brain organoids. This requires multiple coordinated steps, including the generation of cerebral organoids, and the growth and fluorescence tagging of GSCs. We highlight how to recognize optimal organoid generation and how to efficiently mark GSCs, before describing optimized co-culture conditions. We show that GSCs can efficiently integrate into brain organoids and maintain a significant degree of cell fate heterogeneity, paving the way for the analysis of GSC fate behavior and lineage progression. These results establish the 3D culture system as a viable and versatile GBM model for investigating tumor cell biology and GSC heterogeneity.This article has an associated First Person interview with the first author of the paper.


Assuntos
Neoplasias Encefálicas/patologia , Técnicas de Cocultura , Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Organoides , Biomarcadores , Diferenciação Celular , Linhagem Celular Tumoral , Linhagem da Célula/genética , Humanos
6.
Small GTPases ; 12(5-6): 336-357, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33054516

RESUMO

Rnd proteins constitute a subfamily of Rho GTPases represented in mammals by Rnd1, Rnd2 and Rnd3. Despite their GTPase structure, their specific feature is the inability to hydrolyse GTP-bound nucleotide. This aspect makes them atypical among Rho GTPases. Rnds are regulated for their expression at the transcriptional or post-transcriptional levels and they are activated through post-translational modifications and interactions with other proteins. Rnd proteins are mainly involved in the regulation of the actin cytoskeleton and cell proliferation. Whereas Rnd3 is ubiquitously expressed, Rnd1 and 2 are tissue-specific. Increasing data has described their important role during development and diseases. Herein, we describe their involvement in physiological and pathological conditions with a focus on the neuronal and vascular systems, and summarize their implications in tumorigenesis.


Assuntos
Neoplasias/fisiopatologia , Doenças do Sistema Nervoso/fisiopatologia , Doenças Vasculares/fisiopatologia , Proteínas rho de Ligação ao GTP/metabolismo , Humanos , Neoplasias/enzimologia , Doenças do Sistema Nervoso/enzimologia , Doenças Vasculares/enzimologia
7.
Front Cell Dev Biol ; 8: 549533, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072742

RESUMO

Mex3A is an RNA binding protein that can also act as an E3 ubiquitin ligase to control gene expression at the post-transcriptional level. In intestinal adult stem cells, MEX3A is required for cell self-renewal and when overexpressed, MEX3A can contribute to support the proliferation of different cancer cell types. In a completely different context, we found mex3A among the genes expressed in neurogenic niches of the embryonic and adult fish brain and, notably, its expression was downregulated during brain aging. The role of mex3A during embryonic and adult neurogenesis in tetrapods is still unknown. Here, we showed that mex3A is expressed in the proliferative region of the developing brain in both Xenopus and mouse embryos. Using gain and loss of gene function approaches, we showed that, in Xenopus embryos, mex3A is required for neuroblast proliferation and its depletion reduced the neuroblast pool, leading to microcephaly. The tissue-specific overexpression of mex3A in the developing neural plate enhanced the expression of sox2 and msi-1 keeping neuroblasts into a proliferative state. It is now clear that the stemness property of mex3A, already demonstrated in adult intestinal stem cells and cancer cells, is a key feature of mex3a also in developing brain, opening new lines of investigation to better understand its role during brain aging and brain cancer development.

8.
Front Cell Dev Biol ; 8: 220, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32373607

RESUMO

Glioblastoma represents an aggressive form of brain cancer characterized by poor prognosis and a 5-year survival rate of only 3-7%. Despite remarkable advances in brain tumor research in the past decades, very little has changed for patients, due in part to the recurrent nature of the disease and to the lack of suitable models to perform genotype-phenotype association studies and personalized drug screening. In vitro culture of cancer cells derived from patient biopsies has been fundamental in understanding tumor biology and for testing the effect of various drugs. These cultures emphasize the role of in vitro cancer stem cells (CSCs), which fuel tumor growth and are thought to be the cause of relapse after treatment. However, it has become clear over the years that a 2D monolayer culture of these CSCs has certain disadvantages, including the lack of heterogeneous cell-cell and cell-environment interactions, which can now be partially overcome by the introduction of 3D organoid cultures. This is a novel and expanding field of research and in this review, I describe the emerging 3D models of glioblastoma. I also discuss their potential to advance our knowledge of tumor biology and CSC heterogeneity, while debating their current limitations.

9.
Cell Stem Cell ; 23(3): 436-443.e7, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30100168

RESUMO

The intestinal epithelium is largely maintained by self-renewing stem cells but with apparently committed progenitors also contributing, particularly following tissue damage. However, the mechanism of, and requirement for, progenitor plasticity in mediating pathological response remain unknown. Here we show that phosphorylation of the transcription factor Atoh1 is required for both the contribution of secretory progenitors to the stem cell pool and for a robust regenerative response. As confirmed by lineage tracing, Atoh1+ cells (Atoh1(WT)CreERT2 mice) give rise to multilineage intestinal clones both in the steady state and after tissue damage. In a phosphomutant Atoh1(9S/T-A)CreERT2 line, preventing phosphorylation of ATOH1 protein acts to promote secretory differentiation and inhibit the contribution of progenitors to self-renewal. Following chemical colitis, Atoh1+ cells of Atoh1(9S/T-A)CreERT2 mice have reduced clonogenicity that affects overall regeneration. Progenitor plasticity maintains robust self-renewal in the intestinal epithelium, and the balance between stem and progenitor fate is directly coordinated by ATOH1 multisite phosphorylation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Mucosa Intestinal/metabolismo , Regeneração , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação
10.
Development ; 145(10)2018 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-29759978

RESUMO

The development of the nervous system relies on the coordinated regulation of stem cell self-renewal and differentiation. The discovery that brain tumours contain a subpopulation of cells with stem/progenitor characteristics that are capable of sustaining tumour growth has emphasized the importance of understanding the cellular dynamics and the molecular pathways regulating neural stem cell behaviour. By focusing on recent work on glioma and medulloblastoma, we review how lineage tracing contributed to dissecting the embryonic origin of brain tumours and how lineage-specific mechanisms that regulate stem cell behaviour in the embryo may be subverted in cancer to achieve uncontrolled proliferation and suppression of differentiation.


Assuntos
Neoplasias Encefálicas/patologia , Encéfalo/embriologia , Autorrenovação Celular/fisiologia , Glioma/patologia , Meduloblastoma/patologia , Células-Tronco Neoplásicas/citologia , Células-Tronco Neurais/citologia , Encéfalo/citologia , Encéfalo/patologia , Neoplasias Encefálicas/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Transformação Celular Neoplásica/genética , Glioma/genética , Humanos , Meduloblastoma/genética , Transdução de Sinais/fisiologia
11.
Cell Chem Biol ; 24(8): 1017-1028.e7, 2017 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-28807782

RESUMO

Mutations activating KRAS underlie many forms of cancer, but are refractory to therapeutic targeting. Here, we develop Poloppin, an inhibitor of protein-protein interactions via the Polo-box domain (PBD) of the mitotic Polo-like kinases (PLKs), in monotherapeutic and combination strategies to target mutant KRAS. Poloppin engages its targets in biochemical and cellular assays, triggering mitotic arrest with defective chromosome congression. Poloppin kills cells expressing mutant KRAS, selectively enhancing death in mitosis. PLK1 or PLK4 depletion recapitulates these cellular effects, as does PBD overexpression, corroborating Poloppin's mechanism of action. An optimized analog with favorable pharmacokinetics, Poloppin-II, is effective against KRAS-expressing cancer xenografts. Poloppin resistance develops less readily than to an ATP-competitive PLK1 inhibitor; moreover, cross-sensitivity persists. Poloppin sensitizes mutant KRAS-expressing cells to clinical inhibitors of c-MET, opening opportunities for combination therapy. Our findings exemplify the utility of small molecules modulating the protein-protein interactions of PLKs to therapeutically target mutant KRAS-expressing cancers.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mutação , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/química , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Mitose , Estrutura Molecular , Ligação Proteica , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/química , Relação Estrutura-Atividade , Especificidade por Substrato , Quinase 1 Polo-Like
12.
Nat Commun ; 5: 3405, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24572910

RESUMO

A transcriptional programme initiated by the proneural factors Neurog2 and Ascl1 controls successive steps of neurogenesis in the embryonic cerebral cortex. Previous work has shown that proneural factors also confer a migratory behaviour to cortical neurons by inducing the expression of the small GTP-binding proteins such as Rnd2 and Rnd3. However, the directionality of radial migration suggests that migrating neurons also respond to extracellular signal-regulated pathways. Here we show that the Plexin B2 receptor interacts physically and functionally with Rnd3 and stimulates RhoA activity in migrating cortical neurons. Plexin B2 competes with p190RhoGAP for binding to Rnd3, thus blocking the Rnd3-mediated inhibition of RhoA and also recruits RhoGEFs to directly stimulate RhoA activity. Thus, an interaction between the cell-extrinsic Plexin signalling pathway and the cell-intrinsic Ascl1-Rnd3 pathway determines the level of RhoA activity appropriate for cortical neuron migration.


Assuntos
Movimento Celular , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Western Blotting , Células COS , Linhagem Celular Tumoral , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Chlorocebus aethiops , Transferência Ressonante de Energia de Fluorescência , Hibridização In Situ , Camundongos , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Ligação Proteica , Interferência de RNA , Proteínas rho de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/genética
13.
Nat Commun ; 4: 1635, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23535656

RESUMO

The generation of neurons by neural stem cells is a highly choreographed process that requires extensive and dynamic remodelling of the cytoskeleton at each step of the process. The atypical RhoGTPase Rnd3 is expressed by progenitors in the embryonic brain but its role in early steps of neurogenesis has not been addressed. Here we show that silencing Rnd3 in the embryonic cerebral cortex interferes with the interkinetic nuclear migration of radial glial stem cells, disrupts their apical attachment and modifies the orientation of their cleavage plane. These defects are rescued by co-expression of a constitutively active form of cofilin, demonstrating that Rnd3-mediated disassembly of actin filaments coordinates the cellular behaviour of radial glial. Rnd3 also limits the divisions of basal progenitors via a distinct mechanism involving the suppression of cyclin D1 translation. Interestingly, although Rnd3 expression is controlled transcriptionally by Ascl1, this proneural factor is itself required in radial glial progenitors only for proper orientation of cell divisions.


Assuntos
Actinas/fisiologia , Neurogênese/fisiologia , Proteínas rho de Ligação ao GTP/fisiologia , Animais , Proliferação de Células , Ciclina D1/fisiologia , Camundongos , Células-Tronco/citologia , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA