Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Death Dis ; 15(2): 174, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409173

RESUMO

miR-184-knockout mice display perturbed epidermal stem cell differentiation. However, the potential role of miR-184 in skin pathology is unclear. Here, we report that miR-184 controls epidermal stem cell dynamics and that miR-184 ablation enhances skin carcinogenesis in mice. In agreement, repression of miR-184 in human squamous cell carcinoma (SCC) enhances neoplastic hallmarks of human SCC cells in vitro and tumor development in vivo. Characterization of miR-184-regulatory network, suggests that miR-184 inhibits pro-oncogenic pathways, cell proliferation, and epithelial to mesenchymal transformation. Of note, depletion of miR-184 enhances the levels of ß-catenin under homeostasis and following experimental skin carcinogenesis. Finally, the repression of ß-catenin by miR-184, inhibits the neoplastic phenotype of SCC cells. Taken together, miR-184 behaves as an epidermal tumor suppressor, and may provide a potentially useful target for skin SCC therapy.


Assuntos
Carcinoma de Células Escamosas , MicroRNAs , Neoplasias Cutâneas , Animais , Humanos , Camundongos , beta Catenina/genética , beta Catenina/metabolismo , Carcinogênese/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia
2.
Stem Cell Reports ; 18(12): 2313-2327, 2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-38039972

RESUMO

Recently, the murine cornea has reemerged as a robust stem cell (SC) model, allowing individual SC tracing in living animals. The cornea has pioneered seminal discoveries in SC biology and regenerative medicine, from the first corneal transplantation in 1905 to the identification of limbal SCs and their transplantation to successfully restore vision in the early 1990s. Recent experiments have exposed unexpected properties attributed to SCs and progenitors and revealed flexibility in the differentiation program and a key role for the SC niche. Here, we discuss the limbal SC model and its broader relevance to other tissues, disease, and therapy.


Assuntos
Epitélio Corneano , Limbo da Córnea , Camundongos , Animais , Córnea , Células-Tronco , Diferenciação Celular , Transplante de Células-Tronco
3.
Cell Death Differ ; 30(6): 1601-1614, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37095157

RESUMO

The cell fate decisions of stem cells (SCs) largely depend on signals from their microenvironment (niche). However, very little is known about how biochemical niche cues control cell behavior in vivo. To address this question, we focused on the corneal epithelial SC model in which the SC niche, known as the limbus, is spatially segregated from the differentiation compartment. We report that the unique biomechanical property of the limbus supports the nuclear localization and function of Yes-associated protein (YAP), a putative mediator of the mechanotransduction pathway. Perturbation of tissue stiffness or YAP activity affects SC function as well as tissue integrity under homeostasis and significantly inhibited the regeneration of the SC population following SC depletion. In vitro experiments revealed that substrates with the rigidity of the corneal differentiation compartment inhibit nuclear YAP localization and induce differentiation, a mechanism that is mediated by the TGFß-SMAD2/3 pathway. Taken together, these results indicate that SC sense biomechanical niche signals and that manipulation of mechano-sensory machinery or its downstream biochemical output may bear fruits in SC expansion for regenerative therapy.


Assuntos
Epitélio Corneano , Limbo da Córnea , Proteínas de Sinalização YAP , Diferenciação Celular , Epitélio Corneano/metabolismo , Mecanotransdução Celular , Nicho de Células-Tronco , Células-Tronco/metabolismo , Humanos , Proteínas de Sinalização YAP/metabolismo
4.
Cell Stem Cell ; 28(7): 1248-1261.e8, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33984282

RESUMO

The accessibility and transparency of the cornea permit robust stem cell labeling and in vivo cell fate mapping. Limbal epithelial stem cells (LSCs) that renew the cornea are traditionally viewed as rare, slow-cycling cells that follow deterministic rules dictating their self-renewal or differentiation. Here, we combined single-cell RNA sequencing and advanced quantitative lineage tracing for in-depth analysis of the murine limbal epithelium. These analysis revealed the co-existence of two LSC populations localized in separate and well-defined sub-compartments, termed the "outer" and "inner" limbus. The primitive population of quiescent outer LSCs participates in wound healing and boundary formation, and these cells are regulated by T cells, which serve as a niche. In contrast, the inner peri-corneal limbus hosts active LSCs that maintain corneal epithelial homeostasis. Quantitative analyses suggest that LSC populations are abundant, following stochastic rules and neutral drift dynamics. Together these results demonstrate that discrete LSC populations mediate corneal homeostasis and regeneration.


Assuntos
Limbo da Córnea , Células-Tronco , Animais , Córnea , Homeostase , Camundongos , Cicatrização
5.
Stem Cells ; 36(11): 1723-1735, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30157305

RESUMO

One of the main challenges in limbal stem cell (LSC) biology and transplantation is the lack of definitive cell surface markers which can be used to identify and enrich viable LSCs. In this study, expression of 361 cell surface proteins was assessed in ex vivo expanded limbal epithelial cells. One marker, CD200 was selected for further characterization based on expression in a small subset of limbal epithelial cells (2.25% ± 0.69%) and reduced expression through consecutive passaging and calcium induced differentiation. CD200 was localized to a small population of cells at the basal layer of the human and mouse limbal epithelium. CD200+ cells were slow cycling and contained the majority of side population (SP) and all the holoclone forming progenitors. CD200+ cells displayed higher expression of LSCs markers including PAX6, WNT7A, CDH3, CK14, CK15, and ABCB5 and lower expression of Ki67 when compared to CD200- . Downregulation of CD200 abrogated the ability of limbal epithelial cells to form holoclones, suggesting an important function for CD200 in the maintenance and/or self-renewal of LSCs. A second marker, CD109, which was expressed in 56.29% ± 13.96% of limbal epithelial cells, was also found to co-localize with ΔNp63 in both human and mouse cornea, albeit more abundantly than CD200. CD109 expression decreased slowly through calcium induced cell differentiation and CD109+ cells were characterized by higher expression of Ki67, when compared to CD109- subpopulation. Together our data suggest that CD200 expression marks a quiescent population of LSCs with holoclone forming potential, while CD109 expression is associated with a proliferative progenitor phenotype. Stem Cells 2018;36:1723-1735.


Assuntos
Antígenos CD/metabolismo , Células Epiteliais/metabolismo , Limbo da Córnea/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Células Epiteliais/citologia , Feminino , Humanos , Limbo da Córnea/citologia , Masculino , Pessoa de Meia-Idade
6.
Cell Rep ; 22(2): 323-331, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29320729

RESUMO

During morphogenesis, preserving tissue boundaries is essential for cell fate regulation. While embryonic tissues possess high plasticity and repair ability, the questions of whether and how adult tissues cope with acute stem cell (SC) loss or boundary disruption have remained unanswered. Here, we report that K15-GFP transgene labels the murine corneal epithelial boundary and SC niche known as the limbus. K15-GFP+ basal cells expressed SC markers and were located at the corneal regeneration site, as evident by lineage tracing. Remarkably, following surgical deletion of the SC pool, corneal-committed cells dedifferentiated into bona fide limbal SCs that retained normal tissue dynamics and marker expression. Interestingly, however, damage to the limbal stromal niche abolished K15-GFP recovery and led to pathological wound healing. Altogether, this study indicates that committed corneal cells possess plasticity to dedifferentiate, repopulate the SC pool, and correctly re-form the tissue boundary in the presence of intact stroma.


Assuntos
Córnea/metabolismo , Nicho de Células-Tronco/genética , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos , Camundongos Transgênicos
7.
Stem Cell Reports ; 9(6): 1991-2004, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29198823

RESUMO

miR-184 is a highly evolutionary conserved microRNA (miRNA) from fly to human. The importance of miR-184 was underscored by the discovery that point mutations in miR-184 gene led to corneal/lens blinding disease. However, miR-184-related function in vivo remained unclear. Here, we report that the miR-184 knockout mouse model displayed increased p63 expression in line with epidermal hyperplasia, while forced expression of miR-184 by stem/progenitor cells enhanced the Notch pathway and induced epidermal hypoplasia. In line, miR-184 reduced clonogenicity and accelerated differentiation of human epidermal cells. We showed that by directly repressing cytokeratin 15 (K15) and FIH1, miR-184 induces Notch activation and epidermal differentiation. The disease-causing miR-184C57U mutant failed to repress K15 and FIH1 and to induce Notch activation, suggesting a loss-of-function mechanism. Altogether, we propose that, by targeting K15 and FIH1, miR-184 regulates the transition from proliferation to early differentiation, while mis-expression or mutation in miR-184 results in impaired homeostasis.


Assuntos
Cegueira/genética , Diferenciação Celular/genética , Epiderme/crescimento & desenvolvimento , MicroRNAs/genética , Animais , Cegueira/patologia , Proliferação de Células/genética , Epiderme/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Queratina-15/genética , Camundongos , Camundongos Knockout , Oxigenases de Função Mista/genética , Fosfoproteínas/genética , Receptores Notch/genética , Transdução de Sinais/genética , Células-Tronco/metabolismo , Transativadores/genética
8.
J Vis Exp ; (106): e53370, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26709460

RESUMO

Lineage tracing experiments define the origin, fate and behavior of cells in a specific tissue or organism. This technique has been successfully applied for many decades, revealing seminal findings in developmental biology. More recently, it was adopted by stem cell biologists to identify and track different stem cell populations with minimal experimental intervention. The recent developments in mouse genetics, the availability of a large number of mouse strains, and the advancements in fluorescent microscopy allow the straightforward design of powerful lineage tracing systems for various tissues with basic expertise, using commercially available tools. We have recently taken advantage of this powerful methodology to explore the origin and fate of stem cells at the ocular surface using R26R-Confetti mouse. This model offers a multi-color genetic system, for the expression of 4 fluorescent genes in a random manner. Here we describe the principles of this methodology and provide an adaptable protocol for designing lineage tracing experiments; specifically for the corneal epithelium as well as for other tissues.


Assuntos
Córnea/citologia , Proteínas Luminescentes/análise , Células-Tronco/citologia , Alelos , Animais , Linhagem da Célula , Genes Reporter , Integrases/biossíntese , Integrases/genética , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Microscopia Confocal/métodos , Espectrometria de Fluorescência/métodos
9.
Mol Cell ; 59(4): 664-76, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26236014

RESUMO

The most critical stage in initiation of melanoma metastasis is the radial to vertical growth transition, yet the triggers of this transition remain elusive. We suggest that the microenvironment drives melanoma metastasis independently of mutation acquisition. Here we examined the changes in microenvironment that occur during melanoma radial growth. We show that direct contact of melanoma cells with the remote epidermal layer triggers vertical invasion via Notch signaling activation, the latter serving to inhibit MITF function. Briefly, within the native Notch ligand-free microenvironment, MITF, the melanocyte lineage master regulator, binds and represses miR-222/221 promoter in an RBPJK-dependent manner. However, when radial growth brings melanoma cells into contact with distal differentiated keratinocytes that express Notch ligands, the activated Notch intracellular domain impairs MITF binding to miR-222/221 promoter. This de-repression of miR-222/221 expression triggers initiation of invasion. Our findings may direct melanoma prevention opportunities via targeting specific microenvironments.


Assuntos
Queratinócitos/fisiologia , Melanoma Experimental/secundário , Fator de Transcrição Associado à Microftalmia/metabolismo , Neoplasias Cutâneas/patologia , Animais , Sequência de Bases , Sítios de Ligação , Comunicação Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , MicroRNAs/genética , MicroRNAs/metabolismo , Invasividade Neoplásica , Transplante de Neoplasias , Regiões Promotoras Genéticas , Interferência de RNA , Receptores Notch/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo
10.
Stem Cells ; 33(1): 230-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25187087

RESUMO

Accumulating evidence supports the dogma that the corneal epithelium is regenerated by stem cells located exclusively in the limbal niche, at the corneal periphery. Accordingly, limbal stem cells (LSCs) give rise to progenitors that proliferate and migrate centripetally to repopulate the corneal epithelium, which has a short turnover. Moreover, LSC loss leads to corneal opacity and blindness, while limbal grafting restores patients' vision. However, contradicting data suggested that the limbus does not participate in corneal homeostasis and that the cornea contains stem cells. As of today, only indirect evidence for limbal cell migration under homeostasis or injury has been demonstrated. Here, we performed lineage tracing experiments using R26R-Confetti mice to follow K14+ limbal/corneal epithelial cells stochastically induced to express one out of four fluorescent genes. In homeostasis, radial limbal stripes of slow migrating cells proceeded toward the corneal center while, infrequently, slow cycling limbal clones resembling quiescent stem cells were observed. Additionally, rare corneal clones that did not migrate centripetally, but survived for over 4 months, were inspected. In contrast to limbal stripes, corneal clusters had minor contribution to tissue replenishment in homeostasis. Corneal cells, however, significantly contributed to mild wound repair while large limbal streaks appeared within a week following severe wounding that coincided with partial loss of corneal transparency. This data suggest that the mouse limbus largely contributes to corneal renewal while corneal progenitor cells have a long turnover and, therefore, may be able to maintain the corneal epithelium for several months.


Assuntos
Epitélio Corneano/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Epitélio Corneano/metabolismo , Camundongos , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA