Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res ; 79: 103455, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38896969

RESUMO

Calcium indicators are sensitive tools to image neural activity. However, their use in human induced pluripotent stem cells (iPSC)-derived neurons is limited by silencing of the transgene. We generated the iPSC line MSE2336A carrying heterozygous insertion in the safe-harbor locus AAVS1 of the ultrasensitive protein calcium sensor (GCaMP6) under the control of CAG promoter and UCOE to maintain robust transgene expression in differentiated cells. The iPSC exhibited normal cell morphology, expression of pluripotency markers, genome integrity, and the ability to differentiate into the three primary germ layers. This line provides a powerful model to study activity in human neurons.

2.
Nat Cell Biol ; 25(2): 222-234, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36717629

RESUMO

Substantial follicle remodelling during the regression phase of the hair growth cycle is coordinated by the contraction of the dermal sheath smooth muscle, but how dermal-sheath-generated forces are regulated is unclear. Here, we identify spatiotemporally controlled endothelin signalling-a potent vasoconstriction-regulating pathway-as the key activating mechanism of dermal sheath contraction. Pharmacological blocking or genetic ablation of both endothelin receptors, ETA and ETB, impedes dermal sheath contraction and halts follicle regression. Epithelial progenitors at the club hair-epithelial strand bottleneck produce the endothelin ligand ET-1, which is required for follicle regression. ET signalling in dermal sheath cells and downstream contraction is dynamically regulated by cytoplasmic Ca2+ levels through cell membrane and sarcoplasmic reticulum calcium channels. Together, these findings illuminate an epithelial-mesenchymal interaction paradigm in which progenitors-destined to undergo programmed cell death-control the contraction of the surrounding sheath smooth muscle to orchestrate homeostatic tissue regression and reorganization for the next stem cell activation and regeneration cycle.


Assuntos
Endotelinas , Folículo Piloso , Folículo Piloso/metabolismo , Endotelinas/metabolismo , Endotelinas/farmacologia , Receptores de Endotelina/metabolismo , Músculo Liso/metabolismo , Transdução de Sinais , Contração Muscular
3.
Nature ; 592(7854): 428-432, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33790465

RESUMO

Chronic, sustained exposure to stressors can profoundly affect tissue homeostasis, although the mechanisms by which these changes occur are largely unknown. Here we report that the stress hormone corticosterone-which is derived from the adrenal gland and is the rodent equivalent of cortisol in humans-regulates hair follicle stem cell (HFSC) quiescence and hair growth in mice. In the absence of systemic corticosterone, HFSCs enter substantially more rounds of the regeneration cycle throughout life. Conversely, under chronic stress, increased levels of corticosterone prolong HFSC quiescence and maintain hair follicles in an extended resting phase. Mechanistically, corticosterone acts on the dermal papillae to suppress the expression of Gas6, a gene that encodes the secreted factor growth arrest specific 6. Restoring Gas6 expression overcomes the stress-induced inhibition of HFSC activation and hair growth. Our work identifies corticosterone as a systemic inhibitor of HFSC activity through its effect on the niche, and demonstrates that the removal of such inhibition drives HFSCs into frequent regeneration cycles, with no observable defects in the long-term.


Assuntos
Corticosterona/farmacologia , Folículo Piloso/citologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/cirurgia , Adrenalectomia , Animais , Divisão Celular/efeitos dos fármacos , Feminino , Folículo Piloso/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia , Transcriptoma , Regulação para Cima
4.
Science ; 367(6474): 161-166, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31857493

RESUMO

Tissue homeostasis requires the balance of growth by cell production and regression through cell loss. In the hair cycle, during follicle regression, the niche traverses the skin through an unknown mechanism to reach the stem cell reservoir and trigger new growth. Here, we identify the dermal sheath that lines the follicle as the key driver of tissue regression and niche relocation through the smooth muscle contractile machinery that generates centripetal constriction force. We reveal that the calcium-calmodulin-myosin light chain kinase pathway controls sheath contraction. When this pathway is blocked, sheath contraction is inhibited, impeding follicle regression and niche relocation. Thus, our study identifies the dermal sheath as smooth muscle that drives follicle regression for reuniting niche and stem cells in order to regenerate tissue structure during homeostasis.


Assuntos
Derme/fisiologia , Folículo Piloso/fisiologia , Músculo Liso/fisiologia , Regeneração , Nicho de Células-Tronco/fisiologia , Agrecanas/genética , Animais , Humanos , Camundongos , Camundongos Mutantes , Contração Muscular
5.
Dev Cell ; 48(1): 32-48.e5, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30595537

RESUMO

Cell fate transitions are essential for specification of stem cells and their niches, but the precise timing and sequence of molecular events during embryonic development are largely unknown. Here, we identify, with 3D and 4D microscopy, unclustered precursors of dermal condensates (DC), signaling niches for epithelial progenitors in hair placodes. With population-based and single-cell transcriptomics, we define a molecular time-lapse from pre-DC fate specification through DC niche formation and establish the developmental trajectory as the DC lineage emerges from fibroblasts. Co-expression of downregulated fibroblast and upregulated DC genes in niche precursors reveals a transitory molecular state following a proliferation shutdown. Waves of transcription factor and signaling molecule expression then coincide with DC formation. Finally, ablation of epidermal Wnt signaling and placode-derived FGF20 demonstrates their requirement for pre-DC specification. These findings uncover a progenitor-dependent niche precursor fate and the transitory molecular events controlling niche formation and function.


Assuntos
Diferenciação Celular/fisiologia , Derme/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Folículo Piloso/metabolismo , Animais , Fibroblastos/citologia , Folículo Piloso/embriologia , Transdução de Sinais/genética , Pele/metabolismo , Células-Tronco/citologia
6.
Development ; 143(12): 2160-71, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27122169

RESUMO

Primary cilia have been linked to signaling pathways involved in cell proliferation, cell motility and cell polarity. Defects in ciliary function result in developmental abnormalities and multiple ciliopathies. Patients affected by severe ciliopathies, such as Meckel syndrome, present several ocular surface disease conditions of unclear pathogenesis. Here, we show that primary cilia are predominantly present on basal cells of the mouse corneal epithelium (CE) throughout development and in the adult. Conditional ablation of cilia in the CE leads to an increase in proliferation and vertical migration of basal corneal epithelial cells (CECs). A consequent increase in cell density of suprabasal layers results in a thicker than normal CE. Surprisingly, in cilia-deficient CE, cilia-mediated signaling pathways, including Hh and Wnt pathways, were not affected but the intensity of Notch signaling was severely diminished. Although Notch1 and Notch2 receptors were expressed normally, nuclear Notch1 intracellular domain (N1ICD) expression was severely reduced. Postnatal development analysis revealed that in cilia-deficient CECs downregulation of the Notch pathway precedes cell proliferation defects. Thus, we have uncovered a function of the primary cilium in maintaining homeostasis of the CE by balancing proliferation and vertical migration of basal CECs through modulation of Notch signaling.


Assuntos
Cílios/metabolismo , Epitélio Corneano/metabolismo , Homeostase , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Movimento Celular , Proliferação de Células , Cílios/ultraestrutura , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Epitélio Corneano/ultraestrutura , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Proteínas Supressoras de Tumor/metabolismo , Cicatrização
7.
Dev Cell ; 34(5): 577-91, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26256211

RESUMO

Defining the unique molecular features of progenitors and their niche requires a genome-wide, whole-tissue approach with cellular resolution. Here, we co-isolate embryonic hair follicle (HF) placode and dermal condensate cells, precursors of adult HF stem cells and the dermal papilla/sheath niche, along with lineage-related keratinocytes and fibroblasts, Schwann cells, melanocytes, and a population inclusive of all remaining skin cells. With next-generation RNA sequencing, we define gene expression patterns in the context of the entire embryonic skin, and through transcriptome cross-comparisons, we uncover hundreds of enriched genes in cell-type-specific signatures. Axon guidance signaling and many other pathway genes are enriched in multiple signatures, implicating these factors in driving the large-scale cellular rearrangements necessary for HF formation. Finally, we share all data in an interactive, searchable companion website. Our study provides an overarching view of signaling within the entire embryonic skin and captures a molecular snapshot of HF progenitors and their niche.


Assuntos
Folículo Piloso/citologia , Folículo Piloso/embriologia , Queratinócitos/citologia , Pele/metabolismo , Células-Tronco/citologia , Transcriptoma/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Camundongos , Organogênese/fisiologia , Transdução de Sinais/fisiologia , Pele/citologia , Pele/embriologia , Nicho de Células-Tronco
8.
Dev Biol ; 385(2): 179-88, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24309208

RESUMO

Broad dermal Wnt signaling is required for patterned induction of hair follicle placodes and subsequent Wnt signaling in placode stem cells is essential for induction of dermal condensates, cell clusters of precursors for the hair follicle dermal papilla (DP). Progression of hair follicle formation then requires coordinated signal exchange between dermal condensates and placode stem cells. However, it remains unknown whether continued Wnt signaling in DP precursor cells plays a role in this process, largely due to the long-standing inability to specifically target dermal condensates for gene ablation. Here we use the Tbx18(Cre) knockin mouse line to ablate the Wnt-responsive transcription factor ß-catenin specifically in these cells at E14.5 during the first wave of guard hair follicle formation. In the absence of ß-catenin, canonical Wnt signaling is effectively abolished in these cells. Sox2(+) dermal condensates initiate normally; however by E16.5 guard hair follicle numbers are strongly reduced and by E18.5 most whiskers and guard hair follicles are absent, suggesting that active Wnt signaling in dermal condensates is important for hair follicle formation to proceed after induction. To explore the molecular mechanisms by which Wnt signaling in dermal condensates regulates hair follicle formation, we analyze genome-wide the gene expression changes in embryonic ß-catenin null DP precursor cells. We find altered expression of several signaling pathway genes, including Fgfs and Activin, both previously implicated in hair follicle formation. In summary, these data reveal a functional role of Wnt signaling in DP precursors for embryonic hair follicle formation and identify Fgf and Activin signaling as potential effectors of Wnt signaling-regulated events.


Assuntos
Cabelo/crescimento & desenvolvimento , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real
9.
J Invest Dermatol ; 133(10): 2332-2339, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23677168

RESUMO

Systematic ablation of previously identified dermal papilla (DP) signature genes in embryonic DP precursors will reveal their functional roles during hair follicle morphogenesis. In this study, we validate Enpp2/Autotaxin as one of the highest expressed signature genes in postnatal DP, and demonstrate specific expression of this lysophosphatidic acid (LPA)-generating enzyme in embryonic dermal condensates. We further identify dermal and epidermal expression of several LPA receptors, suggesting that LPA signaling could contribute to follicle morphogenesis in both mesenchymal and epithelial compartments. We then use the recently characterized Cre-expressing Tbx18 knock-in line to conditionally ablate Enpp2 in embryonic DP precursors. Despite efficient gene knockout in embryonic day 14.5 (E14.5) dermal condensates, morphogenesis proceeds regularly with normal numbers, lengths, and sizes of all hair follicle types, suggesting that Enpp2 is not required for hair follicle formation. To interrogate DP signature gene expression, we finally isolate control and Enpp2-null DP precursors and identify the expression and upregulation of LIPH, an alternative LPA-producing enzyme, suggesting that this gene could functionally compensate for the absence of Enpp2. We conclude that future coablation of both LPA-producing enzymes or of several LPA receptors may reveal the functional role of LPA signaling during hair follicle morphogenesis.


Assuntos
Derme/embriologia , Folículo Piloso/embriologia , Lipase/genética , Morfogênese/fisiologia , Diester Fosfórico Hidrolases/genética , Animais , Derme/citologia , Derme/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Folículo Piloso/citologia , Folículo Piloso/fisiologia , Lipase/metabolismo , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Diester Fosfórico Hidrolases/metabolismo , Gravidez , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais/fisiologia , Proteínas com Domínio T/genética , Regulação para Cima/fisiologia
10.
J Invest Dermatol ; 133(2): 344-53, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22992803

RESUMO

How cell fate decisions of stem and progenitor cells are regulated by their microenvironment or niche is a central question in stem cell and regenerative biology. Although functional analysis of hair follicle epithelial stem cells by gene targeting is well established, the molecular and genetic characterization of the dermal counterpart during embryonic morphogenesis has been lacking because of the absence of cell type-specific drivers. Here, we report that T-box transcription factor Tbx18 specifically marks dermal papilla (DP) precursor cells during embryonic hair follicle morphogenesis. With Tbx18(LacZ), Tbx18(H2BGFP), and Tbx18(Cre) knock-in mouse models, we demonstrate LacZ and H2BGFP (nuclear green fluorescent protein) expression and Cre activity in dermal condensates of nascent first-wave hair follicles at E14.5. As Tbx18 expression becomes more widespread throughout the dermis at later developmental stages, we use tamoxifen-inducible Cre-expressing mice, Tbx18(MerCreMer), to exclusively target DP precursor cells and their progeny. Finally, we ablate Tbx18 in full knockout mice, but find no perturbations in hair follicle formation, suggesting that Tbx18 is dispensable for normal DP function. In summary, our study establishes Tbx18 as a genetic driver to target for the first time embryonic DP precursors for labeling, isolation, and gene ablation that will greatly enhance investigations into their molecular functions during hair follicle morphogenesis.


Assuntos
Derme/embriologia , Derme/fisiologia , Folículo Piloso/embriologia , Folículo Piloso/fisiologia , Proteínas com Domínio T/genética , Animais , Animais Recém-Nascidos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Antagonistas de Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/genética , Folículo Piloso/citologia , Integrases/genética , Óperon Lac , Camundongos , Camundongos Knockout , Camundongos Nus , Gravidez , Transplante de Pele , Tamoxifeno/farmacologia
11.
Dev Cell ; 23(5): 981-94, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23153495

RESUMO

How dermal papilla (DP) niche cells regulate hair follicle progenitors to control hair growth remains unclear. Using Tbx18(Cre) to target embryonic DP precursors, we ablate the transcription factor Sox2 early and efficiently, resulting in diminished hair shaft outgrowth. We find that DP niche expression of Sox2 controls the migration speed of differentiating hair shaft progenitors. Transcriptional profiling of Sox2 null DPs reveals increased Bmp6 and decreased BMP inhibitor Sostdc1, a direct Sox2 transcriptional target. Subsequently, we identify upregulated BMP signaling in knockout hair shaft progenitors and demonstrate that Bmp6 inhibits cell migration, an effect that can be attenuated by Sostdc1. A shorter and Sox2-negative hair type lacks Sostdc1 in the DP and shows reduced migration and increased BMP activity of hair shaft progenitors. Collectively, our data identify Sox2 as a key regulator of hair growth that controls progenitor migration by fine-tuning BMP-mediated mesenchymal-epithelial crosstalk.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Folículo Piloso/embriologia , Folículo Piloso/metabolismo , Cabelo/crescimento & desenvolvimento , Fatores de Transcrição SOXB1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteína Morfogenética Óssea 6/metabolismo , Proteínas Morfogenéticas Ósseas/deficiência , Movimento Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Folículo Piloso/citologia , Folículo Piloso/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição SOXB1/genética , Transdução de Sinais , Transcriptoma
12.
Int J Dev Biol ; 55(2): 209-14, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21553385

RESUMO

In mammals, the stem cells of spermatogenesis are derived from an embryonic cell population called primordial germ cells (PGCs). Spermatogonial stem cells displaying the "side population" (SP) phenotype have been identified in the immature and adult mouse testis, but noting is known about the expression of the SP phenotype during prenatal development of germ cells. The SP phenotype, defined as the ability of cells to efflux fluorescent dyes such as Hoechst, is common to several stem/progenitor cell types. In the present study, we analyzed and characterized the Hoechst SP via cytofluorimetric analysis of disaggregated gonads at different time points during embryonic development in mice. To directly test the hypothesis that the SP phenotype is a feature of germ cell lineage, experiments were performed on transgenic animals expressing enhanced green fluorescent protein (EGFP) under the control of the Oct4 promoter, to identify early germ cells up to PGCs. We found that prenatal gonads contain a fraction of SP cells at each stage analyzed, and the percentage of cells in the SP fraction decreases as development proceeds. Surprisingly, more than 50% of the PGCs displayed the SP phenotype at 11.5 dpc (days post coitum). The percentage of germ cells with the SP phenotype decreased steadily with development, to less than 1% at 18.5 dpc. Cytofluorimetric analysis along with immunocytochemistry performed on sorted cells indicated that the SP fraction of prenatal gonads, as in the adult testis, was heterogeneous, being composed of both somatic and germ cells. Both cell types expressed the ABC transporters Abcg2, Abcb1a, Abcb1b and Abcc1. These findings provide evidence that the SP phenotype is a common feature of PGCs and identifies a subpopulation of fetal testis cells including prospermatogonia whose differentiation fate remains to be investigated.


Assuntos
Células Germinativas/citologia , Células da Side Population/citologia , Testículo/citologia , Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Diferenciação Celular , Linhagem da Célula , Citometria de Fluxo , Células Germinativas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fator 3 de Transcrição de Octâmero/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Células da Side Population/metabolismo , Espermatogênese , Espermatogônias/citologia , Espermatogônias/metabolismo , Testículo/embriologia , Testículo/metabolismo
13.
Stem Cells ; 28(2): 221-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20014278

RESUMO

Direct reprogramming of somatic cells into induced pluripotent stem (iPS) cells by only four transcription factors (Oct4, Sox2, Klf4, and c-Myc) has great potential for tissue-specific regenerative therapies, eliminating the ethical issues surrounding the use of embryonic stem cells and the rejection problems of using non-autologous cells. The reprogramming efficiency generally is very low, however, and the problems surrounding the introduction of viral genetic material are only partially investigated. Recent efforts to reduce the number of virally expressed transcription factors succeeded at reprogramming neural stem cells into iPS cells by overexpressing Oct4 alone. However, the relative inaccessibility and difficulty of obtaining neural cells in humans remains to be resolved. Here we report that dermal papilla (DP) cells, which are specialized skin fibroblasts thought to instruct hair follicle stem cells, endogenously express high levels of Sox2 and c-Myc, and that these cells can be reprogrammed into iPS cells with only Oct4 and Klf4. Moreover, we show that DP cells are reprogrammed more efficiently than skin and embryonic fibroblasts. iPS cells derived from DP cells expressed pluripotency genes and differentiated into cells from all germ layers in vitro and widely contributed to chimeric mice in vivo, including the germline. Our work establishes DP cells as an easily accessible source to generate iPS cells with efficiency and with less genetic material. This opens up the possibility of streamlined generation of skin-derived, patient-specific pluripotent stem cells and of ultimately replacing the remaining two factors with small molecules for safe generation of transplantable cells.


Assuntos
Reprogramação Celular/fisiologia , Derme/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Fatores de Transcrição Kruppel-Like/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Animais , Células Cultivadas , Reprogramação Celular/genética , Derme/metabolismo , Feminino , Fibroblastos/citologia , Gonadotropinas Equinas , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Fator 3 de Transcrição de Octâmero/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1
14.
Stem Cells ; 27(12): 3043-52, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19711452

RESUMO

Spermatogenesis is maintained by a pool of spermatogonial stem cells (SSCs). Analyses of the molecular profile of SSCs have revealed the existence of subsets, indicating that the stem cell population is more heterogeneous than previously believed. However, SSC subsets are poorly characterized. In rodents, the first steps in spermatogenesis have been extensively investigated, both under physiological conditions and during the regenerative phase that follows germ cell damage. In the widely accepted model, the SSCs are type Asingle (As) spermatogonia. Here, we tested the hypothesis that As spermatogonia are phenotypically heterogeneous by analyzing glial cell line-derived neurotrophic factor (GDNF) family receptor alpha1 (GFRA1) expression in whole-mounted seminiferous tubules, via cytofluorimetric analysis and in vivo colonogenic assays. GFRA1 is a coreceptor for GDNF, a Sertoli cell-derived factor essential for SSC self-renewal and proliferation. Morphometric analysis demonstrated that 10% of As spermatogonia did not express GFRA1 but were colonogenic, as shown by germ cell transplantation assay. In contrast, cells selected for GFRA1 expression were not colonogenic in vivo. In human testes, GFRA1 was also heterogeneously expressed in Adark and in Apale spermatogonia, the earliest spermatogonia. In vivo 5-bromo-2'-deoxyuridine administration showed that both GFRA1(+) and GFRA1(-) As spermatogonia were engaged in the cell cycle, a finding supported by the lack of long-term label-retaining As spermatogonia. GFRA1 expression was asymmetric in 5% of paired cells, suggesting that As subsets may be generated by asymmetric cell division. Our data support the hypothesis of the existence of SSC subsets and reveal a previously unrecognized heterogeneity in the expression profile of As spermatogonia in vivo.


Assuntos
Forma Celular , Espermatogônias/citologia , Células-Tronco/citologia , Envelhecimento , Animais , Separação Celular , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espermatogônias/metabolismo , Células-Tronco/metabolismo
15.
Differentiation ; 78(2-3): 131-6, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19541401

RESUMO

Spermatogonial stem cells (SSC) ensure continuous production of mammalian male gametes. In rodents, the SSC are Asingle spermatogonia (As). Gene loss and gain-of-function mutations have provided some clues into SSC function, but genetic dissection of SSC physiology has not yet been accomplished. The adaptor protein Numb is an evolutionarily conserved protein originally implicated in the control of the fate of sibling cells. Mice homozygous for deficient Numb die before embryonic day 11.5, hampering the analysis of its inactivation in postnatal male germline. Here, we have developed an experimental strategy to conditionally inactivate Numb and its homolog Numblike in the postnatal germline by in vitro delivery of cell-permeant Cre recombinase. Cre-transduced SSC isolated from wild-type mice retained their ability to self-renew and to differentiate in vivo, as shown by their ability to give rise to normal spermatogenic colonies when transplanted in recipient testes. Cre-transduced SSC from conditional mutant mouse line were able to colonize recipient testes upon transplantation. Inactivation of either Numb or Numblike in SSC did not impair the development of normal donor-derived spermatogenesis. However, compared to single-null SSC, double-null SSC generated shorter colonies in which the germ cells failed to differentiate beyond the round spermatid stage, underscoring the essential roles of both Numb and Numblike in spermatogenesis. We demonstrate the feasibility of gene inactivation in adult SSC by ex vivo Cre delivery. This provides a means to analyze the function of genes that operate on a cell-autonomous basis or those that are coupled to signals that SSC receive from bystander cells.


Assuntos
Regulação da Expressão Gênica/fisiologia , Inativação Gênica/fisiologia , Integrases/fisiologia , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Espermatogônias/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espermatogônias/metabolismo , Células-Tronco/metabolismo , Testículo/citologia , Testículo/fisiologia , Transdução Genética
16.
Reproduction ; 132(6): 887-97, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17127749

RESUMO

Numb is an adaptor protein that is asymmetrically inherited at mitosis and controls the fate of sibling cells in different species. The role of m-Numb (mammalian Numb) as an important cell fate-determining factor has extensively been described mostly in neural tissues, particularly in progenitor cells, in the mouse. Biochemical and genetic analyses have shown that Numb acts as an inhibitor of the Notch signaling pathway, an evolutionarily conserved pathway involved in the control of cell proliferation, differentiation, and apoptosis. In the present study, we sought to determine m-Numb distribution in germ cells in the postnatal mouse testis. We show that all four m-Numb isoforms are widely expressed during postnatal testis development. By reverse transcriptase-PCR and western blot analyses, we further identify p71 as the predominantly expressed isoform in germ cells. Moreover, we demonstrate through co-immunoprecipitation studies that m-Numb physically associates with Ap2a1, a component of the endocytotic clathrin-coated vesicles. Finally, we employed confocal immunofluorescence microscopy of whole mount seminiferous tubules and isolated germ cells to gain more insight into the subcellular localization of m-Numb. These morphological analyses confirmed m-Numb and Ap2a1 co-localization. However, we did not observe asymmetric localization of m-Numb neither in mitotic spermatogonial stem cells nor in more differentiated spermatogonial cells, suggesting that spermatogonial stem cell fate in the mouse does not rely on asymmetric partitioning of m-Numb.


Assuntos
Proteínas de Membrana/análise , Proteínas do Tecido Nervoso/análise , Isoformas de Proteínas/análise , Espermatozoides/química , Testículo/química , Complexo 2 de Proteínas Adaptadoras/análise , Complexo 2 de Proteínas Adaptadoras/metabolismo , Subunidades alfa do Complexo de Proteínas Adaptadoras/análise , Subunidades alfa do Complexo de Proteínas Adaptadoras/metabolismo , Processamento Alternativo , Animais , Northern Blotting/métodos , Western Blotting/métodos , Imunoprecipitação/métodos , Masculino , Proteínas de Membrana/genética , Camundongos , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Túbulos Seminíferos/química , Espermatogênese/fisiologia , Espermatogônias/química , Testículo/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA