Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 33(3-4): 209-220, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30692207

RESUMO

Spatiotemporal control of Wnt signaling is essential for the development and homeostasis of many tissues. The transmembrane E3 ubiquitin ligases ZNRF3 (zinc and ring finger 3) and RNF43 (ring finger protein 43) antagonize Wnt signaling by promoting degradation of frizzled receptors. ZNRF3 and RNF43 are frequently inactivated in human cancer, but the molecular and therapeutic implications remain unclear. Here, we demonstrate that adrenocortical-specific loss of ZNRF3, but not RNF43, results in adrenal hyperplasia that depends on Porcupine-mediated Wnt ligand secretion. Furthermore, we discovered a Wnt/ß-catenin signaling gradient in the adrenal cortex that is disrupted upon loss of ZNRF3. Unlike ß-catenin gain-of-function models, which induce high Wnt/ß-catenin activation and expansion of the peripheral cortex, ZNRF3 loss triggers activation of moderate-level Wnt/ß-catenin signaling that drives proliferative expansion of only the histologically and functionally distinct inner cortex. Genetically reducing ß-catenin dosage significantly reverses the ZNRF3-deficient phenotype. Thus, homeostatic maintenance of the adrenal cortex is dependent on varying levels of Wnt/ß-catenin activation, which is regulated by ZNRF3.


Assuntos
Córtex Suprarrenal/metabolismo , Homeostase/genética , Ubiquitina-Proteína Ligases/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Córtex Suprarrenal/citologia , Córtex Suprarrenal/crescimento & desenvolvimento , Doenças do Córtex Suprarrenal/fisiopatologia , Animais , Proliferação de Células/genética , Feminino , Técnicas de Inativação de Genes , Masculino , Camundongos , Modelos Animais , Ativação Transcricional/genética , Ubiquitina-Proteína Ligases/genética
2.
Development ; 148(9)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33914868

RESUMO

In mammalian ovaries, immature oocytes are reserved in primordial follicles until their activation for potential ovulation. Precise control of primordial follicle activation (PFA) is essential for reproduction, but how this is achieved is unclear. Here, we show that canonical wingless-type MMTV integration site family (WNT) signaling is pivotal for pre-granulosa cell (pre-GC) activation during PFA. We identified several WNT ligands expressed in pre-GCs that act in an autocrine manner. Inhibition of WNT secretion from pre-GCs/GCs by conditional knockout (cKO) of the wntless (Wls) gene led to female infertility. In Wls cKO mice, GC layer thickness was greatly reduced in growing follicles, which resulted in impaired oocyte growth with both an abnormal, sustained nuclear localization of forkhead box O3 (FOXO3) and reduced phosphorylation of ribosomal protein S6 (RPS6). Constitutive stabilization of ß-catenin (CTNNB1) in pre-GCs/GCs induced morphological changes of pre-GCs from a squamous into a cuboidal form, though it did not influence oocyte activation. Our results reveal that canonical WNT signaling plays a permissive role in the transition of pre-GCs to GCs, which is an essential step to support oocyte growth.


Assuntos
Fertilidade , Células da Granulosa/metabolismo , Infertilidade Feminina/metabolismo , Ovário/metabolismo , Via de Sinalização Wnt , Animais , Feminino , Camundongos , Camundongos Knockout , Oócitos/metabolismo , Oogênese , Folículo Ovariano/metabolismo , Ovulação , Transcriptoma , Proteínas WT1/genética , beta Catenina/genética
3.
Nature ; 548(7668): 451-455, 2017 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-28813421

RESUMO

The constant regeneration of stomach epithelium is driven by long-lived stem cells, but the mechanism that regulates their turnover is not well understood. We have recently found that the gastric pathogen Helicobacter pylori can activate gastric stem cells and increase epithelial turnover, while Wnt signalling is known to be important for stem cell identity and epithelial regeneration in several tissues. Here we find that antral Wnt signalling, marked by the classic Wnt target gene Axin2, is limited to the base and lower isthmus of gastric glands, where the stem cells reside. Axin2 is expressed by Lgr5+ cells, as well as adjacent, highly proliferative Lgr5- cells that are able to repopulate entire glands, including the base, upon depletion of the Lgr5+ population. Expression of both Axin2 and Lgr5 requires stroma-derived R-spondin 3 produced by gastric myofibroblasts proximal to the stem cell compartment. Exogenous R-spondin administration expands and accelerates proliferation of Axin2+/Lgr5- but not Lgr5+ cells. Consistent with these observations, H. pylori infection increases stromal R-spondin 3 expression and expands the Axin2+ cell pool to cause hyperproliferation and gland hyperplasia. The ability of stromal niche cells to control and adapt epithelial stem cell dynamics constitutes a sophisticated mechanism that orchestrates epithelial regeneration and maintenance of tissue integrity.


Assuntos
Infecções por Helicobacter/metabolismo , Homeostase , Células-Tronco/citologia , Células-Tronco/metabolismo , Estômago/citologia , Células Estromais/metabolismo , Trombospondinas/metabolismo , Animais , Proteína Axina/metabolismo , Proliferação de Células , Células Epiteliais/citologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Antro Pilórico/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células-Tronco , Células Estromais/citologia , Via de Sinalização Wnt
4.
Proc Natl Acad Sci U S A ; 115(26): E5954-E5962, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29891676

RESUMO

In the adult mouse spinal cord, the ependymal cell population that surrounds the central canal is thought to be a promising source of quiescent stem cells to treat spinal cord injury. Relatively little is known about the cellular origin of ependymal cells during spinal cord development, or the molecular mechanisms that regulate ependymal cells during adult homeostasis. Using genetic lineage tracing based on the Wnt target gene Axin2, we have characterized Wnt-responsive cells during spinal cord development. Our results revealed that Wnt-responsive progenitor cells are restricted to the dorsal midline throughout spinal cord development, which gives rise to dorsal ependymal cells in a spatially restricted pattern. This is contrary to previous reports that suggested an exclusively ventral origin of ependymal cells, suggesting that ependymal cells may retain positional identities in relation to their neural progenitors. Our results further demonstrated that in the postnatal and adult spinal cord, all ependymal cells express the Wnt/ß-catenin signaling target gene Axin2, as well as Wnt ligands. Genetic elimination of ß-catenin or inhibition of Wnt secretion in Axin2-expressing ependymal cells in vivo both resulted in impaired proliferation, indicating that Wnt/ß-catenin signaling promotes ependymal cell proliferation. These results demonstrate the continued importance of Wnt/ß-catenin signaling for both ependymal cell formation and regulation. By uncovering the molecular signals underlying the formation and regulation of spinal cord ependymal cells, our findings thus enable further targeting and manipulation of this promising source of quiescent stem cells for therapeutic interventions.


Assuntos
Proteína Axina/metabolismo , Proliferação de Células , Neuroglia/metabolismo , Medula Espinal/crescimento & desenvolvimento , Via de Sinalização Wnt/fisiologia , Animais , Proteína Axina/genética , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Medula Espinal/citologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
5.
Proc Natl Acad Sci U S A ; 113(11): E1489-97, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26929341

RESUMO

Spermatogonial stem cells (SSCs) fuel the production of male germ cells but the mechanisms behind SSC self-renewal, proliferation, and differentiation are still poorly understood. Using the Wnt target gene Axin2 and genetic lineage-tracing experiments, we found that undifferentiated spermatogonia, comprising SSCs and transit amplifying progenitor cells, respond to Wnt/ß-catenin signals. Genetic elimination of ß-catenin indicates that Wnt/ß-catenin signaling promotes the proliferation of these cells. Signaling is likely initiated by Wnt6, which is uniquely expressed by neighboring Sertoli cells, the only somatic cells in the seminiferous tubule that support germ cells and act as a niche for SSCs. Therefore, unlike other stem cell systems where Wnt/ß-catenin signaling is implicated in self-renewal, the Wnt pathway in the testis specifically contributes to the proliferation of SSCs and progenitor cells.


Assuntos
Espermatogônias/citologia , Testículo/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Proteína Axina/genética , Proteína Axina/metabolismo , Diferenciação Celular , Proliferação de Células , Masculino , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Células de Sertoli/metabolismo , Espermatogônias/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Testículo/citologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética
6.
Proc Natl Acad Sci U S A ; 113(11): E1498-505, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26903625

RESUMO

How stem cells maintain their identity and potency as tissues change during growth is not well understood. In mammalian hair, it is unclear how hair follicle stem cells can enter an extended period of quiescence during the resting phase but retain stem cell potential and be subsequently activated for growth. Here, we use lineage tracing and gene expression mapping to show that the Wnt target gene Axin2 is constantly expressed throughout the hair cycle quiescent phase in outer bulge stem cells that produce their own Wnt signals. Ablating Wnt signaling in the bulge cells causes them to lose their stem cell potency to contribute to hair growth and undergo premature differentiation instead. Bulge cells express secreted Wnt inhibitors, including Dickkopf (Dkk) and secreted frizzled-related protein 1 (Sfrp1). However, the Dickkopf 3 (Dkk3) protein becomes localized to the Wnt-inactive inner bulge that contains differentiated cells. We find that Axin2 expression remains confined to the outer bulge, whereas Dkk3 continues to be localized to the inner bulge during the hair cycle growth phase. Our data suggest that autocrine Wnt signaling in the outer bulge maintains stem cell potency throughout hair cycle quiescence and growth, whereas paracrine Wnt inhibition of inner bulge cells reinforces differentiation.


Assuntos
Proteína Axina/metabolismo , Folículo Piloso/citologia , Células-Tronco/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Comunicação Autócrina , Proteína Axina/genética , Regulação da Expressão Gênica , Folículo Piloso/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Mutantes , Camundongos Transgênicos , Células-Tronco/citologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética
7.
Proc Natl Acad Sci U S A ; 111(49): E5262-71, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25422448

RESUMO

Wnt signaling is a critical regulator of bone development, but the identity and role of the Wnt-producing cells are still unclear. We addressed these questions through in situ hybridization, lineage tracing, and genetic experiments. First, we surveyed the expression of all 19 Wnt genes and Wnt target gene Axin2 in the neonatal mouse bone by in situ hybridization, and demonstrated--to our knowledge for the first time--that Osterix-expressing cells coexpress Wnt and Axin2. To track the behavior and cell fate of Axin2-expressing osteolineage cells, we performed lineage tracing and showed that they sustain bone formation over the long term. Finally, to examine the role of Wnts produced by Osterix-expressing cells, we inhibited Wnt secretion in vivo, and observed inappropriate differentiation, impaired proliferation, and diminished Wnt signaling response. Therefore, Osterix-expressing cells produce their own Wnts that in turn induce Wnt signaling response, thereby regulating their proliferation and differentiation.


Assuntos
Fatores de Transcrição/metabolismo , Proteínas Wnt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteína Axina/metabolismo , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Separação Celular , Citometria de Fluxo , Glicoproteínas/metabolismo , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Knockout , Mutação , Osteogênese/fisiologia , Fenótipo , Reação em Cadeia da Polimerase , Fator de Transcrição Sp7 , Células-Tronco , Via de Sinalização Wnt
8.
Gastroenterology ; 148(7): 1392-404.e21, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25725293

RESUMO

BACKGROUND & AIMS: Helicobacter pylori infection is the main risk factor for gastric cancer. We characterized the interactions of H pylori with gastric epithelial progenitor and stem cells in humans and mice and investigated how these interactions contribute to H pylori-induced pathology. METHODS: We used quantitative confocal microscopy and 3-dimensional reconstruction of entire gastric glands to determine the localizations of H pylori in stomach tissues from humans and infected mice. Using lineage tracing to mark cells derived from leucine-rich repeat-containing G-protein coupled receptor 5-positive (Lgr5(+)) stem cells (Lgr5-eGFP-IRES-CreERT2/Rosa26-TdTomato mice) and in situ hybridization, we analyzed gastric stem cell responses to infection. Isogenic H pylori mutants were used to determine the role of specific virulence factors in stem cell activation and pathology. RESULTS: H pylori grow as distinct bacterial microcolonies deep in the stomach glands and interact directly with gastric progenitor and stem cells in tissues from mice and humans. These gland-associated bacteria activate stem cells, increasing the number of stem cells, accelerating Lgr5(+) stem cell proliferation, and up-regulating expression of stem cell-related genes. Mutant bacteria with defects in chemotaxis that are able to colonize the stomach surface but not the antral glands in mice do not activate stem cells. In addition, bacteria that are unable to inject the contact-dependent virulence factor CagA into the epithelium colonized stomach glands in mice, but did not activate stem cells or produce hyperplasia to the same extent as wild-type H pylori. CONCLUSIONS: H pylori colonize and manipulate the progenitor and stem cell compartments, which alters turnover kinetics and glandular hyperplasia. Bacterial ability to alter the stem cells has important implications for gastrointestinal stem cell biology and H pylori-induced gastric pathology.


Assuntos
Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/crescimento & desenvolvimento , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/microbiologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biomarcadores/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Mucosa Gástrica/metabolismo , Genótipo , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/patologia , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Hiperplasia , Cinética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Organoides , Fenótipo , Receptores Acoplados a Proteínas G/genética , Células-Tronco/metabolismo , Células-Tronco/patologia , Técnicas de Cultura de Tecidos , Virulência
9.
Proc Natl Acad Sci U S A ; 110(18): 7324-9, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23589866

RESUMO

Since the discovery of neural stem cells in the mammalian brain, there has been significant interest in understanding their contribution to tissue homeostasis at both the cellular and molecular level. Wnt/ß-catenin signaling is crucial for development of the central nervous system and has been implicated in stem cell maintenance in multiple tissues. Based on this, we hypothesized that the Wnt pathway likely controls neural stem cell maintenance and differentiation along the entire developmental continuum. To test this, we performed lineage tracing experiments using the recently developed tamoxifen-inducible Cre at Axin2 mouse strain to follow the developmental fate of Wnt/ß-catenin-responsive cells in both the embryonic and postnatal mouse brain. From as early as embryonic day 8.5 onwards, Axin2(+) cells can give rise to spatially and functionally restricted populations of adult neural stem cells in the subventricular zone. Similarly, progeny from Axin2(+) cells labeled from E12.5 contribute to both the subventricular zone and the dentate gyrus of the hippocampus. Labeling in the postnatal brain, in turn, demonstrates the persistence of long-lived, Wnt/ß-catenin-responsive stem cells in both of these sites. These results demonstrate the continued importance of Wnt/ß-catenin signaling for neural stem and progenitor cell formation and function throughout developmental time.


Assuntos
Células-Tronco Adultas/citologia , Proteína Axina/metabolismo , Linhagem da Célula , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Células-Tronco Adultas/metabolismo , Animais , Animais Recém-Nascidos , Giro Denteado/citologia , Giro Denteado/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Integrases/metabolismo , Camundongos , Neuroglia/citologia , Neuroglia/metabolismo , Coloração e Rotulagem , Fatores de Tempo
10.
Proc Natl Acad Sci U S A ; 110(17): 6991-6, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23559372

RESUMO

Wnt signaling in mouse mammary development and tumorigenesis has been heavily studied and characterized, but its role in human breast cancer remains elusive. Although Wnt inhibitors are in early clinical development, it is unclear whether they will be of therapeutic benefit to breast cancer patients, and subsequently, to which ones. To address this, we generated a panel of Wnt reporting human breast cancer cell lines and identified a previously unrecognized enrichment for the ability to respond to Wnt in the basal B or claudin-low subtype, which has a poor prognosis and no available targeted therapies. By co-injecting Wnt3A expressing human mammary fibroblasts with human breast cancer cell lines into mouse mammary fat pads, we showed that elevated paracrine Wnt signaling was correlated with accelerated tumor growth. Using this heterotypic system and a dual lentiviral reporter system that enables simultaneous real-time measurement of both Wnt-responsive cells and bulk tumor cells, we analyzed the outcome of elevated Wnt signaling in patient-derived xenograft (PDX) models. Interestingly, the PDX models exhibited responses not observed in the cell lines analyzed. Exogenous WNT3A promoted tumor growth in one human epidermal growth factor receptor 2-overexpressing PDX line but inhibited growth in a second PDX line obtained from a patient with triple-negative breast cancer. Tumor suppression was associated with squamous differentiation in the latter. Thus, our work suggests that paracrine Wnt signaling can either fuel or repress the growth of human breast cancers depending on yet to be determined aspects of the molecular pathways they express.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/fisiopatologia , Comunicação Parácrina/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Fibroblastos/metabolismo , Fibroblastos/transplante , Humanos , Imuno-Histoquímica , Luciferases , Proteínas Luminescentes , Camundongos , Camundongos SCID , Receptor ErbB-2/metabolismo , Fatores de Tempo , Transplante Heterólogo , Proteína Wnt3A/metabolismo , Proteína Vermelha Fluorescente
11.
Proc Natl Acad Sci U S A ; 110(9): 3405-10, 2013 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-23391730

RESUMO

A goal of regenerative medicine is to identify cardiovascular progenitors from human ES cells (hESCs) that can functionally integrate into the human heart. Previous studies to evaluate the developmental potential of candidate hESC-derived progenitors have delivered these cells into murine and porcine cardiac tissue, with inconclusive evidence regarding the capacity of these human cells to physiologically engraft in xenotransplantation assays. Further, the potential of hESC-derived cardiovascular lineage cells to functionally couple to human myocardium remains untested and unknown. Here, we have prospectively identified a population of hESC-derived ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells that give rise to cardiomyocytes, endothelial cells, and vascular smooth muscle cells in vitro at a clonal level. We observed rare clusters of ROR2(+) cells and diffuse expression of KDR and PDGFRα in first-trimester human fetal hearts. We then developed an in vivo transplantation model by transplanting second-trimester human fetal heart tissues s.c. into the ear pinna of a SCID mouse. ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells were delivered into these functioning fetal heart tissues: in contrast to traditional murine heart models for cell transplantation, we show structural and functional integration of hESC-derived cardiovascular progenitors into human heart.


Assuntos
Células-Tronco Embrionárias/citologia , Feto/citologia , Coração/embriologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/transplante , Transplante de Células-Tronco , Animais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem da Célula , Separação Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Endotélio Vascular/citologia , Feto/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/citologia , Camundongos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Linha Primitiva/citologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Sobrevivência de Tecidos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
Proc Natl Acad Sci U S A ; 110(31): 12691-6, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23852729

RESUMO

Developmental biology is challenged to reveal the function of numerous candidate genes implicated by recent genome-scale studies as regulators of organ development and diseases. Recapitulating organogenesis from purified progenitor cells that can be genetically manipulated would provide powerful opportunities to dissect such gene functions. Here we describe systems for reconstructing pancreas development, including islet ß-cell and α-cell differentiation, from single fetal progenitor cells. A strict requirement for native genetic regulators of in vivo pancreas development, such as Ngn3, Arx, and Pax4, revealed the authenticity of differentiation programs in vitro. Efficient genetic screens permitted by this system revealed that Prdm16 is required for pancreatic islet development in vivo. Discovering the function of genes regulating pancreas development with our system should enrich strategies for regenerating islets for treating diabetes mellitus.


Assuntos
Diferenciação Celular , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Células-Tronco/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/terapia , Feminino , Células Secretoras de Glucagon/citologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição Box Pareados/biossíntese , Fatores de Transcrição Box Pareados/genética , Células-Tronco/citologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
13.
Proc Natl Acad Sci U S A ; 109(2): 370-7, 2012 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-22203956

RESUMO

Lipid-modified Wnt/Wingless (Wg) proteins can signal to their target cells in a short- or long-range manner. How these hydrophobic proteins travel through the extracellular environment remains an outstanding question. Here, we report on a Wg binding protein, Secreted Wg-interacting molecule (Swim), that facilitates Wg diffusion through the extracellular matrix. Swim, a putative member of the Lipocalin family of extracellular transport proteins, binds to Wg with nanomolar affinity in a lipid-dependent manner. In quantitative signaling assays, Swim is sufficient to maintain the solubility and activity of purified Wg. In Drosophila, swim RNAi phenotypes resemble wg loss-of-function phenotypes in long-range signaling. We propose that Swim is a cofactor that promotes long-range Wg signaling in vivo by maintaining the solubility of Wg.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Lipocalinas/metabolismo , Transdução de Sinais/fisiologia , Proteína Wnt1/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico/fisiologia , Primers do DNA/genética , Drosophila/genética , Proteínas de Drosophila/genética , Ensaio de Imunoadsorção Enzimática , Matriz Extracelular/metabolismo , Imuno-Histoquímica , Lipocalinas/genética , Dados de Sequência Molecular , Plasmídeos/genética , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Alinhamento de Sequência , Transdução de Sinais/genética , Solubilidade
14.
Proc Natl Acad Sci U S A ; 109(21): 8167-72, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22562792

RESUMO

Inner ear hair cells are specialized sensory cells essential for auditory function. Previous studies have shown that the sensory epithelium is postmitotic, but it harbors cells that can behave as progenitor cells in vitro, including the ability to form new hair cells. Lgr5, a Wnt target gene, marks distinct supporting cell types in the neonatal cochlea. Here, we tested the hypothesis that Lgr5(+) cells are Wnt-responsive sensory precursor cells. In contrast to their quiescent in vivo behavior, Lgr5(+) cells isolated by flow cytometry from neonatal Lgr5(EGFP-CreERT2/+) mice proliferated and formed clonal colonies. After 10 d in culture, new sensory cells formed and displayed specific hair cell markers (myo7a, calretinin, parvalbumin, myo6) and stereocilia-like structures expressing F-actin and espin. In comparison with other supporting cells, Lgr5(+) cells were enriched precursors to myo7a(+) cells, most of which formed without mitotic division. Treatment with Wnt agonists increased proliferation and colony-formation capacity. Conversely, small-molecule inhibitors of Wnt signaling suppressed proliferation without compromising the myo7a(+) cells formed by direct differentiation. In vivo lineage tracing supported the idea that Lgr5(+) cells give rise to myo7a(+) hair cells in the neonatal Lgr5(EGFP-CreERT2/+) cochlea. In addition, overexpression of ß-catenin initiated proliferation and led to transient expansion of Lgr5(+) cells within the cochlear sensory epithelium. These results suggest that Lgr5 marks sensory precursors and that Wnt signaling can promote their proliferation and provide mechanistic insights into Wnt-responsive progenitor cells during sensory organ development.


Assuntos
Cóclea/citologia , Cóclea/crescimento & desenvolvimento , Células Ciliadas Auditivas Internas/metabolismo , Células-Tronco/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Divisão Celular/fisiologia , Linhagem da Célula/fisiologia , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Células Ciliadas Auditivas Internas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Regeneração/fisiologia , Células-Tronco/citologia
15.
MicroPubl Biol ; 20232023.
Artigo em Inglês | MEDLINE | ID: mdl-37260921

RESUMO

The Wnt signaling pathway regulates metazoan development, tissue homeostasis, and regeneration. Many outstanding questions in Wnt signal transduction revolve around the molecular events immediately following Wnt-receptor interactions. To identify binding partners of the Wnt receptor Frizzled 7 (Fzd7) upon pathway activation, we tagged Fzd7 with APEX2, an enzyme that allows biotinylation of proximal interactors with high temporal and spatial resolution. Upon confirming proper localization and signaling activity of APEX2-tagged Fzd7, we labeled proximal interactors of Fzd7 with or without Wnt3a stimulation. Mass spectrometry analysis of biotinylated interactors identified several known Wnt pathway proteins. Top interactors enriched upon Wnt treatment were involved in actin cytoskeleton regulation, vesicle trafficking, or phospholipid modification. Proteins enriched in the Wnt-activated Fzd7 interactome that are without established roles in Wnt signaling warrant further examination.

16.
Front Cell Dev Biol ; 9: 627429, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33634128

RESUMO

Mesenchymal stem cells (MSCs) give rise to adipocytes, osteocytes, and chondrocytes and reside in various tissues, including bone marrow and adipose tissue. The differentiation choices of MSCs are controlled by several signaling pathways, including the Wnt/ß-catenin signaling. When MSCs undergo adipogenesis, they first differentiate into preadipocytes, a proliferative adipocyte precursor cell, after which they undergo terminal differentiation into mature adipocytes. These two steps are controlled by the Wnt/ß-catenin pathway, in such a way that when signaling is abrogated, the next step in adipocyte differentiation can start. This sequence suggests that the main role of Wnt/ß-catenin signaling is to suppress differentiation while increasing MSC and preadipocytes cell mass. During later steps of MSC differentiation, however, active Wnt signaling can promote osteogenesis instead of keeping the MSCs undifferentiated and proliferative. The exact mechanisms behind the various functions of Wnt signaling remain elusive, although recent research has revealed that during lineage commitment of MSCs into preadipocytes, Wnt signaling is inactivated by endogenous Wnt inhibitors. In part, this process is regulated by histone-modifying enzymes, which can lead to increased or decreased Wnt gene expression. The role of Wnt in adipogenesis, as well as in osteogenesis, has implications for metabolic diseases since Wnt signaling may serve as a therapeutic target.

17.
Mol Biol Cell ; 31(13): 1425-1436, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32320321

RESUMO

The Wnt pathway is a key intercellular signaling cascade that regulates development, tissue homeostasis, and regeneration. However, gaps remain in our understanding of the molecular events that take place between ligand-receptor binding and target gene transcription. We used a novel tool for quantitative, real-time assessment of endogenous pathway activation, measured in single cells, to answer an unresolved question in the field-whether receptor endocytosis is required for Wnt signal transduction. We combined knockdown or knockout of essential components of clathrin-mediated endocytosis with quantitative assessment of Wnt signal transduction in mouse embryonic stem cells (mESCs). Disruption of clathrin-mediated endocytosis did not affect accumulation and nuclear translocation of ß-catenin, as measured by single-cell live imaging of endogenous ß-catenin, and subsequent target gene transcription. Disruption of another receptor endocytosis pathway, caveolin-mediated endocytosis, did not affect Wnt pathway activation in mESCs. Additional results in multiple cell lines support that endocytosis is not a requirement for Wnt signal transduction. We show that off-target effects of a drug used to inhibit endocytosis may be one source of the discrepancy among reports on the role of endocytosis in Wnt signaling.


Assuntos
Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Células-Tronco Embrionárias , Endocitose , Regulação da Expressão Gênica , Camundongos , Transporte Proteico
18.
Cell Stem Cell ; 27(5): 840-851.e6, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32818433

RESUMO

Modulation of Wnt signaling has untapped potential in regenerative medicine due to its essential functions in stem cell homeostasis. However, Wnt lipidation and Wnt-Frizzled (Fzd) cross-reactivity have hindered translational Wnt applications. Here, we designed and engineered water-soluble, Fzd subtype-specific "next-generation surrogate" (NGS) Wnts that hetero-dimerize Fzd and Lrp6. NGS Wnt supports long-term expansion of multiple different types of organoids, including kidney, colon, hepatocyte, ovarian, and breast. NGS Wnts are superior to Wnt3a conditioned media in organoid expansion and single-cell organoid outgrowth. Administration of Fzd subtype-specific NGS Wnt in vivo reveals that adult intestinal crypt proliferation can be promoted by agonism of Fzd5 and/or Fzd8 receptors, while a broad spectrum of Fzd receptors can induce liver zonation. Thus, NGS Wnts offer a unified organoid expansion protocol and a laboratory "tool kit" for dissecting the functions of Fzd subtypes in stem cell biology.


Assuntos
Receptores Frizzled , Organoides , Hepatócitos , Células-Tronco , Via de Sinalização Wnt
19.
J Am Heart Assoc ; 5(3): e002457, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-27068625

RESUMO

BACKGROUND: The adult mammalian heart responds to cardiac injury by formation of persistent fibrotic scar that eventually leads to heart failure. In contrast, the neonatal mammalian heart reacts to injury by the development of transient fibrotic tissue that is eventually replaced by regenerated cardiomyocytes. How fibrosis occurs in the neonatal mammalian heart remains unknown. To start elucidating the molecular underpinnings of neonatal cardiac fibrosis, we investigated Wnt signaling in the neonatal heart after cryoinjury. METHODS AND RESULTS: Using expression of the Wnt target gene Axin2 as an indicator of Wnt/ß-catenin signaling activation, we discovered that epicardial cells in the ventricles are responsive to Wnt in the uninjured neonatal heart. Lineage-tracing studies of these Wnt-responsive epicardial cells showed that they undergo epithelial-to-mesenchymal transition and infiltrate into the subepicardial space and exhibit fibroblast phenotypes after injury. In addition, we showed that-similar to adult ischemic injury-neonatal cryoinjury results in activation of Wnt signaling in cardiac fibroblasts near injured areas. Furthermore, through in situ hybridization of all 19 Wnt ligands in injured neonatal hearts, we observed upregulation of Wnt ligands (Wnt2b, Wnt5a, and Wnt9a) that had not been implicated in the adult cardiac injury response. CONCLUSIONS: These results demonstrate that cryoinjury in neonatal heart leads to the formation of fibrotic tissue that involves Wnt-responsive epicardial cells undergoing epithelial-to-mesenchymal transition to give rise to fibroblasts and activation of Wnt signaling in resident cardiac fibroblasts.


Assuntos
Criocirurgia , Transição Epitelial-Mesenquimal , Fibroblastos/metabolismo , Traumatismos Cardíacos/metabolismo , Pericárdio/metabolismo , Via de Sinalização Wnt , Animais , Animais Recém-Nascidos , Proteína Axina/genética , Proteína Axina/metabolismo , Linhagem da Célula , Temperatura Baixa , Modelos Animais de Doenças , Fibroblastos/patologia , Fibrose , Regulação da Expressão Gênica no Desenvolvimento , Traumatismos Cardíacos/patologia , Camundongos Transgênicos , Pericárdio/lesões , Pericárdio/patologia , Fenótipo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA