Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Cell Sci ; 135(21)2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36217793

RESUMO

The gene mutated in colorectal cancer (MCC) encodes a coiled-coil protein implicated, as its name suggests, in the pathogenesis of hereditary human colon cancer. To date, however, the contributions of MCC to intestinal homeostasis and disease remain unclear. Here, we examine the subcellular localization of MCC, both at the mRNA and protein levels, in the adult intestinal epithelium. Our findings reveal that Mcc transcripts are restricted to proliferating crypt cells, including Lgr5+ stem cells, where the Mcc protein is distinctly associated with the centrosome. Upon intestinal cellular differentiation, Mcc is redeployed to the apical domain of polarized villus cells where non-centrosomal microtubule organizing centers (ncMTOCs) are positioned. Using intestinal organoids, we show that the shuttling of the Mcc protein depends on phosphorylation by casein kinases 1δ and ε, which are critical modulators of WNT signaling. Together, our findings support a role for MCC in establishing and maintaining the cellular architecture of the intestinal epithelium as a component of both the centrosome and ncMTOC.


Assuntos
Centrossomo , Centro Organizador dos Microtúbulos , Humanos , Centro Organizador dos Microtúbulos/metabolismo , Centrossomo/metabolismo , Intestinos , Diferenciação Celular , Proteínas/metabolismo , Mucosa Intestinal/metabolismo
2.
Nature ; 561(7722): E7, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29977062

RESUMO

In this Letter, the surname of author Lena Vlaminck was misspelled 'Vlaeminck'. In addition, author Kris Vleminckx should have been associated with affiliation 16 (Center for Medical Genetics, Ghent University, Ghent, Belgium). These have been corrected online.

3.
Nature ; 557(7706): 564-569, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29769720

RESUMO

The four R-spondin secreted ligands (RSPO1-RSPO4) act via their cognate LGR4, LGR5 and LGR6 receptors to amplify WNT signalling1-3. Here we report an allelic series of recessive RSPO2 mutations in humans that cause tetra-amelia syndrome, which is characterized by lung aplasia and a total absence of the four limbs. Functional studies revealed impaired binding to the LGR4/5/6 receptors and the RNF43 and ZNRF3 transmembrane ligases, and reduced WNT potentiation, which correlated with allele severity. Unexpectedly, however, the triple and ubiquitous knockout of Lgr4, Lgr5 and Lgr6 in mice did not recapitulate the known Rspo2 or Rspo3 loss-of-function phenotypes. Moreover, endogenous depletion or addition of exogenous RSPO2 or RSPO3 in triple-knockout Lgr4/5/6 cells could still affect WNT responsiveness. Instead, we found that the concurrent deletion of rnf43 and znrf3 in Xenopus embryos was sufficient to trigger the outgrowth of supernumerary limbs. Our results establish that RSPO2, without the LGR4/5/6 receptors, serves as a direct antagonistic ligand to RNF43 and ZNRF3, which together constitute a master switch that governs limb specification. These findings have direct implications for regenerative medicine and WNT-associated cancers.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Extremidades/embriologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Deformidades Congênitas dos Membros/genética , Receptores Acoplados a Proteínas G/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Animais , Proteínas de Ligação a DNA/metabolismo , Feminino , Fibroblastos , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Camundongos , Proteínas Oncogênicas/antagonistas & inibidores , Proteínas Oncogênicas/metabolismo , Fenótipo , Receptores Acoplados a Proteínas G/deficiência , Ubiquitina-Proteína Ligases/metabolismo , Xenopus/genética
4.
Am J Med Genet A ; 188(6): 1752-1760, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35212137

RESUMO

Huriez syndrome (HRZ, OMIM181600) is a rare genodermatosis characterized by scleroatrophic hands and feet, hypoplastic nails, palmoplantar keratoderma, and predisposition to cutaneous squamous cell carcinoma (cSCC). We report herein three HRZ families from Croatia, the Netherlands, and Germany. Deep sequencing followed by Sanger validation, confirmed the presence of germline causative SMARCAD1 heterozygous pathogenic variants. All seven HRZ patients displayed hypohidrosis, adermatoglyphia, and one patient developed cSCC at 32 years of age. Two novel monoallelic germline mutations were identified which are predicted to disrupt the first exon-intron boundary of the skin-specific SMARCAD1 isoform. On the basis of phenotypic and genotypic convergence with Adermatoglyphia (OMIM136000) and Basan syndrome (OMIM129200), our results lend credence to the notion that these three Mendelian disorders are allelic. We propose adding Huriez syndrome to the previously suggested SMARCAD syndrome designation, which was originally invoked to describe the spectrum of monogenic disorders between Adermatoglyphia and Basan syndrome.


Assuntos
Carcinoma de Células Escamosas , Ceratodermia Palmar e Plantar , Neoplasias Cutâneas , Carcinoma de Células Escamosas/complicações , DNA Helicases/genética , Displasia Ectodérmica , Humanos , Ceratodermia Palmar e Plantar/genética , Ceratose , Unhas Malformadas , Esclerodermia Localizada , Dermatopatias Genéticas , Neoplasias Cutâneas/etiologia , Síndrome
5.
EMBO J ; 34(18): 2321-33, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26240067

RESUMO

Wnt pathway deregulation is a common characteristic of many cancers. Only colorectal cancer predominantly harbours mutations in APC, whereas other cancer types (hepatocellular carcinoma, solid pseudopapillary tumours of the pancreas) have activating mutations in ß-catenin (CTNNB1). We have compared the dynamics and the potency of ß-catenin mutations in vivo. Within the murine small intestine (SI), an activating mutation of ß-catenin took much longer to achieve Wnt deregulation and acquire a crypt-progenitor cell (CPC) phenotype than Apc or Gsk3 loss. Within the colon, a single activating mutation of ß-catenin was unable to drive Wnt deregulation or induce the CPC phenotype. This ability of ß-catenin mutation to differentially transform the SI versus the colon correlated with higher expression of E-cadherin and a higher number of E-cadherin:ß-catenin complexes at the membrane. Reduction in E-cadherin synergised with an activating mutation of ß-catenin resulting in a rapid CPC phenotype within the SI and colon. Thus, there is a threshold of ß-catenin that is required to drive transformation, and E-cadherin can act as a buffer to sequester mutated ß-catenin.


Assuntos
Caderinas/metabolismo , Transformação Celular Neoplásica , Neoplasias do Colo , Mutação , Proteínas de Neoplasias , Via de Sinalização Wnt , beta Catenina , Animais , Caderinas/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
Gut ; 63(8): 1345-54, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24841573

RESUMO

Limited pools of resident adult stem cells are critical effectors of epithelial renewal in the intestine throughout life. Recently, significant progress has been made regarding the isolation and in vitro propagation of fetal and adult intestinal stem cells in mammals. It is now possible to generate ever-expanding, three-dimensional epithelial structures in culture that closely parallel the in vivo epithelium of the intestine. Growing such organotypic epithelium ex vivo facilitates a detailed description of endogenous niche factors or stem-cell characteristics, as they can be monitored in real time. Accordingly, this technology has already greatly contributed to our understanding of intestinal adult stem-cell renewal and differentiation. Transplanted organoids have also been proven to readily integrate into, and effect the long-term repair of, mouse colonic epithelia in vivo, establishing the organoid culture as a promising tool for adult stem cell/gene therapy. In another exciting development, novel genome-editing techniques have been successfully employed to functionally repair disease loci in cultured intestinal stem cells from human patients with a hereditary defect. It is anticipated that this technology will be instrumental in exploiting the regenerative medicine potential of human intestinal stem cells for treating human disorders in the intestinal tract and for creating near-physiological ex vivo models of human gastrointestinal disease.


Assuntos
Mucosa Intestinal/fisiologia , Organoides/fisiologia , Células-Tronco/fisiologia , Técnicas de Cultura de Tecidos , Animais , Diferenciação Celular , Pesquisa Fetal , Humanos , Mucosa Intestinal/citologia , Intestino Delgado/fisiologia , Organoides/anatomia & histologia , Receptores Acoplados a Proteínas G/análise , Células-Tronco/química , Via de Sinalização Wnt
7.
bioRxiv ; 2023 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-38106050

RESUMO

Targeting cancer stem cells (CSCs) is crucial for effective cancer treatment 1 . However, the molecular mechanisms underlying resistance to LGR5 + CSCs depletion in colorectal cancer (CRC) 2,3 remain largely elusive. Here, we unveil the existence of a primitive cell state dubbed the oncofetal (OnF) state, which works in tandem with the LGR5 + stem cells (SCs) to fuel tumor evolution in CRC. OnF cells emerge early during intestinal tumorigenesis and exhibit features of lineage plasticity. Normally suppressed by the Retinoid X Receptor (RXR) in mature SCs, the OnF program is triggered by genetic deletion of the gatekeeper APC. We demonstrate that diminished RXR activity unlocks an epigenetic circuity governed by the cooperative action of YAP and AP1, leading to OnF reprogramming. This high-plasticity state is inherently resistant to conventional chemotherapies and its adoption by LGR5 + CSCs enables them to enter a drug-tolerant state. Furthermore, through phenotypic tracing and ablation experiments, we uncover a functional redundancy between the OnF and stem cell (SC) states and show that targeting both cellular states is essential for sustained tumor regression in vivo . Collectively, these findings establish a mechanistic foundation for developing effective combination therapies with enduring impact on CRC treatment.

8.
Int J Cancer ; 131(10): 2242-52, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22344573

RESUMO

Cellular hierarchies and signals that govern stemness and differentiation of intestinal adenoma cells are not well defined. In this study, we used organotypic culture to investigate the impact of ß-catenin and BMP signals in cells that form intestinal adenoma in the mouse. We found that activation of ß-catenin signaling by loss of APC or transgenic induction of oncogenic mutant ß-catenin (Ctnnb1(mut) ) initiates the conversion of untransformed intestinal cells to tumor cells. These tumor cells display cancer stem cell (CSC) traits such as increased expression of the CSC markers Cd133 and Cd44, a high capacity for self-renewal and unlimited proliferative potential. Subsequent inactivation of transgenic Ctnnb1(mut) results in the reversion of tumor cells to normal intestinal stem cells, which immediately reinstall the cellular hierarchy of the normal intestinal epithelium. Our data demonstrate that oncogenic activation of ß-catenin signaling initiates the early steps of intestinal cellular transformation in the absence of irreversible genetic or epigenetic changes. Interestingly, we found that tumor cells in culture and in adenoma produce BMP4, which counteracts CSC-like traits by initiating irreversible cellular differentiation and loss of self-renewal capacity. We conclude that the opposition of stemness-maintaining oncogenic ß-catenin signals and autocrine differentiating BMP signals within the adenoma cell provides a rationale for the formation of cellular hierarchies in intestinal adenoma and may serve to limit adenoma growth.


Assuntos
Adenoma/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/patologia , Transdução de Sinais , Proteínas Wnt/metabolismo , Adenoma/genética , Animais , Proteínas Morfogenéticas Ósseas/genética , Diferenciação Celular/genética , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Transgênicos , Mutação , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Esferoides Celulares , Células Tumorais Cultivadas , beta Catenina/genética , beta Catenina/metabolismo
9.
Elife ; 92020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32324134

RESUMO

During kidney development, WNT/ß-catenin signalling has to be tightly controlled to ensure proliferation and differentiation of nephron progenitor cells. Here, we show in mice that the signalling molecules RSPO1 and RSPO3 act in a functionally redundant manner to permit WNT/ß-catenin signalling and their genetic deletion leads to a rapid decline of nephron progenitors. By contrast, tissue specific deletion in cap mesenchymal cells abolishes mesenchyme to epithelial transition (MET) that is linked to a loss of Bmp7 expression, absence of SMAD1/5 phosphorylation and a concomitant failure to activate Lef1, Fgf8 and Wnt4, thus explaining the observed phenotype on a molecular level. Surprisingly, the full knockout of LGR4/5/6, the cognate receptors of R-spondins, only mildly affects progenitor numbers, but does not interfere with MET. Taken together our data demonstrate key roles for R-spondins in permitting stem cell maintenance and differentiation and reveal Lgr-dependent and independent functions for these ligands during kidney formation.


Kidneys filter waste out of the bloodstream to produce urine. Each kidney contains many structures called nephrons which separate the waste from the blood. The number of nephrons in a kidney varies between people, and those with low numbers have a higher risk of chronic kidney disease. Nephrons are formed before birth from a specific group of so-called progenitor cells. Each of these cells can either divide to make others like itself, or it can specialize to make nephron cells. At the end of embryonic kidney development, all the progenitor cells become nephron cells. Cells that specialize to become part of a nephron first go through a change called a mesenchyme-to-epithelial transition. Epithelial cells move less than mesenchymal cells, and also develop a clear structure where the two ends of the cell adapt to different roles. Evidence suggests that a cell communication process called WNT/ß-catenin signaling controls this transition. Yet the details of how this transition is controlled are not fully understood. One way to activate WNT/ß-catenin signaling is with R-spondin proteins, which have been found in developing kidneys. Vidal et al. studied R-spondins during the embryonic development of kidneys in mice. Removing R-spondins stopped the progenitor cells from producing more of themselves and increased the number that died. The R-spondins were also needed for the progenitor cells to specialize as nephron cells through the mesenchyme-to-epithelial transition. Further results revealed that R-spondins activate WNT/ß-catenin signaling in these cells, even though the proteins that usually act as R-spondin receptors (called LGR4/5/6) could be removed without affecting the results. This suggests that R-spondins interact with different receptor proteins during kidney development. These findings highlight the role of R-spondins and WNT/ß-catenin signaling in kidney development. Future studies will seek the receptor proteins that R-spondins interact with in kidneys. They may also look to understand how R-spondins balance their different roles in progenitor cells and during cell specialization. These results in mice could also be extended to determine their relevance in human health and disease, including chronic kidney disease, which is responsible for more deaths than breast or prostate cancer.


Assuntos
Rim/embriologia , Néfrons/citologia , Células-Tronco/citologia , Trombospondinas/fisiologia , Animais , Diferenciação Celular , Transição Epitelial-Mesenquimal , Feminino , Camundongos , Néfrons/embriologia , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia , Via de Sinalização Wnt
10.
Nat Cell Biol ; 19(7): 774-786, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28581476

RESUMO

The daily renewal of the corpus epithelium is fuelled by adult stem cells residing within tubular glands, but the identity of these stem cells remains controversial. Lgr5 marks homeostatic stem cells and 'reserve' stem cells in multiple tissues. Here, we report Lgr5 expression in a subpopulation of chief cells in mouse and human corpus glands. Using a non-variegated Lgr5-2A-CreERT2 mouse model, we show by lineage tracing that Lgr5-expressing chief cells do not behave as corpus stem cells during homeostasis, but are recruited to function as stem cells to effect epithelial renewal following injury by activating Wnt signalling. Ablation of Lgr5+ cells severely impairs epithelial homeostasis in the corpus, indicating an essential role for these Lgr5+ cells in maintaining the homeostatic stem cell pool. We additionally define Lgr5+ chief cells as a major cell-of-origin of gastric cancer. These findings reveal clinically relevant insights into homeostasis, repair and cancer in the corpus.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Celulas Principais Gástricas/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células Parietais Gástricas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Regeneração , Neoplasias Gástricas/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem da Célula , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Celulas Principais Gástricas/efeitos dos fármacos , Celulas Principais Gástricas/patologia , Regulação da Expressão Gênica , Genótipo , Humanos , Camundongos Transgênicos , Células-Tronco Neoplásicas/patologia , Organoides , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/patologia , Fenótipo , Regeneração/efeitos dos fármacos , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Tamoxifeno/toxicidade , Técnicas de Cultura de Tecidos , Via de Sinalização Wnt
11.
Sci Rep ; 6: 21923, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26916214

RESUMO

Using in-vivo lineage tracing data we quantified clonal expansion as well as proliferation and differentiation of the Lgr5-positive stem cell population in pyloric gastric glands. Fitting clone expansion models, we estimated that there are five effective Lgr5-positive cells able to give rise to monoclonal glands by replacing each other following a pattern of neutral drift dynamics. This analysis is instrumental to assess stem cell performance; however, stem cell proliferation is not quantified by clone expansion analysis. We identified a suitable mathematical model to quantify proliferation and differentiation of the Lgr5-positive population. As expected for populations in steady-state, the proliferation rate of the Lgr5-positive population was equal to its rate of differentiation. This rate was significantly faster than the rate at which effective cells are replaced, estimated by modelling clone expansion/contraction. This suggests that the majority of Lgr5-positive cell divisions serve to renew epithelial cells and only few result in the effective replacement of a neighbour to effect expansion to the entire gland. The application of the model under altered situations with uncoupled differentiation and proliferation was demonstrated. This methodology represents a valuable tool for quantifying stem cell performance in homeostasis and importantly for deciphering altered stem cell behaviour in disease.


Assuntos
Diferenciação Celular , Proliferação de Células , Epitélio/metabolismo , Piloro/metabolismo , Receptores Acoplados a Proteínas G , Células-Tronco/metabolismo , Animais , Epitélio/fisiologia , Camundongos , Modelos Biológicos , Piloro/fisiologia , Células-Tronco/fisiologia
12.
Oncotarget ; 6(20): 17968-80, 2015 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-26255629

RESUMO

The tumour suppressor p53 is regulated primarily at the protein level. In normal tissues its levels are maintained at a very low level by the action of specific E3 ligases and the ubiquitin proteosome pathway. The mutant p53 protein contributes to transformation, metastasis and drug resistance. High levels of mutant p53 can be found in tumours and the accumulation of mutant p53 has previously been reported in pathologically normal cells in human skin. We show for the first time that similarly elevated levels of mutant p53 can be detected in apparently normal cells in a mutant p53 knock-in mouse model. In fact, in the small intestine, mutant p53 spontaneously accumulates in a manner dependent on gene dosage and cell type. Mutant p53 protein is regulated similarly to wild type p53, which can accumulate rapidly after induction by ionising radiation or Mdm2 inhibitors, however, the clearance of mutant p53 protein is much slower than wild type p53. The accumulation of the protein in the murine small intestine is limited to the cycling, crypt base columnar cells and proliferative zone and is lost as the cells differentiate and exit the cell cycle. Loss of Mdm2 results in even higher levels of p53 expression but p53 is still restricted to proliferating cells in the small intestine. Therefore, the small intestine of these p53 mutant mice is an experimental system in which we can dissect the molecular pathways leading to p53 accumulation, which has important implications for cancer prevention and therapy.


Assuntos
Ciclo Celular , Proliferação de Células , Intestino Delgado/metabolismo , Mutação , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Fatores Etários , Animais , Diferenciação Celular , Dano ao DNA , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Genótipo , Intestino Delgado/diagnóstico por imagem , Intestino Delgado/efeitos dos fármacos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Radiografia , Fatores de Tempo , Técnicas de Cultura de Tecidos
13.
Nat Cell Biol ; 16(8): 745-57, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24997521

RESUMO

The ovary surface epithelium (OSE) undergoes ovulatory tear and remodelling throughout life. Resident stem cells drive such tissue homeostasis in many adult epithelia, but their existence in the ovary has not been definitively proven. Lgr5 marks stem cells in multiple epithelia. Here we use reporter mice and single-molecule fluorescent in situ hybridization to document candidate Lgr5(+) stem cells in the mouse ovary and associated structures. Lgr5 is broadly expressed during ovary organogenesis, but becomes limited to the OSE in neonate life. In adults, Lgr5 expression is predominantly restricted to proliferative regions of the OSE and mesovarian-fimbria junctional epithelia. Using in vivo lineage tracing, we identify embryonic and neonate Lgr5(+) populations as stem/progenitor cells contributing to the development of the OSE cell lineage, as well as epithelia of the mesovarian ligament and oviduct/fimbria. Adult Lgr5(+) populations maintain OSE homeostasis and ovulatory regenerative repair in vivo. Thus, Lgr5 marks stem/progenitor cells of the ovary and tubal epithelia.


Assuntos
Tubas Uterinas/citologia , Tubas Uterinas/metabolismo , Ovário/citologia , Ovário/metabolismo , Receptores Acoplados a Proteínas G/genética , Células-Tronco Adultas/metabolismo , Animais , Animais Recém-Nascidos , Linhagem da Célula/genética , Células-Tronco Embrionárias/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Tubas Uterinas/crescimento & desenvolvimento , Feminino , Expressão Gênica , Marcadores Genéticos , Homeostase/genética , Humanos , Hibridização in Situ Fluorescente , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Ovário/crescimento & desenvolvimento , Regeneração/genética , Transcriptoma
14.
Cell Rep ; 5(2): 349-56, 2013 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-24209744

RESUMO

The pyloric epithelium continuously self-renews throughout life, driven by limited reservoirs of resident Lgr5+ adult stem cells. Here, we characterize the population dynamics of these stem cells during epithelial homeostasis. Using a clonal fate-mapping strategy, we demonstrate that multiple Lgr5+ cells routinely contribute to epithelial renewal in the pyloric gland and, similar to what was previously observed in the intestine, a balanced homeostasis of the glandular epithelium and stem cell pools is predominantly achieved via neutral competition between symmetrically dividing Lgr5+ stem cells. Additionally, we document a lateral expansion of stem cell clones via gland fission under nondamage conditions. These findings represent a major advance in our basic understanding of tissue homeostasis in the stomach and form the foundation for identifying altered stem cell behavior during gastric disease.


Assuntos
Células-Tronco Adultas/metabolismo , Piloro/citologia , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco Adultas/citologia , Animais , Linhagem da Célula , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Homeostase , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética
15.
Cell Rep ; 2(3): 540-52, 2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-22999937

RESUMO

Multipotent stem cells and their lineage-restricted progeny drive nephron formation within the developing kidney. Here, we document expression of the adult stem cell marker Lgr5 in the developing kidney and assess the stem/progenitor identity of Lgr5(+ve) cells via in vivo lineage tracing. The appearance and localization of Lgr5(+ve) cells coincided with that of the S-shaped body around embryonic day 14. Lgr5 expression remained restricted to cell clusters within developing nephrons in the cortex until postnatal day 7, when expression was permanently silenced. In vivo lineage tracing identified Lgr5 as a marker of a stem/progenitor population within nascent nephrons dedicated to generating the thick ascending limb of Henle's loop and distal convoluted tubule. The Lgr5 surface marker and experimental models described here will be invaluable for deciphering the contribution of early nephron stem cells to developmental defects and for isolating human nephron progenitors as a prerequisite to evaluating their therapeutic potential.


Assuntos
Linhagem da Célula/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Alça do Néfron/embriologia , Receptores Acoplados a Proteínas G/biossíntese , Células-Tronco/metabolismo , Animais , Humanos , Córtex Renal/citologia , Córtex Renal/embriologia , Alça do Néfron/citologia , Camundongos , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/genética , Células-Tronco/citologia
16.
PLoS One ; 6(8): e23381, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21853123

RESUMO

In tumor cells, stepwise oncogenic deregulation of signaling cascades induces alterations of cellular morphology and promotes the acquisition of malignant traits. Here, we identified a set of 21 genes, including FGF9, as determinants of tumor cell morphology by an RNA interference phenotypic screen in SW480 colon cancer cells. Using a panel of small molecular inhibitors, we subsequently established phenotypic effects, downstream signaling cascades, and associated gene expression signatures of FGF receptor signals. We found that inhibition of FGF signals induces epithelial cell adhesion and loss of motility in colon cancer cells. These effects are mediated via the mitogen-activated protein kinase (MAPK) and Rho GTPase cascades. In agreement with these findings, inhibition of the MEK1/2 or JNK cascades, but not of the PI3K-AKT signaling axis also induced epithelial cell morphology. Finally, we found that expression of FGF9 was strong in a subset of advanced colon cancers, and overexpression negatively correlated with patients' survival. Our functional and expression analyses suggest that FGF receptor signals can contribute to colon cancer progression.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Progressão da Doença , Fatores de Crescimento de Fibroblastos/metabolismo , Testes Genéticos , Interferência de RNA , Transdução de Sinais , Animais , Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias do Colo/metabolismo , Epitélio/metabolismo , Epitélio/patologia , Fatores de Crescimento de Fibroblastos/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Neoplásicos/genética , Humanos , Sistema de Sinalização das MAP Quinases/genética , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Fenótipo , Transdução de Sinais/genética , Análise de Sobrevida , Proteínas rho de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA