Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(52): 26599-26605, 2019 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-31843916

RESUMO

Cycling intestinal Lgr5+ stem cells are intermingled with their terminally differentiated Paneth cell daughters at crypt bottoms. Paneth cells provide multiple secreted (e.g., Wnt, EGF) as well as surface-bound (Notch ligand) niche signals. Here we show that ablation of Paneth cells in mice, using a diphtheria toxin receptor gene inserted into the P-lysozyme locus, does not affect the maintenance of Lgr5+ stem cells. Flow cytometry, single-cell sequencing, and histological analysis showed that the ablated Paneth cells are replaced by enteroendocrine and tuft cells. As these cells physically occupy Paneth cell positions between Lgr5 stem cells, they serve as an alternative source of Notch signals, which are essential for Lgr5+ stem cell maintenance. Our combined in vivo results underscore the adaptive flexibility of the intestine in maintaining normal tissue homeostasis.

2.
Proc Natl Acad Sci U S A ; 115(52): E12245-E12254, 2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30530645

RESUMO

The significance of cardiac stem cell (CSC) populations for cardiac regeneration remains disputed. Here, we apply the most direct definition of stem cell function (the ability to replace lost tissue through cell division) to interrogate the existence of CSCs. By single-cell mRNA sequencing and genetic lineage tracing using two Ki67 knockin mouse models, we map all proliferating cells and their progeny in homoeostatic and regenerating murine hearts. Cycling cardiomyocytes were only robustly observed in the early postnatal growth phase, while cycling cells in homoeostatic and damaged adult myocardium represented various noncardiomyocyte cell types. Proliferative postdamage fibroblasts expressing follistatin-like protein 1 (FSTL1) closely resemble neonatal cardiac fibroblasts and form the fibrotic scar. Genetic deletion of Fstl1 in cardiac fibroblasts results in postdamage cardiac rupture. We find no evidence for the existence of a quiescent CSC population, for transdifferentiation of other cell types toward cardiomyocytes, or for proliferation of significant numbers of cardiomyocytes in response to cardiac injury.


Assuntos
Proliferação de Células , Traumatismos Cardíacos/fisiopatologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Traumatismos Cardíacos/genética , Traumatismos Cardíacos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Gravidez , Células-Tronco/citologia , Células-Tronco/metabolismo
3.
Cell ; 175(6): 1591-1606.e19, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30500538

RESUMO

The mammalian liver possesses a remarkable regenerative ability. Two modes of damage response have been described: (1) The "oval cell" response emanates from the biliary tree when all hepatocytes are affected by chronic liver disease. (2) A massive, proliferative response of mature hepatocytes occurs upon acute liver damage such as partial hepatectomy (PHx). While the oval cell response has been captured in vitro by growing organoids from cholangiocytes, the hepatocyte proliferative response has not been recapitulated in culture. Here, we describe the establishment of a long-term 3D organoid culture system for mouse and human primary hepatocytes. Organoids can be established from single hepatocytes and grown for multiple months, while retaining key morphological, functional and gene expression features. Transcriptional profiles of the organoids resemble those of proliferating hepatocytes after PHx. Human hepatocyte organoids proliferate extensively after engraftment into mice and thus recapitulate the proliferative damage-response of hepatocytes.


Assuntos
Proliferação de Células , Hepatócitos/metabolismo , Organoides/metabolismo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Hepatócitos/citologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Organoides/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Tempo
4.
Nat Cell Biol ; 20(8): 909-916, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30038251

RESUMO

Enteroendocrine cells (EECs) control a wide range of physiological processes linked to metabolism1. We show that EEC hormones are differentially expressed between crypts (for example, Glp1) and villi (for example, secretin). As demonstrated by single-cell mRNA sequencing using murine Lgr5+ cell-derived organoids, BMP4 signals alter the hormone expression profiles of individual EECs to resemble those found in the villus. Accordingly, BMP4 induces hormone switching of EECs migrating up the crypt-villus axis in vivo. Our findings imply that EEC lineages in the small intestine exhibit a more flexible hormone repertoire than previously proposed. We also describe a protocol to generate human EECs in organoids and demonstrate a similar regulation of hormone expression by BMP signalling. These findings establish alternative strategies to target EECs with therapeutically relevant hormone production through BMP modulation.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Movimento Celular/efeitos dos fármacos , Células Enteroendócrinas/metabolismo , Hormônios Gastrointestinais/metabolismo , Intestino Delgado/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Humanos , Intestino Delgado/citologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Técnicas de Cultura de Tecidos
5.
Proc Natl Acad Sci U S A ; 113(42): 11859-11864, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27708166

RESUMO

Current mouse models for colorectal cancer often differ significantly from human colon cancer, being largely restricted to the small intestine. Here, we aim to develop a colon-specific inducible mouse model that can faithfully recapitulate human colon cancer initiation and progression. Carbonic anhydrase I (Car1) is a gene expressed uniquely in colonic epithelial cells. We generated a colon-specific inducible Car1CreER knock-in (KI) mouse with broad Cre activity in epithelial cells of the proximal colon and cecum. Deletion of the tumor suppressor gene Apc using the Car1CreER KI caused tumor formation in the cecum but did not yield adenomas in the proximal colon. Mutation of both Apc and Kras yielded microadenomas in both the cecum and the proximal colon, which progressed to macroadenomas with significant morbidity. Aggressive carcinomas with some invasion into lymph nodes developed upon combined induction of oncogenic mutations of Apc, Kras, p53, and Smad4 Importantly, no adenomas were observed in the small intestine. Additionally, we observed tumors from differentiated Car1-expressing cells with Apc/Kras mutations, suggesting that a top-down model of intestinal tumorigenesis can occur with multiple mutations. Our results establish the Car1CreER KI as a valuable mouse model to study colon-specific tumorigenesis and metastasis as well as cancer-cell-of-origin questions.


Assuntos
Neoplasias do Colo/etiologia , Regulação da Expressão Gênica , Integrases/genética , Camundongos Transgênicos , Adenoma/etiologia , Adenoma/metabolismo , Adenoma/patologia , Animais , Biomarcadores Tumorais , Anidrase Carbônica I/genética , Anidrase Carbônica I/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Progressão da Doença , Ativação Enzimática , Técnicas de Introdução de Genes , Marcação de Genes , Genes APC , Genes ras , Loci Gênicos , Humanos , Imuno-Histoquímica , Integrases/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Mutação , Especificidade de Órgãos/genética , Pesquisa
6.
Proc Natl Acad Sci U S A ; 113(37): E5399-407, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27573849

RESUMO

Leucine-rich repeat-containing G-protein coupled receptor 5-positive (Lgr5(+)) stem cells reside at crypt bottoms of the small and large intestine. Small intestinal Paneth cells supply Wnt3, EGF, and Notch signals to neighboring Lgr5(+) stem cells. Whereas the colon lacks Paneth cells, deep crypt secretory (DCS) cells are intermingled with Lgr5(+) stem cells at crypt bottoms. Here, we report regenerating islet-derived family member 4 (Reg4) as a marker of DCS cells. To investigate a niche function, we eliminated DCS cells by using the diphtheria-toxin receptor gene knocked into the murine Reg4 locus. Ablation of DCS cells results in loss of stem cells from colonic crypts and disrupts gut homeostasis and colon organoid growth. In agreement, sorted Reg4(+) DCS cells promote organoid formation of single Lgr5(+) colon stem cells. DCS cells can be massively produced from Lgr5(+) colon stem cells in vitro by combined Notch inhibition and Wnt activation. We conclude that Reg4(+) DCS cells serve as Paneth cell equivalents in the colon crypt niche.


Assuntos
Neoplasias do Colo/metabolismo , Proteínas de Neoplasias/genética , Receptores Acoplados a Proteínas G/genética , Células-Tronco/metabolismo , Animais , Colo/citologia , Colo/crescimento & desenvolvimento , Colo/metabolismo , Neoplasias do Colo/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Camundongos , Proteínas de Neoplasias/metabolismo , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Proteínas Associadas a Pancreatite , Celulas de Paneth/citologia , Celulas de Paneth/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Notch/genética , Nicho de Células-Tronco/genética , Células-Tronco/citologia , Via de Sinalização Wnt/genética
7.
Cell Stem Cell ; 19(2): 266-277, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27345837

RESUMO

Adult mitotic tissues like the intestine, skin, and blood undergo constant turnover throughout the life of an organism. Knowing the identity of the stem cell is crucial to understanding tissue homeostasis and its aberrations upon disease. Here we present a computational method for the derivation of a lineage tree from single-cell transcriptome data. By exploiting the tree topology and the transcriptome composition, we establish StemID, an algorithm for identifying stem cells among all detectable cell types within a population. We demonstrate that StemID recovers two known adult stem cell populations, Lgr5+ cells in the small intestine and hematopoietic stem cells in the bone marrow. We apply StemID to predict candidate multipotent cell populations in the human pancreas, a tissue with largely uncharacterized turnover dynamics. We hope that StemID will accelerate the search for novel stem cells by providing concrete markers for biological follow-up and validation.


Assuntos
Análise de Célula Única/métodos , Células-Tronco/citologia , Transcriptoma/genética , Adulto , Algoritmos , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Linhagem da Célula , Entropia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Intestinos/citologia , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Ductos Pancreáticos/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Reprodutibilidade dos Testes
8.
Cell Stem Cell ; 18(2): 203-13, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26831517

RESUMO

Intestinal crypts display robust regeneration upon injury. The relatively rare secretory precursors can replace lost stem cells, but it is unknown if the abundant enterocyte progenitors that express the Alkaline phosphate intestinal (Alpi) gene also have this capacity. We created an Alpi-IRES-CreERT2 (Alpi(CreER)) knockin allele for lineage tracing. Marked clones consist entirely of enterocytes and are all lost from villus tips within days. Genetic fate-mapping of Alpi(+) cells before or during targeted ablation of Lgr5-expressing stem cells generated numerous long-lived crypt-villus "ribbons," indicative of dedifferentiation of enterocyte precursors into Lgr5(+) stems. By single-cell analysis of dedifferentiating enterocytes, we observed the generation of Paneth-like cells and proliferative stem cells. We conclude that the highly proliferative, short-lived enterocyte precursors serve as a large reservoir of potential stem cells during crypt regeneration.


Assuntos
Linhagem da Célula , Enterócitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Biomarcadores/metabolismo , Desdiferenciação Celular , Linhagem Celular , Proliferação de Células , Enterócitos/patologia , Integrases/metabolismo , Neoplasias Intestinais/patologia , Camundongos , Mutação/genética , Organoides , Celulas de Paneth/metabolismo , Celulas de Paneth/patologia , Regeneração/genética , Análise de Célula Única , beta-Galactosidase/metabolismo
9.
Proc Natl Acad Sci U S A ; 112(24): 7548-50, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26023187

RESUMO

Rnf43 (RING finger protein 43) and Znrf3 (zinc/RING finger protein 3) (RZ) are two closely related transmembrane E3 ligases, encoded by Wnt target genes, that remove surface Wnt (wingless-int) receptors. The two genes are mutated in various human cancers. Such tumors are predicted to be hypersensitive to, yet still depend on, secreted Wnts. We previously showed that mutation of RZ in the intestine yields rapidly growing adenomas containing LGR5(+) (leucine-rich repeat-containing G-protein coupled receptor 5) stem cells and Wnt3-producing Paneth cells. We now show that removal of Paneth cells by Math1 mutation inhibits RZ(-/-) tumor formation. Similarly, deletion of Wnt3 inhibits tumorigenesis. Treatment of mice carrying RZ(-/-) intestinal neoplasia with a small molecule Wnt secretion inhibitor (porcupine inhibitor C59) strongly inhibited growth, whereas adjacent normal crypts remained intact. These results establish that paracrine Wnt secretion is an essential driver of RZ(-/-) tumor growth and imply that a therapeutic window exists for the use of porcupine inhibitors for RZ-mutant cancers.


Assuntos
Benzenoacetamidas/farmacologia , Neoplasias Intestinais/tratamento farmacológico , Proteínas de Membrana/antagonistas & inibidores , Piridinas/farmacologia , Ubiquitina-Proteína Ligases/genética , Via de Sinalização Wnt/efeitos dos fármacos , Aciltransferases , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Humanos , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Comunicação Parácrina/efeitos dos fármacos , Ubiquitina-Proteína Ligases/deficiência , Proteína Wnt3/deficiência , Proteína Wnt3/genética , Dedos de Zinco/genética
10.
Nat Commun ; 5: 5103, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-25267258

RESUMO

The transcription factor NF-κB is indispensable for intestinal immune homeostasis, but contributes to chronic inflammation and inflammatory bowel disease (IBD). A20, an inhibitor of both NF-κB and apoptotic signalling, was identified as a susceptibility gene for multiple inflammatory diseases, including IBD. Despite absence of spontaneous intestinal inflammation in intestinal epithelial cell (IEC) specific A20 knockout mice, we found additional myeloid-specific A20 deletion to synergistically drive intestinal pathology through cell-specific mechanisms. A20 ensures intestinal barrier stability by preventing cytokine-induced IEC apoptosis, while A20 prevents excessive cytokine production in myeloid cells. Combining IEC and myeloid A20 deletion induces ileitis and severe colitis, characterized by IEC apoptosis, Paneth and goblet cell loss, epithelial hyperproliferation and intestinal microbiota dysbiosis. Continuous epithelial cell death and regeneration in an inflammatory environment sensitizes cells for neoplastic transformation and the development of colorectal tumours in aged mice.


Assuntos
Cisteína Endopeptidases/metabolismo , Células Epiteliais/enzimologia , Intestinos/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Animais , Apoptose , Colite/enzimologia , Colite/genética , Colite/patologia , Colite/fisiopatologia , Cisteína Endopeptidases/genética , Células Epiteliais/citologia , Células Epiteliais/patologia , Feminino , Células Caliciformes/citologia , Células Caliciformes/enzimologia , Células Caliciformes/patologia , Homeostase , Humanos , Intestinos/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Celulas de Paneth/citologia , Celulas de Paneth/enzimologia , Celulas de Paneth/patologia , Especificidade da Espécie , Proteína 3 Induzida por Fator de Necrose Tumoral alfa
11.
Cell ; 151(7): 1595-607, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23260145

RESUMO

Most studies on TCF7L2 SNP variants in the pathogenesis of type 2 diabetes (T2D) focus on a role of the encoded transcription factor TCF4 in ß cells. Here, a mouse genetics approach shows that removal of TCF4 from ß cells does not affect their function, whereas manipulating TCF4 levels in the liver has major effects on metabolism. In Tcf7l2(-/-) mice, the immediate postnatal surge in liver metabolism does not occur. Consequently, pups die due to hypoglycemia. By combining chromatin immunoprecipitation with gene expression profiling, we identify a TCF4-controlled metabolic gene program that is acutely activated in the postnatal liver. In concordance, adult liver-specific Tcf7l2 knockout mice show reduced hepatic glucose production during fasting and display improved glucose homeostasis when maintained on high-fat diet. Furthermore, liver-specific TCF4 overexpression increases hepatic glucose production. These observations imply that TCF4 directly activates metabolic genes and that inhibition of Wnt signaling may be beneficial in metabolic disease.


Assuntos
Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Glucose/metabolismo , Fígado/metabolismo , Redes e Vias Metabólicas , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Dieta Hiperlipídica , Jejum/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Knockout , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Ativação Transcricional
12.
Nat Cell Biol ; 14(10): 1099-1104, 2012 10.
Artigo em Inglês | MEDLINE | ID: mdl-23000963

RESUMO

Lgr5+ intestinal stem cells generate enterocytes and secretory cells. Secretory lineage commitment requires Notch silencing. The Notch ligand Dll1 is expressed by a subset of immediate stem cell daughters. Lineage tracing in Dll1(GFP-ires-CreERT2) knock-in mice reveals that single Dll1(high) cells generate small, short-lived clones containing all four secretory cell types. Lineage specification thus occurs in immediate stem cell daughters through Notch lateral inhibition. Cultured Dll1(high) cells form long-lived organoids (mini-guts) on brief Wnt3A exposure. When Dll1(high) cells are genetically marked before tissue damage, stem cell tracing events occur. Thus, secretory progenitors exhibit plasticity by regaining stemness on damage.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Intestinal/metabolismo , Células-Tronco/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Linhagem da Célula , Células Cultivadas , Técnicas de Introdução de Genes , Peptídeos e Proteínas de Sinalização Intercelular/análise , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Organoides/metabolismo , Receptores Notch/antagonistas & inibidores , Receptores Notch/metabolismo , Proteína Wnt3A/farmacologia
13.
Science ; 337(6095): 730-5, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22855427

RESUMO

The concept that tumors are maintained by dedicated stem cells, the so-called cancer stem cell hypothesis, has attracted great interest but remains controversial. Studying mouse models, we provide direct, functional evidence for the presence of stem cell activity within primary intestinal adenomas, a precursor to intestinal cancer. By "lineage retracing" using the multicolor Cre-reporter R26R-Confetti, we demonstrate that the crypt stem cell marker Lgr5 (leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5) also marks a subpopulation of adenoma cells that fuel the growth of established intestinal adenomas. These Lgr5(+) cells, which represent about 5 to 10% of the cells in the adenomas, generate additional Lgr5(+) cells as well as all other adenoma cell types. The Lgr5(+) cells are intermingled with Paneth cells near the adenoma base, a pattern reminiscent of the architecture of the normal crypt niche.


Assuntos
Adenoma/patologia , Neoplasias Intestinais/patologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/fisiologia , Receptores Acoplados a Proteínas G/análise , Adenoma/genética , Adenoma/metabolismo , Animais , Biomarcadores/análise , Linhagem da Célula , Transformação Celular Neoplásica , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Genes Reporter , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Neoplasias Intestinais/genética , Camundongos , Células-Tronco Multipotentes/patologia , Células-Tronco Multipotentes/fisiologia , Celulas de Paneth/patologia , Nicho de Células-Tronco , Tamoxifeno/farmacologia , Ensaio Tumoral de Célula-Tronco
14.
Mol Cell Biol ; 32(10): 1918-27, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22393260

RESUMO

Throughout life, intestinal Lgr5+ stem cells give rise to proliferating transient amplifying cells in crypts, which subsequently differentiate into one of the five main cell types and migrate along the crypt-villus axis. These dynamic processes are coordinated by a relatively small number of evolutionarily conserved signaling pathways, which includes the Wnt signaling pathway. The DNA-binding proteins of the T-cell factor family, Tcf1/Tcf7, Lef, Tcf3/Tcf7l1, and Tcf4/Tcf7l2, constitute the downstream effectors of the Wnt signaling pathway. While Tcf4 is the major member active during embryogenesis, the role of these Wnt effectors in the homeostasis of the adult mouse intestinal epithelium is unresolved. Using Tcf1-/-, Tcf3(flox), and novel Tcf4(flox) mice, we demonstrate an essential role for Tcf4 during homeostasis of the adult mouse intestine.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Intestinos/citologia , Células-Tronco/metabolismo , Via de Sinalização Wnt , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Diferenciação Celular , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Camundongos , Células-Tronco/citologia , Fator de Transcrição 4
15.
PLoS One ; 6(8): e22616, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21826198

RESUMO

Follistatin-like 1 (Fstl1) is a secreted protein of the BMP inhibitor class. During development, expression of Fstl1 is already found in cleavage stage embryos and becomes gradually restricted to mesenchymal elements of most organs during subsequent development. Knock down experiments in chicken and zebrafish demonstrated a role as a BMP antagonist in early development. To investigate the role of Fstl1 during mouse development, a conditional Fstl1 KO allele as well as a Fstl1-GFP reporter mouse were created. KO mice die at birth from respiratory distress and show multiple defects in lung development. Also, skeletal development is affected. Endochondral bone development, limb patterning as well as patterning of the axial skeleton are perturbed in the absence of Fstl1. Taken together, these observations show that Fstl1 is a crucial regulator in BMP signalling during mouse development.


Assuntos
Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas Relacionadas à Folistatina/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Organogênese/fisiologia , Animais , Feminino , Proteínas Relacionadas à Folistatina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organogênese/genética
16.
EMBO J ; 30(6): 1104-9, 2011 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-21297579

RESUMO

Somatic cells have been proposed to be limited in the number of cell divisions they can undergo. This is thought to be a mechanism by which stem cells retain their integrity preventing disease. However, we have recently discovered intestinal crypt stem cells that persist for the lifetime of a mouse, yet divide every day. We now demonstrate biochemically that primary isolated Lgr5+ve stem cells contain significant telomerase activity. Telomerase activity rapidly decreases in the undifferentiated progeny of these stem cells and is entirely lost in differentiated villus cells. Conversely, asymmetric segregation of chromosomes has been proposed as a mechanism for stem cells to protect their genomes against damage. We determined the average cell cycle length of Lgr5+ve stem cells at 21.5 h and find that Lgr5+ve intestinal stem cells randomly segregate newly synthesized DNA strands, opposing the 'immortal strand' hypothesis.


Assuntos
Segregação de Cromossomos , Mucosa Intestinal/citologia , Mucosa Intestinal/enzimologia , Nicho de Células-Tronco , Células-Tronco/enzimologia , Células-Tronco/fisiologia , Telomerase/metabolismo , Animais , Ciclo Celular , Diferenciação Celular , Células Epiteliais/enzimologia , Células Epiteliais/fisiologia , Camundongos , Fatores de Tempo
17.
Nature ; 469(7330): 415-8, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21113151

RESUMO

Homeostasis of self-renewing small intestinal crypts results from neutral competition between Lgr5 stem cells, which are small cycling cells located at crypt bottoms. Lgr5 stem cells are interspersed between terminally differentiated Paneth cells that are known to produce bactericidal products such as lysozyme and cryptdins/defensins. Single Lgr5-expressing stem cells can be cultured to form long-lived, self-organizing crypt-villus organoids in the absence of non-epithelial niche cells. Here we find a close physical association of Lgr5 stem cells with Paneth cells in mice, both in vivo and in vitro. CD24(+) Paneth cells express EGF, TGF-α, Wnt3 and the Notch ligand Dll4, all essential signals for stem-cell maintenance in culture. Co-culturing of sorted stem cells with Paneth cells markedly improves organoid formation. This Paneth cell requirement can be substituted by a pulse of exogenous Wnt. Genetic removal of Paneth cells in vivo results in the concomitant loss of Lgr5 stem cells. In colon crypts, CD24(+) cells residing between Lgr5 stem cells may represent the Paneth cell equivalents. We conclude that Lgr5 stem cells compete for essential niche signals provided by a specialized daughter cell, the Paneth cell.


Assuntos
Intestinos/citologia , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Celulas de Paneth/citologia , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células-Tronco/citologia , Animais , Antígeno CD24/metabolismo , Contagem de Células , Proliferação de Células , Técnicas de Cocultura , Humanos , Camundongos , Celulas de Paneth/metabolismo , Nicho de Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt3
18.
Cell ; 143(1): 134-44, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20887898

RESUMO

Intestinal stem cells, characterized by high Lgr5 expression, reside between Paneth cells at the small intestinal crypt base and divide every day. We have carried out fate mapping of individual stem cells by generating a multicolor Cre-reporter. As a population, Lgr5(hi) stem cells persist life-long, yet crypts drift toward clonality within a period of 1-6 months. We have collected short- and long-term clonal tracing data of individual Lgr5(hi) cells. These reveal that most Lgr5(hi) cell divisions occur symmetrically and do not support a model in which two daughter cells resulting from an Lgr5(hi) cell division adopt divergent fates (i.e., one Lgr5(hi) cell and one transit-amplifying [TA] cell per division). The cellular dynamics are consistent with a model in which the resident stem cells double their numbers each day and stochastically adopt stem or TA fates. Quantitative analysis shows that stem cell turnover follows a pattern of neutral drift dynamics.


Assuntos
Linhagem da Célula , Intestino Delgado/citologia , Células-Tronco/citologia , Animais , Células Clonais , Camundongos , Modelos Biológicos , Receptores Acoplados a Proteínas G/metabolismo
19.
Science ; 327(5971): 1385-9, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20223988

RESUMO

Mammalian epidermis consists of three self-renewing compartments: the hair follicle, the sebaceous gland, and the interfollicular epidermis. We generated knock-in alleles of murine Lgr6, a close relative of the Lgr5 stem cell gene. Lgr6 was expressed in the earliest embryonic hair placodes. In adult hair follicles, Lgr6+ cells resided in a previously uncharacterized region directly above the follicle bulge. They expressed none of the known bulge stem cell markers. Prenatal Lgr6+ cells established the hair follicle, sebaceous gland, and interfollicular epidermis. Postnatally, Lgr6+ cells generated sebaceous gland and interfollicular epidermis, whereas contribution to hair lineages gradually diminished with age. Adult Lgr6+ cells executed long-term wound repair, including the formation of new hair follicles. We conclude that Lgr6 marks the most primitive epidermal stem cell.


Assuntos
Linhagem da Célula , Folículo Piloso/citologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Pele/citologia , Células-Tronco/citologia , Animais , Células Epidérmicas , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Cabelo/citologia , Cabelo/embriologia , Cabelo/crescimento & desenvolvimento , Folículo Piloso/embriologia , Folículo Piloso/crescimento & desenvolvimento , Camundongos , Camundongos Nus , Glândulas Sebáceas/citologia , Transdução de Sinais , Transplante de Células-Tronco , Células-Tronco/metabolismo , Cicatrização
20.
Dis Model Mech ; 3(1-2): 104-10, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20075383

RESUMO

Barrett's esophagus (BE) affects approximately 2% of the Western population and progresses to esophageal adenocarcinoma (EAC) in 0.5% of these patients each year. In BE, the stratified epithelium is replaced by an intestinal-type epithelium owing to chronic gastroduodenal reflux. Since self-renewal of intestinal crypts is driven by Notch signaling, we investigated whether this pathway was active in the proliferative crypts of BE. Immunohistochemistry confirmed the presence of an intact and activated Notch signaling pathway in metaplastic BE epithelium, but not in the normal human esophagus. Similar observations were made in two well-known human Barrett's-derived EAC cell lines, OE33 and SKGT-5. We then sought to investigate the effects of Notch inhibition by systemic treatment with a gamma-secretase inhibitor in a well-validated rodent model for BE. As we have shown previously in normal intestinal epithelium, Notch inhibition converted the proliferative Barrett's epithelial cells into terminally differentiated goblet cells, whereas the squamous epithelium remained intact. These data imply that local application of gamma-secretase inhibitors may present a simple therapeutic strategy for this increasingly common pre-malignant condition.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Esôfago de Barrett/enzimologia , Esôfago de Barrett/patologia , Epitélio/patologia , Células Caliciformes/patologia , Mitose , Animais , Biópsia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Dibenzazepinas/farmacologia , Modelos Animais de Doenças , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/metabolismo , Humanos , Metaplasia , Mitose/efeitos dos fármacos , Ratos , Receptores Notch/antagonistas & inibidores , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...