Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(12): 3056-3071.e17, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38848678

RESUMEN

The currently accepted intestinal epithelial cell organization model proposes that Lgr5+ crypt-base columnar (CBC) cells represent the sole intestinal stem cell (ISC) compartment. However, previous studies have indicated that Lgr5+ cells are dispensable for intestinal regeneration, leading to two major hypotheses: one favoring the presence of a quiescent reserve ISC and the other calling for differentiated cell plasticity. To investigate these possibilities, we studied crypt epithelial cells in an unbiased fashion via high-resolution single-cell profiling. These studies, combined with in vivo lineage tracing, show that Lgr5 is not a specific ISC marker and that stemness potential exists beyond the crypt base and resides in the isthmus region, where undifferentiated cells participate in intestinal homeostasis and regeneration following irradiation (IR) injury. Our results provide an alternative model of intestinal epithelial cell organization, suggesting that stemness potential is not restricted to CBC cells, and neither de-differentiation nor reserve ISC are drivers of intestinal regeneration.


Asunto(s)
Homeostasis , Mucosa Intestinal , Receptores Acoplados a Proteínas G , Regeneración , Células Madre , Animales , Células Madre/metabolismo , Células Madre/citología , Ratones , Mucosa Intestinal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Intestinos/citología , Diferenciación Celular , Ratones Endogámicos C57BL , Células Epiteliales/metabolismo , Análisis de la Célula Individual , Masculino
2.
Cell ; 150(2): 351-65, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22817897

RESUMEN

Wnt signaling is involved in self-renewal and maintenance of hematopoietic stem cells (HSCs); however, the particular role of noncanonical Wnt signaling in regulating HSCs in vivo is largely unknown. Here, we show Flamingo (Fmi) and Frizzled (Fz) 8, members of noncanonical Wnt signaling, both express in and functionally maintain quiescent long-term HSCs. Fmi regulates Fz8 distribution at the interface between HSCs and N-cadherin(+) osteoblasts (N-cad(+)OBs that enrich osteoprogenitors) in the niche. We further found that N-cad(+)OBs predominantly express noncanonical Wnt ligands and inhibitors of canonical Wnt signaling under homeostasis. Under stress, noncanonical Wnt signaling is attenuated and canonical Wnt signaling is enhanced in activation of HSCs. Mechanistically, noncanonical Wnt signaling mediated by Fz8 suppresses the Ca(2+)-NFAT- IFNγ pathway, directly or indirectly through the CDC42-CK1α complex and also antagonizes canonical Wnt signaling in HSCs. Taken together, our findings demonstrate that noncanonical Wnt signaling maintains quiescent long-term HSCs through Fmi and Fz8 interaction in the niche.


Asunto(s)
Cadherinas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células Madre , Vía de Señalización Wnt , Animales , Cadherinas/genética , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Transgénicos , Factores de Transcripción NFATC/metabolismo , Receptores Acoplados a Proteínas G/genética
3.
J Hepatol ; 77(3): 619-631, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35452693

RESUMEN

BACKGROUND & AIMS: Vacuole membrane protein 1 (VMP1) is an endoplasmic reticulum (ER) transmembrane protein that regulates the formation of autophagosomes and lipid droplets. Recent evidence suggests that VMP1 plays a critical role in lipoprotein secretion in zebra fish and cultured cells. However, the pathophysiological roles and mechanisms by which VMP1 regulates lipoprotein secretion and lipid accumulation in non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are unknown. METHODS: Liver-specific and hepatocyte-specific Vmp1 knockout mice as well as Vmp1 knock-in mice were generated by crossing Vmp1flox or Vmp1KI mice with albumin-Cre mice or by injecting AAV8-TBG-cre, respectively. Lipid and energy metabolism in these mice were characterized by metabolomic and transcriptome analyses. Mice with hepatic overexpression of VMP1 who were fed a NASH diet were also characterized. RESULTS: Hepatocyte-specific deletion of Vmp1 severely impaired VLDL secretion resulting in massive hepatic steatosis, hepatocyte death, inflammation and fibrosis, which are hallmarks of NASH. Mechanistically, loss of Vmp1 led to decreased hepatic levels of phosphatidylcholine and phosphatidylethanolamine as well as to changes in phospholipid composition. Deletion of Vmp1 in mouse liver also led to the accumulation of neutral lipids in the ER bilayer and impaired mitochondrial beta-oxidation. Overexpression of VMP1 ameliorated steatosis in diet-induced NASH by improving VLDL secretion. Importantly, we also showed that decreased liver VMP1 is associated with NAFLD/NASH in humans. CONCLUSIONS: Our results provide novel insights on the role of VMP1 in regulating hepatic phospholipid synthesis and lipoprotein secretion in the pathogenesis of NAFLD/NASH. LAY SUMMARY: Non-alcoholic fatty liver disease and its more severe form, non-alcoholic steatohepatitis, are associated with a build-up of fat in the liver (steatosis). However, the exact mechanisms that underly steatosis in patients are not completely understood. Herein, the authors identified that the lack of a protein called VMP1 impairs the secretion and metabolism of fats in the liver and could therefore contribute to the development and progression of non-alcoholic fatty liver disease.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Humanos , Lipoproteínas/metabolismo , Hígado/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Fosfolípidos/metabolismo
4.
Sensors (Basel) ; 21(24)2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34960416

RESUMEN

Connected vehicle (CV) technologies are changing the form of traditional traffic models. In the CV driving environment, abundant and accurate information is available to vehicles, promoting the development of control strategies and models. Under these circumstances, this paper proposes a bidirectional vehicles information structure (BDVIS) by making use of the acceleration information of one preceding vehicle and one following vehicle to improve the car-following models. Then, we deduced the derived multiple vehicles information structure (DMVIS), including historical movement information of multiple vehicles, without the acceleration information. Next, the paper embeds the four kinds of basic car-following models into the framework to investigate the stability condition of two structures under the small perturbation of traffic flow and explored traffic response properties with different proportions of forward-looking or backward-looking terms. Under the open boundary condition, simulations on a single lane are conducted to validate the theoretical analysis. The results indicated that BDVIS and the DMVIS perform better than the original car-following model in improving the traffic flow stability, but that they have their own advantages for differently positioned vehicles in the platoon. Moreover, increasing the proportions of the preceding and following vehicles presents a benefit to stability, but if traffic is stable, an increase in any of the parameters would extend the influence time, which reveals that neither ß1 or ß2 is the biggest the best for the traffic.

5.
Am J Physiol Gastrointest Liver Physiol ; 318(4): G796-G802, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32003604

RESUMEN

The gastrointestinal system is arguably one of the most complicated developmental systems in a multicellular organism, as it carries out at least four major functions: digestion of food, absorption of nutrients, excretion of hormones, and defense against pathogens. Anatomically, the fetal gut has a tubular structure with an outer layer of smooth muscle derived from lateral splanchnic mesoderm and an inner lining of epithelium derived from the definitive endoderm. During morphogenesis of the gut tube, the definitive endoderm transforms into a primitive gut tube with a foregut, midgut, and hindgut. During the course of further development, the midgut gives rise to the small and proximal large intestine and the hindgut gives rise to the distal large intestine and rectum. The small intestine is subdivided into three parts: duodenum, jejunum, and ileum, whereas the large intestine is subdivided into the cecum, colon, and rectum.


Asunto(s)
Diferenciación Celular/fisiología , Plasticidad de la Célula/fisiología , Epitelio/fisiología , Regeneración/fisiología , Células Madre/fisiología , Animales , Humanos , Mucosa Intestinal/fisiología , Intestinos
6.
PLoS Genet ; 13(6): e1006771, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28640831

RESUMEN

Ribosomal DNA is one of the most variable regions in the human genome with respect to copy number. Despite the importance of rDNA for cellular function, we know virtually nothing about what governs its copy number, stability, and sequence in the mammalian genome due to challenges associated with mapping and analysis. We applied computational and droplet digital PCR approaches to measure rDNA copy number in normal and cancer states in human and mouse genomes. We find that copy number and sequence can change in cancer genomes. Counterintuitively, human cancer genomes show a loss of copies, accompanied by global copy number co-variation. The sequence can also be more variable in the cancer genome. Cancer genomes with lower copies have mutational evidence of mTOR hyperactivity. The PTEN phosphatase is a tumor suppressor that is critical for genome stability and a negative regulator of the mTOR kinase pathway. Surprisingly, but consistent with the human cancer genomes, hematopoietic cancer stem cells from a Pten-/- mouse model for leukemia have lower rDNA copy number than normal tissue, despite increased proliferation, rRNA production, and protein synthesis. Loss of copies occurs early and is associated with hypersensitivity to DNA damage. Therefore, copy loss is a recurrent feature in cancers associated with mTOR activation. Ribosomal DNA copy number may be a simple and useful indicator of whether a cancer will be sensitive to DNA damaging treatments.


Asunto(s)
Variaciones en el Número de Copia de ADN , Leucemia/genética , ARN Ribosómico/genética , Animales , Células Cultivadas , Daño del ADN , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Mutación , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
7.
Curr Opin Hematol ; 26(4): 258-265, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31170110

RESUMEN

: Hematopoietic stem cells (HSCs) are a unique population of cells with the remarkable ability to replenish themselves through self-renewal and to give rise to differentiated cell lineages. Though having been discovered more than 50 years ago, and having been widely used in bone marrow transplantation to treat blood disorders including leukemia, expansion of HSCs remains an unmet task, thus affecting its more effective usage in clinical practice. PURPOSE OF REVIEW: The purpose of this review article is to summarize past efforts in ex-vivo HSC expansion and to compare recent advances in expanding murine and human HSCs by targeting the N-methyladenosine (mA) pathway. RECENT FINDINGS: Unlike past many efforts that mainly target single or limited pathways and often lead to lineage bias or expansion of progenitor cells or limited long-term HSCs (LT-HSCs), the blocking the degradation of mA pathway has an advantage of stabilizing hundreds of key factors required for maintaining HSCs, thus resulting in expansion of functional LT-HSCs. SUMMARY: The new approach of targeting the mA pathway has a promising application in clinical HSC-based transplantation.


Asunto(s)
Autorrenovación de las Células , Células Madre Hematopoyéticas/citología , Animales , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos
8.
Gastroenterology ; 155(3): 865-879.e12, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29782848

RESUMEN

BACKGROUND & AIMS: Defects in lysosome function and autophagy contribute to the pathogenesis of alcoholic liver disease. We investigated the mechanisms by which alcohol consumption affects these processes by evaluating the functions of transcription factor EB (TFEB), which regulates lysosomal biogenesis. METHODS: We performed studies with GFP-LC3 mice, mice with liver-specific deletion of TFEB, mice with disruption of the transcription factor E3 gene (TFE3-knockout mice), mice with disruption of the Tefb and Tfe3 genes (TFEB and TFE3 double-knockout mice), and Tfebflox/flox albumin cre-negative mice (controls). TFEB was overexpressed from adenoviral vectors or knocked down with small interfering RNAs in mouse livers. Mice were placed on diets of regular ethanol feeding plus an acute binge to induce liver damage (ethanol diet); some mice also were given injections of torin-1, an inhibitor of the kinase activity of the mechanistic target of rapamycin (mTOR). Liver tissues were collected and analyzed by immunohistochemistry, immunoblots, and quantitative real-time polymerase chain reaction to monitor lysosome biogenesis. We analyzed levels of TFEB in liver tissues from patients with alcoholic hepatitis and from healthy donors (controls) by immunohistochemistry. RESULTS: Liver tissues from mice on the ethanol diet had lower levels of total and nuclear TFEB compared with control mice, and hepatocytes had decreased lysosome biogenesis and autophagy. Hepatocytes from mice on the ethanol diet had increased translocation of mTOR into lysosomes, resulting in increased mTOR activation. Administration of torin-1 increased liver levels of TFEB and decreased steatosis and liver injury induced by ethanol. Mice that overexpressed TFEB in the liver developed less severe ethanol-induced liver injury and had increased lysosomal biogenesis and mitochondrial bioenergetics compared with mice carrying a control vector. Mice with knockdown of TFEB and TFEB-TFE3 double-knockout mice developed more severe liver injury in response to the ethanol diet than control mice. Liver tissues from patients with alcohol-induced hepatitis had lower nuclear levels of TFEB than control tissues. CONCLUSIONS: We found that ethanol feeding plus an acute binge decreased hepatic expression of TFEB, which is required for lysosomal biogenesis and autophagy. Strategies to block mTOR activity or increase levels of TFEB might be developed to protect the liver from ethanol-induced damage.


Asunto(s)
Autofagia/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Hígado Graso/genética , Hepatopatías Alcohólicas/genética , Lisosomas/fisiología , Animales , Etanol , Hepatocitos/fisiología , Hígado/metabolismo , Ratones , Ratones Noqueados , Biogénesis de Organelos , Serina-Treonina Quinasas TOR/fisiología
9.
Blood ; 140(5): 411-412, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35925643
10.
Immunity ; 32(3): 392-402, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20303298

RESUMEN

Inflammatory cytokines have been proposed to regulate epithelial homeostasis during intestinal inflammation. We report here that interferon-gamma (IFN-gamma) regulates the crucial homeostatic functions of cell proliferation and apoptosis through serine-threonine protein kinase AKT-beta-catenin and Wingless-Int (Wnt)-beta-catenin signaling pathways. Short-term exposure of intestinal epithelial cells to IFN-gamma resulted in activation of beta-catenin through AKT, followed by induction of the secreted Wnt inhibitor Dkk1. Consequently, we observed an increase in Dkk1-mediated apoptosis upon extended IFN-gamma treatment and reduced proliferation through depletion of the Wnt coreceptor LRP6. These effects were enhanced by tumor necrosis factor-alpha (TNF-alpha), suggesting synergism between the two cytokines. Consistent with these results, colitis in vivo was associated with decreased beta-catenin-T cell factor (TCF) signaling, loss of plasma membrane-associated LRP6, and reduced epithelial cell proliferation. Proliferation was partially restored in IFN-gamma-deficient mice. Thus, we propose that IFN-gamma regulates intestinal epithelial homeostasis by sequential regulation of converging beta-catenin signaling pathways.


Asunto(s)
Células Epiteliales/inmunología , Homeostasis , Interferón gamma/inmunología , Intestinos/inmunología , Transducción de Señal , beta Catenina/metabolismo , Animales , Apoptosis , Línea Celular , Proliferación Celular , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Interferón gamma/deficiencia , Interferón gamma/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Wnt/metabolismo
11.
Nature ; 500(7462): 345-9, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23863936

RESUMEN

The epigenetic regulation of imprinted genes by monoallelic DNA methylation of either maternal or paternal alleles is critical for embryonic growth and development. Imprinted genes were recently shown to be expressed in mammalian adult stem cells to support self-renewal of neural and lung stem cells; however, a role for imprinting per se in adult stem cells remains elusive. Here we show upregulation of growth-restricting imprinted genes, including in the H19-Igf2 locus, in long-term haematopoietic stem cells and their downregulation upon haematopoietic stem cell activation and proliferation. A differentially methylated region upstream of H19 (H19-DMR), serving as the imprinting control region, determines the reciprocal expression of H19 from the maternal allele and Igf2 from the paternal allele. In addition, H19 serves as a source of miR-675, which restricts Igf1r expression. We demonstrate that conditional deletion of the maternal but not the paternal H19-DMR reduces adult haematopoietic stem cell quiescence, a state required for long-term maintenance of haematopoietic stem cells, and compromises haematopoietic stem cell function. Maternal-specific H19-DMR deletion results in activation of the Igf2-Igfr1 pathway, as shown by the translocation of phosphorylated FoxO3 (an inactive form) from nucleus to cytoplasm and the release of FoxO3-mediated cell cycle arrest, thus leading to increased activation, proliferation and eventual exhaustion of haematopoietic stem cells. Mechanistically, maternal-specific H19-DMR deletion leads to Igf2 upregulation and increased translation of Igf1r, which is normally suppressed by H19-derived miR-675. Similarly, genetic inactivation of Igf1r partly rescues the H19-DMR deletion phenotype. Our work establishes a new role for this unique form of epigenetic control at the H19-Igf2 locus in maintaining adult stem cells.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Animales , Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Receptor IGF Tipo 1/genética , Transducción de Señal , Activación Transcripcional
12.
J Assist Reprod Genet ; 36(8): 1761, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31368011

RESUMEN

The original article unfortunately contained a mistake. The 3rd author's name should be Cuilian Zhang. In addition, the 4th affiliation should be updated to "Reproductive Medicine Center, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China".

13.
Genes Dev ; 25(18): 1928-42, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21890648

RESUMEN

Although self-renewal is the central property of stem cells, the underlying mechanism remains inadequately defined. Using a hematopoietic stem and progenitor cell (HSPC)-specific conditional induction line, we generated a compound genetic model bearing both Pten deletion and ß-catenin activation. These double mutant mice exhibit a novel phenotype, including expansion of phenotypic long-term hematopoietic stem cells (LT-HSCs) without extensive differentiation. Unexpectedly, constitutive activation of ß-catenin alone results in apoptosis of HSCs. However, together, the Wnt/ß-catenin and PTEN/PI3k/Akt pathways interact to drive phenotypic LT-HSC expansion by inducing proliferation while simultaneously inhibiting apoptosis and blocking differentiation, demonstrating the necessity of complementary cooperation between the two pathways in promoting self-renewal. Mechanistically, ß-catenin activation reduces multiple differentiation-inducing transcription factors, blocking differentiation partially through up-regulation of Inhibitor of differentiation 2 (Id2). In double mutants, loss of Pten enhances the HSC anti-apoptotic factor Mcl-1. All of these contribute in a complementary way to HSC self-renewal and expansion. While permanent, genetic alteration of both pathways in double mutant mice leads to expansion of phenotypic HSCs, these HSCs cannot function due to blocked differentiation. We developed a pharmacological approach to expand normal, functional HSCs in culture using factors that reversibly activate both Wnt/ß-catenin and PI3K/Akt signaling simultaneously. We show for the first time that activation of either single pathway is insufficient to expand primitive HSCs, but in combination, both pathways drive self-renewal and expansion of HSCs with long-term functional capacity.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Cultivadas , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/enzimología , Ratones , Mutación , Fosfohidrolasa PTEN/genética , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Proteínas Wnt/genética , beta Catenina/genética
14.
Hepatology ; 66(4): 1183-1196, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28407375

RESUMEN

Based on our recent finding that disruption of bile acid (BA) homeostasis in mice results in the induction of hepatic long noncoding RNA H19 expression, we sought to elucidate the role of H19 in cholestatic liver fibrosis. Hepatic overexpression of H19RNA augmented bile duct ligation (BDL)-induced liver fibrosis, which was accompanied by the elevation of serum alanine aminotransferase, aspartate aminotransferase, bilirubin, and BA levels. Multiple genes related to liver fibrosis, inflammation, and biliary hyperplasia were increased in H19-BDL versus null-BDL mice, whereas genes in BA synthesis were decreased. Livers and spleens of H19-BDL mice showed significant enrichment of CD3+γδ+, interleukin-4, and interleukin-17 producing CD4+ and CD8+ immune cell populations. H19 down-regulated hepatic zinc finger E-box-binding homeobox 1 (ZEB1) but up-regulated epithelial cell adhesion molecule (EpCAM) and SRY (sex determining region Y)-box 9 expression. Mechanistically, ZEB1 repressed EpCAM promoter activity and gene transcription. H19RNA impeded ZEB1's inhibitory action by interacting with ZEB1 protein to prevent its binding to the EpCAM promoter. Hepatic overexpression of ZEB1 or knockdown of EpCAM diminished H19-induced fibrosis; the latter was also prevented in H19-/- mice. H19RNA was markedly induced by bile acids in mouse small cholangiocytes and to a lesser extent in mouse large cholangiocytes. The up-regulation of H19RNA and EpCAM correlated positively with the down-regulation of ZEB1 in primary sclerosing cholangitis and primary biliary cirrhosis liver specimens. CONCLUSION: The activation of hepatic H19RNA promoted cholestatic liver fibrosis in mice through the ZEB1/EpCAM signaling pathway. (Hepatology 2017;66:1183-1196).


Asunto(s)
Colestasis/metabolismo , Molécula de Adhesión Celular Epitelial/metabolismo , Cirrosis Hepática/etiología , ARN Largo no Codificante/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Adulto , Anciano , Animales , Ácidos y Sales Biliares , Colestasis/complicaciones , Femenino , Células HEK293 , Células Hep G2 , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Regulación hacia Arriba , Adulto Joven
15.
J Biol Chem ; 291(8): 4166-77, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26565021

RESUMEN

Wnt/ß-catenin signaling is required for crypt structure maintenance. We previously observed nuclear accumulation of Ser-552 phosphorylated ß-catenin (pß-Cat(Ser-552)) in intestinal epithelial cells (IEC) during colitis and colitis-associated cancer. Data here delineate a novel multiprotein cytosolic complex (MCC) involved in ß-catenin signaling in the intestine. The MCC contains p85α, the class IA subunit of PI3K, along with ß-catenin, 14-3-3ζ, Akt, and p110α. MCC levels in IEC increase in colitis and colitis-associated cancer patients. IEC-specific p85α-deficient (p85(ΔIEC)) mice develop more severe dextran sodium sulfate colitis due to delayed ulcer healing and reduced epithelial ß-catenin activation. In colonic IEC, p85α deficiency did not alter PI3K signaling. In vitro shRNA depletion of individual complex members disrupts the MCC and reduces ß-catenin signaling. Despite worse colitis, p85(ΔIEC) mice have reduced tumor burden after azoxymethane/dextran sodium sulfate treatment. Together the data indicate that the ß-catenin MCC is needed for mucosal repair and carcinogenesis. This novel MCC may be an attractive therapeutic target in preventing cancer in colitis patients.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Colitis/metabolismo , Neoplasias del Colon/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Animales , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Colitis/genética , Colitis/patología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Humanos , Ratones , Ratones Transgénicos , Complejos Multiproteicos/genética , Proteínas de Neoplasias/genética , beta Catenina/genética
16.
Blood ; 125(17): 2590-1, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25907901

RESUMEN

In this issue of Blood, Bowers et al report that osteoblasts maintain a subset of quiescent stem cells and that osteoblast ablation converts bone marrow into a proliferation-promoting environment for both normal and malignant stem cells.


Asunto(s)
Células Madre Hematopoyéticas/citología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/patología , Osteoblastos/citología , Animales
18.
J Physiol ; 594(17): 4827-36, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27060879

RESUMEN

The niche constitutes a unique category of cells that support the microenvironment for the maintenance and self-renewal of stem cells. Intestinal stem cells reside at the base of the crypt, which contains adjacent epithelial cells, stromal cells and smooth muscle cells, and soluble and cell-associated growth and differentiation factors. We summarize here recent advances in our understanding of the crucial role of the niche in regulating stem cells. The stem cell niche maintains a balance among quiescence, proliferation and regeneration of intestinal stem cells after injury. Mesenchymal cells, Paneth cells, immune cells, endothelial cells and neural cells are important regulatory components that secrete niche ligands, growth factors and cytokines. Intestinal homeostasis is regulated by niche signalling pathways, specifically Wnt, bone morphogenetic protein, Notch and epidermal growth factor. These insights into the regulatory stem cell niche during homeostasis and post-injury regeneration offer the potential to accelerate development of therapies for intestine-related disorders.


Asunto(s)
Intestinos/citología , Nicho de Células Madre , Células Madre/fisiología , Animales , Células Endoteliales/fisiología , Neuronas/fisiología , Regeneración , Transducción de Señal
19.
Nat Genet ; 39(2): 189-98, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17237784

RESUMEN

Intestinal polyposis, a precancerous neoplasia, results primarily from an abnormal increase in the number of crypts, which contain intestinal stem cells (ISCs). In mice, widespread deletion of the tumor suppressor Phosphatase and tensin homolog (PTEN) generates hamartomatous intestinal polyps with epithelial and stromal involvement. Using this model, we have established the relationship between stem cells and polyp and tumor formation. PTEN helps govern the proliferation rate and number of ISCs and loss of PTEN results in an excess of ISCs. In PTEN-deficient mice, excess ISCs initiate de novo crypt formation and crypt fission, recapitulating crypt production in fetal and neonatal intestine. The PTEN-Akt pathway probably governs stem cell activation by helping control nuclear localization of the Wnt pathway effector beta-catenin. Akt phosphorylates beta-catenin at Ser552, resulting in a nuclear-localized form in ISCs. Our observations show that intestinal polyposis is initiated by PTEN-deficient ISCs that undergo excessive proliferation driven by Akt activation and nuclear localization of beta-catenin.


Asunto(s)
Mucosa Intestinal/metabolismo , Pólipos Intestinales/genética , Fosfohidrolasa PTEN/fisiología , Células Madre/metabolismo , Animales , Recuento de Células , Ciclo Celular , Núcleo Celular/metabolismo , Ratones , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo
20.
J Biol Chem ; 289(34): 23809-16, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-24966324

RESUMEN

Lgr5 is a marker for proliferating stem cells in adult intestine, stomach, and hair follicle. However, Lgr5 is not expressed in adult hematopoietic stem and progenitor cells (HSPCs). Whether Lgr5 is expressed in the embryonic and fetal HSPCs that undergo rapid proliferation is unknown. Here we report the detection of Lgr5 expression in HSPCs in the aorta-gonad-mesonephros (AGM) and fetal liver. We also found that a portion of Lgr5(+) cells expressed the Runx1 gene that is critical for the ontogeny of HSPCs. A small portion of Lgr5(+) cells also expressed HSPC surface markers c-Kit and CD34 in AGM or CD41 in fetal liver. Furthermore, the majority of Lgr5(+) cells expressed Ki67, indicating their proliferating state. Transplantation of fetal liver-derived Lgr5-GFP(+) cells (E12.5) demonstrated that Lgr5-GFP(+) cells were able to reconstitute myeloid and lymphoid lineages in adult recipients, but the engraftment was short-term (4-8 weeks) and 20-fold lower compared with the Lgr5-GFP(-) control. Our data show that Lgr5-expressing cells mark short-term hematopoietic stem and progenitor cells, consistent with the role of Lgr5 in supporting HSPCs rapid proliferation during embryonic and fetal development.


Asunto(s)
Desarrollo Embrionario , Células Madre Hematopoyéticas/citología , Leucina/química , Receptores Acoplados a Proteínas G/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Hígado/embriología , Hígado/metabolismo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA