首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 46 毫秒
1.
The remarkable ability of rapid self-renewal makes the intestinal epithelium an ideal model for the study of adult stem cells. The intestinal epithelium is organized into villus and crypt, and a group of intestinal stem cells located at the base of crypt are responsible for this constant self-renewal throughout the life. Identification of the intestinal stem cell marker Lgr5, isolation and in vitro culture of Lgr5+ intestinal stem cells and the use of transgenic mouse models have significantly facilitated the studies of intestinal stem cell homeostasis and differentiation, therefore greatly expanding our knowledge of the regulatory mechanisms underlying the intestinal stem cell fate determination. In this review, we summarize the current understanding of how signals of Wnt, BMP, Notch and EGF in the stem cell niche modulate the intestinal stem cell fate.  相似文献   

2.
A frequent complication in colorectal cancer (CRC) is regeneration of the tumor after therapy. Here, we report that a gene signature specific for adult intestinal stem cells (ISCs) predicts disease relapse in CRC patients. ISCs are marked by high expression of the EphB2 receptor, which becomes gradually silenced as cells differentiate. Using EphB2 and the ISC marker Lgr5, we have FACS-purified and profiled mouse ISCs, crypt proliferative progenitors, and late transient amplifying cells to define a gene program specific for normal ISCs. Furthermore, we discovered that ISC-specific genes identify a stem-like cell population positioned at the bottom of tumor structures reminiscent of crypts. EphB2 sorted ISC-like tumor cells display robust tumor-initiating capacity in immunodeficient mice as well as long-term self-renewal potential. Taken together, our data suggest that the ISC program defines a cancer stem cell niche within colorectal tumors and plays a central role in CRC relapse.  相似文献   

3.
Cycling Lgr5+ stem cells fuel the rapid turnover of the adult intestinal epithelium. The existence of quiescent Lgr5+ cells has been reported, while an alternative quiescent stem cell population is believed to reside at crypt position +4. Here, we generated a novel Ki67RFP knock-in allele that identifies dividing cells. Using Lgr5-GFP;Ki67RFP mice, we isolated crypt stem and progenitor cells with distinct Wnt signaling levels and cell cycle features and generated their molecular signature using microarrays. Stem cell potential of these populations was further characterized using the intestinal organoid culture. We found that Lgr5high stem cells are continuously in cell cycle, while a fraction of Lgr5low progenitors that reside predominantly at +4 position exit the cell cycle. Unlike fast dividing CBCs, Lgr5low Ki67 cells have lost their ability to initiate organoid cultures, are enriched in secretory differentiation factors, and resemble the Dll1 secretory precursors and the label-retaining cells of Winton and colleagues. Our findings support the cycling stem cell hypothesis and highlight the cell cycle heterogeneity of early progenitors during lineage commitment.  相似文献   

4.
Lgr5 marks adult stem cells in multiple adult organs and is a receptor for the Wnt‐agonistic R‐spondins (RSPOs). Intestinal, stomach and liver Lgr5+ stem cells grow in 3D cultures to form ever‐expanding organoids, which resemble the tissues of origin. Wnt signalling is inactive and Lgr5 is not expressed under physiological conditions in the adult pancreas. However, we now report that the Wnt pathway is robustly activated upon injury by partial duct ligation (PDL), concomitant with the appearance of Lgr5 expression in regenerating pancreatic ducts. In vitro, duct fragments from mouse pancreas initiate Lgr5 expression in RSPO1‐based cultures, and develop into budding cyst‐like structures (organoids) that expand five‐fold weekly for >40 weeks. Single isolated duct cells can also be cultured into pancreatic organoids, containing Lgr5 stem/progenitor cells that can be clonally expanded. Clonal pancreas organoids can be induced to differentiate into duct as well as endocrine cells upon transplantation, thus proving their bi‐potentiality.  相似文献   

5.
小肠上皮具有快速更新的能力,是研究成体干细胞的理想系统.小肠上皮由绒毛和隐窝两部分组成,而位于小肠隐窝底部的小肠干细胞是其持续更新的源泉.近年来,以Lgr5为代表的小肠干细胞标记物的发现、Lgr5+小肠干细胞的分离培养和多种转基因小鼠模型的出现,极大地促进了对小肠干细胞自我更新和分化调控的研究,使得人们可以更加深入地认识小肠干细胞命运决定的分子机制.本文简要综述了近年来人们对Wnt,BMP,Notch和EGF等信号如何在小肠干细胞命运调控中发挥作用的认识.  相似文献   

6.
Roth S  Fodde R 《EMBO reports》2011,12(6):483-484
Mustata et al demonstrate in this issue of EMBO reports that Lgr4 expression in the stem cells and transit amplifying cells of the intestinal crypts is required for the establishment of the stem cell niche and also for the maintenance of intestinal stem cells in ex vivo organoid cultures.EMBO reports 12, 6, 558–564. doi:10.1038/embor.2011.52The ‘nature versus nurture'' debate concerns the relative contributions to an individual''s identity of its nature (that is, its genetic make-up) compared with its nurture, defined as the totality of external, environmental factors. A similar type of debate is ongoing among developmental and stem-cell biologists: is the intrinsic nature (that is, its (epi)genetic make-up) of a stem cell what makes it self-renew and differentiate according to the physiological needs of a given tissue, or is it the immediate environment (nurture) that regulates stemness? Irrespective of the relative weight of each contribution, there is little doubt that both cell-autonomous and environmental factors play crucial roles in the maintenance of homeostasis in self-renewing tissues such as the skin, mammary gland, blood and intestine. In an article published last month in EMBO reports (Mustata et al, 2011), the Lgr4 gene is shown to have a rate-limiting role in establishing the stem-cell niche of the proximal intestinal tract.…the Lgr4 gene is shown to have a rate-limiting role in establishing the stem-cell niche of the proximal intestinal tractThe epithelial lining of the proximal intestine is characterized by a unique tissue architecture consisting of villi and crypts. The intestinal crypt of Lieberkühn is a highly dynamic niche with stem cells in its lower third, which give rise to a population of fast-cycling transit-amplifying cells. Transit-amplifying cells undergo a limited number of cell divisions and eventually differentiate into four specialized cell types of the small intestine: absorptive, enteroendocrine, goblet and Paneth cells. Notably, Paneth cells are the only terminally differentiated cell type of the proximal intestinal tract that (i) move downwards along the crypt–villus axis and (ii) retain canonical Wnt signalling activity upon differentiation (van Es et al, 2005).On the basis of clonal analysis and knock-in experiments, it was shown that the crypt base columnar (CBC) cells—located in the lower third of the crypt and characterized by Lgr5 expression—represent actively cycling stem cells that are able to give rise to all differentiated cell types of the intestinal epithelium (Barker et al, 2007). More recently, it has also been shown that Paneth cells, apart from their well-known bactericidal function, are in close physical association with Lgr5+ stem cells, to which they provide essential niche signals such as EGF, Wnt3a and Dll4 (Sato et al, 2011). This is also important in the light of the observation that single Lgr5+ stem cells, when cultured ex vivo, can generate crypt–villus organoids without a (mesenchymal) niche (Sato et al, 2009). In fact, the latter is only partly true, as these organoids are cultured in matrigel and in the presence of specific growth factors that are probably released by the niche in vivo.Lgr5, together with Lgr4 and Lgr6, belongs to the family of leucine-rich repeat-containing G-protein-coupled seven-transmembrane receptors. Recently, both Lgr5 and Lgr6 have received attention from the stem-cell community: Lgr5 is a downstream Wnt target gene and a marker of cycling stem cells in the intestinal tract and the hair follicle, whereas Lgr6 expression marks adult stem cells in the skin (Barker & Clevers, 2010). However, whether they merely represent stem-cell markers or also have a functional role in stemness is unknown.Mustata et al (2011) report on the functional role of another member of the Lgr family, Lgr4, by studying the effects of a targeted loss-of-function mutation (Lgr4 KO) on the development and differentiation of the mouse small intestine both in vivo and ex vivo. Endogenous Lgr4 expression is detected in transit-amplifying cells above the Paneth-cell zone, in CBC cells, and in rare Paneth cells. Loss of Lgr4 function results in a reduction in crypt depth due to a 50% decrease in epithelial-cell proliferation and, surprisingly, in an 80% reduction in Paneth-cell differentiation. Strikingly, these phenotypic features are apparently antagonistic to those of Lgr5 KO mice, in which premature Paneth-cell development was observed (Garcia et al, 2009). Accordingly, loss of Lgr4 function partly rescues the perinatal lethality of Lgr5 KO mice indicating non-redundancy of their individual functions.Loss of Lgr4 function results in […] an 80% reduction in Paneth-cell differentiationTo further investigate the role of Lgr4 in crypt development, the ex vivo ‘minigut'' culture system (Sato et al, 2009) was used; in contrast to crypts from wild-type mice that give rise to self-renewing structures encompassing all the differentiated cell lineages of the adult gut, organoids derived from age-matched Lgr4 KO animals are initially present as hollow spheres, mainly composed of stem and transit-amplifying cells, which disaggregate within 2–3 days and die within a week in culture. In agreement with their apparently opposite and non-redundant functions, crypt cultures from Lgr5 KO mice survive long-term culture and develop into differentiated organoids comparable with those of normal mice. Whereas loss of Lgr4 function partly rescues the lethality of Lgr5 KO mice in vivo, this is not true ex vivo; compound homozygous Lgr4/5 KO crypts give rise to hollow spheres that collapse and die as observed in Lgr4 KO organoids. Hence, under these experimental conditions—that is, in the absence of a mesenchymal niche—the Lgr4 defect is dominant over the Lgr5 one.Analysis of Paneth-cell differentiation markers and of Wnt targets, including Lgr5, confirmed their downregulation in Lgr4 KO organoids, thus suggesting a role for Lgr4 in Wnt signalling. Notably, lithium chloride treatment partly rescues the ex vivo phenotype of Lgr4 KO crypts, although this is not the case for other Wnt-signalling agonists, such as Wnt3a and Gsk3β inhibitors. On the basis of these observations, the authors conclude that Lgr4 probably has a permissive, rather than a direct and active role in Wnt signalling.In view of this and other studies, a revisitation of the cell-autonomous and niche-independent features of the Lgr5+ cycling stem cell (CBC cells) in the intestinal crypt seems to be necessary (Fig 1). First, the capacity of CBC cells to recapitulate ex vivo the complexity of the crypt–villus unit is mostly dependent on Paneth cells (Sato et al, 2011). When they are sorted as single cells, CBC cells perform poorly in organoid formation, whereas doublets of CBC and Paneth cells show high clonogenicity (Sato et al, 2009, 2011). However, rather than occurring exclusively through the secretion of niche signals in the form of Wnt ligands, the nature of the interdependency between Paneth cells and CBC cells seems to involve additional mechanisms. As shown by Mustata et al, loss of Lgr4 function causes a Paneth-cell differentiation blockade in the presence of wild-type levels of Wnt3a and Wnt11, a defect that can be rescued by lithium chloride, but not by the Wnt3a ligand or Gsk3β inhibitors. This indicates that additional factors secreted by epithelial and possibly mesenchymal cells—for example, stromal myofibroblasts (Vermeulen et al, 2010)—and the physical association of Paneth with Lgr5+ cells underlies their ‘partnership'' in preserving homeostasis within such a highly dynamic tissue. Hence, Paneth cells apparently constitute an essential component of the stem-cell niche in the upper intestinal tract.…rather than occurring exclusively through the secretion of niche signals […] the nature of the interdependency between Paneth cells and CBC cells seems to involve additional mechanismsOpen in a separate windowFigure 1Schematic illustration of the intestinal stem-cell compartment in the upper intestinal tract: Lgr4 (expressed in CBC and TA cells) positively stimulates Paneth-cell differentiation and, indirectly, stem-cell homeostasis, while Lgr5 (expressed in CBC cells) has been reported to inhibit Paneth-cell differentiation (Garcia et al, 2009). CBC, crypt base columnar; Dll4, delta-like 4; EGF, epidermal growth factor; TA, transit amplifying.As it is always the case, good science leads to new questions. Which cell type provides this niche function in the colon where Paneth cells are not present? Of note, it has been shown that in the colon Lgr5+ cells are intermingled with yet uncharacterized CD24+ cells (Sato et al, 2011), a cell-surface antigen known to enrich for Paneth cells in the upper intestinal tract. As CD24 expression does not mark CBC cells, but rather their flanking cells, these observations could again reflect the supportive, niche role of Paneth cells and CD24+ cells in the upper and distal intestinal tract, respectively. This might also be true for colon cancer, where Paneth cells are often present, possibly to provide niche support for cancer stem cells. Alternatively, premature (in the colon) and/or fully differentiated (in the upper intestine) Paneth cells might have a dual function by providing physical and paracrine support for cycling stem cells in homeostasis, as well as representing the hitherto elusive quiescent stem cells that underlie tissue regeneration after tissue insults. Whatever the truth, the intestinal scene is now set to further dissect the complexity of the nature–nurture interaction between intestinal (cancer) stem cells and their niche.  相似文献   

7.
Wnt signaling regulates self-renewal and fate commitment of stem and progenitor cells in development and homeostasis. Leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) is a co-receptor for Wnt signaling that marks highly proliferative stem and progenitor cells in many epithelial tissue types. Wnt signaling instructs neural developmental and homeostatic processes; however, Lgr5 expression in the developing and adult brain has not been characterized. Here we report that Lgr5 is expressed in the postnatal cerebellum during the maturation and synaptogenesis of cerebellar granule neurons (CGNs), processes controlled by Wnt signaling. Using a transgenic reporter mouse for in vivo Lgr5 expression analysis and lineage tracing, we reveal that Lgr5 specifically identified CGNs and was restricted temporally to the CGN maturation phase within the internal granule layer, but absent in the adult brain. Cells marked by Lgr5 were lineage restricted, post-mitotic and long-lived. The ligand for Lgr5, R-spondin, was secreted in a paracrine fashion that evolved during the maturation of CGNs, which coincided with the Lgr5 expression pattern. Our findings provide potential new insight into the critical regulation of Wnt signaling in the developing cerebellum and support a novel role for Lgr5 in the regulation of post-mitotic cells.  相似文献   

8.
9.
Pin C  Watson AJ  Carding SR 《PloS one》2012,7(5):e37115
We developed a slow structural relaxation model to describe cellular dynamics in the crypt of the mouse small intestine. Cells are arranged in a three dimensional spiral the size of which dynamically changes according to cell production demands of adjacent villi. Cell differentiation and proliferation is regulated through Wnt and Notch signals, the strength of which depends on the local cell composition. The highest level of Wnt activity is associated with maintaining equipotent stem cells (SC), Paneth cells and common goblet-Paneth cell progenitors (CGPCPs) intermingling at the crypt bottom. Low levels of Wnt signalling area are associated with stem cells giving rise to secretory cells (CGPCPs, enteroendocrine or Tuft cells) and proliferative absorptive progenitors. Deciding between these two fates, secretory and stem/absorptive cells, depends on Notch signalling. Our model predicts that Notch signalling inhibits secretory fate if more than 50% of cells they are in contact with belong to the secretory lineage. CGPCPs under high Wnt signalling will differentiate into Paneth cells while those migrating out from the crypt bottom differentiate into goblet cells. We have assumed that mature Paneth cells migrating upwards undergo anoikis. Structural relaxation explains the localisation of Paneth cells to the crypt bottom in the absence of active forces. The predicted crypt generation time from one SC is 4-5 days with 10-12 days needed to reach a structural steady state. Our predictions are consistent with experimental observations made under altered Wnt and Notch signalling. Mutations affecting stem cells located at the crypt floor have a 50% chance of being propagated throughout the crypt while mutations in cells above are rarely propagated. The predicted recovery time of an injured crypt losing half of its cells is approximately 2 days.  相似文献   

10.
The Wnt/β-catenin signaling pathbway controls many important biological processes. R-Spondin (RSPO) proteins are a family of secreted molecules that strongly potentiate Wnt/β-catenin signaling, however, the molecular mechanism of RSPO action is not yet fully understood. We performed an unbiased siRNA screen to identify molecules specifically required for RSPO, but not Wnt, induced β-catenin signaling. From this screen, we identified LGR4, then an orphan G protein-coupled receptor (GPCR), as the cognate receptor of RSPO. Depletion of LGR4 completely abolished RSPO-induced β-catenin signaling. The loss of LGR4 could be compensated by overexpression of LGR5, suggesting that LGR4 and LGR5 are functional homologs. We further demonstrated that RSPO binds to the extracellular domain of LGR4 and LGR5, and that overexpression of LGR4 strongly sensitizes cells to RSPO-activated β-catenin signaling. Supporting the physiological significance of RSPO-LGR4 interaction, Lgr4-/- crypt cultures failed to grow in RSPO-containing intestinal crypt culture medium. No coupling between LGR4 and heterotrimeric G proteins could be detected in RSPO-treated cells, suggesting that LGR4 mediates RSPO signaling through a novel mechanism. Identification of LGR4 and its relative LGR5, an adult stem cell marker, as the receptors of RSPO will facilitate the further characterization of these receptor/ligand pairs in regenerative medicine applications.  相似文献   

11.
12.
13.
14.
Gastrointestinal (GI) homeostasis requires the action of multiple pathways. There is some controversy regarding whether small intestine (SI) Paneth cells (PCs) play a central role in orchestrating crypt architecture and their relationship with Lgr5 + ve stem cells. Nevertheless, we previously showed that germline CSF-1 receptor (Csf1r) knock out (KO) or Csf1 mutation is associated with an absence of mature PC, reduced crypt proliferation and lowered stem cell gene, Lgr5 expression. Here we show the additional loss of CD24, Bmi1 and Olfm4 expression in the KO crypts and a high resolution 3D localization of CSF-1R mainly to PC. The induction of GI-specific Csf1r deletion in young adult mice also led to PC loss over a period of weeks, in accord with the anticipated long life span of PC, changed distribution of proliferating cells and this was with a commensurate loss of Lgr5 and other stem cell marker gene expression. By culturing SI organoids, we further show that the Csf1r?/? defect in PC production is intrinsic to epithelial cells as well as definitively affecting stem cell activity. These results show that CSF-1R directly supports PC maturation and that in turn PCs fashion the intestinal stem cell niche.  相似文献   

15.
The R-spondin protein family   总被引:1,自引:0,他引:1  
The four vertebrate R-spondin proteins are secreted agonists of the canonical Wnt/β-catenin signaling pathway. These proteins are approximately 35 kDa, and are characterized by two amino-terminal furin-like repeats, which are necessary and sufficient for Wnt signal potentiation, and a thrombospondin domain situated more towards the carboxyl terminus that can bind matrix glycosaminoglycans and/or proteoglycans. Although R-spondins are unable to initiate Wnt signaling, they can potently enhance responses to low-dose Wnt proteins. In humans, rare disruptions of the gene encoding R-spondin1 cause a syndrome of XX sex reversal (phenotypic male), palmoplantar keratosis (a thickening of the palms and soles caused by excess keratin formation) and predisposition to squamous cell carcinoma of the skin. Mutations in the gene encoding R-spondin4 cause anonychia (absence or hypoplasia of nails on fingers and toes). Recently, leucine-rich repeat-containing G-protein-coupled receptor (Lgr)4, Lgr5 and Lgr6, three closely related orphans of the leucine-rich repeat family of G-protein-coupled receptors, have been identified as receptors for R-spondins. Lgr5 and Lgr6 are markers for adult stem cells. Because R-spondins are potent stimulators of adult stem cell proliferation in vivo and in vitro, these findings might guide the therapeutic use of R-spondins in regenerative medicine.  相似文献   

16.
To explore Lgr5 as the possible stem cell marker in human gastric tissue, 259 normal gastric tissues and dissected gastric adenocarcinoma were analyzed by immunohistochemistry, immunofluorescence double staining and qRT-PCR. The results demonstrated that Lgr5 was expressed in the bottom of the normal gastric gland units, and showed a differential expression in gastric adenocarcinoma with varying differentiation. Lgr5 and Bmi1 were co-expressed within the same cells of gastric glands. CD26 +, CD44 +, ALDH1 + and CD133 + cells co-existed with Lgr5 + cells in the stem cell zone of adjacent normal gastric mucosa, and they were detectable in gastric adenocarcinoma but behaved differently. We concluded that Lgr5 may be the adult stem cell marker in human gastric epithelium; Lgr5 and Bmi1 may belong to the same stem cell population; Lgr5, CD26, CD44, ALDH1, and CD133 may be functionally-associated.  相似文献   

17.
Murine small intestinal crypt development is initiated during the first postnatal week. Soon after formation, overall increases in the number of crypts occurs through a bifurcating process called crypt fission, which is believed to be driven by developmental increases in the number of intestinal stem cells (ISCs). Recent evidence suggests that a heterogeneous population of ISCs exists within the adult intestine. Actively cycling ISCs are labeled by Lgr5, Ascl2 and Olfm4; whereas slowly cycling or quiescent ISC are marked by Bmi1 and mTert. The goal of this study was to correlate the expression of these markers with indirect measures of ISC expansion during development, including quantification of crypt fission and side population (SP) sorting. Significant changes were observed in the percent of crypt fission and SP cells consistent with ISC expansion between postnatal day 14 and 21. Quantitative real-time polymerase chain reaction (RT-PCR) for the various ISC marker mRNAs demonstrated divergent patterns of expression. mTert surged earliest, during the first week of life as crypts are initially being formed, whereas Lgr5 and Bmi1 peaked on day 14. Olfm4 and Ascl2 had variable expression patterns. To assess the number and location of Lgr5-expressing cells during this period, histologic sections from intestines of Lgr5-EGFP mice were subjected to quantitative analysis. There was attenuated Lgr5-EGFP expression at birth and through the first week of life. Once crypts were formed, the overall number and percent of Lgr5-EGFP positive cells per crypt remain stable throughout development and into adulthood. These data were supported by Lgr5 in situ hybridization in wild-type mice. We conclude that heterogeneous populations of ISCs are expanding as measured by SP sorting and mRNA expression at distinct developmental time points.  相似文献   

18.
19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号