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 15 毫秒
1.
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   

2.
3.
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.  相似文献   

4.
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   

5.
6.
An organoid is a complex, multi-cell three-dimensional (3D) structure that contains tissue-specific cells. Epithelial stem cells, which are marked by leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5), have the potential for self-renewal and expansion as organoids. However, in the case of intestinal organoids from Lgr5-EGFP-IRES-CreERT2 transgenic mice, in vitro expansion of the Lgr5 expression is limited in a culture condition supplemented with essential proteins, such as epidermal growth factor (E), noggin (N), and R-spondin 1 (R). In this study, we hypothesized that self-renewal of Lgr5+ stem cells in a 3D culture system can be stimulated by defined compounds (CHIR99021, Valproic acid, Y-27632, and A83-01). Our results demonstrated that dissociated single cells from organoids were organized into a 3D structure in the four compounds containing the ENR culture medium in a 3D and two-dimensional (2D) culture system. Moreover, the Lgr5 expression level of organoids from the ENR- and compound-containing media increased. Furthermore, the conversion of cultured Lgr5+ stem cells from 2D to 3D was confirmed. Therefore, defined compounds promote the expansion of Lgr5+ stem cells in organoids.  相似文献   

7.
8.
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   

9.
Lgr5 was identified as a promising gastrointestinal tract stem cell marker in mice. Lineage tracing indicates that Lgr5 + cells may not only be the cells responsible for the origin of tumors; they may also be the so-called cancer stem cells. In the present study, we investigated the presence of Lgr5 + cells and their biological significance in normal human gastric mucosa and gastric tumors. RNAscope, a newly developed RNA in situ hybridization technique, specifically labeled Lgr5 + cells at the basal glands of the gastric antrum. Notably, the number of Lgr5 + cells was remarkably increased in intestinal metaplasia. In total, 76% of gastric adenomas and 43% of early gastric carcinomas were positive for LGR5. Lgr5 + cells were found more frequently in low-grade tumors with active Wnt signaling and an intestinal gland type, suggesting that LGR5 is likely involved in the very early stages of Wnt-driven tumorigenesis in the stomach. Interestingly, similar to stem cells in normal tissues, Lgr5 + cells were often restricted to the base of the tumor glands, and such Lgr5 + restriction was associated with high levels of intestinal stem cell markers such as EPHB2, OLFM4, and ASCL2. Thus, our findings show that Lgr5 + cells are present at the base of the antral glands in the human stomach and that this cell population significantly expands in intestinal metaplasias. Furthermore, Lgr5 + cells are seen in a large number of gastric tumors ; their frequent basal arrangements and coexpression of ISC markers support the idea that Lgr5 + cells act as stem cells during the early stage of intestinal-type gastric tumorigenesis.  相似文献   

10.
Small stem cells, such as spore-like cells, blastomere-like stem cells (BLSCs), and very-small embryonic-like stem cells (VSELs) have been described in recent studies, although their multipotency in human tissues has not yet been confirmed. Here, we report the discovery of adult multipotent stem cells derived from human bone marrow, which we call StemBios (SB) cells. These isolated SB cells are smaller than 6 ìm and are DAPI+ and Lgr5+ (Leucine-Rich Repeat Containing G Protein-Coupled Receptor 5). Because Lgr5 has been characterized as a stem cell marker in the intestine, we hypothesized that SB cells may have a similar function. In vivo cell tracking assays confirmed that SB cells give rise to three types of cells, and in vitro studies demonstrated that SB cells cultured in proprietary media are able to grow to 6–25 ìm in size. Once the SB cells have attached to the wells, they differentiate into different cell lineages upon exposure to specific differentiation media. We are the first to demonstrate that stem cells smaller than 6 ìm can differentiate both in vivo and in vitro. In the future, we hope that SB cells will be used therapeutically to cure degenerative diseases.  相似文献   

11.
12.
13.
14.
15.
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   

16.
Factors regulating the proliferation and apoptosis of intestinal stem cells (ISCs) remain incompletely understood. Because ISCs exist among microbial ligands, immune receptors such as toll-like receptor 4 (TLR4) could play a role. We now hypothesize that ISCs express TLR4 and that the activation of TLR4 directly on the intestinal stem cells regulates their ability to proliferate or to undergo apoptosis. Using flow cytometry and fluorescent in situ hybridization for the intestinal stem cell marker Lgr5, we demonstrate that TLR4 is expressed on the Lgr5-positive intestinal stem cells. TLR4 activation reduced proliferation and increased apoptosis in ISCs both in vivo and in ISC organoids, a finding not observed in mice lacking TLR4 in the Lgr5-positive ISCs, confirming the in vivo significance of this effect. To define molecular mechanisms involved, TLR4 inhibited ISC proliferation and increased apoptosis via the p53-up-regulated modulator of apoptosis (PUMA), as TLR4 did not affect crypt proliferation or apoptosis in organoids or mice lacking PUMA. In vivo effects of TLR4 on ISCs required TIR-domain-containing adapter-inducing interferon-β (TRIF) but were independent of myeloid-differentiation primary response-gene 88 (MYD88) and TNFα. Physiological relevance was suggested, as TLR4 activation in necrotizing enterocolitis led to reduced proliferation and increased apoptosis of the intestinal crypts in a manner that could be reversed by inhibition of PUMA, both globally or restricted to the intestinal epithelium. These findings illustrate that TLR4 is expressed on ISCs where it regulates their proliferation and apoptosis through activation of PUMA and that TLR4 regulation of ISCs contributes to the pathogenesis of necrotizing enterocolitis.  相似文献   

17.
Several studies have suggested ERBB3/HER3 may be a useful prognostic marker for colorectal cancer. Tumours with an intestinal stem cell signature have also been shown to be more aggressive. Here, we investigate whether ERBB3 is associated with intestinal stem cell markers in colorectal cancer and if cancer stem cells within tumours are marked by expression of ERBB3. Expression of ERBB3 and intestinal stem cell markers (LGR5, EPHB2, CD44s and CD44v6) was assessed by qRT-PCR in primary colorectal tumours (stages 0 to IV) and matched normal tissues from 53 patients. The localisation of ERBB3, EPHB2 and KI-67 within tumours was investigated using co-immunofluorescence. Expression of ERBB3 and intestinal stem cell markers were significantly elevated in adenomas and colorectal tumours compared to normal tissue. Positive correlations were found between ERBB3 and intestinal stem cell markers. However, co-immunofluorescence analysis showed that ERBB3 and EPHB2 marked specific cell populations that were mutually exclusive within tumours with distinct proliferative potentials, the majority of ERBB3+ve cells being non-proliferative. This pattern resembles cellular organisation within normal colonic epithelium where EPHB2 labelled proliferative cells reside at the crypt base and ERBB3+ve cells mark differentiated cells at the top of crypts. Our results show that ERBB3 and intestinal stem cell markers correlate in colorectal cancers. ERBB3 localises to differentiated cell populations within tumours that are non-proliferative and distinct from cancer stem cells. These data support the concept that tumours contain discrete stem, proliferative and differentiation compartments similar to that present in normal crypts.  相似文献   

18.
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   

19.
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   

20.
Adult stem cell niches are often co-inhabited by cycling and quiescent stem cells. In the intestine, lineage tracing has identified Lgr5(+) cells as frequently cycling stem cells, whereas Bmi1(+), mTert(+), Hopx(+) and Lrig1(+) cells appear to be more quiescent. Here, we have applied a non-mutagenic and cell cycle independent approach to isolate and characterize small intestinal label-retaining cells (LRCs) persisting in the lower third of the crypt of Lieberkühn for up to 100 days. LRCs do not express markers of proliferation and of enterocyte, goblet or enteroendocrine differentiation, but are positive for Paneth cell markers. While during homeostasis, LR/Paneth cells appear to play a supportive role for Lgr5(+) stem cells as previously shown, upon tissue injury they switch to a proliferating state and in the process activate Bmi1 expression while silencing Paneth-specific genes. Hence, they are likely to contribute to the regenerative process following tissue insults such as chronic inflammation.  相似文献   

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