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 31 毫秒
1.
2.
Intestine is the organ for food digestion, nutrient absorption and pathogen defense, in which processes intestinal epithelium plays a central role. Intestinal epithelium undergoes fast turnover, and its homeostasis is regulated by multiple signaling pathways, including Wnt, Notch, Hippo and BMP pathways. BMP signaling has been shown to negatively regulate self-renewal of Lgr5+ intestinal stem cells, constrains the expansion of intestinal epithelium, therefore attenuating colorectal cancer formation. BMPs and their receptors are expressed in both epithelial and mesenchymal cells, suggesting a two-way interaction between the mesenchyme and epithelium. In this review, we summarize the current understanding of the function of BMP signaling in homeostasis, cancerous transformation and inflammatory response of intestinal epithelium.  相似文献   

3.
The small intestine consists of two histological compartments composed of the crypts and the villi. The function of the adult small intestinal epithelium is mediated by four different types of mature cells: enterocytes, goblet, enteroendocrine and Paneth. Undifferentiated cells reside in the crypts and produce these four types of mature cells. The niche-related Wnt and Bmp signaling pathways have been suggested to be involved in the regulation and maintenance of the stem cell microenvironment. In our laboratory, we isolated the first normal human intestinal epithelial crypt (HIEC) cell model from the human fetal intestine and in this study we investigated the expression of a panel of intestinal stem cell markers in HIEC cells under normal culture parameters as well as under conditions that mimic the stem cell microenvironment. The results showed that short term stimulation of HIEC cells with R-spondin 1 and Wnt-3a±SB-216763, a glycogen synthase kinase 3β (GSK3β) inhibitor, induced β-catenin/TCF activity and expression of the WNT target genes, cyclin D2 and LGR5. Treatment of HIEC cells with noggin, an antagonist of BMP signaling, abolished SMAD2/5/8 phosphorylation. Inducing a switch from inactive WNT/active BMP toward active WNT/inactive BMP pathways was sufficient to trigger a robust intestinal primordial stem-like cell signature with predominant LGR5, PHLDA1, PROM1, SMOC2 and OLFM4 expression. These findings demonstrate that even fully established cultures of intestinal cells can be prompted toward a CBC stem cell-like phenotype. This model should be useful for studying the regulation of human intestinal stem cell self-renewal and differentiation.  相似文献   

4.
Lgr4/Gpr48 is one of the newly identified R-spondins receptors and potentiates Wnt signaling, which regulates intestinal homeostasis. We used a hypomorphic mouse strain to determine the role of Lgr4 in intestinal inflammation and recovery. Intestinal inflammation was induced with dextran sulfate sodium (DSS) followed by a recovery period. Intestinal inflammation symptoms and molecular mechanisms were examined. We found that Lgr4−/− mice exhibited dramatically higher susceptibility to and mortality from DSS-induced inflammatory bowel disease than WT mice. Lgr4 deficiency resulted in greatly reduced numbers of either Paneth cells or stem cells in the intestine. During the intestinal regeneration process, cell proliferation but not apoptosis of intestinal epithelial cells was significantly impaired in Lgr4−/− mice. When Wnt/β-catenin signaling was reactivated by crossing with APCmin/+ mice or by treating with a GSK-3β inhibitor, the number of Paneth cells was partially restored and the mortality caused by DSS-induced inflammatory bowel disease was strikingly reduced in Lgr4-deficient animals. Thus, Lgr4 is critically involved in the maintenance of intestinal homeostasis and protection against inflammatory bowel disease through modulation of the Wnt/β-catenin signaling pathway.  相似文献   

5.
肠道是最复杂的器官之一,负责营养的吸收和消化。肠道具有多层结构保护整个肠道免受病原体的侵害。肠道上皮是由单层柱状上皮细胞组成,是抵抗病原体的第一道屏障。因此,肠上皮必须保持完整性以保护肠免受感染和毒性剂的侵害。上皮细胞分为两个谱系(吸收型与分泌型),并且每隔3~4天脱落至肠腔中。细胞的快速更替是由于肠道干细胞的存在,肠道干细胞排列在隐窝底部终极分化的潘氏细胞之间并沿隐窝绒毛轴分化成不同的上皮细胞。一旦肠道干细胞受到损伤,潘氏细胞将通过提供WNT配体和Notch刺激来补充肠道干细胞。因此,潘氏细胞充当辅助细胞以维持干细胞微环境,即生态位。该综述探讨了干细胞和潘氏细胞之间的相互作用,进一步探讨了维持肠道稳态的信号通路。  相似文献   

6.
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   

7.
8.
The concept of ‘field cancerization’ describes the clonal expansion of genetically altered, but morphologically normal cells that predisposes a tissue to cancer development. Here, we demonstrate that biased stem cell competition in the mouse small intestine can initiate the expansion of such clones. We quantitatively analyze how the activation of oncogenic K-ras in individual Lgr5+ stem cells accelerates their cell division rate and creates a biased drift towards crypt clonality. K-ras mutant crypts then clonally expand within the epithelium through enhanced crypt fission, which distributes the existing Paneth cell niche over the two new crypts. Thus, an unequal competition between wild-type and mutant intestinal stem cells initiates a biased drift that leads to the clonal expansion of crypts carrying oncogenic mutations.  相似文献   

9.
Wnt signalling induces maturation of Paneth cells in intestinal crypts   总被引:9,自引:0,他引:9  
Wnt signalling, which is transduced through beta-catenin/TCF4, maintains the undifferentiated state of intestinal crypt progenitor cells. Mutational activation of the pathway initiates the adenomacarcinoma sequence. Whereas all other differentiated epithelial cells migrate from the crypt onto the villus, Paneth cells home towards the source of Wnt signals--that is, the crypt bottom. Here, we show that expression of a Paneth gene programme is critically dependent on TCF4 in embryonic intestine. Moreover, conditional deletion of the Wnt receptor Frizzled-5 abrogates expression of these genes in Paneth cells in the adult intestine. Conversely, adenomas in Apc-mutant mice and colorectal cancers in humans inappropriately express these Paneth-cell genes. These observations imply that Wnt signals in the crypt can separately drive a stem-cell/progenitor gene programme and a Paneth-cell maturation programme. In intestinal cancer, both gene programmes are activated simultaneously.  相似文献   

10.
Wnt/β-catenin signalling plays a key role in the homeostasis of the intestinal epithelium. Whereas its role in the maintenance of the stem cell compartment has been clearly demonstrated, its role in the Paneth cell fate remains unclear. We performed genetic studies to elucidate the functions of the Wnt/β-catenin pathway in Paneth cell differentiation. We analysed mice with inducible gain-of-function mutations in the Wnt/β-catenin pathway and mice with a hypomorphic β-catenin allele that have not been previously described. We demonstrated that acute activation of Wnt/β-catenin signalling induces de novo specification of Paneth cells in both the small intestine and colon and that colon cancers resulting from Apc mutations expressed many genes involved in Paneth cell differentiation. This suggests a key role for the Wnt/β-catenin pathway in Paneth cell differentiation. We also showed that a slight decrease in β-catenin gene dosage induced a major defect in Paneth cell differentiation, but only a modest effect on crypt morphogenesis. Overall, our findings show that a high level of β-catenin activation is required to determine Paneth cell fate and that fine tuning of β-catenin signalling is critical for correct Paneth cell lineage.  相似文献   

11.
12.
Bone Morphogenetic Proteins (BMPs) form a group of secreted factors that belongs to the TGF-β superfamily. Among different roles in a number of immune cell types, BMPs are known to regulate T cell development within the thymus, although the role of BMP signaling in human mature T cells remains elusive. In this study, we demonstrate that canonical BMP signaling is necessary during two critical events that regulate the size and function of human naive CD4+ T cell population: activation and homeostasis. Upon stimulation via TCR, naive CD4+ T cells upregulate the expression of BMP ligands triggering canonical BMP signaling in CD25+ cells. Blockade of BMP signaling severely impairs CD4+ T cell proliferation after activation mainly through regulation of IL-2, since the addition of this cytokine recuperates normal T cell expansion after inhibition of BMP signaling. Similarly, activation of canonical BMP pathway is required for both the maintenance of cell survival and the homeostatic proliferation induced by IL-7, a key factor for T cell homeostasis. Moreover, upregulation of two critical receptors for T cell homeostasis, CXCR4 and CCR9, triggered by IL-7 is also abrogated in the absence of BMP signaling. Collectively, we describe important roles of the canonical BMP signaling in human naive CD4+ T cell activation and homeostasis that could be valuable for clinical application.  相似文献   

13.
Nemeth MJ  Bodine DM 《Cell research》2007,17(9):746-758
Hematopoietic stem cells (HSCs) are a rare population of cells that are responsible for life-long generation of blood cells of all lineages. In order to maintain their numbers, HSCs must establish a balance between the opposing cell fates of self-renewal (in which the ability to function as HSCs is retained) and initiation of hematopoietic differentiation. Multiple signaling pathways have been implicated in the regulation of HSC cell fate. One such set of pathways are those activated by the Wnt family of ligands. Wnt signaling pathways play a crucial role during embryogenesis and deregulation of these pathways has been implicated in the formation of solid tumors. Wnt signaling also plays a role in the regulation of stem cells from multiple tissues, such as embryonic, epidermal, and intestinal stem cells. However, the function of Wnt signaling in HSC biology is still controversial. In this review, we will discuss the basic characteristics of the adult HSC and its regulatory microenvironment, the "niche", focusing on the regulation of the HSC and its niche by the Wnt signaling pathways.  相似文献   

14.
Nat Cell Biol 14 4, 401–408 March042012The intestine represents the most vigorously renewing, adult epithelial tissue that makes maintenance of its homeostasis a delicate balance between proliferation, cell cycle arrest, migration, differentiation, and cell death. These processes are precisely controlled by a network of developmental signalling cascades, which include Wnt, Notch, BMP/TGFβ, and Hedgehog pathways. A new, elegant study by Wong et al (2012) now adds Lrig1 as a key player in the control of intestinal homeostasis. As for epidermal stem cells, Lrig1 limits the size of the intestinal progenitor compartment by dampening EGF/ErbB-triggered stem cell expansion.The epithelium of the small intestine is separated into two distinct compartments: a proliferative crypt, containing tissue-specific stem cells, and a villus with differentiated, short-lived cells, which are replenished by a constant stream of cell migration from the underlying crypt (Scoville et al, 2008). In particular, the canonical Wnt pathway in combination with Notch signals control stem cell maintenance and proliferation in the crypt. In addition, both pathways direct differentiation into the Paneth and the absorptive cell lineage, respectively. Intensive cross-talk between the epithelium and the underlying mesenchyme helps to define the crypt–villus boundary. This relies on epithelial-derived Hedgehog and Wnt ligands that trigger stromal BMP production, which in turn signals back to the epithelium to restrict proliferation to the crypt. A gradient of BMP antagonists produced by mesenchymal cells at the bottom of the crypts supports compartmentalization. In addition, a Wnt gradient in the crypt defines EphB expression and establishes repulsion-mediated separation into Paneth cell, proliferative, and differentiation zones along the crypt–villus axis (Figure 1A).Open in a separate windowFigure 1(A) The epithelium of the small intestine contains two populations of multipotent stem cells that reside at the bottom of the crypts. These give rise to transit-amplifying progenitors, which rapidly divide while migrating upwards. Cell cycle arrest and functional differentiation occur when these cells pass from the upper part of the crypt into the villus where they continue their upward movement until they finally undergo apoptosis. Only long-living Paneth cells follow a different path as they migrate downwards to populate the base of the crypt. Control of proliferation and lineage specification of all intestinal epithelial cells is directed in a self-organizing, dynamically regulated process based on cell–cell and cell–environment interactions. Among them, Wnt and Notch signalling have been defined as major determinants for stem cell maintenance, for proliferation of stem cells in the crypt and lineage specification. Epithelial-derived Hedgehog ligands and reciprocal stromal BMP ligands establish a connection between the epithelium and the stroma that regulates the crypt–villus boundary. In addition, repulsive interactions mediated by the Eph/ephrin family allow establishment of stable compartments. Importantly, ErbB signalling, which is partially suppressed by Lrig1 at the base of the crypt, is now shown to be a new key player in the control of stem and progenitor cell expansion. (B) Cross-talk of signalling pathways in intestinal homeostasis with an emphasis on ErbB signalling. A negative feedback loop via Lrig1 helps to fine-tune population size and proliferative activity of intestinal progenitor cells. Lrig1 has been identified as a direct target of Myc and is known to repress ErbB signalling. Myc itself is a main target of the ErbB and Wnt pathways implicated in intestinal stem and progenitor cell expansion. Moreover, Lrig1 has been found to promote BMP signalling, which interferes with intestinal proliferation by restricting AKT activation via PTEN.In the small intestine, two stem cell (SC) populations coexist: Lgr5+crypt base columnar cells (CBCs) that cycle every 24 h and are interspersed between Paneth cells, and slower dividing SCs concentrated above (around position +4 relative to the crypt bottom) the Lgr5+position (Takeda et al, 2011). The localization of these Hopx+mTert+slowly cycling SCs partly overlaps with that of quiescent cells, which show long-term label retention upon irradiation damage and pulse labelling with BrdU. Lgr5+CBCs are, however, dispensable (Tian et al, 2008) and can be replaced by the second stem cell population, which also shows greater activity during damage repair. The relationship between these two stem cell populations, which can reciprocally generate each other, and the mechanisms that govern quiescence are being elucidated. Importantly, leucine-rich repeats and Ig-like domains 1 (Lrig1), a transmembrane protein that interacts with ErbBs and promotes its degradation, has now been found to be enriched at the crypt base and in the progenitor compartment of the small intestine and colon (Wong et al, 2012). Lrig1 is highly expressed in Lgr5+, Musashi1+, Ascl2+, and Olfm4+CBCs, and shows an inverse relation to the pattern of activated, phosphorylated EGFR above the crypt base (Figure 1A). In line with these patterns, deletion of Lrig1 in the mouse causes a dramatic crypt expansion and increased numbers of CBCs, transit-amplifying and Paneth cells. Whether the increase of Paneth cells, which actually do not express Lrig1, is a secondary effect due to the progenitor expansion remains open. Importantly, reduction of EGFR signalling by pharmacological (Gefitinib) and genetic modulation (Egfrwa-2 mice) is able to partially normalize all Lrig1 phenotypes. These data establish EGF/ErbB signalling, as an important regulator of the crypt compartment, and suggest Lrig1 as a central control that dampens the expansion of stem cells during normal intestinal homeostasis.Lrig1 was initially identified in the skin and proposed to maintain epidermal stem cells in a quiescent state (Watt and Jensen, 2009). Lrig1 marks human interfollicular epidermal stem cells, which can give rise to all epithelial lineages including hair follicle cells in skin reconstitution assays. However, during normal homeostasis, these cells are only bipotent, contributing to the sebaceous gland and the interfollicular epidermis. In contrast to quiescent Lrig1+SCs in the skin, Lrig1+ intestinal SCs are rapidly dividing and Lrig1 appears to only reduce their proliferative capacity. However, similar to the situation in the skin, Lrig1 and EGF signalling may play an important role during damage repair. Earlier experiments analysed the phenotype of mice lacking major EGF family members (Egger et al, 1997; Troyer et al, 2001). While these mice display some duodenal lesions during normal homeostasis, further experiments established EGF signalling as a key protective component that ameliorates mucosal damage. It remains to be seen whether activation of intestinal SCs during damage repair involves mitigation of Lrig1 dampening.Lrig1 is known to repress ErbB signalling by mediating ubiquitinylation and degradation of activated receptors, thereby limiting the amplitude of EGF signalling (Watt and Jensen, 2009). Consequently, Lrig1 deletion in the intestine induced upregulation of EGFR, ErbB2, and ErbB3, promoting downstream activation of c-Myc within intestinal stem and progenitor cells (Wong et al, 2012). Importantly, Lrig1 is a direct Myc target gene, and thereby part of a negative feedback loop that helps to fine-tune the population size and proliferative activity of intestinal progenitor cells (Figure 1B).Since the rescue of the Lrig1−/− phenotype by EGFR deficiency was only partial (Wong et al, 2012), other mechanisms may contribute. Intriguingly, Lrig1 has been shown to promote BMP signalling by direct binding to Type I (ALK6) and Type II (ALK1, ALK2, ALK3, and ActRIB) BMP receptors (Gumienny et al, 2010). BMPR1A inactivation, deficiency of its downstream effector PTEN, and transgenic overexpression of the BMP inhibitor Noggin display crypt expansion and increased SC numbers. Inhibition of BMP signalling in these genetic models enhanced AKT activation and increased Wnt signalling, promoting proliferation and adenoma formation (Figure 1B; Scoville et al, 2008). Future work will reveal a potential involvement of BMP and Wnt signalling in the Lrig1 knockout phenotype.The ErbB pathway has been linked to inflammatory bowel disease, and progression and metastatic potential of colorectal cancer. EGFR inhibition blocks adenoma formation in preclinical models, and ErbB pathway inhibition is currently being evaluated in clinical trials with colorectal cancer patients, where promising results have been reported (Cunningham et al, 2004). In contrast, Lrig1 is expressed at low levels in several cancer types but is overexpressed in some prostate and colorectal tumours (Hedman and Henriksson, 2007). Given this heterogeneity, the Lrig1 function in tumours appears to be cell- and context-dependent. Due to early postnatal lethality of Lrig1 knockout mice, the exciting possibility that Lrig1 may act as an intestinal tumour suppressor could not be answered by the current study but clearly deserves further attention.  相似文献   

15.
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   

16.
17.

Background

In celiac disease (CD), intestinal epithelium damage occurs secondary to an immune insult and is characterized by blunting of the villi and crypt hyperplasia. Similarities between Hedgehog (Hh)/BMP4 downregulation, as reported in a mouse model, and CD histopathology, suggest mechanistic involvement of Hh/BMP4/WNT pathways in proliferation and differentiation of immature epithelial cells in the context of human intestinal homeostasis and regeneration after damage. Herein we examined the nature of intestinal crypt hyperplasia and involvement of Hh/BMP4 in CD histopathology.

Methods and Findings

Immunohistochemistry, qPCR and in situ hybridization were used to study a cohort of 24 healthy controls (HC) and 24 patients with diagnosed acute celiac disease (A-CD) intestinal biopsies. In A-CD we observed an increase in cells positive for Leucin-rich repeat-containing G protein-coupled receptor 5 (LGR5), an epithelial stem cell specific marker and expansion of WNT responding compartment. Further, we observed alteration in number and distribution of mesenchymal cells, predicted to be part of the intestinal stem cells niche. At the molecular level we found downregulation of indian hedgehog (IHH) and other components of the Hh pathway, but we did not observe a concurrent downregulation of BMP4. However, we observed upregulation of BMPs antagonists, gremlin 1 and gremlin 2.

Conclusions

Our data suggest that acute CD histopathology partially recapitulates the phenotype reported in Hh knockdown models. Specifically, Hh/BMP4 paradigm appears to be decoupled in CD, as the expansion of the immature cell population does not occur consequent to downregulation of BMP4. Instead, we provide evidence that upregulation of BMP antagonists play a key role in intestinal crypt hyperplasia. This study sheds light on the molecular mechanisms underlying CD histopathology and the limitations in the use of mouse models for celiac disease.  相似文献   

18.
Snail family members regulate epithelial‐to‐mesenchymal transition (EMT) during invasion of intestinal tumours, but their role in normal intestinal homeostasis is unknown. Studies in breast and skin epithelia indicate that Snail proteins promote an undifferentiated state. Here, we demonstrate that conditional knockout of Snai1 in the intestinal epithelium results in apoptotic loss of crypt base columnar stem cells and bias towards differentiation of secretory lineages. In vitro organoid cultures derived from Snai1 conditional knockout mice also undergo apoptosis when Snai1 is deleted. Conversely, ectopic expression of Snai1 in the intestinal epithelium in vivo results in the expansion of the crypt base columnar cell pool and a decrease in secretory enteroendocrine and Paneth cells. Following conditional deletion of Snai1, the intestinal epithelium fails to produce a proliferative response following radiation‐induced damage indicating a fundamental requirement for Snai1 in epithelial regeneration. These results demonstrate that Snai1 is required for regulation of lineage choice, maintenance of CBC stem cells and regeneration of the intestinal epithelium following damage.  相似文献   

19.
During intestinal regeneration, opposing gradients of Wnt and BMP signaling ensure successful differentiation along the crypt/villus axis. In this issue of The EMBO Journal, Horiguchi et al ( 2017 ) show how intestinal subepithelial myofibroblasts can influence cell fate decisions in the regenerating intestine via autocrine secretion of angiopoietin‐like protein 2 (ANGPTL2).  相似文献   

20.
Aging and carcinogenesis coincide with the accumulation of DNA damage and mutations in stem and progenitor cells. Molecular mechanisms that influence responses of stem and progenitor cells to DNA damage remain to be delineated. Here, we show that niche positioning and Wnt signaling activity modulate the sensitivity of intestinal stem and progenitor cells (ISPCs) to DNA damage. ISPCs at the crypt bottom with high Wnt/β‐catenin activity are more sensitive to DNA damage compared to ISPCs in position 4 with low Wnt activity. These differences are not induced by differences in cell cycle activity but relate to DNA damage‐dependent activation of Wnt signaling, which in turn amplifies DNA damage checkpoint activation. The study shows that instructed enhancement of Wnt signaling increases radio‐sensitivity of ISPCs, while inhibition of Wnt signaling decreases it. These results provide a proof of concept that cell intrinsic levels of Wnt signaling modulate the sensitivity of ISPCs to DNA damage and heterogeneity in Wnt activation in the stem cell niche contributes to the selection of ISPCs in the context of DNA damage.  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