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.

Background

Secreted Frizzled related proteins (SFRPs) are extracellular regulators of Wnt signaling. These proteins contain an N-terminal cysteine rich domain (CRD) highly similar to the CRDs of the Frizzled family of seven-transmembrane proteins that act as Wnt receptors. SFRPs can bind to Wnts and prevent their interaction with the Frizzled receptor. Recently it has been reported that a splice variant of human Frizzled-4 (FZD4S) lacking the transmembrane and the cytoplasmic domains of Frizzled-4 can activate rather than inhibit Wnt-8 activity in Xenopus embryos. This indicates that secreted CRD containing proteins such as Frizzled ecto-domains and SFRPs may not always act as Wnt inhibitors. It is not known how FZD4S can activate Wnt/β-catenin signaling and what biological role this molecule plays in vivo.

Results

Here we report that the Xenopus frizzled-4 is alternatively spliced to give rise to a putative secreted protein that lacks the seven-transmembrane and the cytoplasmic domains. We performed functional experiments in Xenopus embryos to investigate how this novel splicing variant, Xfz4S, can modulate the Wnt/β-catenin pathway. We show that Xfz4S as well as the extracellular domain of Xfz8 (ECD8) can act as both activators and inhibitors of Wnt/β-catenin signaling dependent on the Wnt ligand presented. The positive regulation of Wnt/β-catenin signaling by the extracellular domains of Frizzled receptors is mediated by the members of low density lipoprotein receptor-related protein (LRP-5/6) that act as Wnt coreceptors.

Conclusion

This work provides evidence that the secreted extracellular domains of Frizzled receptors may act as both inhibitors and activators of Wnt signaling dependent on the Wnt ligand presented.  相似文献   

2.
ABSTRACT: BACKGROUND: Secreted frizzled-related proteins (SFRPs) are antagonists of the Wnt signaling pathway, which plays a central role in stem cell maintenance and differentiation of stem cells and hematopoietic progenitors. Epigenetic downregulation of SFRPs by promoter hypermethylation has been described to be involved in the pathogenesis of hematopoietic malignancies. There is an association between aberrant Wnt signaling and the established cancer stem cell concept. In contrast to BCR-ABL1-positive chronic myeloid leukemia, BCR-ABL1-negative myeloproliferative neoplasms (Ph-MPN) are characterized by the frequent occurrence of an autoactivating mutation in the JAK2 tyrosine kinase (JAK2V617F) or other mutations in the JAK-STAT pathway. However, pathogenetic mechanisms of JAK2 mutated or unmutated Ph-MPN remain not completely understood.We determined the promoter methylation status of SFRP-1, -2, -4, and [MINUS SIGN]5 in 57 MPN patient samples by methylation-specific polymerase chain reaction (PCR) (MSP). JAK2V617F was assessed by allele-specific PCR. RESULTS: Aberrant methylation among primary MPN samples was 4% for SFRP-1, 25% for SFRP-2, 2% for SFRP-4, and 0% for SFRP-5. Hypermethylation of SFRP-2, which was the most frequently hypermethylated gene in our study, could not be correlated to any specific MPN subtype. However, we detected a significant correlation between SFRP-2 methylation and presence of a JAK2V617F mutation (P = 0.008). None of the 10 CML samples showed any SFRP-methylation. CONCLUSIONS: Our data indicate that epigenetic dysregulation of the Wnt signaling pathway is a common event in MPN with aberrant methylation of at least one SFRP being detected in 25% of the primary patient samples and in 30% if only accounting for Ph-MPN. A significant correlation between SFRP-2 methylation and presence of JAK2V617F in our data support the hypothesis that epigenetic dysregulation may be a complementary mechanism to genetic aberrations. Aberrant methylation of crucial stem cell maintenance genes seems to contribute to disease pathogenesis in Ph-MPN.  相似文献   

3.

Background

Conserved Wnt ligands are critical for signalling during development; however, various factors modulate their activity. Among these factors are the Secreted Frizzled-Related Proteins (SFRP). We previously isolated the SFRP-4 gene from an involuting rat mammary gland and later showed that transgenic mice inappropriately expressing SFRP-4 during lactation exhibited a high level of apoptosis with reduced survival of progeny.

Results

In order to address the questions related to the mechanism of Wnt signalling and its inhibition by SFRP-4 which we report here, we employed partially-purified Wnt-3a in a co-culture model system. Ectopic expression of SFRP-4 was accomplished by infection with a pBabepuro construct. The co-cultures comprised Line 31E mouse mammary secretory epithelial cells and Line 30F, undifferentiated, fibroblast-like mouse mammary cells. In vitro differentiation of such co-cultures can be demonstrated by induction of the β-casein gene in response to lactogenic hormones. We show here that treatment of cells with partially-purified Wnt-3a initiates Dvl-3, Akt/PKB and GSK-3β hyperphosphorylation and β-catenin activation. Furthermore, while up-regulating the cyclin D1 and connexin-43 genes and elevating transepithelial resistance of Line 31E cell monolayers, Wnt-3a treatment abrogates differentiation of co-cultures in response to the lactogenic hormones prolactin, insulin and glucocorticoid. Cells which express SFRP-4, however, are largely unaffected by Wnt-3a stimulation. Since a physical association between Wnt-3a and SFRP-4 could be demonstrated with immunoprecipitation/Western blotting experiments, this interaction, presumably owing to the Frizzled homology region typical of all SFRPs, explains the refractory response to Wnt-3a which was observed.

Conclusion

This study demonstrates that Wnt-3a treatment activates the Wnt signalling pathway and interferes with in vitro differentiation of mammary co-cultures to β-casein production in response to lactogenic hormones. Similarly, in another measure of differentiation, following Wnt-3a treatment mammary epithelial cells could be shown to up-regulate the cyclin D1 and connexin-43 genes while phenotypically they show increased transepithelial resistance across the cell monolayer. All these behavioural changes can be blocked in mammary epithelial cells expressing SFRP-4. Thus, our data illustrate in an in vitro model a mechanism by which SFRP-4 can modulate a differentiation response to Wnt-3a.  相似文献   

4.
The Wnt family of secreted glycolipoproteins plays pivotal roles in development and human diseases. Tiki family proteins were identified as novel Wnt inhibitors that act by cleaving the Wnt amino-terminal region to inactivate specific Wnt ligands. Tiki represents a new metalloprotease family that is dependent on Mn2+/Co2+ but lacks known metalloprotease motifs. The Tiki extracellular domain shares homology with bacterial TraB/PrgY proteins, known for their roles in the inhibition of mating pheromones. The TIKI/TraB fold is predicted to be distantly related to structures of additional bacterial proteins and may use a core β-sheet within an α+β-fold to coordinate conserved residues for catalysis. In this study, using assays for Wnt3a cleavage and signaling inhibition, we performed mutagenesis analyses of human TIKI2 to examine the structural prediction and identify the active site residues. We also established an in vitro assay for TIKI2 protease activity using FRET peptide substrates derived from the cleavage motifs of Wnt3a and Xenopus wnt8 (Xwnt8). We further identified two pairs of potential disulfide bonds that reside outside the β-sheet catalytic core but likely assist the folding of the TIKI domain. Finally, we systematically analyzed TIKI2 cleavage of the 19 human WNT proteins, of which we identified 10 as potential TIKI2 substrates, revealing the hydrophobic nature of Tiki cleavage sites. Our study provides insights into the Tiki family of proteases and its Wnt substrates.  相似文献   

5.
6.
Hematopoietic stem cells (HSCs) can self-renew and differentiate into all cell types of the blood. This is therapeutically important as HSC transplants can provide a curative effect for blood cancers and disorders. The process by which HSCs develop has been the subject of extensive research in a variety of model organisms; however, efforts to produce bonafide HSCs from pluripotent precursors capable of long-term multilineage reconstitution have fallen short. Studies in zebrafish, chicken, and mice have been instrumental in guiding efforts to derive HSCs from human pluripotent stem cells and have identified a complex set of molecular signals and cellular interactions mediated by such developmental regulators as fibroblast growth factor, Notch, transforming growth factor beta (TGFβ), and Wnt, which collectively promote the stepwise developmental progression toward mature HSCs. Tight temporal and spatial control of these signals is critical to generate the appropriate numbers of HSCs needed for the life of the organism. The role of the Wnt family of signaling proteins in hematopoietic development has been the subject of many studies owing in part to the complex nature of its signaling mechanisms. By integrating cell fate specification with cell polarity establishment, Wnt is uniquely capable of controlling complex biological processes, including at multiple stages of embryonic HSC development, from HSC specification to emergence from the hemogenic epithelium to subsequent expansion. This review highlights key signaling events where specific Wnt signals instruct and guide hematopoietic development in both zebrafish and mice and extend these findings to current efforts of generating HSCs in vitro.  相似文献   

7.
Wnt proteins and their receptors, members of the frizzled protein family, play a key role in regulating a wide range of developmental processes. Recently, putative regulators of Wnt signaling known as secreted frizzled-related proteins (SFRPs) have been identified in several vertebrates. Here, we describe the cloning of a novel SFRP (suSFRP1) from the sea urchin, Strongylocentrotus purpuratus. SuSFRP1 contains a putative signal sequence, four cysteine-rich domains and a single Ig domain. The developmental expression of suSFRP1 mRNA is highly dynamic and can be separated into three phases: (1) abrupt accumulation in most or all cells of the embryo at the early blastula stage; (2) restriction of expression to the prospective endoderm and animal pole region of the gastrula; and (3) expression in prospective muscle cells of the coelomic pouches during late embryogenesis.  相似文献   

8.
The Wnt (wingless-type) signaling pathway plays an important role in embryonic development, tissue homeostasis, and tumor progression becaluse of its effect on cell proliferation, migration, and differentiation. Secreted frizzled-related proteins (SFRPs) are extracellular inhibitors of Wnt signaling that act by binding directly to Wnt ligands or to Frizzled receptors. In recent years, aberrant expression of SFRPs has been reported to be associated with numerous cancers. As gene expression of SFRP members is often lost through promoter hypermethylation, inhibition of methylation through the use of epigenetic modifying agents could renew the expression of SFRP members and further antagonize deleterious Wnt signaling. Several reports have described epigenetic silencing of these Wnt signaling antagonists in various human cancers, suggesting their possible role as tumor suppressors. SFRP family members thus come across as potential tools in combating Wnt-driven tumorigenesis. However, little is known about SFRP family members and their role in different cancers. This review comprehensively covers all the available information on the role of SFRP molecules in various human cancers.  相似文献   

9.
Hematopoietic stem cells (HSCs) reside in hypoxic areas of the bone marrow. However, the role of hypoxia in the maintenance of HSCs has not been fully characterized. We performed xenotransplantation of human cord blood cells cultured in hypoxic or normoxic conditions into adult NOD/SCID/IL-2Rγnull (NOG) mice. Hypoxic culture (1% O2) for 6 days efficiently supported the maintenance of HSCs, although cell proliferation was suppressed compared to the normoxic culture. In contrast, hypoxia did not affect in vitro colony-forming ability. Upregulation of a cell cycle inhibitor, p21, was observed in hypoxic culture. Immunohistochemical analysis of recipient bone marrow revealed that engrafted CD34+CD38 cord blood HSCs were hypoxic. Taken together, these results demonstrate the significance of hypoxia in the maintenance of quiescent human cord blood HSCs.  相似文献   

10.
11.
Inhibitors of phosphodiesterase 5 (PDE5) are widely used to treat erectile dysfunction and pulmonary hypertension in clinics. PDE5, cyclic guanosine monophosphate (cGMP), and protein kinase G (PKG) are important components of the non-canonical Wnt signaling. This study aimed to investigate the effect of PDE5 inhibition on canonical Wnt signaling and osteoblastogenesis, using both in vitro cell culture and in vivo animal models. In the in vitro experiments, PDE5 inhibition resulted in activation of cGMP-dependent protein kinase 2 and consequent inhibition of glycogen synthase kinase 3β phosphorylation, destabilization of cytosolic β-catenin and the ultimate suppression of canonical Wnt signaling and reduced osteoblastic differentiation in HEK293T and C3H10T1/2 cells. In animal experiments, systemic inhibition of PDE5 suppressed the activity of canonical Wnt signaling and osteoblastogenesis in bone marrow-derived stromal cells, resulting in the reduction of bone mass in wild-type adult C57B/6 mice, significantly attenuated secreted Frizzled-related protein-1 (SFRP1) deletion-induced activation of canonical Wnt signaling and excessive bone growth in adult SFRP1−/− mice. Together, these results uncover a hitherto uncharacterized role of PDE5/cGMP/PKG signaling in bone homeostasis and provide the evidence that long-term treatment with PDE5 inhibitors at a high dosage may potentially cause bone catabolism.In the canonical Wnt (Wnt/β-catenin (β-cat)) signaling cascade, Wnt binds to Frizzled (Frz) receptors and the low-density lipoprotein receptor-related protein (LRP) 5 or 6, thereby activating dishevelled, suppressing the glycogen synthase kinase 3β (GSK3β) activity and inhibiting phosphorylation of β-cat at Thr41, Ser37, and Ser33 sites. The stabilized cytosolic β-cat enters the nucleus and consequently activates its downstream target genes via lymphoid enhancer-binding factor-1 (Lef-1) and T-cell factors.1, 2 This signaling is fine-tuned in part via a negative feedback mechanism involving secreted and transmembrane Wnt inhibitors and activators, secreted Frz-related proteins (SFRPs), and Dickkopf-1 (Dkk1).3, 4Canonical Wnt signaling is critical not only to bone development in embryogenesis but also to the maintenance of bone mass during adult life.5 The initial evidence came from the discoveries that in humans loss- or gain-of-function mutations in LRP5 were linked with the osteoporosis-pseudoglioma syndrome and a high-bone-density syndrome, respectively.6, 7, 8 Subsequent studies in mice showed that Wnt signaling might promote ossification by inducing the differentiation of bone-forming osteoblasts, suppressing the development of bone-resorbing osteoclasts, and driving the differentiation of multi-potent stem cells toward an osteoblast cell fate.9Non-canonical Wnt signaling is β-cat independent and consists of two main pathways: the Rho small GTPases-mediated planar cell polarity pathway and the Wnt/Ca2+ pathway,10 involved in various aspects of cell fate differentiation and cell movement. Non-canonical Wnt signaling has profound effects on tissue morphogenesis in a variety of vertebrate species.10 The potential role for non-canonical Wnt signaling in bone formation has been investigated recently in limited studies, which have shown that the non-canonical Wnt-Gαq/11-PKC pathway operates in mammalian osteoprogenitors to promote osteoblast development, and that Wnt16 exhibits a stimulatory effect on bone metabolism.11, 12, 13 Nevertheless, the molecular events in the non-canonical Wnt signaling regulation of bone development and homeostasis have yet to be further elucidated.Phosphodiesterases (PDEs) are a large family of enzymes that cleave cyclic nucleotides. To date, 11 PDE subtypes have been identified, among which PDE5 has been most extensively studied. PDE5, cyclic guanosine monophosphate (cGMP), and cGMP-dependent protein kinase (PKG) are among the major components of the non-canonical Wnt signaling pathway and are involved in the regulation of intracellular Ca2+ concentration.14, 15 It is now well established that PDE5 degrades 3''-5′- cGMP and its inhibition leads to an increase in intracellular cGMP levels and activation of protein kinase G (PKG), resulting in a decrease in Ca2+ influx and consequent relaxation of smooth muscles, which produces the therapeutic effects in clinical erectile dysfunction (ED) and pulmonary hypertension (PH).14 Currently, little is known regarding the involvement of PDE5 in Wnt signaling regulation of bone formation and homeostasis. The objective of this study was to determine the effect of PDE5 inhibition on canonical Wnt signaling and bone mass.  相似文献   

12.
13.
Functional decline of the hematopoietic system occurs during aging and contributes to clinical consequences, including reduced competence of adaptive immunity and increased incidence of myeloid diseases. This has been linked to aging of the hematopoietic stem cell (HSC) compartment and has implications for clinical hematopoietic cell transplantation as prolonged periods of T‐cell deficiency follow transplantation of adult mobilized peripheral blood (PB), the primary transplant source. Here, we examined the gene expression profiles of young and aged HSCs from human cord blood and adult mobilized PB, respectively, and found that Wnt signaling genes are differentially expressed between young and aged human HSCs, with less activation of Wnt signaling in aged HSCs. Utilizing the OP9‐DL1 in vitro co‐culture system to promote T‐cell development under stable Notch signaling conditions, we found that Wnt signaling activity is important for T‐lineage differentiation. Examination of Wnt signaling components and target gene activation in young and aged human HSCs during T‐lineage differentiation revealed an association between reduced Wnt signal transduction, increasing age, and impaired or delayed T‐cell differentiation. This defect in Wnt signal activation of aged HSCs appeared to occur in the early T‐progenitor cell subset derived during in vitro T‐lineage differentiation. Our results reveal that reduced Wnt signaling activity may play a role in the age‐related intrinsic defects of aged HSCs and early hematopoietic progenitors and suggest that manipulation of this pathway could contribute to the end goal of improving T‐cell generation and immune reconstitution following clinical transplantation.  相似文献   

14.
Kremen1 and Kremen2 (Krm1 and Krm2) are transmembrane coreceptors for Dickkopf1 (Dkk1), an antagonist of Wnt/β-catenin signaling. The physiological relevance of Kremen proteins in mammals as Wnt modulators is unresolved. We generated and characterized Krm mutant mice and found that double mutants show enhanced Wnt signaling accompanied by ectopic postaxial forelimb digits and expanded apical ectodermal ridges. Triple mutant Krm1−/ Krm2−/ Dkk1+/ mice show enhanced growth of ectopic digits, indicating that Dkk1 and Krm genes genetically interact during limb development. Wnt/β-catenin signaling also plays a critical role in bone formation. Single Krm mutants show normal bone formation and bone mass, while double mutants show increased bone volume and bone formation parameters. Our study provides the first genetic evidence for a functional interaction of Kremen proteins with Dkk1 as negative regulators of Wnt/β-catenin signaling and reveals that Kremen proteins are not universally required for Dkk1 function.  相似文献   

15.
Metazoans display remarkable conservation of gene families, including growth factors, yet somehow these genes are used in different ways to generate tremendous morphological diversity. While variations in the magnitude and spatio-temporal aspects of signaling by a growth factor can generate different body patterns, how these signaling variations are organized and coordinated during development is unclear. Basic body plans are organized by the end of gastrulation and are refined as limbs, organs, and nervous systems co-develop. Despite their proximity to developing tissues, neurons are primarily thought to act after development, on behavior. Here, we show that in Caenorhabditis elegans, the axonal projections of neurons regulate tissue progenitor responses to Wnts so that certain organs develop with the correct morphology at the right axial positions. We find that foreshortening of the posteriorly directed axons of the two canal-associated neurons (CANs) disrupts mid-body vulval morphology, and produces ectopic vulval tissue in the posterior epidermis, in a Wnt-dependent manner. We also provide evidence that suggests that the posterior CAN axons modulate the location and strength of Wnt signaling along the anterior–posterior axis by employing a Ror family Wnt receptor to bind posteriorly derived Wnts, and hence, refine their distributions. Surprisingly, despite high levels of Ror expression in many other cells, these cells cannot substitute for the CAN axons in patterning the epidermis, nor can cells expressing a secreted Wnt inhibitor, SFRP-1. Thus, unmyelinated axon tracts are critical for patterning the C. elegans body. Our findings suggest that the evolution of neurons not only improved metazoans by increasing behavioral complexity, but also by expanding the diversity of developmental patterns generated by growth factors such as Wnts.  相似文献   

16.
Corneal epithelial wound repair involves the migration of epithelial cells to cover the defect followed by the proliferation of the cells to restore thickness. Heparan sulfate proteoglycans (HSPGs) are ubiquitous extracellular molecules that bind to a plethora of growth factors, cytokines, and morphogens and thereby regulate their signaling functions. Ligand binding by HS chains depends on the pattern of four sulfation modifications, one of which is 6-O-sulfation of glucosamine (6OS). SULF1 and SULF2 are highly homologous, extracellular endosulfatases, which post-synthetically edit the sulfation status of HS by removing 6OS from intact chains. The SULFs thereby modulate multiple signaling pathways including the augmentation of Wnt/ß-catenin signaling. We found that wounding of mouse corneal epithelium stimulated SULF1 expression in superficial epithelial cells proximal to the wound edge. Sulf1−/−, but not Sulf2−/−, mice, exhibited a marked delay in healing. Furthermore, corneal epithelial cells derived from Sulf1−/− mice exhibited a reduced rate of migration in repair of a scratched monolayer compared to wild-type cells. In contrast, human primary corneal epithelial cells expressed SULF2, as did a human corneal epithelial cell line (THCE). Knockdown of SULF2 in THCE cells also slowed migration, which was restored by overexpression of either mouse SULF2 or human SULF1. The interchangeability of the two SULFs establishes their capacity for functional redundancy. Knockdown of SULF2 decreased Wnt/ß-catenin signaling in THCE cells. Extracellular antagonists of Wnt signaling reduced migration of THCE cells. However in SULF2- knockdown cells, these antagonists exerted no further effects on migration, consistent with the SULF functioning as an upstream regulator of Wnt signaling. Further understanding of the mechanistic action of the SULFs in promoting corneal repair may lead to new therapeutic approaches for the treatment of corneal injuries.  相似文献   

17.
Wnt signaling negatively regulates adipocyte differentiation, and ectopic expression of Wnt-1 in 3T3-L1 cells induces several downstream molecules of Wnt signaling, including Wnt-1 inducible signaling pathway protein (WISP)-2. In this study, we examined the role of WISP-2 in the process of adipocyte differentiation using an in vitro cell culture system. In the differentiation of 3T3-L1 cells, WISP-2 expression was observed in growing cells and declined thereafter. In the mitotic clonal expansion phase of adipocyte differentiation, WISP-2 expression was transiently down-regulated concurrently with up-regulation of CCAAT/enhancer–binding protein δ expression. Treatment of 3T3-L1 cells in the differentiation medium with lithium, an activator of Wnt signaling, inhibited the differentiation process with concomitant induction of WISP-2. Treatment of differentiated cells with lithium induced de-differentiation as evidenced by profound reduction of peroxisome proliferator–activator receptor γ expression and concomitant induction of WISP-2. However, de-differentiation of differentiated cells induced by tumor necrosis factor-α did not induce WISP-2 expression. To directly examine the effect of WISP-2 on adipocyte differentiation, 3T3-L1 cells were infected with a retrovirus carrying WISP-2. Although forced expression of WISP-2 inhibited preadipocyte proliferation, it had no effect on adipocyte differentiation. Thus, although WISP-2 is a downstream protein of Wnt signaling, the role of WISP-2 on adipocyte differentiation may be marginal, at least in this in vitro culture model.  相似文献   

18.
Previously we reported that Wnt5a is highly expressed in the murine urogenital ridge-derived UG26-1B6 cells but not embryonic liver-derived EL08-1D2 cells. Mouse long-term repopulating hematopoietic stem cells (LTR-HSC) were maintained in non-contact UG26-1B6 cultures but not EL08-1D2 non-contact cultures, unless Wnt5a was also added to the cultures, suggesting a role for Wnt5a in the in vitro maintenance of LTR-HSC. Here, we investigated if the effect of Wnt5a on adult LTR-HSC activity is HSC-autonomous. To test the effect of Wnt5a on maintenance of LTR-HSC, we performed limiting dilution competitive transplantation assays of murine Lin-Sca1+ c-kit+ (LSK) cells cultured for 5 days with TPO and SCF with and without Wnt5a. The effect of Wnt5a on the generation of colony forming units (CFU) and the homing ability of LSK progeny was also tested. No effects were found of Wnt5a on total cell expansion, the number of CFU, or homing ability of day 5 LSK progeny. Furthermore, addition of Wnt5a did not improve, but may have impeded maintenance of LTR-HSC. In conclusion, our data indicate that Wnt5a does not enhance the maintenance and expansion of adult murine LTR-HSCs or committed progenitors cultured in vitro in serum- and stroma-free conditions.  相似文献   

19.
Maintenance of hematopoietic stem cells (HSCs) in vitro has been believed to be difficult due to a lack of complete understanding of HSC quiescence maintained by the niche. Recent evidence suggests that in vitro maintenance of human and mouse long-term HSCs (LT-HSCs) is possible through dual inhibition (2i) of both GSK-3 and mTOR in the absence of cytokines, serum, or feeder cells.Hematopoietic stem cells (HSCs) are generally quiescent, and have the ability to self-renew or to differentiate into mature blood cells. Despite recent advances, it has not been possible to maintain functional long-term HSCs (LT-HSCs) outside the hematopoietic niche, because mechanisms by which HSC quiescence is maintained by the niche1 have not been fully understood. There have been many attempts to expand HSCs and hematopoietic progenitor cells in vitro using hematopoietic cytokines combined with factors, including Wnt activators2,3,4, glycogen synthase kinase 3 (GSK-3) inhibitors5, Notch ligand, HoxB4, prostaglandin E2, aryl hydrocarbon receptor antagonists, angiopoietin-like proteins, or pleiotrophin6,7. However, all studies have required hematopoietic cytokines, which may promote lineage commitment at the expense of LT-HSC maintenance.Huang et al.8 previously reported that disruption of GSK-3 in hematopoietic cells in mice leads to an increase in the number of HSCs through Wnt activation, and that the subsequent depletion of LT-HSCs occurs because inhibition of GSK-3 also activates mammalian target of rapamycin (mTOR) (Figure 1A). The mTOR pathway is recognized as an established nutrient sensor, and nutrient-sensing systems are associated with HSC homeostasis. Indeed, HSCs reside in a low-perfusion environment in the bone marrow with low oxygen and low nutrition. Activation of mTOR has been shown to increase the proliferation of committed progenitors at the cost of HSC maintenance (Figure 1A), indicating that low nutrient availability is an essential characteristic of the niche. Thus, Huang et al. hypothesized that low nutrient availability might contribute to HSC maintenance.Open in a separate windowFigure 1Schematic diagrams of maintenance of LT-HSCs. (A) Disruption of GSK-3 results in HSC self-renewal through Wnt activation in a β-catenin-dependent manner. However, in assays of LT-HSC function, disruption of GSK-3 leads to depletion of HSCs through activation of mTOR. GSK-3 regulates both Wnt and mTOR signalings in HSCs8. (B) GSK-3 inhibition-induced mTOR activation is attributable to HSC depletion, which can be prevented by mTOR inhibition9. (C) In vitro maintenance of LT-HSCs is possible by dual inhibition (2i) of GSK-3 and mTOR under cytokine-free, serum-free, feeder-free conditions9. CHIR99021 and lithium are GSK-3 inhibitors, and rapamycin is mTOR inhibitor.Recently, Huang et al.9 clearly demonstrate that human and mouse LT-HSCs can be maintained in vitro by inhibiting both GSK-3 and mTOR, in the absence of cytokines, serum, or feeder cells (Figure 1B). Moreover, the combination of two clinically approved inhibitors, lithium (GSK-3) and rapamycin (mTOR) (Figure 1C), increases the number of functional LT-HSCs in mice. First, Huang et al.9 determined whether dual inhibition (2i) of GSK-3 and mTOR would be sufficient for maintaining HSCs in vitro. They cultured mouse c-Kit+ or LinSca1+c-Kit+ (LSK) cells in X-VIVO 15 (Lonza) (which is chemically defined, serum-free, hematopoietic cell medium) supplemented with inhibitors of GSK-3 (CHIR99021 or lithium) and mTOR (rapamycin) for 7 days in the absence of cytokines, serum, or feeder cells. They subsequently assessed the hematopoietic potential of the cultured HSCs by competitive repopulation assay. It was confirmed that HSCs cultured with 2i maintained long-term reconstitution potential, and that the frequency of HSCs was similar to that in uncultured c-Kit+ cells. Similarly, they confirmed that the effects of 2i on LT-HSCs were recapitulated in human HSCs that are present in umbilical cord blood CD34+ cells. To explore the mechanism by which 2i preserves HSCs, they also investigated cell cycle status in mouse LSK cells. They found an increased percentage of quiescent cells by 2i, suggesting that the maintenance of LSK cells by 2i is the result of increased dormancy in vitro. Finally, they demonstrated that GSK-3 and mTOR inhibition increases mouse LT-HSCs in vivo. They treated mice with lithium and rapamycin for 2 weeks, and found that both the overall bone marrow cellularity and the absolute number of LT-HSCs increased in the treatment group. In a competitive repopulation assay, the absolute number of competitive rescue units was increased by 2-fold in bone marrow of treated mice.The above findings by Huang et al.9 are outstanding, but many questions need to be answered in future studies. i) The authors examined 2i cultures for 7 days in vitro (and 2 weeks in vivo), and it would be interesting to examine how long it is possible to maintain LT-HSCs in vitro under 2i condition. However, as the authors mentioned9, prolonged activation of Wnt signaling might be associated with transformation in vitro, and might have the risk of inducing colorectal cancers and leukemias when GSK-3 inhibitors are administered in vivo10. Nevertheless, lithium (GSK-3 inhibitor) has been used to treat bipolar disorder for over 50 years and is not associated with an increased risk of malignancies11, as the authors pointed out9. ii) In cytokine-free medium, is there cytokine production by HSCs or progenitor cells? It may be possible that cytokine production would contribute to the maintenance of LT-HSCs in an autocrine or paracrine manner. iii) Although feeder cells and/or serum are not defined factors for culture, it would be of interest to investigate whether 2i culture in the presence of supporting cells would further improve the maintenance of LT-HSCs. Some extrinsic regulators for HSC quiescence12, such as N-cadherins, could contribute to LT-HSC maintenance in cooperation with 2i. iv) In addition to iii), hypoxic environment is known to be an extrinsic regulator for HSC quiescence12, as the bone marrow niche is a low-perfusion environment. Hypoxic culture might synergize with 2i. v) When human ESCs/iPSCs are induced to differentiate into HSCs, it is difficult to capture true human LT-HSCs in vitro. If this is due to inability to maintain human LT-HSCs in vitro, it would be interesting to examine whether 2i culture would enable in vitro induction and maintenance of transplantable LT-HSCs derived from human ESCs/iPSCs.In summary, dual inhibition (2i) of GSK-3 and mTOR allows for the maintenance of human and mouse LT-HSCs in vitro (Figure 1C), and this may resolve the difficulty in culturing HSCs, which in turn, may improve basic research of HSCs (e.g., gene editing in vitro) and human HSC transplantation outcomes. Furthermore, although the effect of 2i on expansion of HSCs is relatively small, a combination of 2i drugs may increase human clinical trials1,6,7 that use 2i in vivo for the aim of increasing the number of LT-HSCs, since 2i drugs are known as clinically tolerated medications. Insights gained from the discovery of 2i for HSC maintenance may lead to great benefits for patients with hematologic disorders, hopefully in the near future.  相似文献   

20.
Dishevelled (Dvl) proteins are intracellular effectors of Wnt signaling that have essential roles in both canonical and noncanonical Wnt pathways. It has long been known that Wnts stimulate Dvl phosphorylation, but relatively little is known about its functional significance. We have previously reported that both Wnt3a and Wnt5a induce Dvl2 phosphorylation that is associated with an electrophoretic mobility shift and loss of recognition by monoclonal antibody 10B5. In the present study, we mapped the 10B5 epitope to a 16-amino acid segment of human Dvl2 (residues 594–609) that contains four Ser/Thr residues. Alanine substitution of these residues (P4m) eliminated the mobility shift induced by either Wnt3a or Wnt5a. The Dvl2 P4m mutant showed a modest increase in canonical Wnt/β-catenin signaling activity relative to wild type. Consistent with this finding, Dvl2 4Pm preferentially localized to cytoplasmic puncta. In contrast to wild-type Dvl2, however, the P4m mutant was unable to rescue Wnt3a-dependent neurite outgrowth in TC-32 cells following suppression of endogenous Dvl2/3. Earlier work has implicated casein kinase 1δ/ϵ as responsible for the Dvl mobility shift, and a CK1δ in vitro kinase assay confirmed that Ser594, Thr595, and Ser597 of Dvl2 are CK1 targets. Alanine substitution of these three residues was sufficient to abrogate the Wnt-dependent mobility shift. Thus, we have identified a cluster of Ser/Thr residues in the C-terminal domain of Dvl2 that are Wnt-induced phosphorylation (WIP) sites. Our results indicate that phosphorylation at the WIP sites reduces Dvl accumulation in puncta and attenuates β-catenin signaling, whereas it enables noncanonical signaling that is required for neurite outgrowth.  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