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 375 毫秒
1.
<正>Since the first murine and human embryonic stem cell lines were established by Drs. Evans and Kaufman [1] and Thomson et al. [2], respectively, great progress has been make in the field of  相似文献   

2.
Neural stem cells (NSC) have been implicated not only in brain development and neurogenesis but also in tumourigenesis. Brain tumour stem cells (BTSC) have been isolated from several paediatric or adult human brain tumours, however their origin is still disputed. This review discusses the normal role of NSC in the adult mammalian brain and their anatomical location. It compares the molecular characteristics and the biological behaviour of NSC/BTSC, and describes the molecular pathways involved in controlling self-renewal and maintenance of adult NSC/BTSC and brain tumour development. It also assesses the current hypotheses about the origin of BTSC and the clinical consequences.  相似文献   

3.
Zhang QB  Ji XY  Huang Q  Dong J  Zhu YD  Lan Q 《Cell research》2006,16(12):909-915
Understanding of the differentiation profile of brain tumor stem cells (BTSCs), the key ones among tumor cell population, through comparison with neural stem cells (NSCs) would lend insight into the origin of glioma and ultimately yield new approaches to fight this intractable disease. Here, we cultured and purified BTSCs from surgical glioma specimens and NSCs from human fetal brain tissue, and further analyzed their cellular biological behaviors, especially their differentiation property. As expected, NSCs differentiated into mature neural phenotypes. In the same differentiation condition, however, BTSCs exhibited distinguished differences. Morphologically, cells grew flattened and attached for the first week, but gradually aggregated and reformed floating tumor sphere thereafter. During the corresponding period, the expression rate of undifferentiated cell marker CD 133 and nestin in BTSCs kept decreasing, but 1 week later, they regained ascending tendency. Interestingly, the differentiated cell markers GFAP and β-tubulinlII showed an expression change inverse to that of undifferentiated cell markers. Taken together, BTSCs were revealed to possess a capacity to resist differentiation, which actually represents the malignant behaviors of glioma.  相似文献   

4.
Placenta-derived stem cells (PDSCs) have gained interest as an alternative source of stem cells for regenerative medicine because of their potential for self-renewal and differentiation and their immunomodulatory properties. Although many studies have characterized various PDSCs biologically, the properties of the self-renewal and differentiation potential among PDSCs have not yet been directly compared. We consider the characterization of chorionic-plate-derived mesenchymal stem cells (CP-MSCs) and Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) among various PDSCs and the assessment of their differentiation potential to be important for future studies into the applicability and effectiveness of PDSCs in cell therapy. In the present study, the capacities for self-renewal and multipotent differentiation of CP-MSCs and WJ-MSC isolated from normal term placentas were compared. CP-MSCs and WJ-MSCs expressed mRNAs for the pluripotent stem cell markers Oct-4, Nanog, and Sox-2. Additionally, HLA-G for immunomodulatory effects was found to be expressed at both the mRNA and protein levels in both cell types. The CP-MSCs and WJ-MSCs also had the capacities to differentiate into cells of mesodermal (adipogenic and osteogenic) and endodermal (hepatogenic) lineages. Expression of adipogenesis-related genes was higher in CP-MSCs than in WJ-MSCs, whereas WJ-MSCs accumulated more mineralized matrix than CP-MSCs. The WJ-MSCs expressed more of CYP3A4 mRNA, a marker for mature hepatocytes, than CP-MSCs. Thus, we propose that CP-MSCs and WJ-MSCs are useful sources of cells for appropriate clinical applications in the treatment of various degenerative diseases.  相似文献   

5.
Embryonic stem cells without embryos?   总被引:2,自引:0,他引:2  
Grompe M 《Nature biotechnology》2005,23(12):1496-1497
  相似文献   

6.
7.
8.
Multipotent hematopoietic stem cells are maintained by the bone marrow niche, but how niche-derived membrane-bound stem cell factor (mSCF) regulates HSCs remains unclear. In this issue, Hao et al. (2021. J. Cell Biol. https://doi.org/10.1083/jcb.202010118) describe that mSCF, synergistically with VCAM-1, induces large, polarized protrusions that serve as anchors for HSCs to their niche.

Hematopoietic stem cells (HSCs) generate all blood and immune cells throughout life via self-renewal and multilineage differentiation within the bone marrow niche. HSCs are the basis for bone marrow transplantation, saving thousands of lives yearly. The bone marrow niche often serves as a paradigm for studying stem cell biology. In addition, elucidating the underlying mechanism in the niche helps devise strategies to expand functional HSCs for clinical use. Within the niche, leptin receptor–positive perisinusoidal stromal cells and endothelial cells are the major source of essential cytokines for HSC maintenance, including vascular cell adhesion molecule 1 (VCAM-1) and stem cell factor (SCF; 1, 2). Locally produced soluble and membrane-bound cytokines preserve the unique localization and anchorage of HSCs to stromal cells within their niche. Consistent with this notion, mouse genetic data have shown that membrane-bound SCF (mSCF) is important for HSC maintenance in vivo (3). However, given that both soluble and membrane-bound forms of SCF can engage with the cognate cKIT receptors, the mechanisms by which mSCF sustains HSCs function in vivo remain elusive. Likewise, it is unclear why the expansion and maintenance of HSCs ex vivo by adding SCF to culture as an either soluble or immobilized form has only been achieved with limited success.In this issue, Hao et al. addressed this question by using a supported lipid bilayer (SLB) system to model the interaction between HSCs and membrane-bound cytokines, including SCF (4). SLBs present an advantage over conventional immobilization methods; they allow the lateral mobility of membrane-bound proteins and clustering of receptors and signaling complexes, thus resembling the lipid bilayer of plasma membrane in vivo. Focusing on HSC cytokines that may be presented as membrane-bound forms in the bone marrow niche, the authors performed an imaging screen in vitro using SLBs and found that mSCF but not soluble SCF (sSCF) induced mSCF/cKIT clustering and the formation of membrane protrusions on HSCs. While mSCF alone was sufficient to promote cell protrusions, HSCs required both mSCF and VCAM-1 for large, polarized protrusions. They followed HSCs at different time points after exposure to VCAM-1 and mSCF by scanning electron microscopy and observed that HSCs first formed diffuse mSCF clusters and multifocal thin protrusions and then proceeded to a polarized, clustered morphology with larger and thicker protrusions. Using a controlled sheer stress device, Hao et al. showed that these polarized protrusions had a functional consequence on the adhesion strength of HSCs. mSCF and VCAM-1 dramatically increased the adhesion of HSCs to SLB compared with VCAM-1 or mSCF alone. Interestingly, the effect was more prominent in HSCs compared with their immediate downstream progenies, multipotent progenitors. This phenotype was also specific to ligands presented on SLB because the effect was canceled when the cytokines were directly immobilized onto the glass surface. Then, they had a close look into the cytoskeletal organization of HSCs in the presence of both mSCF and VCAM-1 on SLB. They found that F-actin and myosin IIa concentrated at the protrusion, which led them to speculate that the cytoskeleton remodeling mediates the formation of the polarized morphology. Indeed, chemical inhibitors blocking myosin contraction, actin polymerization, or Rho-associated protein kinase disrupted the formation of the large and polarized protrusion. The authors noted that phosphatidylinositol 3-kinase (PI3K) also localized with mSCF/cKIT clusters, so they further assessed the contribution of the PI3K/Akt pathway to the polarized morphology of HSCs by using total internal reflection fluorescence microscopy and PI3K and Akt chemical inhibitors. PI3K/Akt activation contributed downstream of the mSCF–VCAM-1 synergy to regulating HSC cell adhesion and polarized mSCF/cKIT distribution. In addition, PI3K signaling enhanced the nuclear retention of FOXO3a, a crucial factor for HSC self-renewal; this enhancement was induced by mSCF but lessened by sSCF. Intriguingly, sSCF also competed with mSCF and abrogated the effect of the mSCF–VCAM-1 synergy on polarized protrusion formation. However, whether and how PI3K transmits the mSCF–VCAM-1 synergy into proliferation or quiescence cues in HSCs requires further investigation. Taken together, these data suggest that mSCF and VCAM-1 synergize to induce polarized protrusions on HSCs, which regulates their adhesion to the niche (Fig. 1). These protrusions share many features with the immunological synapse (5), which points toward the existence of a similar model for stem cells, “stem cell synapse,” where HSCs interact with and receive a variety of signals from their niche cells.Open in a separate windowFigure 1.VCAM-1 and mSCF synergistically promote the formation of polarized protrusions (stem cell synapse) on HSCs. (A and B) VCAM-1 or mSCF alone does not induce apparent polarized morphology on HSCs. The signaling and adhesion of HSCs to the niche is not at its full potential. (C) VCAM-1 and mSCF together induce robust receptor clustering on HSCs, optimal signaling, and strong adhesion. (D) sSCF can competitively disrupt the polarized protrusions on HSCs. The figure was created with BioRender.com.While the study by Hao et al. sheds light on how niche signals, particularly mSCF, regulate HSCs, several outstanding questions remain. First, even though many hematopoietic cells express cKIT (some of them even express higher levels than HSCs), HSCs respond to mSCF + VCAM-1 the strongest by recruiting the most mSCF to clusters. What is the specific mechanism in HSCs underlying this specificity? Second, SCF is produced both as mSCF and sSCF in vivo, through alternative splicing and proteolytic cleavage; if mSCF is mainly responsible for anchoring HSCs in the niche, what is the function of sSCF in vivo? Does sSCF modulate the available pool of mSCF? Third, robust maintenance of HSCs in culture has been challenging. HSCs can be maintained in a system composed of sSCF, thromopoietin (TPO), fibronectin, and polyvinyl alcohol (6). Tethering cytokines to SLB elicits more physiological response from HSCs compared with soluble cytokines or direct immobilization. Does SLB improve maintenance of HSCs in in vitro culture? Fourth, some cytokines, such as TPO, act on HSCs in a long-range manner (7). How do these systemic cytokines induce robust signaling in HSCs? Do they participate in the stem cell synapse even if they are not the initiators? Finally, do stem cells and their niche interact by forming similar synapses in other stem cell systems? Answering these questions will deepen our understanding of the stem cell niche and help integrate the niche component into potential, more successful applications in regenerative medicine.  相似文献   

9.
10.

Background

The diversity of cell types and tissue types that originate throughout development derives from the differentiation potential of embryonic stem cells and somatic stem cells. While the former are pluripotent, and thus can give rise to a full differentiation spectrum, the latter have limited differentiation potential but drive tissue remodeling. Additionally cancer tissues also have a small population of self-renewing cells with stem cell properties. These cancer stem cells may arise through dedifferentiation from non-stem cells in cancer tissues, illustrating their plasticity, and may greatly contribute to the resistance of cancers to chemotherapies.

Scope of review

The capacity of the different types of stem cells for self-renewal, the establishment and maintenance of their differentiation potential, and the selection of differentiation programs are greatly defined by the interplay of signaling molecules provided by both the stem cells themselves, and their microenvironment, the niche. Here we discuss common and divergent roles of TGF-β family signaling in the regulation of embryonic, reprogrammed pluripotent, somatic, and cancer stem cells.

Major conclusions

Increasing evidence highlights the similarities between responses of normal and cancer stem cells to signaling molecules, provided or activated by their microenvironment. While TGF-β family signaling regulates stemness of normal and cancer stem cells, its effects are diverse and depend on the cell types and physiological state of the cells.

General significance

Further mechanistic studies will provide a better understanding of the roles of TGF-β family signaling in the regulation of stem cells. These basic studies may lead to the development of a new therapeutic or prognostic strategies for the treatment of cancers. This article is part of a Special Issue entitled Biochemistry of Stem Cells.  相似文献   

11.

Background

The discovery of markers to identify the intestinal stem cell population and the generation of powerful transgenic mouse models to study stem cell physiology have led to seminal discoveries in stem cell biology.

Scope of review

In this review we give an overview of the current knowledge in the field of intestinal stem cells (ISCs) highlighting the most recent progress on markers defining the ISC population and pathways governing intestinal stem cell maintenance and differentiation. Furthermore we review their interaction with other stem cell related pathways. Finally we give an overview of alteration of these pathways in human inflammatory gastrointestinal diseases.

Major conclusions

We highlight the complex network of interactions occurring among different pathways and put in perspective the many layers of regulation that occur in maintaining the intestinal homeostasis.

General significance

Understanding the involvement of ISCs in inflammatory diseases can potentially lead to new therapeutic approaches to treat inflammatory GI pathologies such as IBD and celiac disease and could reveal the molecular mechanisms leading to the pathogenesis of dysplasia and cancer in inflammatory chronic conditions. This article is part of a Special Issue entitled Biochemistry of Stem Cells.  相似文献   

12.
Currently, many gastrointestinal diseases are a major reason for the increased mortality rate of children and adults every year. Additionally, these patients may cope with the high cost of the parenteral nutrition (PN), which aids in the long-term survival of the patients. Other treatment options include surgical lengthening, which is not sufficient in many cases, and intestinal transplantation. However, intestinal transplantation is still accompanied by many challenges, including immune rejection and donor availability, which may limit the transplant’s success. The development of more safe and promising alternative treatments for intestinal diseases is still ongoing. Stem cell-based therapy (SCT) and tissue engineering (TE) appear to be the next promising choices for the regeneration of the damaged intestine. However, suitable stem cell source is required for the SCT and TE process. Thus, in this review we discuss how intestinal stem cells (ISCs) are a promising cell source for small intestine diseases. We will also discuss the different markers were used to identify ISCs. Moreover, we discuss the dominant Wnt signaling pathway in the ISC niche and its involvement in some intestinal diseases. Additionally, we discuss ISC culture and expansion, which are critical to providing enough cells for SCT and TE. Finally, we conclude and recommend that ISC isolation, culture and expansion should be considered when SCT is a treatment option for intestinal disorders. Therefore, we believe that ISCs should be considered a cell source for SCT for many gastrointestinal diseases and should be highlighted in future clinical applications.  相似文献   

13.
Intestinal stem cells (ISCs) play an important role in maintaining intestinal homeostasis via promoting a healthy gut barrier. Within the stem cell niche, gut microbiota linking the crosstalk of dietary influence and host response has been identified as a key regulator of ISCs. Emerging insights from recent research reveal that ISC and gut microbiota interplay regulates epithelial self-renewal. This article reviews the recent knowledge on the key role of ISC in their local environment (stem cell niche) associating with gut microbiota and their metabolites as well as the signaling pathways. The current progress of intestinal organoid culture is further summarized. Subsequently, the key challenges and future directions are discussed.  相似文献   

14.
目的探索骨髓间充质干细胞(MSCs)对放射性肠炎(RE)肠黏膜修复的途径。 方法体外分离、培养大鼠骨髓MSCs。将RE模型的大鼠采用随机数字表法分为治疗组[经尾静脉注射干细胞悬液1 mL(细胞浓度为1×106个/mL)]和对照组(注射等量生理盐水,每日1次,连续3 d),每组10只。每日观察两组大鼠的活动量、进食和进水量、体质量变化等。1周后处死大鼠获取小肠标本,HE染色观察肠黏膜的修复情况,电镜观察上皮细胞的超微结构,免疫组化染色观察小肠隐窝Bmi-1阳性干细胞增殖情况,采用Image-Pro Plus 6.0软件对Bmi-1阳性细胞数量进行分析。组间差异的比较采用t检验。 结果成功分离得到大鼠骨髓MSCs,流式细胞仪鉴定:CD29、CD90、CD34、CD45阳性细胞比例分别为98.6﹪、99.6﹪、0.56﹪、0.89﹪。与对照组相比,治疗组大鼠移植1周后,体质量增加(190.30 g ± 13.23 g比235.00 g±14.30 g);大鼠小肠黏膜上皮得到修复,绒毛高度增高(627.50 μm ± 40.55 μm比984.33 μm ± 61.80 μm);上皮细胞超微结构较完整、清晰,隐窝Bmi-1阳性干细胞增殖数量增多[(60.67±9.63)个/mm2比(87.33 ±5.47)个/mm2],差异具有统计学意义(P < 0.05)。 结论骨髓MSCs能促进RE模型的大鼠肠黏膜的修复,这一作用可能是通过促进小肠隐窝干细胞的增殖发挥。  相似文献   

15.
Calorie restriction has been recently shown to increase intestinal stem cell competition and to reduce mutation fixation in young mice. However, the impact of aging on this process is unknown. By employing Confetti reporter mice, here we show that, unexpectedly, old mice have more intestinal stem cell (ISC) competition than young mice. Moreover, differently from what observed in young mice, calorie restriction, when applied at late-life, decreases this process. Importantly, we also observed a strong correlation between the ISC competition and Paneth cell number. In vivo analysis and in vitro organoid experiments indicated that Paneth cells play a major role in driving intestinal stem cell competition and crypt clonality. Taken together, our results provide evidence that increasing the number of Paneth cells can increase the number of competitive ISCs, representing a valuable therapeutic target to delay fixation of mutated intestinal stem cells.  相似文献   

16.
17.
It is well known that exposure of double-stranded RNA (dsRNA) to intestine immediately induces villus damage with severe diarrhea, which is mediated by toll-like receptor 3 signaling activation. However, the role of intestinal stem cells (ISCs) remains obscure during the pathology. In the present study, polyinosinic-polycytidylic acid (poly[I:C]), mimicking viral dsRNA, was used to establish intestinal damage model. Mice were acutely and chronically exposed to poly(I:C), and ISCs in jejunum were analyzed. The results showed that the height of villus was shorter 48 hr after acute poly(I:C) exposure compared with that of controls, while chronic poly(I:C) treatment increased both villus height and crypt depth in jejunum compared with control animals. The numbers of ISCs in jejunum were significantly increased after acute and chronic poly(I:C) exposure. Poly (I:C)-stimulated ISCs have stronger capacities to differentiate into intestine endocrine cells. Mechanistically, poly(I:C) treatment increased expression of Stat1 and Axin2 in the intestinal crypt, which was along with increased expression of Myc, Bcl2, and ISC proliferation. These findings suggest that dsRNA exposure could induce ISC proliferation to ameliorate dsRNA-induced intestinal injury.  相似文献   

18.
A frequent complication in colorectal cancer (CRC) is regeneration of the tumor after therapy. Here, we report that a gene signature specific for adult intestinal stem cells (ISCs) predicts disease relapse in CRC patients. ISCs are marked by high expression of the EphB2 receptor, which becomes gradually silenced as cells differentiate. Using EphB2 and the ISC marker Lgr5, we have FACS-purified and profiled mouse ISCs, crypt proliferative progenitors, and late transient amplifying cells to define a gene program specific for normal ISCs. Furthermore, we discovered that ISC-specific genes identify a stem-like cell population positioned at the bottom of tumor structures reminiscent of crypts. EphB2 sorted ISC-like tumor cells display robust tumor-initiating capacity in immunodeficient mice as well as long-term self-renewal potential. Taken together, our data suggest that the ISC program defines a cancer stem cell niche within colorectal tumors and plays a central role in CRC relapse.  相似文献   

19.
New nonimmortalized fibroblast-like cell lines SC5-MSC and SC3a-MSC, FetMSC, FRSN were obtained from human embryonic stem cells (ESC), bone marrow of a 5-6-days embryo and foreskin of a 3-years-old boy, respectively. All the lines are successfully used as the feeder at human ESC cultivation. It is determined that the average cell population doublings time varies from 25.5 h for ISC5-MSC to 38.8 h for SC3a-MSC. Active proliferation of all the lines is also shown by the corresponding growth curves. Numerical and structural karyotypic analysis showed that these lines had normal karyotype: 46,XX (SC5-MSC and SC3a-MSC) and 46,XY (FetMSC and FRSN). To determine the status of the lines, their cell surface markers were analyzed by flow cytometry. This analysis revealed the presence of surface antigens CD44, CD73, CD90, CD105 and HLA-ABC, characteristic of human MSC, and the absence of CD34 and HLA-DR. Different lines were found to express CD117(c-kit) to a different level. Immunofluorescence and flow cytometry analysis did not detect TRA-1-60 and Oct-4, characteristic of human embryonic stem cells, and revealed interlinear variations in the level of SSEA, which did not depend on the cell origin. It is not clear yet whether these interlinear variations affect functional MSC status. In all the lines, immunofluorescence analysis showed the presence of the markers of early differentiation in the derivates of three germ layers which may allow MSC to be useful, in corresponding microenvironments, for reparation of tissue injures. Adipogenic and osteogenic differentiatiation of all cell lines has been shown.  相似文献   

20.
Adult stem cells are the most primitive cells of a lineage and are distinguished by the properties of self-renewal and multipotency. Coordinated control of stem cell proliferation and multilineage differentiation is essential to ensure a steady output of differentiated daughter cells necessary to maintain tissue homeostasis. However, little is known about the signals that coordinate stem cell proliferation and daughter cell differentiation. Here we investigate the role of the conserved JAK/STAT signaling pathway in the Drosophila intestinal stem cell (ISC) lineage. We show first, that JAK/STAT signaling is normally active in both ISCs and their newly formed daughters, but not in terminally differentiated enteroendocrine (ee) cells or enterocyte (EC) cells. Second, analysis of ISC lineages shows that JAK/STAT signaling is necessary but not sufficient for daughter cell differentiation, indicating that competence to undergo multilineage differentiation depends upon JAK/STAT. Finally, our analysis reveals JAK/STAT signaling to be a potent regulator of ISC proliferation, but not ISC self-renewal. On the basis of these findings, we suggest a model in which JAK/STAT signaling coordinates the processes of stem cell proliferation with the competence of daughter cells to undergo multilineage differentiation, ensuring a robust cellular output in the lineage.  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