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.
3.
During vessel sprouting, endothelial “tip” cells migrate at the forefront, while the endothelial “stalk” cells elongate the sprout; endothelial “phalanx” cells line quiescent vessels. Tip and stalk cells can dynamically switch phenotypes under the control of VEGF and Notch signaling. Novel findings now show that in addition to signaling cascades, metabolism coregulates the formation of the new vasculature. Recent studies demonstrated that ECs rely primarily on glycolysis for ATP production, that glycolysis is further enhanced in angiogenic ECs, and that the key glycolytic regulator PFKFB3 codetermines angiogenesis by controlling the balance of tip versus stalk cells and promoting a migratory tip cell phenotype. On the other hand, FAO regulates endothelial stalk cell proliferation by providing carbon sources for biosynthetic processes, more particularly for de novo nucleotide synthesis for DNA replication. Here, we overview the current understanding of the various metabolic pathways in ECs and their impact on vessel formation in health and disease.  相似文献   

4.
The functional shift of quiescent endothelial cells into tip cells that migrate and stalk cells that proliferate is a key event during sprouting angiogenesis. We previously showed that the sialomucin CD34 is expressed in a small subset of cultured endothelial cells and that these cells extend filopodia: a hallmark of tip cells in vivo. In the present study, we characterized endothelial cells expressing CD34 in endothelial monolayers in vitro. We found that CD34-positive human umbilical vein endothelial cells show low proliferation activity and increased mRNA expression of all known tip cell markers, as compared to CD34-negative cells. Genome-wide mRNA profiling analysis of CD34-positive endothelial cells demonstrated enrichment for biological functions related to angiogenesis and migration, whereas CD34-negative cells were enriched for functions related to proliferation. In addition, we found an increase or decrease of CD34-positive cells in vitro upon exposure to stimuli that enhance or limit the number of tip cells in vivo, respectively. Our findings suggest cells with virtually all known properties of tip cells are present in vascular endothelial cell cultures and that they can be isolated based on expression of CD34. This novel strategy may open alternative avenues for future studies of molecular processes and functions in tip cells in angiogenesis.  相似文献   

5.
OBJECTIVE: To develop an adenovirus-mediated angiogenesis model, dependent on VEGF, in a system amenable to functional characterization. METHODS: Adenovirus (AdV) expressing VEGF (Ad-VEGF) or GFP (Ad-EGFP) (1-3.3 x 10(8) TCID50/mL), and Monastral blue were injected into the fat pad on either side of a mesenteric connective tissue panel of halothane-anesthetized male Wistar rats after intravital microscopic imaging. The intestine was replaced in the animal, the laparotomy was sutured, and the animal was allowed to recover. Six days later, the same connective tissue panel was identified from the Monastral blue depot and the mesentery was imaged as before, and then excised, fixed, and stained for endothelial cells (GSL isolectin-1B4), proliferating cells, VEGF, VEGF-R2, and actin. The increase in fractional microvascular area (FVA) was measured, and proliferating cell density, sprout density, vessel branch point density, vessel density, and mean vessel length were determined. RESULTS: AdVEGF injection significantly increased the fractional vessel area (2.9 +/- 0.4-fold), proliferating endothelial cell density (1.7 +/- 0.1-fold), sprout density (3.1 +/- 0.3-fold), branch point density (1.9 +/- 0.1-fold), and the microvessel density (1.4 +/- 0.1-fold), and decreased the mean vessel length (0.69 +/- 0.04-fold). VEGF-R2 staining was evident near sprouting tips of endothelial cells, but not on the tip itself, and evidence for arteriogenesis was observed as well as clear evidence of angiogenesis. CONCLUSIONS: Ad-VEGF injection into the fat pad of the rat induced significant angiogenesis in the mesentery. This two-dimensional model of VEGF-induced angiogenesis is amenable to physiological, biochemical, and molecular assessment and may be a useful tool to help understand mechanisms of angiogenesis.  相似文献   

6.
Considered during the past decades as cell dust, microparticles are now deemed true biomarkers and vectors of biological information between cells. Depending on their origin, the composition of microparticles varies and the subsequent message transported by them, such as proteins, mRNA, or miRNA, can differ. Recent studies have described microparticles as "cargos" of deleterious information in blood vessel wall under pathological situations such as hypertension, myocardial infarction, and metabolic syndrome. In addition, it has been reported that depending on their origin, microparticles also possess a therapeutic potential regarding angiogenesis. Microparticles can act directly through the interaction ligand/receptor or indirectly on angiogenesis by modulating soluble factor production involved in endothelial cell differentiation, proliferation, migration, and adhesion; by reprogramming endothelial mature cells; and by inducing changes in levels, phenotype, and function of endothelial progenitor cells. This results in an increase in formation of in vitro capillary-like tubes and the generation of new vessels in vivo under ischemic conditions, for instance. Taking into consideration these properties of microparticles, recent evidence provides new basis to expand the possibility that microparticles might be used as therapeutic tools in pathologies associated with an alteration of angiogenesis.  相似文献   

7.
Angiogenesis, the formation of new blood vessels, is regulated by vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs). VEGFR2 is abundant in the tip cells of angiogenic sprouts, where VEGF/VEGFR2 functions upstream of the delta-like ligand 4 (DLL4)/Notch signal transduction pathway. VEGFR3 is expressed in all endothelia and is indispensable for angiogenesis during early embryonic development. In adults, VEGFR3 is expressed in angiogenic blood vessels and some fenestrated endothelia. VEGFR3 is abundant in endothelial tip cells, where it activates Notch signaling, facilitating the conversion of tip cells to stalk cells during the stabilization of vascular branches. Subsequently, Notch activation suppresses VEGFR3 expression in a negative feedback loop. Here we used conditional deletions and a Notch pathway inhibitor to investigate the cross-talk between VEGFR2, VEGFR3, and Notch in vivo. We show that postnatal angiogenesis requires VEGFR2 signaling also in the absence of Notch or VEGFR3, and that even small amounts of VEGFR2 are able to sustain angiogenesis to some extent. We found that VEGFR2 is required independently of VEGFR3 for endothelial DLL4 up-regulation and angiogenic sprouting, and for VEGFR3 functions in angiogenesis. In contrast, VEGFR2 deletion had no effect, whereas VEGFR3 was essential for postnatal lymphangiogenesis, and even for lymphatic vessel maintenance in adult skin. Knowledge of these interactions and the signaling functions of VEGFRs in blood vessels and lymphatic vessels is essential for the therapeutic manipulation of the vascular system, especially when considering multitargeted antiangiogenic treatments.The VEGF/VEGFR and Notch signal transduction pathways are key elements of blood vessel formation in physiological and pathological conditions. VEGFR2 is expressed in blood vascular endothelial cells, where it stimulates endothelial proliferation, migration, survival, and vascular permeability (1). Vegfr2 gene-targeted mice die at embryonic day (E) 8.5 because of lack of vasculogenesis (2) and inhibitors blocking the VEGF/VEGFR2 pathway are used to inhibit pathological angiogenesis in patients with cancer or age-related macular degeneration (3). VEGFR3 is also required for developmental angiogenesis and Vegfr3−/− mice die at E9.5 because of cardiovascular remodeling defects (4). When development progresses, VEGFR3 becomes restricted to lymphatic endothelial cells (5). In adults, VEGFR3 expression is very low or absent in most blood vessels, but detectable in e.g., high endothelial venules and fenestrated capillaries (6). VEGFR3 expression is up-regulated in angiogenic endothelial cells, for example in the tumor vasculature, and it is often highly expressed in endothelial tip cells (710).Several studies have highlighted the importance of Notch signaling in arteriovenous differentiation and tip cell selection during angiogenesis in mice and zebrafish (11), and VEGFR and Notch signals are tightly coordinated during angiogenic sprouting. VEGFR2, which is expressed in endothelial tip cells, activates Notch in adjacent endothelial cells via VEGF-induced up-regulation of delta-like ligand 4 (DLL4), but it is not clear to what extent VEGFR2 expression is suppressed by the DLL4/Notch signals from neighboring cells (1215). VEGFC/VEGFR3 signaling activates Notch in blood vascular endothelial cells, facilitating the conversion of tip cells to stalk cells during the stabilization of vascular loops (16). Notch, in turn, regulates VEGFR3 expression in zebrafish and in postnatal mice (7, 8, 17).An important question regarding VEGFR signaling is the cross-talk between VEGFR2 and VEGFR3 during vessel morphogenesis. VEGFR2/VEGFR3 heterodimers in endothelial cells are important for angiogenic sprouting in mice and arteriogenesis in zebrafish (18, 19). VEGFR2 activation induces VEGFR3 expression in blood vessels and silencing of the VEGFR3 gene prolongs VEGFR2 phosphorylation in human blood vascular endothelial cells in culture (7, 16). Previous studies used VEGFR2 deletion in combination with poorly characterized tyrosine kinase inhibitors to probe the cross-talk of the receptors (20). However, the functions and interactions of the VEGFRs have not been validated by simultaneous genetic deletions in mice so far. To circumvent the embryonic lethality in Vegfr2−/− and Vegfr3−/− mice, we deleted VEGFR2 and VEGFR3 conditionally in newborn pups to analyze postnatal sprouting angiogenesis.  相似文献   

8.
Vascular development and angiogenesis initially depend on endothelial tip cell invasion, which is followed by a series of maturation steps, including lumen formation and recruitment of perivascular cells. Notch ligands expressed on the endothelium and their cognate receptors expressed on perivascular cells are involved in blood vessel maturation, though little is known regarding the Notch-dependent effectors that facilitate perivascular coverage of nascent vessels. Here, we report that vascular smooth muscle cell (VSMC) recognition of the Notch ligand Jagged1 on endothelial cells leads to expression of integrin αvβ3 on VSMCs. Once expressed, integrin αvβ3 facilitates VSMC adhesion to VWF in the endothelial basement membrane of developing retinal arteries, leading to vessel maturation. Genetic or pharmacologic disruption of Jagged1, Notch, αvβ3, or VWF suppresses VSMC coverage of nascent vessels and arterial maturation during vascular development. Therefore, we define a Notch-mediated interaction between the developing endothelium and VSMCs leading to adhesion of VSMCs to the endothelial basement membrane and arterial maturation.  相似文献   

9.
The recruitment of mural cells such as pericytes to patent vessels with an endothelial lumen is a key factor for the maturation of blood vessels and the prevention of hemorrhage in pathological angiogenesis. To date, our understanding of the specific trigger underlying the transition from cell growth to the maturation phase remains incomplete. Since rapid endothelial cell growth causes pericyte loss, we hypothesized that suppression of endothelial growth factors would both promote pericyte recruitment, in addition to inhibiting pathological angiogenesis. Here, we demonstrate that targeted knockdown of apelin in endothelial cells using siRNA induced the expression of monocyte chemoattractant protein-1 (MCP-1) through activation of Smad3, via suppression of the PI3K/Akt pathway. The conditioned medium of endothelial cells treated with apelin siRNA enhanced the migration of vascular smooth muscle cells, through MCP-1 and its receptor pathway. Moreover, in vivo delivery of siRNA targeting apelin, which causes exuberant endothelial cell proliferation and pathological angiogenesis through its receptor APJ, led to increased pericyte coverage and suppressed pathological angiogenesis in an oxygen-induced retinopathy model. These data demonstrate that apelin is not only a potent endothelial growth factor, but also restricts pericyte recruitment, establishing a new connection between endothelial cell proliferation signaling and a trigger of mural recruitment.  相似文献   

10.
Delta-like 4 (Dll4) is a transmembrane ligand for Notch receptors that is expressed in arterial blood vessels and sprouting endothelial cells. Here we show that Dll4 regulates vessel branching during development by inhibiting endothelial tip cell formation. Heterozygous deletion of dll4 or pharmacological inhibition of Notch signaling using gamma-secretase inhibitor revealed a striking vascular phenotype, with greatly increased numbers of filopodia-extending endothelial tip cells and increased expression of tip cell marker genes compared with controls. Filopodia extension in dll4(+/-) retinal vessels required the vascular growth factor VEGF and was inhibited when VEGF signaling was blocked. Although VEGF expression was not significantly altered in dll4(+/-) retinas, dll4(+/-) vessels showed increased expression of VEGF receptor 2 and decreased expression of VEGF receptor 1 compared with wild-type, suggesting they could be more responsive to VEGF stimulation. In addition, expression of dll4 in wild-type tip cells was itself decreased when VEGF signaling was blocked, indicating that dll4 may act downstream of VEGF as a "brake" on VEGF-mediated angiogenic sprouting. Taken together, these data reveal Dll4 as a negative regulator of vascular sprouting and vessel branching that is required for normal vascular network formation during development.  相似文献   

11.
Development of the retinal vasculature   总被引:1,自引:0,他引:1  
Blood vessels that supply the inner portion of the retina are extensively reorganized during development. The vessel regression, sprouting angiogenesis, vascular remodelling and vessel differentiation events involved critically depend on cell–cell signalling between different cellular components such as neurons, glia, endothelial cells, pericytes and immune cells. Studies in mice using transgenic and gene deletion approaches have started to unravel the genetic basis of some of these signalling pathways and have lead to a much improved understanding of the molecular mechanisms controlling retinal blood vessel behaviour both during development and under pathological conditions. Such insight will provide the basis of future therapeutic approaches aimed at manipulating retinal blood vessels.  相似文献   

12.
Angiogenesis entails new vessel formation from preexisting vessels. It follows vasculogenesis during embryo development. In post-natal life, it occurs both in physiological conditions (wound repair and cyclically in the female genital system) and pathological conditions such as tumors. Several sequential steps are involved, including basement membrane degradation by proteolytic enzymes secreted by the endothelial cells, chemotaxis toward the stimulus and proliferation of these cells, canalization, branching and formation of vascular loops, stabilization and functional maturation of neovessels following perivascular apposition of pericytes and smooth muscle cells, and neosynthesis of basement membrane constituents. Tumor angiogenesis is regulated by several factors, mainly growth factors for the endothelial cells secreted by both the tumor and host inflammatory cells, and mobilized from extracellular matrix stores by proteases secreted by tumor cells. Regulatory factors also include the extracellular matrix components and endothelial cell integrins, hypoxia, oncogenes and tumor suppressor genes. Angiogenesis is mandatory to the process of tumor progression (growth, invasion and metastasis), since it conveys oxygen and metabolites, whereas endothelial cells secrete growth factors for tumor cells and a variety of proteinases which facilitate invasion and increase opportunities for tumor cells to enter the circulation. We present our results concerning the relationship between angiogenesis and progression in patients with melanoma, multiple myeloma, B-cell non-Hodgkin's lymphomas and mycosis fungoides. Lastly, it is becoming increasingly evident that agents interfering with blood vessel formation also interfere with tumor progression. These include antagonists of angiogenic growth factors, angiogenic receptors, endothelial cell integrins, and proteolytic enzymes, as well as non-specific toxic agents for vessels and low-dose chemotherapeutic agents. Their recent applications in preclinical models and in neoplastic patients are reviewed.  相似文献   

13.
Fluid forces control endothelial sprouting   总被引:1,自引:0,他引:1  
During angiogenesis, endothelial cells (ECs) from intact blood vessels quickly infiltrate avascular regions via vascular sprouting. This process is fundamental to many normal and pathological processes such as wound healing and tumor growth, but its initiation and control are poorly understood. Vascular endothelial cell growth factor (VEGF) can promote vessel dilation and angiogenic sprouting, but given the complex nature of vascular morphogenesis, additional signals are likely necessary to determine, for example, which vessel segments sprout, which dilate, and which remain quiescent. Fluid forces exerted by blood and plasma are prime candidates that might codirect these processes, but it is not known whether VEGF cooperates with mechanical fluid forces to mediate angiogenesis. Using a microfluidic tissue analog of angiogenic sprouting, we found that fluid shear stress, such as exerted by flowing blood, attenuates EC sprouting in a nitric oxide-dependent manner and that interstitial flow, such as produced by extravasating plasma, directs endothelial morphogenesis and sprout formation. Furthermore, positive VEGF gradients initiated sprouting but negative gradients inhibited sprouting, promoting instead sheet-like migration analogous to vessel dilation. These results suggest that ECs integrate signals from fluid forces and local VEGF gradients to achieve such varied goals as vessel dilation and sprouting.  相似文献   

14.
Contribution from diverse tissue-specific stem cell types is required to create the cell populations necessary for the activation of angiogenesis and neovascular growth in cancer. Bone marrow (BM)-derived circulating endothelial progenitors (EPCs) that would differentiate to bona fide endothelial cells (ECs) were previously believed to be necessary for tumor angiogenesis. However, numerous recent studies demonstrate that EPCs are not needed for tumor angiogenesis and indicate EPCs to be artifactual rather than physiological. It is evident that tumor infiltrating hematopoietic cells produced by BM-residing hematopoietic stem cells (HSCs) may contribute to tumor angiogenesis in a paracrine manner by stimulating ECs or by remodeling the extracellular matrix. Therefore, identification of the various hematopoietic cell subpopulations that are critical for tumor angiogenesis and better understanding of their proangiogenic functions and mechanisms of action have potential therapeutic significance. Stem and progenitor cell subsets for also other vascular or perivascular cell types such as pericytes or mesenchymal/stromal cells may provide critical contributions to the growing neovasculature. Furthermore, we hypothesize that the existence of a yet undiscovered—and largely unsearched—tissue-specific adult vascular endothelial stem cell (VESC) would provide completely novel targeted approaches to block pathological angiogenesis and cancer growth. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".  相似文献   

15.
It has been more than 20 years since it was first demonstrated that endothelial cells will rapidly form capillary-like structures in vitro when plated on top of a reconstituted basement membrane extracellular matrix (BME, Matrigel, EHS matrix, etc.). Subsequently, this morphological differentiation has been demonstrated with a variety of endothelial cells; with endothelial progenitor cells; and with transformed/immortalized endothelial cells. The differentiation process involves several steps in blood vessel formation, including cell adhesion, migration, alignment, protease secretion, and tubule formation. Because the formation of vessel structures is rapid and quantifiable, endothelial cell differentiation on basement membrane has found numerous applications in assays. Such differentiation has been used (1) to study angiogenic and antiangiogenic factors, (2) to define mechanisms and pathways involved in angiogenesis, and (3) to define endothelial cell populations. Further, the endothelial cell differentiation assay has been successfully used to study processes ranging from wound repair and reproduction to development and tumor growth. The assay is easy to perform and is the most widely used in vitro angiogenesis assay.  相似文献   

16.
Tip cells, the leading cells of angiogenic sprouts, were identified in cultures of human umbilical vein endothelial cells (HUVECs) by using CD34 as a marker. Here, we show that tip cells are also present in primary human microvascular endothelial cells (hMVECs), a more relevant endothelial cell type for angiogenesis. By means of flow cytometry, immunocytochemistry, and qPCR, it is shown that endothelial cell cultures contain a dynamic population of CD34+ cells with many hallmarks of tip cells, including filopodia-like extensions, elevated mRNA levels of known tip cell genes, and responsiveness to stimulation with VEGF and inhibition by DLL4. Furthermore, we demonstrate that our in vitro tip cell model can be exploited to investigate cellular and molecular mechanisms in tip cells and to discover novel targets for anti-angiogenesis therapy in patients. Small interfering RNA (siRNA) was used to knockdown gene expression of the known tip cell genes angiopoietin 2 (ANGPT2) and tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1), which resulted in similar effects on tip cells and sprouting as compared to inhibition of tip cells in vivo. Finally, we identified two novel tip cell-specific genes in CD34+ tip cells in vitro: insulin-like growth factor 2 (IGF2) and IGF-1-receptor (IGF1R). Knockdown of these genes resulted in a significant decrease in the fraction of tip cells and in the extent of sprouting in vitro and in vivo. In conclusion, this study shows that by using our in vitro tip cell model, two novel essential tip cells genes are identified.  相似文献   

17.
Angiogenesis factors   总被引:1,自引:0,他引:1  
Angiogenesis is a recent highlight in the medical field; the developmental process and pathological conditions for various diseases can be understood from the novel aspect of "angiogenesis". Many angiogenesis-related factors are involved in the development of vessels during embryogenesis (vasculogenesis), as well as the induction of new vessels in response to physiological or pathological stimuli. In particular, the appearance of hemangioblasts, precursor cells for vascular endothelial cells and blood cells, and blood islands are expected to play a "prelude" role in tubulogenesis. Gene knock out mice of vascular endothelial growth factor (VEGF)/VEGF receptor, ephrin-B2, and angiopoietin-1 results in a failure of normal vessels production. Dormant factors derived from proteolytic cleavage of various physiological substrates are expected to balance a homeostasis of "angiogenic states" in the host. Furthermore, angiogenesis under various pathological conditions of malignant tumors, ocular diseases, psoriasis, rheumatoid arthritis, atherosclerosis and other diseases is associated with complex angiogenesis networks, suggesting pleiotropic mechanisms for angiogenesis.  相似文献   

18.
血管内皮生长因子与心肌缺血   总被引:1,自引:0,他引:1  
血管内皮生长因子(VEGF)是内皮细胞特异性的促有丝分裂原,属分泌性的糖蛋白。有促进内皮细胞增生,增强血管通透性;加速新血管形成的作用。VEGF通过与其特异性受体结合发挥生物学作用。VEGF在生理和病理状态下新生血管形成中起重要作用。VEGF及其受体在缺血和/或缺 氧的 心肌细胞中表达升高。VEGF蛋白应用于心肌缺血可以通过促进缺血区新生血管形成,提高侧枝血流量而改善心功能。VEGF基因治疗心肌缺血具有广阔应用前景。  相似文献   

19.
Retinal angiogenesis in diabetes may lead to visual impairment and even irreversible blindness in people of working age group worldwide. The main pathological feature of proliferative diabetic retinopathy (PDR) is hypoxia, and overproduction of growth factors like vascular endothelial growth factor (VEGF) and erythropoietin (Epo). This results in pathological proliferation of retinal endothelial cells (RECs), leading to new vessel formation (angiogenesis). Inhibition of angiogenesis is a promising strategy for treatment of PDR and other retinal neovascular disorders. Pigment epithelium-derived factor (PEDF), a 50-kDa protein secreted by retinal pigment epithelium, inhibits the growth of new blood vessel induced in the eye in a variety of ways with a yet elusive mechanism. Here, we investigated the possible mechanism by which PEDF inhibits VEGF- and Epo-induced angiogenic effects in RECs is mediated through PI3K/Akt pathway. PEDF treatment induced the apoptosis in RECs by activating caspase-3 and DNA fragmentation. We found a dose-dependent increase in cell survival with VEGF or Epo, which was attenuated in the presence of PEDF. In addition, PEDF significantly (P < 0.05) inhibited migration and in vitro tube formation in RECs in the presence of VEGF as like PI3K/Akt inhibitor. Of interest, PEDF effectively abrogated VEGF-mediated phosphorylation of PI3K/Akt. Further studies using RECs transfected with constitutively active and dominant-negative forms of Akt suggest that PEDF could inhibit VEGF- and also Epo-induced angiogenesis by disruption of PI3K/Akt signaling.  相似文献   

20.
OBJECTIVE: To examine the roles of apoptosis, macrophages, and endothelial cell migration in vascular remodeling during development of the central nervous system. METHODS: The terminal deoxynucleotide transferase-mediated dUTP nick end labeling (TUNEL) technique was combined with Griffonia simplicifolia isolectin B4 histochemistry to detect apoptotic endothelial cells in retinal whole-mount preparations derived from rats at various stages of postnatal development as well as from rat pups exposed to hyperoxia. Macrophages were detected by immunohistochemistry with the monoclonal antibody ED1, and proliferating endothelial cells were identified by incorporation of bromodeoxyuridine. RESULTS: Only small numbers of TUNEL-positive endothelial cells were detected during normal development of the retinal vasculature, with the apoptotic cell density in the inner plexus peaking during the first postnatal week and decreasing markedly during the second week, at a time when vessel retraction was widespread. Neither apoptotic endothelial cells nor macrophages were apparent at sites of initiation of vessel retraction. TUNEL-positive endothelial cells were observed in vessels destined to remain. Hyperoxia induced excessive vessel withdrawal, resulting in the generation of isolated vascular segments containing apoptotic endothelial cells. A topographical analysis showed low numbers of proliferating endothelial cells at sites of angiogenesis whereas vascular proliferation was increased in the adjacent inner plexus. CONCLUSIONS: Endothelial cell apoptosis and macrophages do not initiate vessel retraction, but rather contribute to the removal of redundant cells throughout the vasculature. We suggest that vessel retraction is mediated by endothelial cell migration and that endothelial cells derived from retracting vascular segments are redeployed in the formation of new vessels. Only when retraction results in compromised circulation and redeployment is not possible, does endothelial cell apoptosis occur.  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