首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
M Saari 《Acta anatomica》1975,91(3):376-379
As remnants of pupillary membrane, some albino rat eyes revealed vascular loops starting from arteriovenous bridges near the pupillary margin and crossing the pupil. These vascular loops bend in miosis and straighten in mydriasis, which prevents them from being broken during pupillary movement. The vessel wall reveals endothelial cells and pericytes. The lumen contains red blood cells, which means that they are functional vessels. They may function as vascular shunts between opposite sides of the albino rat iris.  相似文献   

2.
小鼠胚胎干细胞分化为血管内皮细胞的永生化研究   总被引:9,自引:0,他引:9  
本文探讨了小鼠胚胎干细胞(ES细胞)、诱导分化的血管内皮细胞永生化。在体外培养系统中,以维甲酸(RA)和转化生长因子-β1(TGF-β1)诱导小鼠胚胎干细胞(ES细胞)的拟胚体(EB)分化为“圆形细胞”和由这些“圆形细胞”组成的血管样结构。经光学和扫描电镜及免疫荧光等法分析检测,证明组成血管样结构的细胞具有专一性vWF荧光染色,表明是血管内皮样细胞。利用脂质体将人端粒酶催化亚基逆转录酶(hTERT)基因转染诱导分化中的“圆形细胞”。应用Dot-blot,RT-PCR,Western blot及免疫组织化学等方法分析、观察和证明了诱导分化的组成血管样结构的园形细胞和被hTERT基因转染的“圆形”细胞的形态和生物学特性。结果表明,携带hTERT基因的从ES细胞分化来的圆形细胞在体外可大量增殖,持续传代,95%具有血管内皮细胞的一些特有标志和管道化生长特征。因此,通过人端粒酶基因的转染途径可解决由ES细胞诱导分化而来的内皮细胞扩增和永生化问题,为构建组织工程化血管及其它人工血管的内皮化提供种子细胞来源打下基础。  相似文献   

3.
The infiltration of leukocytes into the central nervous system (CNS) is an essential step in the neuropathogenesis of multiple sclerosis (MS). Leukocyte extravasation from the bloodstream is a multistep process that depends on several factors including fluid dynamics within the vasculature and molecular interactions between circulating leukocytes and the vascular endothelium. An important step in this cascade is the presence of chemokines on the vascular endothelial cell surface. Chemokines displayed along the endothelial lumen bind chemokine receptors on circulating leukocytes, initiating intracellular signaling that culminates in integrin activation, leukocyte arrest, and extravasation. The presence of chemokines at the endothelial lumen can help guide the movement of leukocytes through peripheral tissues during normal immune surveillance, host defense or inflammation. The expression and display of homeostatic or inflammatory chemokines therefore critically determine which leukocyte subsets extravasate and enter the peripheral tissues. Within the CNS, however, infiltrating leukocytes that cross the endothelium face additional boundaries to parenchymal entry, including the abluminal presence of localizing cues that prevent egress from perivascular spaces. This review focuses on the differential display of chemokines along endothelial surfaces and how they impact leukocyte extravasation into parenchymal tissues, especially within the CNS. In particular, the display of chemokines by endothelial cells of the blood brain barrier may be altered during CNS autoimmune disease, promoting leukocyte entry into this immunologically distinct site. Recent advances in microscopic techniques, including two-photon and intravital imaging have provided new insights into the mechanisms of chemokine-mediated capture of leukocytes within the CNS.  相似文献   

4.
The surface ultrastructure of Kupffer cells in the rat liver has been studied by scanning electron microscopy (SEM). The results demonstrate that Kupffer cells are both significantly different and clearly distinct from endothelial cells. Kupffer cells have neither pores (and/or "sieve plates") nor fenestrations, all of which are present in endothelial cells. They possess a stellate shape, and only indirectly, with slender and irregular evaginations, contribute to the lining of the sinusoidal wall. Furthermore, the luminal surface in some areas contains a large population of short microvilli, microphicae and invaginations. These elements form a kind of microlabyrinth which may correpond to the "worm-like" structures described by transmission electron microscopy (TEM). In the present study, transition forms between endothelial and Kupffer cells were never found. On the contrary, considering the highly fenestrated nature of the endothelial cells, the Kupffer cells may, by ameboid movements, easily cross the overlapping barrier of the sinusoid and protrude into the lumen. Thus, acting as activated macrophages, the Kupffer cells might function to prevent the entrance of foreign material into the tissues of the liver through the fragile and highly fenestrated endothelium. Finally, the topographical reconstruction of the sinusoid by correlated SEM and TEM studies demonstrates the Kupffer cells, with their protruding cytoplasm and ability to extend into the lumen of the sinusoid, may actually change the caliber of the vessel, and thus function as a "sphincter" which causes a temporary arrest of the blood flow when the diameter of the sinusoidal lumen is reduced.  相似文献   

5.
Capillaries derived from the perineural vascular plexus invade brain tissue early in embryonic development. Considerably later they differentiate into blood-brain barrier (BBB)-forming blood vessels. In the chick, the BBB as defined by impermeability for the protein horseradish peroxidase develops around embryonic day 13. We have previously found that brain endothelial cells start to express a number of proteins at around the same time, suggesting that these proteins play a role in BBB function. Here we describe a 74 kd protein defined by the monoclonal antibody HT7 that is expressed on the surface of chick embryonic blood cells and brain endothelial but on no other endothelial cells. This protein is not detectable on early embryonic brain endothelium, but is expressed by these cells on embryonic day 10. It is absent in choroid plexus endothelial cells which represent permeable fenestrated endothelial cells. The antigen is expressed on choroid plexus epithelium which is the site of the blood-cerebrospinal fluid barrier. Since it is also found in basolateral membranes of kidney tubules, it may be involved in specific carrier mechanisms. Embryonic mouse brain tissue transplanted on the chick chorio-allantoic membrane induces the expression of this antigen on endothelial cells derived from the chorio-allantois. Brain tissue can therefore induce in endothelial cells in vivo the expression of a molecule characteristic of brain endothelium.  相似文献   

6.
The regulation of vascular tone, vascular permeability, and thromboresistance is essential to maintain blood circulation and therefore tissue environments under physiological conditions. Atherogenic stimuli, including diabetes, dyslipidemia, and oxidative stress, induce vascular dysfunction, leading to atherosclerosis, which is a key pathological basis for cardiovascular diseases such as ischemic heart disease and stroke. We have proposed a novel concept termed "vascular failure" to comprehensively recognize the vascular dysfunction that contributes to the development of cardiovascular diseases. Vascular endothelial cells form the vascular endothelium as a monolayer that covers the vascular lumen and serves as an interface between circulating blood and immune cells. Endothelial cells regulate vascular function in collaboration with smooth muscle cells. Endothelial dysfunction under pathophysiological conditions contributes to the development of vascular dysfunction. Here, we address the barrier function and microtubule function of endothelial cells. Endothelial barrier function, mediated by cell-to-cell junctions between endothelial cells, is regulated by small GTPases and kinases. Microtubule function, regulated by the acetylation of tubulin, a component of the microtubules, is a target of atherogenic stimuli. The elucidation of the molecular mechanisms of endothelial dysfunction as a cellular mechanism for vascular failure could provide novel therapeutic targets of cardiovascular diseases.  相似文献   

7.
8.
Summary Developing blood vessels in rat cerebral cortex were studied at a number of stages between 3 and 28 days postnatal, in an attempt to obtain data on the mechanisms by which the lumen is established within cords of mesodermal cells. A combination of techniques was utilized in an attempt to elucidate these mechanisms. These were: (a) aldehyde fixation and block staining with phosphotungstic acid; (b) aldehyde perfusion followed by perfusion of a lead solution and post-fixation in osmium tetroxide; (c) conventional preparation of tissue with aldehyde and osmium fixation.Support for interendothelial lumen formation was readily forthcoming, including vessels with junctions between two or more endothelial cells cut transversely. There was some support for intraendothelial lumen formation, in the form of seamless endothelial cells. Other features noted included the presence of free ribosomes and vacuoles in the endothelial cells, endothelial flaps, sprouts and tendrils, intraluminal debris, endothelial degeneration and a junction with a nonendothelial cell.Large numbers of endothelial vacuoles were noted, many of them occurring at the abluminal edge of the cells. These vacuoles may be involved in the formation of intraendothelial lumina and also in the enlargement of both types of lumina. This study provides evidence that besides the well-established inter-endothelial lumen formation, intraendothelial mechanisms may also be operative in rat cerebral cortex. The techniques employed in this study offer the potential for clarifying these and related issues.We would like to acknowledge the financial assistance of the Nuffield Foundation  相似文献   

9.
There is controversy concerning whether intravascular activation of neutrophils during acute inflammation injures contiguous endothelial cells in vivo. Several physiologic defense mechanisms tend to limit such injury. In this paper we have examined evidence for one of these putative protective mechanisms: endothelial cell modulation of the activation responses of neutrophils during adherence and diapedesis. In vitro, endothelial cells co-incubated with neutrophils inhibit the release of superoxide anion when stimulated by receptor-mediated activators. The possible mechanisms include contact-linked down-regulation of neutrophil activation, the release from endothelial cells of soluble mediators which attenuate neutrophil activation responses, and the presence of free radical scavengers in endothelial cells which are active at the interface between endothelial cells and adherent neutrophils. There may be a broad spectrum of mechanisms by which intercellular interactions protect the lining cells of the vascular lumen from 'inadvertent' destruction by phagocytes which become activated while in an intravascular location.  相似文献   

10.
Neural stem cell proliferation and differentiation are regulated by external cues from their microenvironment. As endothelial cells are closely associated with neural stem cell in brain germinal zones, we investigated whether endothelial cells may interfere with neurogenesis. Neural precursor cells (NPC) from telencephalon of EGFP mouse embryos were cocultured in direct contact with endothelial cells. Endothelial cells did not modify the overall proliferation and apoptosis of neural cells, albeit they transiently delayed spontaneous apoptosis. These effects appeared to be specific to endothelial cells since a decrease in proliferation and a raise in apoptosis were observed in cocultures with fibroblasts. Endothelial cells stimulated the differentiation of NPC into astrocytes and into neurons, whereas they reduced differentiation into oligodendrocytes in comparison to adherent cultures on polyornithine. Determination of NPC clonogenicity and quantification of LeX expression, a marker for NPC, showed that endothelial cells decreased the number of cycling NPC. On the other hand, the presence of endothelial cells increased the number of neural cells having "side population" phenotype, another marker reported on NPC, which we have shown to contain quiescent cells. Thus, we show that endothelial cells may regulate neurogenesis by acting at different level of NPC differentiation, proliferation and quiescence.  相似文献   

11.
内皮祖细胞(EPCs)研究进展   总被引:2,自引:0,他引:2  
组织工程血管以及组织工程化组织的血管化因目前内皮种子细胞扩增能力和生物活力的不足而受到限制。EPCs(内皮祖细胞)是内皮细胞的前体细胞。在胚胎期,内皮细胞系与造血细胞系来源于血岛内共同的祖先细胞;出生后,EPCs存在于骨髓,并可被转移至外周血,参与缺血组织的血管重建和血管的内膜化。因此EPCs有望成为今后组织工程内皮种子细胞的重要来源。  相似文献   

12.
The formation of intersegmental blood vessels (ISVs) in the zebrafish embryo serves as a paradigm to study angiogenesis in vivo. ISV formation is thought to occur in discrete steps. First, endothelial cells of the dorsal aorta migrate out and align along the dorsoventral axis. The dorsal-most cell, also called tip cell, then joins with its anterior and posterior neighbours, thus establishing a simple vascular network. The vascular lumen is then established via formation of vacuoles, which eventually fuse with those of adjacent endothelial cells to generate a seamless tube with an intracellular lumen. To investigate the cellular architecture and the development of ISVs in detail, we have analysed the arrangement of endothelial cell junctions and have performed single cell live imaging. In contrast to previous reports, we find that endothelial cells are not arranged in a linear head-to-tail configuration but overlap extensively and form a multicellular tube, which contains an extracellular lumen. Our studies demonstrate that a number of cellular behaviours, such as cell divisions, cell rearrangements and dynamic alterations in cell-cell contacts, have to be considered when studying the morphological and molecular processes involved in ISV and endothelial lumen formation in vivo.  相似文献   

13.
Clathrin-independent endocytosis (CIE) is a form of bulk plasma membrane (PM) endocytosis that allows cells to sample and evaluate PM composition. Once in endosomes, the internalized proteins and lipids can be recycled back to the PM or delivered to lysosomes for degradation. Endosomes arising from CIE contain lipid and signaling molecules suggesting that they might be involved in important biological processes. During vasculogenesis, new blood vessels are formed from precursor cells in a process involving internalization and accumulation of endocytic vesicles. Here, we found that CIE has a role in endothelial lumen formation. Specifically, we found that human vascular endothelial cells (HUVECs) utilize CIE for internalization of distinct cargo molecules and that in three-dimensional cultures CIE membranes are delivered to the newly formed lumen.  相似文献   

14.
Vessel walls are comprised of several different cell populations residing in and on complex extracellular matrices. Each of the vascular cell types has diverse and sometimes unique functions and morphologies, and each has roles in repair processes following injury. Large vessel endothelial cells are known to respond to denudation injury by sheet migration and proliferation. This is in contrast to the migration through soft tissues with tube formation and subsequent lumen formation exhibited by microvascular endothelial cells in response to injury. Vascular smooth muscle cells of larger vessels respond to injury by migration from the arterial media into the intima, proliferation, and matrix biosynthesis, ultimately causing intimal thickening. Both these cell types exhibit "dysfunctional" phenotypes during their responses to injury. Microvascular cell responses to injury, while extremely variable, are less well documented. Specifically, responses to injury by microvascular endothelial vascular cells appear to be modulated, in part, by the composition and organization of the surrounding matrix as well as by the various soluble factors and cytokines found at sites of injury, suggesting that the extracellular matrix and soluble factors modulate each other's effects on local vascular cell populations following injury.  相似文献   

15.
The blood-brain barrier in a reptile, Anolis carolinensis   总被引:1,自引:0,他引:1  
An electron microscopic study was made of the ultrastructure and permeability of the capillaries in the cerebral hemispheres of the lizard, Anolis carolinensis. The brain of Anolis is vascularized by a loop-type pattern consisting exclusively of arteriovenous capillary loops. The ultrastructure of the endothelium and the arrangement of the various layers from the capillary lumen to the central nervous tissue is similar to that of mammals. The endothelial cells form a continuous layer around the lumen and are joined by tight interendothelial junctions. The basal lamina of the endothelium is also continuous and encloses pericyte processes. The cells of the nervous tissue rest directly on the basal lamina of the capillary and are separated from each other by a 200 Å space. Intravenously injected horseradish peroxidase (MW 40,000) and ferritin (MW 500,000) were used to study the permeability of the capillaries. The entry of horseradish peroxidase and ferritin into the intercellular spaces of the brain is restricted by the tightness of the interendothelial junctions. No vesicular transport of either tracer occurs; however, ferritin does enter the endothelial cells in vacuoles. No tracer molecules are present in the basal lamina, pericytes, or nervous tissue. The different responses of the endothelial cell to the tracers used in this study suggest that endocytotic activities of endothelial cells involve different processes. Vacuoles formed by marginal folds, vacuoles formed by endothelial surface projections or deep invaginations of the plasma membrane, 600–800 Å vesicles, and coated vesicles all seem to differ in the nature of the substances which they endocytose.  相似文献   

16.
Duffy SL  Murphy JT 《BioTechniques》2001,31(3):495-6, 498, 500-1
Endothelial "capillary leak", the loss of vascular integrity in response to noxious stimuli, is characterized by extravasation of protein-richfluidfrom capillary lumen into surrounding tissue interstitium. This increase in vascular permeability, in response to inflammatory mediators, correlates with endothelial cell contraction and the formation of intercellular gaps within the monolayer. However, in vivo assessment of paracellular solute flow between endothelial cells may be complicated by multiple uncontrolled parameters. In vitro examinations of endothelial barrier leak have relied on electrical impedence or macromolecule diffusion techniques to determine the details pertinent to capillary barrier function. In this report, a simple, sensitive, nonradioactive, colorimetric assay to quantify the leak of a labeled protein marker across endothelial monolayers is described. This procedure avoids the hazards of radioisotope labels and the technical limitations of electrical resistance technology.  相似文献   

17.
Long-chain fatty acids are an important source of energy in vascular endothelium. Their oxidation is stimulated by carnitine and inhibited by blockage of the mitochondrial respiratory chain. Excess fatty acid can be reversibly stored as triacylglycerol in the cells. Cultured vascular endothelial cells, in contrast to cardiac vascular endothelium in the intact heart, take up and intracellularly degrade artificial chylomicrons (intralipid enriched with apolipoprotein C-II) but not natural chylomicrons. Fatty acids not bound to albumin, such as those generated from chylomicrons in the lipoprotein lipase reaction, although initially a good source of substrate for beta-oxidation, endanger heart function. Fatty acid excess initiates the breakdown of the endothelial barrier between the vascular lumen and interstitium; it may precipitate edema formation, lead to insufficient oxygenation and finally cause loss of heart function.  相似文献   

18.
Microsphere embolism (ME)-induced up-regulation of endothelial nitric oxide synthase (eNOS) in endothelial cells of brain microvessels was observed 2-48 h after ischemia. eNOS induction preceded disruption of the blood-brain barrier (BBB) observed 6-72 h after ischemia. In vascular endothelial cells, ME-induced eNOS expression was closely associated with protein tyrosine nitration, which is a marker of generation of peroxynitrite. Leakage of rabbit IgG from microvessels was also evident around protein tyrosine nitration-immunoreactive microvessels. To determine whether eNOS expression and protein tyrosine nitration in vascular endothelial cells mediates BBB disruption in the ME brain, we tested the effect of a novel calmodulin-dependent NOS inhibitor, 3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate (DY-9760e), which inhibits eNOS activity and, in turn, protein tyrosine nitration. Concomitant with inhibition of protein tyrosine nitration in vascular endothelial cells, DY-9760e significantly inhibited BBB disruption as assessed by Evans blue (EB) excretion. DY-9760e also inhibited cleavage of poly (ADP-ribose) polymerase as a marker of the apoptotic pathway in vascular endothelial cells. Taken together with previous evidence in which DY-9760e inhibited brain edema, ME-induced eNOS expression in vascular endothelial cells likely mediates BBB disruption and, in turn, brain edema.  相似文献   

19.
Blood vascular networks in vertebrates are essential to tissue survival. Establishment of a fully functional vasculature is complex and requires a number of steps including vasculogenesis and angiogenesis that are followed by differentiation into specialized vascular tissues (i.e., arteries, veins, and lymphatics) and organ-specific differentiation. However, an equally essential step in this process is the pruning of excessive blood vessels. Recent studies have shown that pruning is critical for the effective perfusion of blood into tissues. Despite its significance, vessel pruning is the least understood process in vascular differentiation and development. Two recently published PLOS Biology papers provide important new information about cellular dynamics of vascular regression.Vascular biology is a rapidly emerging field of research. Given the critical role the vasculature frequently plays in a wide range of common and serious diseases such as arteriosclerosis, ischemic diseases, cancer, and chronic inflammatory diseases, a better understanding of the formation, maintenance, and remodeling of blood vessels is of major importance.A mature vascular network is a highly anisotropic, hierarchical, and dynamic structure that has evolved to provide optimal oxygen delivery to tissues under a variety of conditions. Whilst much has been learned about early steps in vascular development such as vasculogenesis and angiogenesis, we still know relatively little about how such anatomical and functional organization is achieved. Furthermore, the dynamic nature of mature vascular networks, with its potential for extensive remodeling and a continuing need for stability and maintenance, is even less understood. The issue of optimal vascular density in tissue is of particular importance as several recent studies demonstrated that excessive vascularity may, in fact, reduce effective perfusion [13]. Since all neovascularization processes initially result in the formation of excessive amounts of vasculature, be that capillaries, arterioles, or venules, pruning must occur to return the vascular density to its optimal value in order to achieve effective tissue perfusion.Yet despite its functional importance, little is known about how regression of the once formed vasculature actually happens. While several potential mechanisms have been proposed including apoptosis of endothelial cells, intussusception vascular pruning, and endothelial cell migration away from the regressing vessel, cellular and molecular understanding of how this might happen is conspicuously lacking. Two articles recently published in PLOS Biology describe migration of endothelial cells as the key mechanism of apoptosis-independent vascular pruning and place it in a specific biologic context. This important advance offers not only a new understanding of a poorly understood aspect of vascular biology but may also prove to be of considerable importance in the development of pro- and anti-angiogenic therapies.To put vessel regression in context, it helps to briefly outline the current understanding of vessel formation. During embryonic development, vasculature forms in several distinct steps that begin with vasculogenesis, a step that involves differentiation of stem cells into primitive endothelial cells that then form initial undifferentiated and nonhierarchically organized lumenized vascular structures termed the primary plexus [4]. The primary plexus is then remodeled, by the process termed angiogenesis, into a more mature vascular network [5]. This remodeling event involves both formation of new vessels accomplished either by branching angiogenesis, a process dependent on tip cell-driven formation of new branches [6], or intussusception, a poorly understood process of splitting an existing vessel into two [7]. This incompletely differentiated and still nonhierarchical vasculature then further remodels into a number of distinctly different types of vessels such as capillaries, arteries, and veins. This requires fate specification, differentiation, and incorporation of various mural cells into evolving vascular structures. Finally, additional specialization of the vascular network occur in an organ-specific manner.Once formed, vascular networks require active maintenance as withdrawal of key signals, such as of ongoing fibroblast growth factor (FGF) or vascular endothelial growth factor (VEGF) stimulation, can lead to a rapid loss of vascular integrity and even changes in endothelial cell fate [812]. In addition, mature vessels retain the capacity for extensive remodeling and new growth as can be seen in a number of conditions from cancer to myocardial infarction and wound healing responses, among many others [5].A key issue common to both embryonic and adult vessel remodeling is how an existing lumenized vessel connected to the rest of the vasculature undergoes a change that results in its remodeling into something else. Such a change may involve either a new branch formation or regression of an existing branch, while the patency and integrity of the remaining circulation is maintained. Two types of cellular process leading to branching have been described—sprouting and intussusception. Formation of vascular branches by sprouting involves VEGF-A-induced expression of high levels of delta-like ligand 4 (Dll4) in a subset of endothelial cells at the leading edge of the vascular sprouts that are lying closest to the source of VEGF, thus converting them to a “tip cell” phenotype. Some of the key features of tip cells include the presence of cytoplasmic processes that extend into avascular (or hypoxic) tissue that form nascent branches. Dll4 expressed on tip cells binds Notch-1 receptor in neighboring endothelial cells, thereby activating their downstream Notch signaling. In turn, Notch signaling shuts down the formation of additional filopodia processes, converting these cells to a “stalk cell” phenotype and thereby avoiding excessive branching [1315]. The bone morphogenetic protein signaling pathway provides further input in determining stalk cell fate [16]. Importantly, tip cells are only partially lumenized; only once they have converted to a stalk phenotype does the lumen extend to what was a tip cell and its sprouts.An alternative mechanism of branching involves intussusception, a process by which a tissue pillar from the surrounding tissue splits the existing endothelial tube into two along its long axis, creating two adjusting vessels. While this process has been described morphologically, virtually nothing is known about its molecular and cellular regulation. In development, angiogenesis by intussusception occurs in vessels previously formed by sprouting angiogenesis [17,18]. Importantly, however, both sprouting angiogenesis and intussusception allow growth and remodeling of vascular network without any integrity compromise, thereby avoiding bleeding and related complications.There are certain parallels between vessel formation and branching and vessel regression. While growth occurs either via sprouting (a process linked to endothelial cell-migration) or intussusception, regression involves either “reverse intussusception,” endothelial migration-dependent regression, or apoptosis. The latter is the primary means of regression of the hyaloid vasculature in the eye and of the vascular loss seen in oxygen-induced retinopathy (OIR). In the case of hyaloid vasculature, secretion of WNT7b by macrophages invading the hyaloid membrane induces apoptosis of hyaloid endothelial cells leading to the regression of the entire hyaloid vasculature [19]. This total apoptosis-induced loss of hyaloid blood vessels contrasts with a less extensive vascular regression seen in the setting of OIR. In this condition, exposure of the developing retinal vasculature to abnormally high oxygen levels leads to vascular damage characterized by capillary pruning [20]. The pruning is the consequence of apoptosis of endothelial cells due to the toxic effect of a combination of high oxygen and low VEGF level. Interestingly, larger vessels and mature capillaries are not sensitive to hyperoxia [21].Intussusception vascular pruning was also described in a low VEGF level context in the chick chorioallantoic membrane. Application of VEGF-releasing hydrogels to the membrane surface results in formation of an excessive vasculature. Removal or degradation of the hydrogel induces an abrupt VEGF withdrawal. In this context, formation of transluminal pillars, similar to the ones seen in intussusception angiogenesis, is observed in vessels undergoing pruning [22]. The same process is observed in the tumor vasculature in the setting of anti-angiogenic therapy [23]. Finally, apoptosis-independent vascular regression, driven by endothelial cell migration, has been described in the mouse retina, yolk vessels of the chick and mouse embryos, branchial arches, and the zebrafish brain [2428].In all of these cases, only a subset of vessels is designated for pruning, and the selection of these vessels is highly regulated. Yet, factors involved in choosing a particular vascular branch for pruning remain ill-defined. One such factor is low blood flow [27,28]. Another is Notch signaling that has been shown to at least partially control vascular pruning in mouse retina and in intersegmental vessels (ISVs) in zebrafish [24]. Loss of Notch-regulated ankyrin repeat protein (Nrarp), target gene of Notch signaling, leads to an increase in vascular regression in these tissues due to a decrease in Wnt signaling-induced stalk cell proliferation. Similarly, in Dll4 +/- mice, developmental retinal vascular regression and OIR-induced vascular pruning are reduced [29], confirming the involvement of the Notch pathway in the control of vascular regression.The two factors may be linked, as low flow can affect endothelial shear stress and lead to a decrease in Notch activation. Such a link is suggested by studies on vascular regression in mice with endothelial expression of dominant negative NFκB pathway inhibitor that demonstrate excessive vascular growth but reduced tissue perfusion [2]. Molecular studies showed inhibition of flow- or cytokine-induced NFκB activation results in decreased Dll4 expression [2].Another important issue is the fate of endothelial cells from vessels undergoing pruning. In PLOS Biology, two groups recently described endothelial cell behavior during vascular pruning in three different models: the mouse retina, the ISVs in zebrafish, and the subintestinal vessel in zebrafish [30,31]. Using a high resolution time-lapse microscopy technique, Lenard and collaborators showed that vascular pruning during the subintestinal vessel formation occurs in two different ways. In type I pruning, the first step is the collapse of the lumen. Once that occurs, endothelial cells migrate and incorporate into the neighboring vessels. In type II pruning, the lumen is maintained. One endothelial cell in the center of the pruning vessel undergoes self-fusion, leading to a unicellular lumenized vessel. At the same time, other endothelial cells migrate away and incorporate into the neighboring vessels. The eventual lumen collapse is the last step after which the remaining single endothelial cell migrates and incorporates into one of the major vessels.Franco and collaborators described a pruning mechanism similar to the type I pruning described by Lenard et al., showing lumen disruption as an initial step in pruning of retinal vasculature in mice and ISVs in zebrafish [31]. By analyzing the first axial polarity map of endothelial cells in these models, they demonstrated that axial orientation predicts endothelial cell migration, and that migration-driven pruning occurs in vessels with low flow. Interestingly, migrating endothelial cells in regressing vessel display a tip cell phenotype with filopodia.The cellular dynamic of vessel pruning described here is the reverse of the cellular dynamic during anastomosis and angiogenesis [32]. Given the crucial role of factors as VEGF for the migration of endothelial cells during angiogenesis, can we go further and propose that other cytokines or cell–cell signaling may be involved in the migration of these endothelial cells? Indeed, low blood flow seems to be the cause of vessel pruning, but how can we explain the direction of endothelial cell migration, moreover with a tip cell morphology? Also, what determines the choice between type I and type II pruning? The collapse of lumen suggests a reorganization of the cytoskeleton, and a loss of polarity and electrostatic repulsion of endothelial cells. Molecular mechanisms leading from low shear stress to loss of endothelial cell polarity need further investigation. As defective vascular pruning could be involved in poor recovery after injury or ischemic accident, a better understanding of the molecular control of this phenomenon appears to have medical consequences. Another question that is still unanswered is the fate of mural cells that surrounded the pruned vessels. Small vessels are covered by pericytes, which have strong interaction with endothelial cells. How and when are these interactions disrupted? Are pericytes integrated into the neighboring vessel, or do they undergo apoptosis? Further studies are needed to understand the molecular and cellular mechanisms by which vasculature can adapt, even at the adult stage, to support the nutrient and oxygen needs of each cell.Overall, taking the results of these studies together with other recent developments in this field, the following picture is emerging (Fig 1). Under conditions of low blood flow in certain vascular tree branches, pruning will occur via endothelial cell migration out of these branches to the neighboring (presumably higher blood flow) vessels. This results in decreased total vascular cross-sectional area and increased average blood flow, thereby terminating further pruning. Importantly, this occurs without the loss of luminal integrity and without reduction in the total endothelial cell mass. At the same time, vessels that suddenly find themselves in a low VEGF environment will regress either by apoptosis of endothelial cells or by intussusception. In both cases, there is a reduction in the total vasculature without an increase in blood flow to this tissue. Thus, the local context determines the mechanism: migratory regression and remodeling in low shear stress versus apoptotic pruning in low VEGF milieu.Open in a separate windowFig 1Vessel regression under low flow versus low VEGF conditions.Vessel regression under low flow conditions proceeds by endothelial cell (EC) migration-driven regression, resulting in a decrease in total vessel areas but an increase in blood flow (left panel). Vessel regression under low VEGF conditions proceeds by EC apoptosis or intussusception regression, resulting in decreased vessel number and decreased flow to tissues subtended by the regressing vasculature (right panel). Image credit: Nicolas Ricard & Michael Simons.This distinction is likely to be of a significant practical importance, in particular in the context of therapies designed to facilitate vessel normalization in tumors after VEGF-targeting treatments and therapies designed to promote vascularization of mildly ischemic tissues as occurs, for example, in the setting of chronic stable angina and other similar conditions. In the former case, a precipitous drop in VEGF levels is likely to induce vascular regression by induction of endothelial apoptosis, and further promotion of apoptosis may facilitate this process. In contrast, in the latter case, low flow in newly formed collateral arteries may induce their regression by stimulating outmigration of endothelial cells, thereby limiting their beneficial functional impact. Therapies designed to inhibit this mechanism, therefore, may promote growth of the new functional vasculature.  相似文献   

20.
Endothelin (ET)-1 was originally characterized as a potent vasoconstrictor peptide secreted by vascular endothelial cells. It possesses a wide range of biological activities within the cardiovascular system and in other organs, including the brain. Also secreted by endothelial cells, nitric oxide (NO), has recently been identified as a relaxing factor, as well as a pleiotropic mediator, second messenger, immune defence molecule, and neurotransmitter. Most of the data concerning the secretion of these two agents in vitro has been collected from studies on macrovascular endothelial cells. Given the remarkable heterogeneity of endothelia in terms of morphology and function, we have analyzed the ability of brain microvessel endothelial cells in vitro to release ET-1 and NO, which, at the level of the blood-brain barrier, have perivascular astrocytes as potential targets. The present study was performed with immortalized rat brain microvessel endothelial cells, which display in culture a non transformed phenotype. Our data demonstrate that: (1) these cells release NO when induced by IFNγ and TNFα, (2) they constitutively secrete ET-1, and (3) cAMP potentiates the cytokine-induced NO release and exerts a biphasic regulation on ET-1 secretion: micromolar concentrations of 8-Br-cAMP inhibit and higher doses stimulate ET-1 secretion. This stimulation is blocked by EGTA and the calmodulin antagonist W7, but not by protein kinase C inhibitors, suggesting the involvement of the calmodulin branch of the calcium messenger system. These results suggest that cerebral microvessel endothelial cells may participate in vivo to the regulation of glial activity in the brain through the release of NO and ET-1. © 1993 Wiley-Liss, Inc.  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