Fact-checked by Grok 2 weeks ago

Angiogenesis

Angiogenesis is the physiological process through which new blood vessels form from pre-existing vasculature, enabling the growth of networks essential for delivering oxygen and nutrients to tissues. This process occurs throughout life, beginning during embryonic development and continuing in adulthood to support , tissue repair, , and adaptation to physiological demands such as exercise-induced changes in and cardiac tissue. In healthy contexts, angiogenesis maintains tissue by ensuring no is more than a few hundred micrometers from a , thus preventing and supporting metabolic functions. Key mechanisms include sprouting angiogenesis, where endothelial tip cells migrate in response to gradients of (VEGF), followed by stalk and lumen formation guided by Delta-Notch signaling, as well as intussusceptive angiogenesis involving vessel splitting for rapid network expansion. Major regulators encompass pro-angiogenic factors like VEGF and (bFGF), which promote endothelial and migration, balanced by inhibitors such as endostatin and thrombospondin-1 that induce and limit vessel growth. Pathologically, dysregulated angiogenesis contributes to diseases including cancer, where tumors induce an "angiogenic switch" to sustain growth and ; ocular disorders like age-related macular degeneration; and chronic inflammatory conditions such as . Therapeutic strategies exploit these pathways, with anti-angiogenic agents like —a targeting VEGF—approved by the FDA in 2004 for treating and later expanded to other malignancies and neovascular eye diseases. Recent advances include combining anti-angiogenic agents with immunotherapies to enhance efficacy in treating various cancers. Conversely, pro-angiogenic therapies, such as recombinant bFGF, aim to stimulate vessel growth in ischemic conditions like . Research in this field, pioneered by in the 1970s with his hypothesis linking tumor progression to angiogenesis, has grown exponentially, with over 5,200 articles published in 2009 and informing ongoing clinical trials.

Introduction

Definition and Basic Process

Angiogenesis is the physiological process through which new blood vessels form from pre-existing vasculature, primarily involving the proliferation, migration, and reorganization of endothelial cells to create tubular structures that integrate into the vascular network. This process is essential for expanding the circulatory system in response to tissue demands, contrasting with vasculogenesis, which involves de novo vessel formation from endothelial precursor cells known as angioblasts during embryonic development. Unlike vasculogenesis, angiogenesis relies on the remodeling and extension of established vessels rather than initial assembly from isolated precursors. The basic process of angiogenesis unfolds in a series of coordinated steps, beginning with the degradation of the surrounding existing capillaries. Endothelial cells release proteases to break down this barrier, allowing cells to protrude and initiate vessel sprouting. A specialized endothelial cell is then selected as the "tip cell," which extends to sense environmental cues and lead directional migration toward angiogenic stimuli, such as (VEGF), which serves as a primary initiator of this response. Behind the tip cell, "stalk cells" proliferate to elongate the sprout, forming a multicellular column that maintains connectivity with the parent vessel. Subsequently, lumen formation occurs as endothelial cells rearrange to create hollow tubes, involving intracellular fusion and remodeling to establish a conduit for blood flow. The nascent vessels then undergo , where sprouts from different sites connect to form functional loops, enabling circulation. Finally, vessel maturation stabilizes the structure, with recruited to the endothelial tubes to deposit components and regulate , while cells contribute to vessel wall reinforcement, ensuring long-term integrity and contractility. Endothelial cells remain central throughout, forming the inner lining and responding to signals for both sprouting and stabilization.

Biological Importance

Angiogenesis plays a fundamental role in normal by enabling the delivery of oxygen and nutrients to tissues during , growth, and repair processes. In embryonic , it supports the expansion of metabolically active tissues by forming new blood vessels from existing ones, ensuring adequate vascularization proportional to metabolic demands. This process is crucial for , where new capillary networks facilitate the influx of immune cells, nutrients, and oxygen to promote tissue regeneration and formation. Furthermore, angiogenesis maintains tissue by dynamically adjusting vascular density in response to physiological changes, such as increased capillary formation in during exercise or in with weight gain. Dysregulation of angiogenesis leads to significant pathological consequences, with excessive vessel formation contributing to diseases like cancer, chronic inflammation, and retinopathy. In tumors, angiogenesis is essential for growth beyond a microscopic size, as it provides the necessary blood supply for nutrient delivery and waste removal, a concept first proposed by in 1971. Overexpression of pro-angiogenic factors like VEGF drives aberrant neovascularization in chronic inflammatory conditions such as and , exacerbating tissue damage. In diabetic retinopathy, excessive retinal angiogenesis results in leaky, fragile vessels that cause vision loss through hemorrhage and edema. Conversely, insufficient angiogenesis impairs recovery in ischemic conditions, such as , where limited vessel growth restricts oxygen supply to hypoxic tissues, and delays in chronic ulcers by hindering nutrient delivery. The process of angiogenesis exhibits remarkable evolutionary conservation across vertebrates, including jawless fish, and shares mechanistic roots with hypoxia-sensing pathways in . Recent studies using have revealed heterogeneity in endothelial cell responses, further elucidating adaptive vascular remodeling (as of ). This conservation underscores its fundamental role in vascular adaptation across species and holds promise for , where harnessing conserved pathways could enhance tissue repair in humans. Angiogenesis operates in balance with vascular regression, forming a dynamic remodeling system where unused vessels regress—such as in muscle after disuse—while new ones form in response to stimuli, ensuring efficient resource allocation.

Types of Angiogenesis

Sprouting Angiogenesis

Sprouting angiogenesis represents the primary mechanism by which new blood vessels form from pre-existing capillaries, involving the directed outgrowth of endothelial cells to expand vascular networks. This process begins with the activation of endothelial cells in response to angiogenic stimuli, leading to the degradation of the and the (ECM) via matrix metalloproteinases and other proteases. The selected leading endothelial cells, known as tip cells, extend dynamic to sense and migrate toward pro-angiogenic cues, such as (VEGF) gradients, while trailing stalk cells proliferate to elongate the sprout. Central to sprout initiation is the selection of tip cells through lateral inhibition mediated by Delta-Notch signaling. Endothelial cells compete for tip cell fate; those with higher VEGF receptor 2 (VEGFR2) expression upregulate Delta-like 4 (Dll4), activating in neighboring cells to suppress their tip cell characteristics and promote stalk cell identity instead. This results in a patterned arrangement of alternating tip and stalk cells, ensuring organized branching and preventing excessive density. Delta-Notch interactions thus maintain a balance between at the tip and in the stalk, with disruptions leading to hyper- or hypo-branching phenotypes observed in developmental models. As tip cells advance, they invade the surrounding using filopodia protrusions stabilized by dynamics and , guiding the sprout through the tissue. Stalk cells follow, forming a tubular structure via lumenogenesis, where intracellular vacuoles to create a vessel . These sprouts eventually anastomose—connecting tip cells from adjacent sprouts—to form functional vascular loops that enable flow and . are recruited to stabilize the nascent vessels, depositing new components to mature the network. This mode of angiogenesis predominates in embryonic vascular development, where it establishes the primary through patterned in structures like the and intersomitic vessels. It is also critical during , facilitating rapid to supply oxygen and nutrients to repairing tissues. In pathological contexts, such as solid tumors, angiogenesis drives aberrant vessel growth, supporting tumor expansion by providing metabolic support despite often resulting in leaky, tortuous vessels.

Intussusceptive Angiogenesis

Intussusceptive angiogenesis, also known as splitting angiogenesis, is a mode of vessel formation characterized by the internal division of existing without the need for endothelial cell or significant . This process was first morphologically identified in the developing rat lung, where small transluminal pillars were observed perforating capillary walls. Unlike angiogenesis, it relies on the remodeling of preexisting vascular structures to rapidly expand microvascular networks. The mechanism begins with the formation of intravascular tissue pillars that span the of a , typically initiated by points of contact between opposing endothelial cells. These pillars, often 1-1.5 μm in diameter, arise in regions of altered , such as low zones, and are influenced by forces like blood flow convergence at . The process unfolds in distinct phases: initial endothelial cell contact and protrusion formation, followed by perforation of the to create a transluminal bridge, involvement of to stabilize the structure, and finally deposition of components like for pillar maturation. As pillars grow and align in rows, they fuse into that bisect the , leading to and the creation of two parallel daughter vessels from a single parent , all without requiring extensive degradation or endothelial migration. Structurally, intussusceptive angiogenesis results in a rapid increase in capillary density through this internal partitioning, transforming two-dimensional networks into more complex three-dimensional architectures. is often reversible, allowing for pillar regression and vessel pruning to optimize network efficiency in response to changing demands. For instance, in models of adaptation, it can elevate the capillary-to-fiber ratio by 15-20%, enhancing tissue oxygenation without net cell addition. This form of angiogenesis is prevalent in contexts requiring swift vascular adaptation, such as alveolarization during postnatal development, where it facilitates a 20- to 30-fold increase in capillary volume from birth to adulthood in rats. It also occurs in tumor microenvironments adapting to , enabling rapid vessel duplication within hours to days, and in inflammatory conditions like murine , where it supports tissue perfusion during acute responses. Shear stress variations, such as those from altered blood flow, can trigger pillar initiation in these settings. Compared to sprouting angiogenesis, intussusceptive angiogenesis offers key advantages in speed and efficiency, completing vessel splitting within hours or even minutes rather than days, and demanding minimal energy since it bypasses the need for endothelial and major extracellular remodeling. This makes it particularly suited for scenarios of rapid tissue expansion or adaptation under physiological constraints.

Coalescent Angiogenesis

Coalescent angiogenesis represents a distinct of vascular characterized by the fusion of existing segments to form larger conduits, thereby optimizing blood flow efficiency without relying on from pre-existing vessels. This process involves the longitudinal of two or more smaller vessels, which merge along their axes to create a single, wider vessel capable of handling increased hemodynamic demands. Triggered primarily by detected through endothelial mechanoreceptors, the mechanism entails dynamic remodeling where endothelial cells align and fuse, often modulated by signaling pathways such as VEGF-induced Delta-like ligand 4 expression or inhibition to facilitate cell-cell and expansion. A key aspect of coalescent angiogenesis is the concurrent of underperfused or unnecessary vessels, which accompanies to sculpt a hierarchical vascular network from an initial isotropic . This remodeling reduces the total number of vessels while increasing their diameters, transforming a low-resistance, inefficient into a tree-like structure that supports convective transport of nutrients and oxygen. Structural adaptations include the elimination of internal pillars within fused segments, ensuring seamless and preventing flow disruptions, as evidenced by intravital studies in embryonic models showing phased progression from mesh formation to stabilized conduits over hours to days. This type of angiogenesis plays a critical role in embryonic vascular patterning, where it contributes to the maturation of major arteries like the dorsal aorta through symmetric fusion of primitive vessels in avian and mammalian models. In retinal development, coalescent processes aid in refining the superficial vascular plexus by merging nascent capillaries into deeper, more robust networks during early postnatal stages. Guidance by pro-maturational factors such as angiopoietins supports these fusions, promoting vessel stability during network reorganization.81426-9) Following fusion, recruitment is essential for stabilizing the newly formed larger vessels, where these mural cells invest along the to enhance structural integrity and regulate permeability. This post-fusion stabilization prevents regression of the remodeled conduits and ensures long-term functionality in the hierarchical network, as respond to PDGF-B signaling from endothelial cells to migrate and envelop the fused segments.

Regulation of Angiogenesis

Mechanical Factors

Mechanical forces play a pivotal role in regulating angiogenesis by influencing endothelial cell behavior and vascular network architecture independent of chemical mediators. These forces arise from hemodynamic conditions, extracellular matrix (ECM) interactions, and tissue deformations, guiding processes such as sprouting initiation and vessel stabilization. Key mechanical cues include hemodynamic shear stress from blood flow, interstitial flow through tissues, and tensile forces within the ECM, each modulating endothelial responses through mechanotransduction pathways. Hemodynamic , generated by blood flow along vessel walls, promotes angiogenic branching at low magnitudes (less than 10 dyn/cm²) while inhibiting excessive at physiological levels (10–70 dyn/cm²) to ensure vessel maturation and alignment. Interstitial flow, occurring at velocities up to 2 µm/s in perivascular spaces, directs endothelial via durotaxis along gradients and enhances tip polarization, facilitating sprout elongation and . Tensile forces, often from cyclic stretching of the (5–15% ), increase traction and cytoskeleton remodeling, thereby boosting and capillary on matrices with in the range of 500–2500 Pa. In contexts such as exercise-induced blood flow, these forces drive adaptive vascular remodeling in ; elevated and stretch in contribute to aberrant vessel thickening; and injury-related triggers matrix stiffening to support . Endothelial cells sense these mechanical stimuli through mechanotransduction complexes involving , , and . (e.g., αvβ3 and α5β1) link the to the , activating focal adhesion kinase (FAK) and Rho-associated kinase (ROCK) pathways to upregulate and in response to and . , localized at cell-cell junctions, transmits signals via 3-kinase (PI3K)/Akt activation, promoting cell survival and directed sprouting. mediates stretch-induced junctional remodeling, weakening adherens junctions under high to enable tip cell specification and collective during branching. These pathways allow mechanics to amplify underlying chemical signals, such as enhancing (VEGF) responsiveness in one coordinated process.

Pro-angiogenic Chemical Signals

Pro-angiogenic chemical signals encompass a diverse array of soluble factors and matrix-associated proteins that orchestrate activation, migration, proliferation, and vessel maturation during angiogenesis. These molecules are primarily secreted by hypoxic , inflammatory cells, and endothelial cells themselves, responding to cues like tissue injury or growth demands. Key families include vascular endothelial growth factors (VEGFs), fibroblast growth factors (FGFs), angiopoietins, matrix metalloproteinases (MMPs), and select signaling pathways such as Dll4-Notch and class 3 semaphorins, alongside platelet-derived growth factors (PDGFs), each contributing distinct mechanisms to vascular expansion. The VEGF family stands as the cornerstone of pro-angiogenic signaling, with VEGF-A being the predominant isoform that binds primarily to receptors (VEGFRs) 1 and 2 on endothelial cells. VEGF-A exists in multiple isoforms generated by , such as VEGF-A_{165} and VEGF-A_{121}, which differ in heparin-binding domains affecting their and . Upon binding to VEGFR-2, a receptor, VEGF-A triggers downstream pathways like PI3K/Akt and MAPK/ERK, promoting endothelial , , , and increased essential for sprout invasion into the (). VEGFR-1 modulates these effects by sequestering VEGF-A or facilitating fine-tuned signaling, while VEGF-C and VEGF-D, though more lymphangiogenic, support angiogenesis via VEGFR-2 under certain conditions. Hypoxia-inducible factor (HIF)-1α upregulates VEGF expression in low-oxygen environments, initiating angiogenic cascades. Fibroblast growth factors, particularly FGF-1 and FGF-2 (basic FGF), exert potent mitogenic effects on endothelial cells by binding to fibroblast growth factor receptors (FGFRs), which are tyrosine kinases expressed on vascular . FGF-2, often released from damaged or cells, activates FGFR-1 and FGFR-2, stimulating and MAPK pathways that enhance endothelial , migration, and production for remodeling. These factors synergize with VEGFs to amplify angiogenesis, as evidenced in models of and tumor vascularization where FGF blockade impairs vessel formation. Unlike VEGFs, FGFs also recruit and smooth muscle cells to stabilize nascent vessels. Angiopoietins, including Ang-1 and Ang-2, modulate vessel maturation and remodeling through the Tie2 on endothelial cells. Ang-1, produced by and cells, acts as a Tie2 that promotes endothelial junction integrity, suppresses permeability, and recruits mural cells to stabilize mature vessels post-sprouting. In contrast, Ang-2, stored in Weibel-Palade bodies of endothelial cells and released upon stimulation, antagonizes Ang-1 at Tie2, destabilizing junctions to enable remodeling and responsiveness to pro-angiogenic cues like VEGF. This dynamic balance ensures appropriate vessel branching and during physiological angiogenesis. Matrix metalloproteinases, such as MMP-2 (gelatinase A) and MMP-9 (gelatinase B), facilitate angiogenesis by degrading components like collagen IV and , creating paths for endothelial migration and sprout elongation. These zinc-dependent endopeptidases are secreted as pro-enzymes and activated by or other MMPs, with endothelial cells and inflammatory infiltrates as primary sources. MMP-2 and MMP-9 not only liberate -bound growth factors like VEGF but also expose cryptic pro-angiogenic sites on matrix proteins, enhancing endothelial invasion without excessive tissue disruption. Among other regulators, the Dll4-Notch signaling pathway refines angiogenic branching by in endothelial tip cells. Dll4, a membrane-bound upregulated by VEGF on leading tip cells, activates Notch receptors on adjacent stalk cells, suppressing their responsiveness to VEGF and limiting excessive to maintain organized vessel patterns. Class 3 semaphorins, such as Sema3A and Sema3C, provide guidance cues during vascular patterning by interacting with neuropilin-1 and plexin receptors, promoting directed endothelial and fine-tuning branch orientation in developmental and reparative angiogenesis. (PDGF), particularly PDGF-BB, supports recruitment and vessel maturation by binding PDGF receptor-β on , indirectly enhancing endothelial stability and preventing regression of new vessels.

Anti-angiogenic Chemical Signals

Anti-angiogenic chemical signals are essential endogenous molecules that counteract pro-angiogenic factors to regulate vascular and prevent excessive formation. These inhibitors maintain a delicate balance during physiological processes such as embryonic development, where uncontrolled angiogenesis could lead to malformed vasculature, and in adulthood, where they suppress aberrant vessel growth in pathological conditions like tumor progression. Among the key endogenous inhibitors, thrombospondin-1 (TSP-1), a large , plays a pivotal role by binding to the receptor on endothelial cells, thereby activating signaling pathways that induce and inhibit and . Originally identified as the first natural , TSP-1 is secreted by various cell types including endothelial cells and platelets, and its expression is upregulated in response to tissue remodeling needs. Endostatin, a 20-kDa fragment derived from the C-terminal noncollagenous domain (NC1) of XVIII, potently suppresses endothelial cell proliferation, migration, and survival by disrupting -mediated signaling, particularly through α5β1 , and interfering with (VEGF) pathways. This inhibitor is generated via proteolytic cleavage by enzymes such as cathepsin L and is present in the of various tissues, contributing to the suppression of in normal .00005-0) Similarly, angiostatin, a 38-kDa internal fragment cleaved from plasminogen by urokinase-type , inhibits endothelial cell and by binding to αvβ3 and F1F0 on the cell surface, thereby blocking ATP production necessary for angiogenic responses. Circulating in as part of the fibrinolytic , angiostatin helps regulate vessel remodeling during and prevents pathological overgrowth. Soluble receptor-1 (sVEGFR-1, also known as sFlt-1) acts as a receptor that sequesters VEGF and (PlGF) in the extracellular space, preventing their interaction with membrane-bound VEGFR-2 on endothelial cells and thus dampening pro-angiogenic signaling. Produced by of the FLT1 gene in endothelial cells, monocytes, and trophoblasts, sFlt-1 levels increase under hypoxic conditions to fine-tune VEGF . Other notable inhibitors include interferon-alpha (IFN-α), a that downregulates the expression of pro-angiogenic factors such as (bFGF), interleukin-8 (IL-8), and matrix metalloproteinase-9 (MMP-9) in endothelial and tumor cells, thereby suppressing vessel sprouting. Interleukin-4 (IL-4), an immune-modulatory , inhibits bFGF-induced endothelial proliferation and tube formation, often through upregulation of anti-angiogenic genes. Tissue inhibitors of metalloproteinases (TIMPs), particularly TIMP-2 and TIMP-3, block matrix degradation required for endothelial invasion; for instance, TIMP-2 binds α3β1 to halt independently of its MMP-inhibitory function, while TIMP-3 directly antagonizes VEGF binding to VEGFR-2. These anti-angiogenic signals operate through feedback mechanisms that sense local cues like or tissue stress, upregulating inhibitor production to curb excessive vessel growth; for example, in embryonic development, TSP-1 and endostatin limit vascular overexpansion to ensure proper organ patterning, while in , their downregulation allows unchecked angiogenesis in tumors. This loop, often involving proteolytic generation of inhibitors from larger precursors, maintains vascular quiescence and prevents disorders arising from imbalanced angiogenesis.

Physiological Roles

Embryonic Development and Growth

Angiogenesis is essential for embryonic development, enabling the expansion and patterning of the vascular system to support tissue growth and organogenesis. In mouse embryos, the process initiates shortly after implantation, with the first signs of vascular development appearing in the extraembryonic yolk sac around embryonic day 7.5 (E7.5), where endothelial precursors undergo vasculogenesis to form blood islands. By E8.5, these coalesce into a primitive capillary plexus, marking the onset of angiogenic remodeling that refines the network into larger vessels. This early yolk sac angiogenesis is critical for nutrient exchange and provides a scaffold for intraembryonic vascular extension. The transition from vasculogenesis to angiogenesis occurs progressively from E8.5 onward, involving the sprouting of new vessels from the initial plexus to establish a hierarchical circulatory system. This includes arterial-venous specification, where endothelial cells differentiate into artery- or vein-specific subtypes based on cues like blood flow hemodynamics and molecular signals such as ephrin-B2 and Coup-TFII, beginning around E9.5 in the yolk sac and embryo proper. Neural guidance cues, including semaphorins (e.g., Sema3A) and netrins, play a pivotal role in this patterning by directing endothelial tip cell migration and filopodial extension, ensuring precise vascular alignment with developing tissues. Sprouting angiogenesis, the dominant mechanism here, relies on VEGF gradients to drive tip cell selection and stalk cell proliferation. Organ-specific vascularization exemplifies angiogenesis's role in embryogenesis. In the , vessels sprout from a perineural around E9.0-E10.5, invading the under VEGF and Wnt signaling to form a ramified network and initiate blood- barrier formation by E12.5. Coronary angiogenesis in the heart begins at E11.5, with endothelial cells from and sprouting into the myocardium to vascularize the compact layer; recent lineage tracing confirms contributions from multiple progenitors including and , with minor input from epicardial cells, dependent on factors like PDGF-B and supported by epicardial cues. In limb buds, initial vascular ingress occurs at E9.5 via angiogenic sprouts from the intersomitic vessels and , forming a primitive that patterns the limb's arterial arches and venous drainage, influenced by FGF and BMP gradients from the apical ectodermal ridge. Genetic models underscore the indispensability of angiogenesis regulators. Homozygous null mutations in VEGF lead to embryonic lethality between E8.5 and E9.5, characterized by arrested endothelial cell differentiation and failure to form vessels or embryonic . Similarly, Tie2 (Tek) knockout mice succumb at E9.5 with disorganized endothelial clusters in the and embryo, lacking proper vessel remodeling and integrity due to impaired signaling. These phenotypes highlight how disruptions in core pathways halt developmental progression, preventing organ vascularization and tissue viability.

Wound Healing and Tissue Repair

Angiogenesis plays a crucial role in by supplying oxygen and nutrients to the injured tissue, facilitating the repair process through the formation of new blood vessels from existing ones. In the inflammatory , signals from immune cells trigger angiogenesis, where endothelial cells from nearby vessels proliferate and migrate into the site to form sprouts that invade the fibrin-rich clot. This process is essential for establishing a provisional vascular that supports subsequent repair stages. During the proliferative phase, these sprouts organize into a mature microvascular network within the , a fibrovascular matrix composed of fibroblasts, , and new vessels that fills the wound bed and promotes re-epithelialization. Key drivers include in the wound bed, which stabilizes hypoxia-inducible factor-1α (HIF-1α) to upregulate pro-angiogenic factors like (VEGF), and macrophage-secreted factors such as VEGF and (bFGF), which amplify endothelial cell proliferation and migration. In the remodeling phase, excess vessels undergo regression through , the network to restore normal tissue architecture and prevent excessive scarring. In chronic wounds, such as diabetic ulcers, angiogenesis is often impaired due to an excess of anti-angiogenic inhibitors like endostatin and thrombospondin-1, alongside reduced pro-angiogenic signaling from hyperglycemia-induced , leading to persistent and stalled formation. This dysregulation results in non-healing ulcers that affect millions annually and increase risk. Conversely, regenerative potential is highlighted in scarless fetal , where angiogenesis is enhanced with a 2-fold increase in vessel density and elevated VEGF expression at early gestational stages, enabling rapid regeneration without . Matrix metalloproteinases (MMPs) contribute to this by remodeling the to support endothelial invasion, as detailed in pro-angiogenic signaling pathways.

Exercise-Induced Vascular Adaptation

Physical exercise stimulates angiogenesis primarily in to meet heightened metabolic demands, enhancing tissue and oxygen delivery during activity. This adaptive response involves the formation of new capillaries through mechanisms such as and intussusceptive angiogenesis, driven by both hemodynamic and biochemical cues. In endurance-trained athletes, these changes result in a more efficient vascular network, supporting prolonged performance without excessive fatigue. The primary mechanisms triggering exercise-induced capillary growth include shear stress from increased blood flow and metabolic perturbations like and accumulation. , a mechanical force exerted on endothelial cells by elevated blood velocity during exercise, promotes angiogenesis by upregulating (NO) production and (VEGF) expression, as demonstrated in models where blocking VEGF abolished shear-dependent vessel formation. Concurrently, metabolic demand signals such as —produced during —act via the HCAR1 receptor to stabilize hypoxia-inducible factor-1α (HIF-1α), thereby enhancing VEGF from muscle fibers and fostering sprouting. These processes, while interconnected, differ from general mechanical factors in their exercise-specific integration of flow dynamics with tissue-level . Key adaptations include an elevated -to-fiber ratio, which improves oxygen diffusion and nutrient supply to muscle cells. In untrained individuals, can increase this ratio by 10-20% within weeks, while in athletes, it may rise by up to 30%, correlating with enhanced aerobic capacity and reduced reliance on pathways. This vascular remodeling optimizes oxygen delivery, as evidenced by higher VO₂max and capillary density in trained versus sedentary populations. At the molecular level, coactivator 1-alpha (PGC-1α) plays a central role by upregulating VEGF transcription in response to exercise-induced , independent of HIF-1α in some contexts. PGC-1α , triggered by AMPK signaling during contraction, not only drives angiogenesis but also coordinates , ensuring matched vascular and metabolic adaptations; knockout studies show 60-80% reductions in VEGF protein levels and ~20% reductions in capillary-to-fiber ratios without PGC-1α. VEGF, released primarily from myofibers, binds endothelial receptors to initiate endothelial and . Despite these benefits, exercise-induced angiogenesis exhibits limits, including plateau effects where capillary growth stabilizes after initial phases, as seen in studies showing no further increases beyond 4-8 weeks of endurance exercise. Sustained is required to maintain these adaptations and prevent , with detraining leading to rapid capillary .

Pathological Implications

Tumor Angiogenesis and Vessel Formation

Tumors initially grow as avascular masses limited to approximately 1-2 mm in diameter, relying on for nutrient and oxygen supply, beyond which ensues without vascularization. This constraint prompts the "angiogenic switch," where tumor cells transition to an angiogenic , secreting pro-angiogenic factors to induce vessel formation and support further expansion. In many solid tumors, this process predominantly involves angiogenesis, adapting normal vascular mechanisms to the pathological tumor environment. Hypoxia within the expanding avascular tumor core activates hypoxia-inducible factor-1 (HIF-1), which transcriptionally upregulates (VEGF) expression in tumor cells. Secreted VEGF binds to endothelial receptors on nearby vessels, promoting endothelial , , and tube formation to generate new tumor vasculature. Concurrently, an imbalance favoring angiopoietin-2 (Ang-2) over angiopoietin-1 destabilizes existing vessels by disrupting pericyte-endothelial interactions, resulting in immature, leaky vessels that facilitate initial tumor but exhibit structural disarray. Tumor-induced vessels are characteristically abnormal, featuring tortuous, dilated, and irregularly branched structures with increased permeability due to fenestrations and discontinuous membranes. These irregularities lead to heterogeneous blood flow, regions of poor , and persistent intratumoral , which paradoxically sustains further VEGF production and angiogenic drive. The hyperpermeable enables extravasation of plasma proteins and s, creating a fibrin-rich matrix that supports tumor invasion and facilitates by providing routes for tumor intravasation. Vascular heterogeneity in tumors arises from inconsistent perivascular coverage, with often deficient or loosely associated, failing to stabilize vessels and contributing to their immaturity. This deficit exacerbates vessel leakiness and susceptibility, fostering a microenvironment that promotes tumor progression and resistance to physiological constraints on growth.

Ocular Diseases

Excessive angiogenesis plays a central role in several ocular diseases, leading to pathological vessel growth that disrupts normal retinal and choroidal function, ultimately causing vision impairment or blindness. In conditions such as wet age-related macular degeneration (AMD) and proliferative diabetic retinopathy (PDR), aberrant arises from imbalances in angiogenic signaling, primarily driven by (VEGF), which promotes endothelial and vessel permeability. These disorders highlight how local environmental stressors in the eye trigger uncontrolled vascularization, distinct from physiological angiogenesis due to the fragile and leaky nature of the new vessels. Wet , the neovascular form of age-related , is characterized by (CNV), where new blood vessels grow from the into the sub-retinal pigment epithelium space or the sub-retinal space. This process is initiated by local in the aging and , compounded by , which upregulates VEGF expression from retinal pigment epithelial cells and other sources, driving endothelial migration and tube formation. The resulting vessels are immature and permeable, leading to fluid leakage, , hemorrhage, and that distort the and cause central vision loss. Progression typically begins with accumulation and outer creating hypoxic avascular zones, followed by invasive CNV that breaches and the blood- barrier, exacerbating leakage into the neurosensory . A unique aspect of CNV in wet is the disruption of the outer blood- barrier at , allowing choroidal vessels to invade the avascular outer , which is normally shielded from systemic circulation. Diabetic retinopathy, particularly its proliferative stage (PDR), involves where fragile new vessels sprout from the or retinal veins into the vitreous or along the retinal surface. Hyperglycemia-induced retinal ischemia creates hypoxic areas of non-perfusion, triggering and the release of pro-angiogenic factors like VEGF from Müller glial cells and , which stimulate endothelial and vascular invasion. These vessels are prone to rupture, causing vitreous hemorrhage, tractional , and neovascular glaucoma, all contributing to severe vision loss. The disease progresses from initial microvasculopathy with capillary dropout forming avascular hypoxic zones to aggressive preretinal or intravitreal neovascularization that breaches the inner blood- barrier, allowing leakage and inflammatory infiltration into the neural . Distinctively, the inner blood-retinal barrier's tight junctions in PDR are compromised by VEGF-mediated downregulation of and claudins, facilitating pathological vessel growth into normally avascular vitreous spaces. VEGF, as a dominant pro-angiogenic signal, underscores these mechanisms across both conditions.

Cardiovascular Disorders

In cardiovascular disorders, angiogenesis is often dysregulated, manifesting as either insufficient vessel formation in ischemic conditions or excessive, aberrant that exacerbates disease progression. This imbalance contributes to poor , plaque instability, and adverse cardiac remodeling, highlighting the critical role of angiogenic processes in maintaining vascular . In ischemic contexts, such as post-myocardial (MI), insufficient angiogenesis severely limits recovery by failing to restore adequate blood supply to the infarcted myocardium. Following acute MI, the hypoxic environment triggers angiogenic responses starting from the peri-infarcted border zone, but inadequate leads to persistent ischemia, increased infarct size, and reduced cardiomyocyte survival. Mechanisms include disrupted signaling pathways like HIF-1α/VEGF activation and impaired endothelial cell proliferation due to excessive (ROS) or dysregulated microRNAs (e.g., miR-19a-3p inhibiting VEGF expression), which collectively hinder vessel maturation and . This deficiency promotes adverse left , characterized by and , ultimately progressing to chronic . Conversely, in , excessive plaque neovascularization promotes vessel instability through the formation of immature, leaky microvessels within the arterial wall. within advanced plaques induces angiogenesis primarily from adventitial , driven by HIF-1α and VEGF-A, resulting in fragile vessels that facilitate infiltration and intraplaque hemorrhage. These leaky structures extravasate red blood cells, leading to deposition, iron-mediated , and , which correlate strongly with plaque vulnerability (e.g., microvessel density r=0.99 with leakage). Ruptured plaques exhibit the highest neovessel density, often 2-3 times that of stable lesions, exacerbating accumulation and activity that precipitate acute events like . Underlying these dysregulations are key mechanisms such as impaired of endothelial progenitor cells (EPCs) and , which compromise angiogenic capacity across cardiovascular diseases. EPCs, essential for postnatal and vascular repair, show reduced numbers and functionality in conditions like , , and , correlating with endothelial injury and limited collateral formation. and risk factors disrupt EPC mobilization and integration into nascent vessels, while —marked by reduced bioavailability—further impairs EC migration and tube formation, perpetuating ischemia. Angiopoietins, for instance, modulate this process by stabilizing vessels, but their imbalance exacerbates dysfunction in ischemic settings. The outcomes of these angiogenic impairments often culminate in due to poor collateral vessel formation, which fails to compensate for chronic . In patients with stable and chronic total occlusion, factors like exposure inhibit angiogenesis by disrupting VEGFR2/PI3K/Akt signaling, reducing endothelial and recovery, and independently predicting inadequate collaterals (OR 1.043). This leads to diminished density in ischemic myocardium, worsening ventricular function and increasing mortality risk, as evidenced by lower limb ischemia models showing reduced blood flow with impaired growth. Overall, such deficiencies in collateral angiogenesis contribute to progressive by sustaining myocardial and remodeling.

Therapeutic Applications

Promoting Angiogenesis

Promoting angiogenesis involves therapeutic strategies aimed at stimulating new formation to restore in ischemic tissues, particularly for conditions like (PAD) and myocardial ischemia. These approaches leverage angiogenic factors such as (VEGF) to enhance vascularization, addressing limitations in natural repair mechanisms. Clinical applications focus on delivering these factors through targeted methods to improve outcomes in tissue repair and regeneration. Gene therapy represents a key approach for promoting angiogenesis, primarily through VEGF delivery to upregulate local angiogenic signaling in ischemic regions. Adenoviral or plasmid-based vectors encoding VEGF-A have been used to stimulate endothelial , migration, and recruitment, showing efficacy in preclinical models of limb and cardiac ischemia. For instance, intramuscular VEGF transfer in PAD patients has demonstrated improved collateral vessel formation and limb in phase II trials. Protein administration offers a direct method to enhance angiogenesis by infusing recombinant VEGF or (FGF) proteins into affected areas. In PAD, intra-arterial VEGF-165 delivery has increased capillary density and walking distance in clinical studies, while in myocardial ischemia, intracoronary FGF-2 administration has promoted collateral growth and reduced symptoms. These therapies provide rapid onset but require repeated dosing due to short half-lives. Stem cell and endothelial (EPC) transplantation further augments angiogenesis by mobilizing cells that secrete angiogenic factors and integrate into nascent vessels. Autologous bone marrow-derived mononuclear cells or EPCs, when injected into ischemic limbs or hearts, enhance and in patients with critical limb ischemia and post-myocardial damage. A randomized trial of intramuscular EPC transplantation in severe limb ischemia reported significant healing and reduced rates at 24 weeks. In applications for PAD and myocardial ischemia, these strategies collectively improve blood flow and functional recovery; for example, combined and delivery has shown synergistic effects in restoring hindlimb perfusion in animal models of . Wound healing augmentation benefits similarly, with VEGF protein or accelerating granulation tissue formation and epithelialization in chronic ulcers by boosting microvascular networks. Tissue engineering integrates these approaches using scaffolds embedded with growth factors like VEGF to support angiogenesis in and transplants. or decellularized scaffolds releasing controlled doses of angiogenic proteins promote vascular infiltration and maturation within engineered tissues, enabling viable organoid development for applications in skin grafts and vascularized implants. Preclinical studies demonstrate that VEGF-loaded scaffolds enhance vessel density in subcutaneously implanted , facilitating nutrient delivery for larger-scale tissue constructs. Despite these advances, challenges persist, including the transient effects of VEGF-based therapies, which often lead to short-lived vessel formation due to rapid protein or immune clearance of vectors. Off-target growth poses additional risks, such as aberrant vessel leakage or from excessive VEGF signaling, contributing to inconsistent clinical outcomes. Recent progress, such as recombinant CCL28 protein administration, addresses some limitations by promoting stable angiogenesis via CCR10+ endothelial cells, improving cardiac repair and in models. Exercise serves as a natural promoter of angiogenesis through shear stress-induced VEGF expression, complementing therapeutic interventions in ischemic rehabilitation.

Inhibiting Angiogenesis

Anti-angiogenic therapies aim to suppress pathological neovascularization, particularly in cancers where tumor growth relies on sustained blood vessel formation. These approaches target key signaling pathways to starve tumors of nutrients and oxygen, thereby inhibiting proliferation and metastasis. Bevacizumab, a humanized monoclonal antibody that binds vascular endothelial growth factor A (VEGF-A), was the first approved angiogenesis inhibitor and is used in combination with chemotherapy for various solid tumors, including metastatic colorectal, non-small cell lung, and renal cell carcinomas. Sunitinib, an oral multitargeted tyrosine kinase inhibitor, blocks receptors such as VEGF receptors, platelet-derived growth factor receptors, and others involved in angiogenesis, demonstrating efficacy in advanced renal cell carcinoma and gastrointestinal stromal tumors by reducing tumor vascularization and progression. In ocular diseases, anti-angiogenic agents address aberrant vessel growth in conditions like neovascular age-related (). , a recombinant humanized fragment that neutralizes all isoforms of VEGF-A, is administered via intravitreal injection to inhibit , preserving in wet patients as shown in multicenter trials. Emerging strategies expand beyond traditional VEGF inhibition to enhance therapeutic durability. Immunotherapies modulating tumor-associated macrophages () reprogram these cells from pro-angiogenic M2 to anti-angiogenic M1 phenotypes, with 2024 advances in engineered macrophages demonstrating improved tumor infiltration and vascular suppression in preclinical models. Targets like delta-like ligand 4 (DLL4), a Notch pathway component, offer promise in VEGF-resistant tumors; DLL4 blockade disrupts tip cell selection in sprouting vessels, reducing tumor angiogenesis independently or synergistically with agents. Additionally, ivonescimab, a bispecific targeting PD-1 and VEGF-A, was approved in in May 2024 for advanced non-small cell in combination with , showing promise in ongoing global trials as of November 2025. Despite successes, challenges persist in clinical application. Resistance often arises through alternative pro-angiogenic pathways, such as or signaling, leading to tumor adaptation and relapse. Common side effects include due to systemic VEGF inhibition affecting normal vasculature, as well as proteinuria and increased risk. Some pro-angiogenic herbal compounds, like those from medicines, may inadvertently counter by enhancing vessel formation, though their clinical impact remains underexplored.

History

Early Discoveries

The earliest scientific insights into angiogenesis emerged in the 18th century through the work of Scottish surgeon John Hunter, who in the 1760s observed the dynamic growth of s in embryos, noting their proportionality to the metabolic demands of developing tissues. Hunter's observations underscored the adaptive nature of vascular expansion during rapid physiological processes, such as and embryonic development, laying foundational concepts for later research. The term "angiogenesis" was first used by John Hunter in 1787 to describe blood vessel growth in antlers. In the mid-19th century, advanced the cellular theory of , which laid groundwork for understanding its role in pathological processes including tissue remodeling and repair. Toward the end of the , Moritz Ribbert's 1880 experiments demonstrated that tumors actively induce from adjacent host tissues, revealing a tumor-host interaction essential for growth. In 1907, Edwin Goldmann demonstrated, using transparent chamber models in rabbits, that tumors induce from adjacent host tissues and that central tumor regions undergo due to limited vascular supply, underscoring the need for angiogenesis to support tumor growth beyond initial sizes. Early 20th-century tumor implantation studies reinforced these findings; for example, implants of tumor fragments in animal models, such as those conducted in the using ear chambers, illustrated that viable tumor growth is strictly dependent on rapid vascular ingrowth, without which tumors remained dormant or necrotic. In 1935, Arthur T. Hertig applied the term to blood vessel formation in the developing of monkeys.

Key Milestones in Research

The molecular era of angiogenesis research began with Judah Folkman's seminal 1971 hypothesis, which posited that tumor growth is dependent on angiogenesis and that tumors secrete a factor to induce , laying the groundwork for targeting this process therapeutically. This idea shifted the focus from tumor cells alone to the , predicting that inhibiting angiogenesis could restrict tumor expansion beyond microscopic sizes. A major breakthrough came in the 1980s with the isolation of vascular permeability factor (VPF) from tumor fluid in 1983 by Harold Dvorak and colleagues, a potent inducer of vascular leakage later recognized as a key angiogenic mediator. Building on this, between 1989 and 1990, Napoleone Ferrara's team at isolated and cloned (VEGF) from bovine pituitary cells and tumor cells, identifying it as a specific for endothelial cells and establishing VEGF as the primary driver of pathological angiogenesis. These discoveries enabled the molecular characterization of angiogenesis signaling pathways, transforming it from a descriptive phenomenon into a targetable process. In the , research expanded to endogenous inhibitors, with Michael O'Reilly in Folkman's laboratory isolating angiostatin in 1994 from the conditioned medium of a Lewis lung , revealing it as a kringle-domain fragment of plasminogen that selectively suppresses endothelial and . Shortly after, in 1997, the same group discovered endostatin, a XVIII fragment extracted from a tumor, which potently inhibits angiogenesis and tumor growth in preclinical models by disrupting endothelial cell survival and migration. These findings demonstrated that tumors produce their own angiogenesis inhibitors, explaining dormancy mechanisms and inspiring a new class of anti-angiogenic agents. During the same decade, Peter Burri and colleagues elucidated intussusceptive angiogenesis, a non- of vessel formation first observed in the developing rat lung in 1986 and mechanistically detailed in 1990, where existing capillaries divide internally via pillar formation to rapidly expand networks without endothelial proliferation. This process, distinct from traditional , was shown to contribute to adaptive vascular remodeling in physiological and pathological contexts, broadening the understanding of angiogenic diversity. Therapeutic translation advanced significantly with the 2004 FDA approval of (Avastin), the first anti-angiogenic drug, a targeting VEGF, which extended survival in metastatic when combined with in pivotal trials. This milestone validated Folkman's hypothesis clinically, establishing anti-VEGF therapy as a cornerstone for and spurring approvals in other cancers. In the 2010s and 2020s, single-cell RNA sequencing (scRNA-seq) has unveiled profound endothelial cell heterogeneity in angiogenesis, with seminal work by Kalucka et al. in 2020 mapping transcriptomic profiles across murine tissues and identifying distinct angiogenic subtypes responsive to stimuli like VEGF. These studies revealed context-specific endothelial populations, such as tip, stalk, and cells, with varying proliferative and migratory potentials, enhancing insights into therapeutic resistance and vascular normalization.

Measurement and Quantification

In Vitro and In Vivo Assays

In vitro assays provide controlled environments to study individual steps of angiogenesis, such as endothelial , migration, and differentiation, using isolated cells or simplified matrices. These models allow for of pro- and anti-angiogenic factors but simplify the complex multicellular interactions present . One widely adopted is the endothelial tube formation on , where human umbilical vein endothelial cells (HUVECs) or other endothelial cells are seeded onto a extract like , a gelled matrix derived from . Within 4-16 hours, the cells reorganize into capillary-like tube structures mimicking vascular formation, enabling assessment of angiogenic potential in response to stimuli such as (VEGF). This is valued for its simplicity and reproducibility, with tube formation quantified by parameters including total branch length and number of branching nodes. The scratch wound migration assay evaluates endothelial cell motility, a key early step in angiogenesis. In this method, a confluent monolayer of endothelial cells is scratched with a sterile tool to create a denuded area, and cell migration into the wound is monitored over 24-48 hours using time-lapse imaging. Factors like VEGF can enhance closure rates, reflecting chemotactic responses. Migration speed and wound closure percentage serve as primary metrics. Ex vivo assays bridge simplicity and complexity by using intact tissue explants. The aortic ring sprouting assay involves embedding transverse sections of or aorta in a three-dimensional or matrix, where microvessels sprout outward over 7-14 days, recapitulating sprouting angiogenesis with contributions from and fibroblasts. Sprout length, vessel density, and invasion area are common metrics to gauge angiogenic activity. Another prominent ex vivo model is the chick chorioallantoic membrane (CAM) assay, utilizing the vascularized extra-embryonic membrane of 8-10 day old chicken embryos. Implants such as tumor fragments or pellets are placed on the , inducing vessel invasion and remodeling observable over 3-5 days, providing insights into angiogenic responses in a developing vascular bed. Metrics include the area of vessel invasion and density around the implant. Across these assays, key quantitative metrics focus on structural features to assess angiogenic extent, such as cumulative branch length (total elongation in micrometers), node count (number of junctions or endpoints indicating network complexity), and invasion area (spatial coverage of sprouts in square millimeters). These parameters are often analyzed using software like ImageJ's Angiogenesis Analyzer for automated, reproducible measurements. Despite their utility, and assays have limitations, primarily the absence of full physiological context including blood flow, immune interactions, and systemic hormonal influences, which can lead to discrepancies with in vivo outcomes. Additionally, variability in matrix composition and sourcing affects reproducibility, and these models often overlook long-term vessel maturation and stability.

Imaging and Molecular Techniques

Imaging techniques play a crucial role in visualizing and quantifying angiogenesis in vivo, enabling non-invasive assessment of vascular development and response to therapies. Magnetic resonance imaging (MRI) enhanced with gadolinium (Gd)-based probes has emerged as a powerful tool for monitoring anti-angiogenic effects, particularly in tumor models. Recent advances in 2025 introduced Gd-DOTA-G3CNGRC, a targeted probe that binds specifically to aminopeptidase N (APN/CD13) on angiogenic endothelial cells, allowing early detection of therapeutic efficacy through enhanced contrast in T1-weighted images. This probe demonstrated superior specificity compared to non-targeted Gd agents, with signal intensity reductions correlating to decreased vascular permeability post-treatment in preclinical studies. Intravital microscopy provides dynamic, real-time visualization of angiogenic processes at the cellular level, capturing , sprout formation, and vascular remodeling in living tissues. By employing multiphoton or confocal techniques, researchers can track multi-cellular interactions during angiogenesis, such as guidance and extension, with resolutions down to micrometers. This method has revealed flow-directed endothelial behaviors in models like the wounded , where serial imaging over days highlights temporal changes in vessel and maturation. Molecular techniques offer precise quantification of angiogenic activity through gene and protein expression . Quantitative polymerase chain reaction (qPCR) is widely used to measure mRNA levels of key angiogenic factors like (VEGF) and angiopoietins, providing insights into transcriptional regulation during hypoxia-induced angiogenesis. (ELISA) detects soluble markers such as circulating VEGF and (sFlt-1), which reflect systemic angiogenic states and therapeutic modulation in serum samples. Single-cell RNA sequencing (scRNA-seq) unveils endothelial cell heterogeneity in angiogenic niches, identifying subpopulations with distinct or profiles that drive vessel instability. In vivo models leverage species-specific advantages for imaging angiogenesis. The transparency of zebrafish larvae facilitates high-resolution optical imaging of subintestinal vessels, allowing non-invasive tracking of angiogenic sprouting in response to genetic or pharmacological perturbations without pigmentation interference. In mice, the hindlimb ischemia model induces robust angiogenesis via ligation, enabling longitudinal assessment of collateral vessel formation and over weeks. Quantitative metrics derived from these techniques standardize angiogenesis evaluation. Microvessel density (MVD), often assessed via immunostaining, serves as a for angiogenic extent, with elevated counts indicating proliferative vascular networks in ischemic tissues. Perfusion rates, measured through dynamic contrast-enhanced imaging or laser Doppler flowmetry, quantify functional blood flow, revealing improvements in vessel patency post-angiogenic stimulation. Integration of () in image analysis, as advanced in 2024 protocols, automates vessel segmentation and density calculations from data, enhancing and reducing manual bias in large datasets.

References

  1. [1]
    Overview of Angiogenesis - NCBI - NIH
    Angiogenesis is the growth of blood vessels from the existing vasculature. It occurs throughout life in both health and disease, beginning in utero and ...
  2. [2]
    Angiogenesis and Its Therapeutic Opportunities - PMC
    Angiogenesis plays critical roles in human physiology that range from reproduction and fetal growth to wound healing and tissue repair.
  3. [3]
    Angiogenesis - PMC - PubMed Central - NIH
    Angiogenesis begins with endothelial cell (EC) activation, degradation of vascular basement membrane, and vascular sprouting within interstitial matrix.Angiogenesis · Integrins In Embryonic... · Ecm Remodeling During...
  4. [4]
    ADGRL4/ELTD1 is a highly conserved angiogenesis-associated ...
    Jul 12, 2019 · We identified ADGRL4/ELTD1 as a highly conserved early angiogenic gene which emerged in the first true vertebrates (bony fish) approximately 435 million years ...
  5. [5]
    VEGF and Notch in Tip and Stalk Cell Selection - PMC
    During angiogenesis, tip and stalk endothelial cells expand existing blood vessel networks. This is controlled by the VEGF and Notch signaling pathways, which ...Missing: review | Show results with:review
  6. [6]
    Notch mediates the sprouting of tip cells - Nature
    During sprouting angiogenesis, tip cells are selected from the endothelial cells of existing vessels to pave the way for new blood vessels.
  7. [7]
    The modes of angiogenesis: an updated perspective - PMC - NIH
    Aug 28, 2023 · These different modes of angiogenesis involve, for example, increasing microvasculature density by sprouting of endothelial cells, splitting of vessels.Missing: seminal | Show results with:seminal
  8. [8]
    Endothelial Cell Self-fusion during Vascular Pruning | PLOS Biology
    Apr 17, 2015 · New vascular sprouts connect to each other to form functional loops with blood flow, a process termed anastomosis.
  9. [9]
    Tumour vascularization: sprouting angiogenesis and beyond - PMC
    This review summarizes the different mechanisms of tumour vascularization, the molecular players that are involved and their relevance in clinical practice.Missing: seminal | Show results with:seminal
  10. [10]
    Tortuous Microvessels Contribute to Wound Healing via Sprouting ...
    Aug 24, 2017 · A new type of transient wound vessel, tortuous microvessels, sprout dynamically and disproportionately contribute to wound-healing neoangiogenesis.
  11. [11]
    Intussusceptive Angiogenesis | Circulation Research
    Intussusceptive angiogenesis is a novel mode of blood vessel formation and remodeling, which occurs by internal division of the preexisting capillary plexus ...Missing: seminal | Show results with:seminal
  12. [12]
  13. [13]
    Intussusceptive angiogenesis: Its emergence, its characteristics, and ...
    Sep 16, 2004 · This review shall familiarize the reader with the various aspects of intussusceptive angiogenesis (IA). The basic event in IA is the ...Missing: seminal | Show results with:seminal
  14. [14]
    Intussusceptive Angiogenesis: Expansion and Remodeling of ...
    Abstract. Intussusceptive angiogenesis is a dynamic intravascular process capable of dramatically modifying the structure of the microcirculation.Missing: seminal papers
  15. [15]
    Intussusceptive Angiogenesis: A Biologically Relevant Form of ...
    Jun 26, 2012 · Here, the mechanism of intussusceptive angiogenesis is reviewed and the current techniques and models for investigating intussusceptive ...Missing: seminal | Show results with:seminal
  16. [16]
  17. [17]
  18. [18]
  19. [19]
  20. [20]
  21. [21]
  22. [22]
  23. [23]
    Endogenous Inhibitors of Angiogenesis - AACR Journals
    Thrombospondin-1 (TSP-1) was the first protein to be recognized as a naturally occurring inhibitor of angiogenesis (71). It is a large multifunctional ECM ...
  24. [24]
    Endogenous inhibitors of angiogenesis in malignant gliomas
    In this review, we focus on angiostatin, endostatin, PEX, pigment epithelial-derived factor, and thrombospondin (TSP)-1 and -2, either because these ...
  25. [25]
    Angiostatin and endostatin: endogenous inhibitors of tumor growth
    Angiostatin and Endostatin, endogenous angiogenesis inhibitors that have been shown to regress tumors in murine models.Missing: thrombospondin- | Show results with:thrombospondin-
  26. [26]
    Soluble VEGF receptor 1 (sFLT1) induces non-apoptotic death in ...
    Apr 22, 2016 · Soluble Vascular Endothelial Growth Factor Receptor 1 (sVEGFR1/sFLT1) is an angiogenesis inhibitor that competes with angiogenic factors ...
  27. [27]
    Interferon-alpha as angiogenesis inhibitor: learning from tumor models
    The effects of IFN-alpha on the vasculature have been mainly attributed to inhibition of basic fibroblast growth factor production by tumor cells or ...
  28. [28]
    Inhibition of Angiogenesis by Interleukin 4 - PMC - NIH
    Interleukin (IL)-4, a crucial modulator of the immune system and an active antitumor agent, is also a potent inhibitor of angiogenesis.
  29. [29]
    The tissue inhibitors of metalloproteinases (TIMPs) - PubMed Central
    ... inhibition of MMPs by TIMPs can prevent angiogenesis [117]. However, TIMP-2 and TIMP-3 also exhibit anti-angiogenic activity that is independent of their ...
  30. [30]
    Endogenous inhibitors of angiogenesis: A historical review
    Endogenous inhibitors of angiogenesis are defined as proteins or fragments of proteins that are formed in the body and can inhibit the formation of blood ...
  31. [31]
    Vascular development and hemodynamic force in the mouse yolk sac
    Vascular remodeling of the mouse embryonic yolk sac is a highly dynamic process dependent on multiple genetic signaling pathways as well as biomechanical ...
  32. [32]
    The murine allantois: a model system for the study of blood vessel ...
    Sep 27, 2012 · These vessels form the primary vascular plexus, evident at the 4s stage, which joins the embryonic and yolk sac circulation at the base of the ...Ecm In The Allantois · Wnt And Bmp Signaling In... · Allantois Culture Ex Vivo...Missing: timeline | Show results with:timeline<|control11|><|separator|>
  33. [33]
    Arterial–Venous Specification During Development
    Mar 13, 2009 · Initial embryonic determination of artery or vein identity is regulated by variety of genetic factors that work in concert to specify endothelial cell fate.Missing: timeline | Show results with:timeline
  34. [34]
    Neural guidance molecules regulate vascular remodeling and ...
    Emerging evidence summarized below suggests that axon growth cones and capillary tip cells use common signaling cues including netrins, slits, and semaphorins ...Ephrins And Eph Receptors... · Netrins As Repulsive Vessel... · Neuropilin Receptors And The...
  35. [35]
    Developmental Angiogenesis of the Central Nervous System - PMC
    During development, the vasculature of the CNS develops exclusively via angiogenesis—the growth of new blood vessels from pre-existing ones—as opposed to de ...Vegf/vegfr/neuropilin-1 · Notch/dlll4 · Tgfβ/endoglin/alk1
  36. [36]
    Three-Dimensional Analysis of Vascular Development in the Mouse ...
    Key vasculogenic (de-novo vessel forming) and angiogenic (vessel remodelling) events occur in the mouse embryo between embryonic days (E) 8.0 and 10.0 of ...
  37. [37]
    Vascular Endothelial Growth Factor A (VEGF-A) Is Involved in ... - NIH
    In VEGF-A gene knockout studies, even VEGF-A heterozygous (+/−) mice showed embryonic lethality due to multiple defects in vascular structure formation (6, 13).Results · An Inhibitor Of Vegf... · Vegfr-Positive Cells Migrate...
  38. [38]
    Rescue of the early vascular defects in Tek/Tie2 null mice reveals an ...
    Gene targeting studies in mice have revealed that the Tek receptor and its ligands appear to be required for the angiogenic remodeling and vessel stabilization ...
  39. [39]
    Angiogenesis in embryonic development--a review - PubMed
    ... Tie2 system, were identified as central regulators of embryonic angiogenesis. The complex interactions between these and other endothelial signalling ...
  40. [40]
    Angiogenesis in Wound Healing - ScienceDirect.com
    During wound healing, angiogenic capillary sprouts invade the fibrin/fibronectin-rich wound clot and within a few days organize into a microvascular network.
  41. [41]
    Skin Acute Wound Healing: A Comprehensive Review
    Jun 2, 2019 · Wound healing angiogenesis is thought to be an ongoing process in two phases: the proliferation of new blood vessels and the pruning and ...
  42. [42]
    Wound angiogenesis as a function of tissue oxygen tension - PNAS
    Inflammatory cells (i.e., macrophages and neutrophils) assist in wound healing by migrating to the injury site where they release angiogenic growth factors ...
  43. [43]
    The Role of Hypoxia-Inducible Factor in Wound Healing - PMC
    Hypoxia has been traditionally regarded as an important stimulus for fibroblast growth and angiogenesis. Both adaptive responses occur through the activation of ...
  44. [44]
    Diabetes and Wound Angiogenesis - PMC - NIH
    Jul 3, 2017 · Insufficient angiogenesis plays a significant role in the pathogenesis of diabetic wound healing and micro and microvascular disease.
  45. [45]
    Increased angiogenesis and expression of vascular endothelial ...
    Increased angiogenesis and expression of vascular endothelial growth factor during scarless repair. Plast Reconstr Surg. 2005 Jan;115(1):204-12. Authors. Amy ...
  46. [46]
    Exercise-induced skeletal muscle angiogenesis: impact of age, sex ...
    Jan 30, 2023 · This review will detail the biochemical, cellular and biomechanical signals for exercise-induced skeletal muscle angiogenesis, including recent ...
  47. [47]
    Peripheral limitations for performance: Muscle capillarization
    Sep 28, 2023 · Exercise training increases the number of capillaries per muscle fiber by about 10%–20% within a few weeks in untrained subjects, whereas ...
  48. [48]
    VEGF, shear stress and muscle angiogenesis: a complicated triangle
    Apr 24, 2015 · 2013). The effects of exercise on muscle angiogenesis are complex and involve both metabolic (reduced tissue levels of oxygen and nutrients) and ...<|control11|><|separator|>
  49. [49]
    Metabolic regulation of exercise-induced angiogenesis - PMC - NIH
    In this review, we provide a concise overview on how exercise-induced alterations in muscle metabolism can evoke metabolic changes in endothelial cells (ECs) ...Missing: demand | Show results with:demand
  50. [50]
    PGC-1alpha mediates exercise-induced skeletal muscle VEGF ...
    In skeletal muscle of PGC-1alpha KO mice, VEGF protein expression was approximately 60-80% lower and the capillary-to-fiber ratio approximately 20% lower than ...Missing: angiogenesis athletes
  51. [51]
    Tumor angiogenesis: therapeutic implications - PubMed
    Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971 Nov 18;285(21):1182-6. doi: 10.1056/NEJM197111182852108. Author. J Folkman. PMID: 4938153; DOI ...
  52. [52]
    Angiogenesis in cancer and other diseases - PubMed
    Pathological angiogenesis is a hallmark of cancer and various ischaemic and inflammatory diseases. Concentrated efforts in this area of research are leading ...
  53. [53]
    Vessel Cooption, Regression, and Growth in Tumors Mediated by ...
    The VEGFs and the angiopoietins seem to play complementary and coordinated roles in vascular development (9, 11). During development, VEGF acts via the Flk1/KDR ...Missing: Holash | Show results with:Holash
  54. [54]
    Novel targets beyond vascular endothelial growth factor-A inhibition
    Oct 11, 2025 · VEGF-A and the VEGF-A/VEGFR-2 axis is widely recognised for its promotion of angiogenesis and vascular permeability, the two hallmark features ...
  55. [55]
    VEGF in Diabetic Retinopathy and Age-Related Macular Degeneration
    The release of VEGF is known to mediate the abnormal angiogenesis seen in wet AMD, and long-term anti-VEGF therapy has remained the gold standard treatment to ...Vegf In Diabetic Retinopathy... · 4. Anti-Vegf Therapy For Dr... · 4.4. Anti-Vegf Therapy For...
  56. [56]
    A Linkage between Angiogenesis and Inflammation in Neovascular ...
    Nov 1, 2022 · It is observed that during aging and early stages of AMD development, choroidal blood flow drops and both choroid and choriocapillaries become ...
  57. [57]
    Application and mechanism of anti-VEGF drugs in age-related ...
    Sep 22, 2022 · Wet AMD is mainly characterized by choroidal neovascularization (CNV) formation, retinal pigment epithelium (RPE) detachment, macular hemorrhage ...
  58. [58]
    Novel approach to antiangiogenic factors in age-related macular ...
    VEGF leads to vascular leakage and neovascularization [7, 13, 14]. Pathological angiogenesis in eye tissues is induced by VEGF formation in tissues such as ...
  59. [59]
    Mechanisms of Acquired Resistance to Anti-VEGF Therapy ... - IOVS
    This progresses to the angiogenic late-stage of the disease, termed neovascular (or wet) AMD, when hypoxia triggers overexpression and release of VEGF causing ...<|control11|><|separator|>
  60. [60]
    Vascular endothelial growth factor and its inhibitor in age-related ...
    As a possible path for pathogenesis of CNV in AMD, it is suggested that VEGF plays a major part at the initial stage of CNV by promoting angiogenesis and is a ...
  61. [61]
    Role of oxidative stress, inflammation, hypoxia and angiogenesis in ...
    May 29, 2018 · Diabetes induced hypoxia retinal tissues stimulate angiogenesis by modulating the balance of pro-angiogenic and anti-angiogenic mediators this ...
  62. [62]
    Angiogenesis-Inflammation Cross Talk in Diabetic Retinopathy
    Pathological angiogenesis of the retina is a key component of irreversible causes of blindness, as observed in proliferative diabetic retinopathy (PDR).
  63. [63]
    Molecular Mediators and Regulators of Retinal Angiogenesis
    Dec 19, 2022 · The pathological condition like hypoxia/ischemia leads to initiation of angiogenesis from center to periphery of retina. While VEGF is a main ...
  64. [64]
    Diabetic Retinopathy: Vascular and Inflammatory Disease - PMC
    Conversely, the angiogenic factors VEGF and angiopoietin-1 may elicit proinflammatory responses in endothelial cells by upregulating the expression of cell ...
  65. [65]
    Proliferative diabetic retinopathy transcriptomes reveal ... - Nature
    Sep 22, 2021 · In this study we identify potential molecular mechanisms and target candidates in the pathogenesis of PDR fibrovascular tissue formation.
  66. [66]
  67. [67]
  68. [68]
  69. [69]
  70. [70]
  71. [71]
  72. [72]
    Endothelial Progenitor Cell Dysfunction in Cardiovascular Diseases ...
    Dec 30, 2012 · Patients with cardiovascular diseases have been shown to exhibit reduced EPC number and function. It has become increasingly apparent that these ...<|separator|>
  73. [73]
    Therapeutic angiogenesis: controlled delivery of angiogenic factors
    For therapeutic purposes, local levels of angiogenic VEGF can be upregulated by using genes, proteins and cells. Thus far, vegf gene delivery has comprised the ...
  74. [74]
    Gene-Therapeutic Strategies Targeting Angiogenesis in Peripheral ...
    Mar 30, 2018 · It has been described that VEGF-A stimulates endothelial cell proliferation and migration, as well as endothelial progenitor cell recruitment to ...
  75. [75]
    Vascular Endothelial Growth Factor (VEGF) and Its Role in the ... - NIH
    May 10, 2024 · Angiogenesis stimulation can be physiological or therapeutic and it can accelerate collateral circulation formation and increase ...2. Vegf Role In Angiogenesis... · 4. Vegf As A Biomarker In... · 6. Vegf Inhibition In...<|control11|><|separator|>
  76. [76]
    Therapeutic angiogenesis-based strategy for peripheral artery disease
    Therapeutic angiogenesis, which aims to induce endogenous vessel formation and blood perfusion recovery in the ischemic sites, is considered one of the most ...Peripheral Artery Disease · Gene-Based Therapy · Cell-Based Therapy
  77. [77]
    Stem cell-based therapies to promote angiogenesis in ischemic ...
    Stem cell therapy is a promising approach for the treatment of tissue ischemia associated with myocardial infarction and peripheral arterial disease.
  78. [78]
    Therapeutic Angiogenesis with Somatic Stem Cell Transplantation
    Oct 8, 2019 · Therapeutic angiogenesis is an important strategy to rescue ischemic tissues in patients with critical limb ischemia having no other ...
  79. [79]
    Therapeutic Strategies for Enhancing Angiogenesis in Wound Healing
    Epidermal growth factor (EGF) is another promising growth factor for wound healing therapies and a known mediator of angiogenesis. Early clinical trials showed ...
  80. [80]
    Angiogenesis in Tissue Engineering: As Nature Intended? - Frontiers
    Mar 19, 2020 · Similarly, angiogenesis within tissue engineered scaffolds is traditionally stimulated by the addition of single growth factors (GF) to the ...
  81. [81]
    Angiogenesis induction using organoid-tissue modules
    Oct 16, 2025 · Accordingly, extensive efforts in tissue engineering have focused on developing biomaterial scaffolds, angiogenic growth factor delivery ...
  82. [82]
    CCL28 contributes to angiogenesis and cardiac repair through ...
    Oct 20, 2025 · The deletion of CCL28 results in impaired angiogenesis, whereas the use of recombinant CCL28 protein has therapeutic potential for myocardial ...
  83. [83]
    Bevacizumab - StatPearls - NCBI Bookshelf - NIH
    Bevacizumab is a humanized monoclonal antibody against vascular endothelial growth factor A (VEGF-A). It is used to treat several different cancers.
  84. [84]
    Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of ...
    Bevacizumab (Avastin®), a VEGF-A targeting monoclonal antibody, was the first approved angiogenesis inhibitor. •. Approved in a range of solid tumor indications ...Anti-Tumour Treatment · Introduction · Lessons Learnt And Future...
  85. [85]
    Sunitinib: the antiangiogenic effects and beyond - PMC - NIH
    Sep 8, 2016 · Mechanistically, sunitinib inhibits multiple receptor tyrosine kinases, especially those involved in angiogenesis, that is, vascular endothelial ...
  86. [86]
    A preclinical review of sunitinib, a multitargeted receptor tyrosine ...
    Sunitinib malate is an oral, multitargeted tyrosine kinase inhibitor that targets both angiogenic pathways (i.e., vascular endothelial growth factor ...
  87. [87]
    Ranibizumab for neovascular age-related macular degeneration
    Oct 5, 2006 · Methods: In this multicenter, 2-year, double-blind, sham-controlled study, we randomly assigned patients with age-related macular degeneration ...
  88. [88]
    Ranibizumab and Bevacizumab for Neovascular Age-Related ...
    Apr 28, 2011 · Clinical trials have established the efficacy of ranibizumab for the treatment of neovascular age-related macular degeneration (AMD).
  89. [89]
    Emerging macrophage-based therapies for cancer: a review of ...
    Recognizing its critical role, therapeutic strategies have been developed to target immune and stromal components, angiogenesis, and metabolic pathways (9–13).
  90. [90]
    Advances in Engineered Macrophages: A New Frontier in Cancer ...
    Apr 1, 2024 · Current research on macrophages in cancer immunotherapy is centred on unravelling their intricate functions within the tumour microenvironment ...
  91. [91]
    Dll4-Notch signaling as a therapeutic target in tumor angiogenesis
    Sep 18, 2011 · The endothelial Notch ligand Dll4 (delta-like 4) has recently emerged as a critical regulator of tumor angiogenesis and thus as a promising new therapeutic ...
  92. [92]
    The role of Notch ligand, Delta-like ligand 4 (DLL4), in cancer ...
    Apr 26, 2021 · DLL4 seems to be a promising target in anti-cancer therapy. ... DLL4-Notch signaling mediates tumor resistance to anti-VEGF therapy in vivo.
  93. [93]
    Current Challenges of Cancer Anti-angiogenic Therapy and the ...
    In this review, we shed light on tumor vessels and their microenvironment and consider the current directions of anti-angiogenic and nanotherapeutic treatments.Missing: transient | Show results with:transient<|separator|>
  94. [94]
    Anti-angiogenesis in cancer therapeutics: the magic bullet
    Jul 2, 2021 · Tumour resistance and cardiovascular toxicity are important challenges which limit the efficacy and long-term use of anti-angiogenic agents in ...
  95. [95]
    Targeting angiogenesis in gastrointestinal tumors: current challenges
    Sep 6, 2016 · The most common adverse effects of anti-angiogenic therapy are hypertension, proteinuria, thromboembolism, gastrointestinal perforation ...<|separator|>
  96. [96]
    Exploring the pro-angiogenic potential of Chinese herbal medicines
    Jul 24, 2025 · This review aims to summarise current research on the pro-angiogenic effects and underlying molecular mechanisms of CHM, including herbal extracts, traditional ...
  97. [97]
    The History of Tumour Angiogenesis as a Therapeutic Target
    Aug 6, 2025 · 11 On October 23 rd 1907, Professor Edwin Goldmann (1862- 1913) co-discoverer of the blood-brain barrier in 1913, 12 and a student of the ...Missing: Ernst | Show results with:Ernst
  98. [98]
    Judah Folkman, a pioneer in the study of angiogenesis - PMC - NIH
    Judah Folkman found a revolutionary new way to think about cancer. He postulated that in order to survive and grow, tumors require blood vessels.
  99. [99]
    Historical Highlights - The Angiogenesis Foundation
    1787: British surgeon Dr. John Hunter first uses the term “angiogenesis” (new blood vessel growth) to describe blood vessels growing in the reindeer antler.Missing: 1760s chick Virchow 1880 1907 diffusion limits implantation
  100. [100]
    Intussusceptive angiogenesis--the alternative to capillary sprouting
    It was first discovered in the lung as a means of capillary network growth (intussusceptive microvascular growth). The mechanism consists in the repeated ...
  101. [101]
    A critical analysis of current in vitro and in vivo angiogenesis assays
    The limitation of all the assays is the time consuming nature of the analysis, which means that although the most reliable results are likely to be obtained by ...
  102. [102]
    Tube formation: an in vitro matrigel angiogenesis assay - PubMed
    This chapter describes an easy-to-perform assay that is based on the differentiation of endothelial cells and the formation of tube-like structures on an ...
  103. [103]
    Endothelial Cell Tube Formation Assay for the In Vitro Study of ... - NIH
    This assay can be used to identify genes and pathways that are involved in the promotion or inhibition of angiogenesis in a rapid, reproducible, and ...
  104. [104]
    Angiogenesis Analyzer for ImageJ — A comparative morphometric ...
    Jul 14, 2020 · Endothelial tube formation assay. Network formation in the ETFA was carried out by seeding HUVEC (105 cells/well) on Matrigel (250 µl/well) ...Missing: seminal | Show results with:seminal
  105. [105]
    Investigating Angiogenesis on a Functional and Molecular Level by ...
    May 31, 2024 · We detail a scratch wound migration assay utilizing a live cell imaging system measuring endothelial cell migration in a 2D setting and the ...
  106. [106]
  107. [107]
    A microfluidic wound-healing assay for quantifying endothelial cell ...
    The wound-healing assay works by growing endothelial cells in a confluent monolayer and physically introducing a wound by scratching the layer. Cells move into ...
  108. [108]
    The aortic ring model of angiogenesis: a quarter century of ... - NIH
    The aortic ring model has become one of the most widely used methods to study angiogenesis and its mechanisms.
  109. [109]
    Aortic Ring Assay - PMC - NIH
    The aortic ring assay is an angiogenesis model that is based on organ culture. In this assay, angiogenic vessels grow from a segment of the aorta.Missing: review | Show results with:review
  110. [110]
    Improved quantification of angiogenesis in the rat aortic ring assay
    The improved method uses computer-assisted analysis to measure aortic ring area, microvessel number, branchings, length, distribution, and fibroblast-like cell ...Missing: review | Show results with:review<|separator|>
  111. [111]
    The chorioallantoic membrane assay is a promising ex vivo model ...
    The chorioallantoic membrane (CAM) assay is a tool for studying vascular anomalies, and is a suitable model system for studying them.
  112. [112]
    The Chick Embryo Chorioallantoic Membrane Model: A Research ...
    The chick CAM model is used for toxicology, angiogenesis, cancer research, and testing new therapies, acting as an intermediate between in vitro and animal ...
  113. [113]
    Chick chorioallantoic membrane assay as an in vivo model to study ...
    Jun 4, 2018 · In this model, human cancer cells are transplanted onto the CAM membrane that surrounds chicken embryo within the fertilized chicken egg. After ...<|separator|>
  114. [114]
    A Comparison of Methods for Quantifying Angiogenesis in the ... - NIH
    Various quantification parameters have been used to measure angiogenesis such as counting branching points, tubule area, tubule numbers, average tubule length, ...<|separator|>
  115. [115]
    Evaluation of Angiogenesis Assays - MDPI
    May 16, 2019 · A disadvantage of this model is the lack of blood flow, circulating endothelial progenitors, and hormonal factors that a have key role in ...
  116. [116]
    Early evaluation of anti-angiogenic effects with gadolinium(III ...
    Jul 1, 2025 · In this study, we developed a magnetic resonance (MR) probe specifically targeting angiogenesis to facilitate targeted imaging for the early evaluation of anti ...
  117. [117]
    Characterization of multi-cellular dynamics of angiogenesis and ...
    Jul 13, 2018 · Time-lapse intra vital microscopy reveals specific EC migratory behaviour in correlation with blood flow direction and arterio-venous identity.
  118. [118]
    Serial intravital imaging captures dynamic and functional endothelial ...
    Apr 13, 2021 · We aimed to develop a direct visual approach to track the fate and function of single endothelial cells over several days and weeks in the same vascular bed in ...
  119. [119]
    Determination of Microvessel Density by Quantitative Real-time PCR ...
    To address this issue, we did quantitative PCR (qPCR) analyses of several angiogenic and lymphangiogenic markers (VEGFR-2, VEGFR-3, VE-cadherin, P1H12, Tie2, ...
  120. [120]
    Angiogenesis markers (VEGF, soluble receptor of VEGF and ...
    Levels of Vascular Endothelial Growth Factor (VEGF), angiopoietin-1 (Ang-1), and soluble Fms-like tyrosine kinase-1 (sFlt-1) were measured by ELISA in serum ...
  121. [121]
    Angiogenesis-on-a-chip coupled with single-cell RNA sequencing ...
    Jan 3, 2024 · We perform single-cell RNA sequencing of sprouting endothelial cells from our chip to reveal high activation of autophagy in two endothelial cell populations.
  122. [122]
    Zebrafish as an Emerging Model Organism to Study Angiogenesis in ...
    Mar 8, 2016 · In fish, only the embryo and larva are transparent, making studies in adults just as difficult as in mammals. The small size of embryos makes ...
  123. [123]
    Multimodal Investigation of Angiogenesis and Its Prevention by ...
    Jun 25, 2025 · Due to the optical clarity of zebrafish larvae, confocal fluorescence laser scanning microscopy (cLSM) is the gold standard imaging method to ...
  124. [124]
    Systematic Interrogation of Angiogenesis in the Ischemic Mouse ...
    A common approach to investigating ischemia-induced angiogenesis is a mouse model of hind limb ischemia. In 1998, Couffinhal et al reported such a model ...
  125. [125]
    Tumor Microvessel Density as a Potential Predictive Marker for ...
    Microvessel density (MVD) quantified based on CD31-positive endothelial cells in tumor samples assayed by IHC is often used as a surrogate for tumoral ...
  126. [126]
    Quantitative assessment of tumor angiogenesis using real-time ...
    Recent studies have shown that MIP ultrasound imaging allows reliable and accurate quantification and monitoring of tumor angiogenesis during anti-angiogenic ...
  127. [127]
    Falsifying computational models of angiogenesis through ... - bioRxiv
    Aug 22, 2024 · Here we present new dynamic time-lapse videos of tube formation assays and a new analysis pipeline based on ImageJ and demonstrate its use for ...