Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Diabetologia ; 67(6): 1138-1154, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38489029

ABSTRACT

AIMS/HYPOTHESIS: A hallmark chronic complication of type 2 diabetes mellitus is vascular hyperpermeability, which encompasses dysfunction of the cerebrovascular endothelium and the subsequent development of associated cognitive impairment. The present study tested the hypothesis that during type 2 diabetes circulating small extracellular vesicles (sEVs) exhibit phenotypic changes that facilitate pathogenic disruption of the vascular barrier. METHODS: sEVs isolated from the plasma of a mouse model of type 2 diabetes and from diabetic human individuals were characterised for their ability to disrupt the endothelial cell (EC) barrier. The contents of sEVs and their effect on recipient ECs were assessed by proteomics and identified pathways were functionally interrogated with small molecule inhibitors. RESULTS: Using intravital imaging, we found that diabetic mice (Leprdb/db) displayed hyperpermeability of the cerebrovasculature. Enhanced vascular leakiness was recapitulated following i.v. injection of sEVs from diabetic mice into non-diabetic recipient mice. Characterisation of circulating sEV populations from the plasma of diabetic mice and humans demonstrated increased quantity and size of sEVs compared with those isolated from non-diabetic counterparts. Functional experiments revealed that sEVs from diabetic mice or humans induced the rapid and sustained disruption of the EC barrier through enhanced paracellular and transcellular leak but did not induce inflammation. Subsequent sEV proteome and recipient EC phospho-proteome analysis suggested that extracellular vesicles (sEVs) from diabetic mice and humans modulate the MAPK/MAPK kinase (MEK) and Rho-associated protein kinase (ROCK) pathways, cell-cell junctions and actin dynamics. This was confirmed experimentally. Treatment of sEVs with proteinase K or pre-treatment of recipient cells with MEK or ROCK inhibitors reduced the hyperpermeability-inducing effects of circulating sEVs in the diabetic state. CONCLUSIONS/INTERPRETATION: Diabetes is associated with marked increases in the concentration and size of circulating sEVs. The modulation of sEV-associated proteins under diabetic conditions can induce vascular leak through activation of the MEK/ROCK pathway. These data identify a new paradigm by which diabetes can induce hyperpermeability and dysfunction of the cerebrovasculature and may implicate sEVs in the pathogenesis of cognitive decline during type 2 diabetes.


Subject(s)
Capillary Permeability , Diabetes Mellitus, Type 2 , Extracellular Vesicles , Animals , Extracellular Vesicles/metabolism , Mice , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Humans , Male , Diabetes Mellitus, Experimental/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Proteomics , Mice, Inbred C57BL
2.
Nat Commun ; 15(1): 1037, 2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38310100

ABSTRACT

Liver failure causes breakdown of the Blood CNS Barrier (BCB) leading to damages of the Central-Nervous-System (CNS), however the mechanisms whereby the liver influences BCB-integrity remain elusive. One possibility is that the liver secretes an as-yet to be identified molecule(s) that circulate in the serum to directly promote BCB-integrity. To study BCB-integrity, we developed light-sheet imaging for three-dimensional analysis. We show that liver- or muscle-specific knockout of Hfe2/Rgmc induces BCB-breakdown, leading to accumulation of toxic-blood-derived fibrinogen in the brain, lower cortical neuron numbers, and behavioral deficits in mice. Soluble HFE2 competes with its homologue RGMa for binding to Neogenin, thereby blocking RGMa-induced downregulation of PDGF-B and Claudin-5 in endothelial cells, triggering BCB-disruption. HFE2 administration in female mice with experimental autoimmune encephalomyelitis, a model for multiple sclerosis, prevented paralysis and immune cell infiltration by inhibiting RGMa-mediated BCB alteration. This study has implications for the pathogenesis and potential treatment of diseases associated with BCB-dysfunction.


Subject(s)
Blood-Brain Barrier , Encephalomyelitis, Autoimmune, Experimental , Animals , Female , Mice , Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Endothelial Cells/metabolism , Liver/metabolism , Muscles/metabolism
3.
Circ Res ; 127(6): 727-743, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32552404

ABSTRACT

RATIONALE: We previously identified somatic activating mutations in the KRAS (Kirsten rat sarcoma viral oncogene homologue) gene in the endothelium of the majority of human sporadic brain arteriovenous malformations; a disorder characterized by direct connections between arteries and veins. However, whether this genetic abnormality alone is sufficient for lesion formation, as well as how active KRAS signaling contributes to arteriovenous malformations, remains unknown. OBJECTIVE: To establish the first in vivo models of somatic KRAS gain of function in the endothelium in both mice and zebrafish to directly observe the phenotypic consequences of constitutive KRAS activity at a cellular level in vivo, and to test potential therapeutic interventions for arteriovenous malformations. METHODS AND RESULTS: Using both postnatal and adult mice, as well as embryonic zebrafish, we demonstrate that endothelial-specific gain of function mutations in Kras (G12D or G12V) are sufficient to induce brain arteriovenous malformations. Active KRAS signaling leads to altered endothelial cell morphogenesis and increased cell size, ectopic sprouting, expanded vessel lumen diameter, and direct connections between arteries and veins. Furthermore, we show that these lesions are not associated with altered endothelial growth dynamics or a lack of proper arteriovenous identity but instead seem to feature exuberant angiogenic signaling. Finally, we demonstrate that KRAS-dependent arteriovenous malformations in zebrafish are refractory to inhibition of the downstream effector PI3K but instead require active MEK (mitogen-activated protein kinase kinase 1) signaling. CONCLUSIONS: We demonstrate that active KRAS expression in the endothelium is sufficient for brain arteriovenous malformations, even in the setting of uninjured adult vasculature. Furthermore, the finding that KRAS-dependent lesions are reversible in zebrafish suggests that MEK inhibition may represent a promising therapeutic treatment for arteriovenous malformation patients. Graphical Abstract: A graphical abstract is available for this article.


Subject(s)
Endothelial Cells/enzymology , Gain of Function Mutation , Intracranial Arteriovenous Malformations/genetics , MAP Kinase Kinase 1/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Female , Genetic Predisposition to Disease , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Intracranial Arteriovenous Malformations/enzymology , Intracranial Arteriovenous Malformations/pathology , Intracranial Hemorrhages/enzymology , Intracranial Hemorrhages/genetics , Intracranial Hemorrhages/pathology , MAP Kinase Kinase 1/antagonists & inhibitors , Male , Mice, Transgenic , Permeability , Phenotype , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Signal Transduction , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins
4.
Aging Cell ; 19(3): e13103, 2020 03.
Article in English | MEDLINE | ID: mdl-31960578

ABSTRACT

Extracellular vesicles (EVs) have emerged as important regulators of inter-cellular and inter-organ communication, in part via the transfer of their cargo to recipient cells. Although circulating EVs have been previously studied as biomarkers of aging, how circulating EVs change with age and the underlying mechanisms that contribute to these changes are poorly understood. Here, we demonstrate that aging has a profound effect on the circulating EV pool, as evidenced by changes in concentration, size, and cargo. Aging also alters particle function; treatment of cells with EV fractions isolated from old plasma reduces macrophage responses to lipopolysaccharide, increases phagocytosis, and reduces endothelial cell responses to vascular endothelial growth factor compared to cells treated with young EV fractions. Depletion studies indicate that CD63+ particles mediate these effects. Treatment of macrophages with EV-like particles revealed that old particles increased the expression of EV miRNAs in recipient cells. Transfection of cells with microRNA mimics recapitulated some of the effects seen with old EV-like particles. Investigation into the underlying mechanisms using bone marrow transplant studies revealed circulating cell age does not substantially affect the expression of aging-associated circulating EV miRNAs in old mice. Instead, we show that cellular senescence contributes to changes in particle cargo and function. Notably, senolytic treatment of old mice shifted plasma particle cargo and function toward that of a younger phenotype. Collectively, these results demonstrate that senescent cells contribute to changes in plasma EVs with age and suggest a new mechanism by which senescent cells can affect cellular functions throughout the body.


Subject(s)
Aging/blood , Cellular Senescence/genetics , Extracellular Vesicles/metabolism , Aging/genetics , Animals , Biomarkers/metabolism , Bone Marrow Transplantation , Cellular Senescence/drug effects , Extracellular Vesicles/drug effects , Female , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Jurkat Cells , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Phagocytosis/drug effects , Phagocytosis/genetics , Transfection , Vascular Endothelial Growth Factor A/pharmacology
5.
J Cell Mol Med ; 24(1): 632-639, 2020 01.
Article in English | MEDLINE | ID: mdl-31746130

ABSTRACT

The exact molecular mechanisms underlying CCM pathogenesis remain a complicated and controversial topic. Our previous work illustrated an important VEGF signalling loop in KRIT1 depleted endothelial cells. As VEGF is a major mediator of many vascular pathologies, we asked whether the increased VEGF signalling downstream of KRIT1 depletion was involved in CCM formation. Using an inducible KRIT1 endothelial-specific knockout mouse that models CCM, we show that VEGFR2 activation plays a role in CCM pathogenesis in mice. Inhibition of VEGFR2 using a specific inhibitor, SU5416, significantly decreased the number of lesions formed and slightly lowered the average lesion size. Notably, VEGFR2 inhibition also decreased the appearance of lesion haemorrhage as denoted by the presence of free iron in adjacent tissues. The presence of free iron correlated with increased microvessel permeability in both skeletal muscle and brain, which was completely reversed by SU5416 treatment. Finally, we show that VEGFR2 activation is a common downstream consequence of KRIT1, CCM2 and CCM3 loss of function, though the mechanism by which VEGFR2 activation occurs likely varies. Thus, our study clearly shows that VEGFR2 activation downstream of KRIT1 depletion enhances the severity of CCM formation in mice, and suggests that targeting VEGF signalling may be a potential future therapy for CCM.


Subject(s)
Endothelial Cells/pathology , Hemangioma, Cavernous, Central Nervous System/pathology , Hemorrhagic Stroke/pathology , KRIT1 Protein/physiology , Pulmonary Artery/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/metabolism , Hemorrhagic Stroke/etiology , Hemorrhagic Stroke/metabolism , Male , Mice , Mice, Knockout , Pulmonary Artery/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
Proc Natl Acad Sci U S A ; 116(33): 16410-16419, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31350345

ABSTRACT

Atherosclerosis is a chronic inflammatory disease that is driven, in part, by activation of vascular endothelial cells (ECs). In response to inflammatory stimuli, the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway orchestrates the expression of a network of EC genes that contribute to monocyte recruitment and diapedesis across the endothelium. Although many long noncoding RNAs (lncRNAs) are dysregulated in atherosclerosis, they remain poorly characterized, especially in the context of human vascular inflammation. Prior studies have illustrated that lncRNAs can regulate their neighboring protein-coding genes via interaction with protein complexes. We therefore identified and characterized neighboring interleukin-1ß (IL-1ß)-regulated messenger RNA (mRNA)-lncRNA pairs in ECs. We found these pairs to be highly correlated in expression, especially when located within the same chromatin territory. Additionally, these pairs were predominantly divergently transcribed and shared common gene regulatory elements, characterized by active histone marks and NF-κB binding. Further analysis was performed on lncRNA-CCL2, which is transcribed divergently to the gene, CCL2, encoding a proatherosclerotic chemokine. LncRNA-CCL2 and CCL2 showed coordinate up-regulation in response to inflammatory stimuli, and their expression was correlated in unstable symptomatic human atherosclerotic plaques. Knock-down experiments revealed that lncRNA-CCL2 positively regulated CCL2 mRNA levels in multiple primary ECs and EC cell lines. This regulation appeared to involve the interaction of lncRNA-CCL2 with RNA binding proteins, including HNRNPU and IGF2BP2. Hence, our approach has uncovered a network of neighboring mRNA-lncRNA pairs in the setting of inflammation and identified the function of an lncRNA, lncRNA-CCL2, which may contribute to atherogenesis in humans.


Subject(s)
Atherosclerosis/genetics , Chemokine CCL2/genetics , Inflammation/genetics , RNA, Long Noncoding/genetics , Atherosclerosis/pathology , Cell Line , Chromatin/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , Histone Code/genetics , Humans , Inflammation/pathology , Interleukin-1beta/genetics , NF-kappa B/genetics , RNA, Messenger/genetics , RNA-Binding Proteins , Signal Transduction/genetics
7.
J Biol Chem ; 293(22): 8449-8461, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29610276

ABSTRACT

The CCCTC-binding factor (CTCF) is a versatile transcriptional regulator required for embryogenesis, but its function in vascular development or in diseases with a vascular component is poorly understood. Here, we found that endothelial Ctcf is essential for mouse vascular development and limits accumulation of reactive oxygen species (ROS). Conditional knockout of Ctcf in endothelial progenitors and their descendants affected embryonic growth, and caused lethality at embryonic day 10.5 because of defective yolk sac and placental vascular development. Analysis of global gene expression revealed Frataxin (Fxn), the gene mutated in Friedreich's ataxia (FRDA), as the most strongly down-regulated gene in Ctcf-deficient placental endothelial cells. Moreover, in vitro reporter assays showed that Ctcf activates the Fxn promoter in endothelial cells. ROS are known to accumulate in the endothelium of FRDA patients. Importantly, Ctcf deficiency induced ROS-mediated DNA damage in endothelial cells in vitro, and in placental endothelium in vivo Taken together, our findings indicate that Ctcf promotes vascular development and limits oxidative stress in endothelial cells. These results reveal a function for Ctcf in vascular development, and suggest a potential mechanism for endothelial dysfunction in FRDA.


Subject(s)
CCCTC-Binding Factor/physiology , Embryo, Mammalian/pathology , Endothelium, Vascular/pathology , Friedreich Ataxia/pathology , Gene Expression Regulation , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/metabolism , Endothelium, Vascular/metabolism , Female , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Male , Mice , Mice, Knockout , Frataxin
8.
N Engl J Med ; 378(3): 250-261, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29298116

ABSTRACT

BACKGROUND: Sporadic arteriovenous malformations of the brain, which are morphologically abnormal connections between arteries and veins in the brain vasculature, are a leading cause of hemorrhagic stroke in young adults and children. The genetic cause of this rare focal disorder is unknown. METHODS: We analyzed tissue and blood samples from patients with arteriovenous malformations of the brain to detect somatic mutations. We performed exome DNA sequencing of tissue samples of arteriovenous malformations of the brain from 26 patients in the main study group and of paired blood samples from 17 of those patients. To confirm our findings, we performed droplet digital polymerase-chain-reaction (PCR) analysis of tissue samples from 39 patients in the main study group (21 with matching blood samples) and from 33 patients in an independent validation group. We interrogated the downstream signaling pathways, changes in gene expression, and cellular phenotype that were induced by activating KRAS mutations, which we had discovered in tissue samples. RESULTS: We detected somatic activating KRAS mutations in tissue samples from 45 of the 72 patients and in none of the 21 paired blood samples. In endothelial cell-enriched cultures derived from arteriovenous malformations of the brain, we detected KRAS mutations and observed that expression of mutant KRAS (KRASG12V) in endothelial cells in vitro induced increased ERK (extracellular signal-regulated kinase) activity, increased expression of genes related to angiogenesis and Notch signaling, and enhanced migratory behavior. These processes were reversed by inhibition of MAPK (mitogen-activated protein kinase)-ERK signaling. CONCLUSIONS: We identified activating KRAS mutations in the majority of tissue samples of arteriovenous malformations of the brain that we analyzed. We propose that these malformations develop as a result of KRAS-induced activation of the MAPK-ERK signaling pathway in brain endothelial cells. (Funded by the Swiss Cancer League and others.).


Subject(s)
Intracranial Arteriovenous Malformations/genetics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Adult , Cells, Cultured , DNA Mutational Analysis , Exome , Gene Expression , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intracranial Arteriovenous Malformations/etiology , Intracranial Arteriovenous Malformations/pathology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Phosphorylation , Proto-Oncogene Proteins p21(ras)/metabolism
9.
Sci Rep ; 7(1): 8296, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28811547

ABSTRACT

The intracellular scaffold KRIT1/CCM1 is an established regulator of vascular barrier function. Loss of KRIT1 leads to decreased microvessel barrier function and to the development of the vascular disorder Cerebral Cavernous Malformation (CCM). However, how loss of KRIT1 causes the subsequent deficit in barrier function remains undefined. Previous studies have shown that loss of KRIT1 increases the production of reactive oxygen species (ROS) and exacerbates vascular permeability triggered by several inflammatory stimuli, but not TNF-α. We now show that endothelial ROS production directly contributes to the loss of barrier function in KRIT1 deficient animals and cells, as targeted antioxidant enzymes reversed the increase in permeability in KRIT1 heterozygous mice as shown by intravital microscopy. Rescue of the redox state restored responsiveness to TNF-α in KRIT1 deficient arterioles, but not venules. In vitro, KRIT1 depletion increased endothelial ROS production via NADPH oxidase signaling, up-regulated Nox4 expression, and promoted NF-κB dependent promoter activity. Recombinant yeast avenanthramide I, an antioxidant and inhibitor of NF-κB signaling, rescued barrier function in KRIT1 deficient cells. However, KRIT1 depletion blunted ROS production in response to TNF-α. Together, our data indicate that ROS signaling is critical for the loss of barrier function following genetic deletion of KRIT1.


Subject(s)
Endothelium/metabolism , KRIT1 Protein/deficiency , NADPH Oxidases/metabolism , Oxidation-Reduction , Signal Transduction , Animals , Antioxidants/metabolism , Capillary Permeability/drug effects , Capillary Permeability/genetics , Gene Expression Regulation , KRIT1 Protein/genetics , KRIT1 Protein/metabolism , Mice , Mice, Knockout , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
PLoS One ; 11(9): e0162338, 2016.
Article in English | MEDLINE | ID: mdl-27612188

ABSTRACT

The phosphoinositide-specific phospholipase C, PLCε, is a unique signaling protein with known roles in regulating cardiac myocyte growth, astrocyte inflammatory signaling, and tumor formation. PLCε is also expressed in endothelial cells, however its role in endothelial regulation is not fully established. We show that endothelial cells of multiple origins, including human pulmonary artery (HPAEC), human umbilical vein (HUVEC), and immortalized brain microvascular (hCMEC/D3) endothelial cells, express PLCε. Knockdown of PLCε in arterial endothelial monolayers decreased the effectiveness of the endothelial barrier. Concomitantly, RhoA activity and stress fiber formation were increased. PLCε-deficient arterial endothelial cells also exhibited decreased Rap1-GTP levels, which could be restored by activation of the Rap1 GEF, Epac, to rescue the increase in monolayer leak. Reintroduction of PLCε rescued monolayer leak with both the CDC25 GEF domain and the lipase domain of PLCε required to fully activate Rap1 and to rescue endothelial barrier function. Finally, we demonstrate that the barrier promoting effects PLCε are dependent on Rap1 signaling through the Rap1 effector, KRIT1, which we have previously shown is vital for maintaining endothelial barrier stability. Thus we have described a novel role for PLCε PIP2 hydrolytic and Rap GEF activities in arterial endothelial cells, where PLCε-dependent activation of Rap1/KRIT1 signaling promotes endothelial barrier stability.


Subject(s)
Endothelial Cells/metabolism , Phosphoinositide Phospholipase C/metabolism , Animals , Cell Line , Endothelium, Vascular/physiology , Gene Knockdown Techniques , Humans , KRIT1 Protein , Lipase/metabolism , Microtubule-Associated Proteins/metabolism , Phosphoinositide Phospholipase C/genetics , Proto-Oncogene Proteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , cdc25 Phosphatases/metabolism , rhoA GTP-Binding Protein/metabolism
11.
J Biol Chem ; 289(47): 33054-65, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25320085

ABSTRACT

Disruption of endothelial cell-cell contact is a key event in many cardiovascular diseases and a characteristic of pathologically activated vascular endothelium. The CCM (cerebral cavernous malformation) family of proteins (KRIT1 (Krev-interaction trapped 1), PDCD10, and CCM2) are critical regulators of endothelial cell-cell contact and vascular homeostasis. Here we show novel regulation of vascular endothelial growth factor (VEGF) signaling in KRIT1-depleted endothelial cells. Loss of KRIT1 and PDCD10, but not CCM2, increases nuclear ß-catenin signaling and up-regulates VEGF-A protein expression. In KRIT1-depleted cells, increased VEGF-A levels led to increased VEGF receptor 2 (VEGFR2) activation and subsequent alteration of cytoskeletal organization, migration, and barrier function and to in vivo endothelial permeability in KRIT1-deficient animals. VEGFR2 activation also increases ß-catenin phosphorylation but is only partially responsible for KRIT1 depletion-dependent disruption of cell-cell contacts. Thus, VEGF signaling contributes to modifying endothelial function in KRIT1-deficient cells and microvessel permeability in Krit1(+/-) mice; however, VEGF signaling is likely not the only contributor to disrupted endothelial cell-cell contacts in the absence of KRIT1.


Subject(s)
Endothelial Cells/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Capillary Permeability , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cattle , Cell Communication , Cell Membrane Permeability , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , KRIT1 Protein , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , RNA Interference , Vascular Endothelial Growth Factor Receptor-2/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...