Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Van N. Pham is active.

Publication


Featured researches published by Van N. Pham.


Nature Medicine | 2012

ApoB-containing lipoproteins regulate angiogenesis by modulating expression of VEGF receptor 1.

Inbal Avraham-Davidi; Yona Ely; Van N. Pham; Daniel Castranova; Moshe Grunspan; Guy Malkinson; Liron Gibbs-Bar; Oded Mayseless; Gabriella Allmog; Brigid Lo; Carmen M. Warren; Thomas T. Chen; Josette M. Ungos; Kameha R. Kidd; Kenna Shaw; Ilana Rogachev; Wuzhou Wan; Philip M Murphy; Steven A. Farber; Liran Carmel; Gregory S. Shelness; M. Luisa Iruela-Arispe; Brant M. Weinstein; Karina Yaniv

Despite the clear major contribution of hyperlipidemia to the prevalence of cardiovascular disease in the developed world, the direct effects of lipoproteins on endothelial cells have remained obscure and are under debate. Here we report a previously uncharacterized mechanism of vessel growth modulation by lipoprotein availability. Using a genetic screen for vascular defects in zebrafish, we initially identified a mutation, stalactite (stl), in the gene encoding microsomal triglyceride transfer protein (mtp), which is involved in the biosynthesis of apolipoprotein B (ApoB)-containing lipoproteins. By manipulating lipoprotein concentrations in zebrafish, we found that ApoB negatively regulates angiogenesis and that it is the ApoB protein particle, rather than lipid moieties within ApoB-containing lipoproteins, that is primarily responsible for this effect. Mechanistically, we identified downregulation of vascular endothelial growth factor receptor 1 (VEGFR1), which acts as a decoy receptor for VEGF, as a key mediator of the endothelial response to lipoproteins, and we observed VEGFR1 downregulation in hyperlipidemic mice. These findings may open new avenues for the treatment of lipoprotein-related vascular disorders.


American Journal of Human Genetics | 2011

Loss of BRCC3 Deubiquitinating Enzyme Leads to Abnormal Angiogenesis and Is Associated with Syndromic Moyamoya

Snaigune Miskinyte; Matthew G. Butler; Dominique Hervé; Catherine Sarret; Marc Nicolino; Jacob D. Petralia; Françoise Bergametti; Minh Arnould; Van N. Pham; Aniket V. Gore; Konstantinos Spengos; Steven Gazal; Gary K. Steinberg; Brant M. Weinstein; Elisabeth Tournier-Lasserve

Moyamoya is a cerebrovascular angiopathy characterized by a progressive stenosis of the terminal part of the intracranial carotid arteries and the compensatory development of abnormal and fragile collateral vessels, also called moyamoya vessels, leading to ischemic and hemorrhagic stroke. Moyamoya angiopathy can either be the sole manifestation of the disease (moyamoya disease) or be associated with various conditions, including neurofibromatosis, Down syndrome, TAAD (autosomal-dominant thoracic aortic aneurysm), and radiotherapy of head tumors (moyamoya syndromes). Its prevalence is ten times higher in Japan than in Europe, and an estimated 6%-12% of moyamoya disease is familial in Japan. The pathophysiological mechanisms of this condition remain obscure. Here, we report on three unrelated families affected with an X-linked moyamoya syndrome characterized by the association of a moyamoya angiopathy, short stature, and a stereotyped facial dysmorphism. Other symptoms include an hypergonadotropic hypogonadism, hypertension, dilated cardiomyopathy, premature coronary heart disease, premature hair graying, and early bilateral acquired cataract. We show that this syndromic moyamoya is caused by Xq28 deletions removing MTCP1/MTCP1NB and BRCC3. We also show that brcc3 morphant zebrafish display angiogenesis defects that are rescued by endothelium-specific expression of brcc3. Altogether, these data strongly suggest that BRCC3, a deubiquitinating enzyme that is part of the cellular BRCA1 and BRISC complexes, is an important player in angiogenesis and that BRCC3 loss-of-function mutations are associated with moyamoya angiopathy.


Development | 2011

Assembly and patterning of the vascular network of the vertebrate hindbrain

Misato Fujita; Young R. Cha; Van N. Pham; Atsuko Sakurai; Beth L. Roman; J. Silvio Gutkind; Brant M. Weinstein

The cranial vasculature is essential for the survival and development of the central nervous system and is important in stroke and other brain pathologies. Cranial vessels form in a reproducible and evolutionarily conserved manner, but the process by which these vessels assemble and acquire their stereotypic patterning remains unclear. Here, we examine the stepwise assembly and patterning of the vascular network of the zebrafish hindbrain. The major artery supplying the hindbrain, the basilar artery, runs along the ventral keel of the hindbrain in all vertebrates. We show that this artery forms by a novel process of medial sprouting and migration of endothelial cells from a bilateral pair of primitive veins, the primordial hindbrain channels. Subsequently, a second wave of dorsal sprouting from the primordial hindbrain channels gives rise to angiogenic central arteries that penetrate into and innervate the hindbrain. The chemokine receptor cxcr4a is expressed in migrating endothelial cells of the primordial hindbrain channels, whereas its ligand cxcl12b is expressed in the hindbrain neural keel immediately adjacent to the assembling basilar artery. Knockdown of either cxcl12b or cxcr4a results in defects in basilar artery formation, showing that the assembly and patterning of this crucial artery depends on chemokine signaling.


Developmental Dynamics | 2001

Isolation and expression analysis of three zebrafish angiopoietin genes

Van N. Pham; Beth L. Roman; Brant M. Weinstein

The Tie1 and Tie2 receptor tyrosine kinases and the Tie2 ligands, the angiopoietins, play critical roles in vertebrate vascular embryogenesis, helping to mediate the interaction between endothelial cells and the pericytes or vascular smooth muscle cells that envelop and support them. We have obtained full‐length cDNA sequences for zebrafish orthologs of angiopoietin‐1 (ang1), angiopoietin‐2 (ang2), and angiopoietin‐like‐3 (angptl3). Ang1 is expressed in head ventral mesenchyme, in the ventromedial region of somites, in mesenchyme surrounding trunk axial vessels, and in the hypochord, a transient embryonic structure of endodermal origin that has been implicated in dorsal aorta assembly in both zebrafish and Xenopus. Ang2 is expressed in head and anterior trunk ventral mesenchyme and the developing pronephric glomeruli. Angptl3 is expressed in the yolk syncytial layer.


Developmental Biology | 2008

Loss of unc45a precipitates arteriovenous shunting in the aortic arches

Matthew John Anderson; Van N. Pham; Andreas M. Vogel; Brant M. Weinstein; Beth L. Roman

Aortic arch malformations are common congenital disorders that are frequently of unknown etiology. To gain insight into the factors that guide branchial aortic arch development, we examined the process by which these vessels assemble in wild type zebrafish embryos and in kurzschluss(tr12) (kus(tr12)) mutants. In wild type embryos, each branchial aortic arch first appears as an island of angioblasts in the lateral pharyngeal mesoderm, then elaborates by angiogenesis to connect to the lateral dorsal aorta and ventral aorta. In kus(tr12) mutants, angioblast formation and initial sprouting are normal, but aortic arches 5 and 6 fail to form a lumenized connection to the lateral dorsal aorta. Blood enters these blind-ending vessels from the ventral aorta, distending the arteries and precipitating fusion with an adjacent vein. This arteriovenous malformation (AVM), which shunts nearly all blood directly back to the heart, is not exclusively genetically programmed, as its formation correlates with blood flow and aortic arch enlargement. By positional cloning, we have identified a nonsense mutation in unc45a in kus(tr12) mutants. Our results are the first to ascribe a role for Unc45a, a putative myosin chaperone, in vertebrate development, and identify a novel mechanism by which an AVM can form.


Developmental Biology | 2014

SoxF factors and Notch regulate nr2f2 gene expression during venous differentiation in zebrafish

Matthew R. Swift; Van N. Pham; Daniel Castranova; Kameha Bell; Richard J. Poole; Brant M. Weinstein

Initial embryonic determination of artery or vein identity is regulated by genetic factors that work in concert to specify the endothelial cell׳s (EC) fate, giving rise to two structurally unique components of the circulatory loop. The Shh/VEGF/Notch pathway is critical for arterial specification, while the orphan receptor nr2f2 (COUP-TFII) has been implicated in venous specification. Studies in mice have shown that nr2f2 is expressed in venous but not arterial ECs, and that it preferentially induces markers of venous cell fate. We have examined the role of nr2f2 during early arterial-venous development in the zebrafish trunk. We show that expression of a subset of markers of venous endothelial identity requires nr2f2, while the expression of nr2f2 itself requires sox7 and sox18 gene function. However, while sox7 and sox18 are expressed in both the cardinal vein and the dorsal aorta during early trunk development, nr2f2 is expressed only in the cardinal vein. We show that Notch signaling activity present in the dorsal aorta suppresses expression of nr2f2, restricting nr2f2-dependent promotion of venous differentiation to the cardinal vein.


Developmental Dynamics | 2006

fused-somites–like mutants exhibit defects in trunk vessel patterning†

Kenna M. Shaw; Daniel Castranova; Van N. Pham; Makoto Kamei; Kameha R. Kidd; Brigid Lo; Jesus Torres-Vasquez; Alexander Ruby; Brant M. Weinstein

We identified four mutants in two distinct loci exhibiting similar trunk vascular patterning defects in an F3 genetic screen for zebrafish vascular mutants. Initial vasculogenesis is not affected in these mutants, with proper specification and differentiation of endothelial cells. However, all four display severe defects in the growth and patterning of angiogenic vessels in the trunk, with ectopic branching and disoriented migration of intersegmental vessels. The four mutants are allelic to previously characterized mutants at the fused‐somites (fss) and beamter (bea) loci, and they exhibit comparable defects in trunk somite boundary formation. The fss locus has been shown to correspond to tbx24; we show here that bea mutants are defective in the zebrafish dlC gene. Somitic expression of known vascular guidance factors efnb2a, sema3a1, and sema3a2 is aberrantly patterned in fss and bea mutants, suggesting that the vascular phenotype is due to loss of proper guidance cues provided by these factors. Developmental Dynamics 235:1753–1760, 2006. Published 2006 Wiley‐Liss, Inc.


Blood | 2012

CDP-diacylglycerol synthetase-controlled phosphoinositide availability limits VEGFA signaling and vascular morphogenesis

Weijun Pan; Van N. Pham; Amber N. Stratman; Daniel Castranova; Makoto Kamei; Kameha R. Kidd; Brigid Lo; Kenna M. Shaw; Jesús Torres-Vázquez; Constantinos M. Mikelis; Gutkind Js; George E. Davis; Brant M. Weinstein

Understanding the mechanisms that regulate angiogenesis and translating these into effective therapies are of enormous scientific and clinical interests. In this report, we demonstrate the central role of CDP-diacylglycerol synthetase (CDS) in the regulation of VEGFA signaling and angiogenesis. CDS activity maintains phosphoinositide 4,5 bisphosphate (PIP2) availability through resynthesis of phosphoinositides, whereas VEGFA, mainly through phospholipase Cγ1, consumes PIP2 for signal transduction. Loss of CDS2, 1 of 2 vertebrate CDS enzymes, results in vascular-specific defects in zebrafish in vivo and failure of VEGFA-induced angiogenesis in endothelial cells in vitro. Absence of CDS2 also results in reduced arterial differentiation and reduced angiogenic signaling. CDS2 deficit-caused phenotypes can be successfully rescued by artificial elevation of PIP2 levels, and excess PIP2 or increased CDS2 activity can promote excess angiogenesis. These results suggest that availability of CDS-controlled resynthesis of phosphoinositides is essential for angiogenesis.


Development | 2015

Single-cell analysis of endothelial morphogenesis in vivo.

Jianxin A. Yu; Daniel Castranova; Van N. Pham; Brant M. Weinstein

Vessel formation has been extensively studied at the tissue level, but the difficulty in imaging the endothelium with cellular resolution has hampered study of the morphogenesis and behavior of endothelial cells (ECs) in vivo. We are using endothelial-specific transgenes and high-resolution imaging to examine single ECs in zebrafish. By generating mosaics with transgenes that simultaneously mark endothelial nuclei and membranes we are able to definitively identify and study the morphology and behavior of individual ECs during vessel sprouting and lumen formation. Using these methods, we show that developing trunk vessels are composed of ECs of varying morphology, and that single-cell analysis can be used to quantitate alterations in morphology and dynamics in ECs that are defective in proper guidance and patterning. Finally, we use single-cell analysis of intersegmental vessels undergoing lumen formation to demonstrate the coexistence of seamless transcellular lumens and single or multicellular enclosed lumens with autocellular or intercellular junctions, suggesting that heterogeneous mechanisms contribute to vascular lumen formation in vivo. The tools that we have developed for single EC analysis should facilitate further rigorous qualitative and quantitative analysis of EC morphology and behavior in vivo. Highlighted article: Following the behaviour of single cells during vessel formation at high spatio-temporal resolution provides insights into endothelial cell migration and lumen formation.


Development | 2016

Reck enables cerebrovascular development by promoting canonical Wnt signaling

Florian Ulrich; Jorge Carretero-Ortega; Javier Menéndez; Carlos Narvaez; Belinda Sun; Eva Lancaster; Valerie Pershad; Sean Trzaska; Evelyn Véliz; Makoto Kamei; Andrew Prendergast; Kameha R. Kidd; Kenna M. Shaw; Daniel Castranova; Van N. Pham; Brigid Lo; Benjamin L. Martin; David W. Raible; Brant M. Weinstein; Jesús Torres-Vázquez

The cerebral vasculature provides the massive blood supply that the brain needs to grow and survive. By acquiring distinctive cellular and molecular characteristics it becomes the blood-brain barrier (BBB), a selectively permeable and protective interface between the brain and the peripheral circulation that maintains the extracellular milieu permissive for neuronal activity. Accordingly, there is great interest in uncovering the mechanisms that modulate the formation and differentiation of the brain vasculature. By performing a forward genetic screen in zebrafish we isolated no food for thought (nft y72), a recessive late-lethal mutant that lacks most of the intracerebral central arteries (CtAs), but not other brain blood vessels. We found that the cerebral vascularization deficit of nft y72 mutants is caused by an inactivating lesion in reversion-inducing cysteine-rich protein with Kazal motifs [reck; also known as suppressor of tumorigenicity 15 protein (ST15)], which encodes a membrane-anchored tumor suppressor glycoprotein. Our findings highlight Reck as a novel and pivotal modulator of the canonical Wnt signaling pathway that acts in endothelial cells to enable intracerebral vascularization and proper expression of molecular markers associated with BBB formation. Additional studies with cultured endothelial cells suggest that, in other contexts, Reck impacts vascular biology via the vascular endothelial growth factor (VEGF) cascade. Together, our findings have broad implications for both vascular and cancer biology. Summary: A zebrafish screen identifies reck as a key modulator of Wnt signaling required in the brain endothelium for intracerebral vascularisation and proper expression of barriergenesis markers.

Collaboration


Dive into the Van N. Pham's collaboration.

Top Co-Authors

Avatar

Brant M. Weinstein

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Daniel Castranova

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Beth L. Roman

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Brigid Lo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Makoto Kamei

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kameha R. Kidd

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kenna M. Shaw

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Matthew G. Butler

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Matthew R. Swift

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge