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










Publication year range
1.
Curr Opin Hematol ; 26(3): 161-169, 2019 05.
Article in English | MEDLINE | ID: mdl-30855334

ABSTRACT

PURPOSE OF REVIEW: The TGFß (transforming growth factor ß) superfamily - a large group of structurally related and evolutionarily conserved proteins - profoundly shapes and organizes the vasculature during normal development and adult homeostasis. Mutations inactivating several of its ligands, receptors, or signal transducers set off hereditary hemorrhagic telangiectasia (HHT), a disorder that causes capillary networks to form incorrectly. Drosha, an essential microRNA-processing enzyme, also interfaces with TGFß signal transducers, but its involvement in vascular conditions had not been tested until recently. This review summarizes current evidence that links mutations of Drosha to HHT. RECENT FINDINGS: Genetic studies have revealed that rare missense mutations in the Drosha gene occur more commonly among HHT patients than in healthy people. Molecular analyses also indicated that Drosha enzymes with HHT-associated mutations generate microRNAs less efficiently than their wild-type counterpart when stimulated by TGFß ligands. In zebrafish or mouse, mutant Drosha proteins cause the formation of dilated, leaky blood vessels deprived of capillaries, similar to those typically found in patients with HHT. SUMMARY: Recent evidence suggests that Drosha-mediated microRNA biogenesis contributes significantly to the control of vascular development and homeostasis by TGFß. Loss or reduction of Drosha function may predispose carriers to HHT and possibly other vascular diseases.


Subject(s)
MicroRNAs , Mutation, Missense , Ribonuclease III , Signal Transduction , Telangiectasia, Hereditary Hemorrhagic , Animals , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Telangiectasia, Hereditary Hemorrhagic/enzymology , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Zebrafish
2.
Arterioscler Thromb Vasc Biol ; 38(5): 1216-1229, 2018 05.
Article in English | MEDLINE | ID: mdl-29449337

ABSTRACT

OBJECTIVE: ALK1 (activin-receptor like kinase 1) is an endothelial cell-restricted receptor with high affinity for BMP (bone morphogenetic protein) 9 TGF-ß (transforming growth factor-ß) family member. Loss-of-function mutations in ALK1 cause a subtype of hereditary hemorrhagic telangiectasia-a rare disease characterized by vasculature malformations. Therapeutic strategies are aimed at reducing potential complications because of vascular malformations, but currently, there is no curative treatment for hereditary hemorrhagic telangiectasia. APPROACH AND RESULTS: In this work, we report that a reduction in ALK1 gene dosage (heterozygous ALK1+/- mice) results in enhanced retinal endothelial cell proliferation and vascular hyperplasia at the sprouting front. We found that BMP9/ALK1 represses VEGF (vascular endothelial growth factor)-mediated PI3K (phosphatidylinositol 3-kinase) by promoting the activity of the PTEN (phosphatase and tensin homolog). Consequently, loss of ALK1 function in endothelial cells results in increased activity of the PI3K pathway. These results were confirmed in cutaneous telangiectasia biopsies of patients with hereditary hemorrhagic telangiectasia 2, in which we also detected an increase in endothelial cell proliferation linked to an increase on the PI3K pathway. In mice, genetic and pharmacological inhibition of PI3K is sufficient to abolish the vascular hyperplasia of ALK1+/- retinas and in turn normalize the vasculature. CONCLUSIONS: Overall, our results indicate that the BMP9/ALK1 hub critically mediates vascular quiescence by limiting PI3K signaling and suggest that PI3K inhibitors could be used as novel therapeutic agents to treat hereditary hemorrhagic telangiectasia.


Subject(s)
Activin Receptors, Type II/genetics , Activin Receptors, Type I/genetics , Endothelial Cells/enzymology , Mutation , Neovascularization, Pathologic , Phosphatidylinositol 3-Kinase/metabolism , Retinal Telangiectasis/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Activin Receptors, Type I/deficiency , Angiogenesis Inhibitors/pharmacology , Animals , Case-Control Studies , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Activation , Gene Deletion , Genetic Predisposition to Disease , Growth Differentiation Factor 2/pharmacology , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Hyperplasia , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Retinal Telangiectasis/drug therapy , Retinal Telangiectasis/enzymology , Retinal Telangiectasis/pathology , Signal Transduction , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Telangiectasia, Hereditary Hemorrhagic/enzymology , Telangiectasia, Hereditary Hemorrhagic/pathology , Vascular Endothelial Growth Factor A/pharmacology
3.
Arterioscler Thromb Vasc Biol ; 36(4): 707-17, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26821948

ABSTRACT

OBJECTIVE: To determine the role of Gja5 that encodes for the gap junction protein connexin40 in the generation of arteriovenous malformations in the hereditary hemorrhagic telangiectasia type 2 (HHT2) mouse model. APPROACH AND RESULTS: We identified GJA5 as a target gene of the bone morphogenetic protein-9/activin receptor-like kinase 1 signaling pathway in human aortic endothelial cells and importantly found that connexin40 levels were particularly low in a small group of patients with HHT2. We next took advantage of the Acvrl1(+/-) mutant mice that develop lesions similar to those in patients with HHT2 and generated Acvrl1(+/-); Gja5(EGFP/+) mice. Gja5 haploinsufficiency led to vasodilation of the arteries and rarefaction of the capillary bed in Acvrl1(+/-) mice. At the molecular level, we found that reduced Gja5 in Acvrl1(+/-) mice stimulated the production of reactive oxygen species, an important mediator of vessel remodeling. To normalize the altered hemodynamic forces in Acvrl1(+/-); Gja5(EGFP/+) mice, capillaries formed transient arteriovenous shunts that could develop into large malformations when exposed to environmental insults. CONCLUSIONS: We identified GJA5 as a potential modifier gene for HHT2. Our findings demonstrate that Acvrl1 haploinsufficiency combined with the effects of modifier genes that regulate vessel caliber is responsible for the heterogeneity and severity of the disease. The mouse models of HHT have led to the proposal that 3 events-heterozygosity, loss of heterozygosity, and angiogenic stimulation-are necessary for arteriovenous malformation formation. Here, we present a novel 3-step model in which pathological vessel caliber and consequent altered blood flow are necessary events for arteriovenous malformation development.


Subject(s)
Activin Receptors, Type II/metabolism , Activin Receptors, Type I/metabolism , Arteriovenous Malformations/enzymology , Connexins/metabolism , Endothelial Cells/enzymology , Retinal Vessels/enzymology , Telangiectasia, Hereditary Hemorrhagic/enzymology , Activin Receptors, Type I/genetics , Activin Receptors, Type II/genetics , Animals , Arteriovenous Malformations/genetics , Arteriovenous Malformations/pathology , Cells, Cultured , Connexins/genetics , Disease Models, Animal , Genetic Predisposition to Disease , Haploinsufficiency , Humans , Mice, Mutant Strains , Mice, Transgenic , Neovascularization, Pathologic , Phenotype , RNA Interference , Reactive Oxygen Species/metabolism , Retinal Vessels/pathology , Signal Transduction , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Transfection , Vascular Remodeling , Gap Junction alpha-5 Protein
4.
Med Hypotheses ; 83(3): 302-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24986705

ABSTRACT

BACKGROUND: Hereditary hemorrhagic telangiectasia (HHT) and hepatopulmonary syndrome are disorders characterized by the development of multiple pulmonary arteriovenous malformations (PAVM). PRESENTATION OF THE HYPOTHESIS: COX2 may be at the origin of a cascade of pro inflammatory events to favour angiogenesis and PAVM development. TESTING THE HYPOTHESIS: HHT and hepatopulmonary syndrome mouse models may be used to show its effects on PAVM formation. Anti COX-2 therapy could also be tested in human individuals, particularly in patients presenting a hepatopulmonary syndrome or HHT with small PAVM. IMPLICATION OF THE HYPOTHESIS: PAVMs are one of the main causes of morbidity in patients presenting with HHT disease, owing to the risks of rupture as well as paradoxical embolism exposing to stroke and/or cerebral abscess. Percutaneous embolization has become the treatment of choice of PAVM. Anti COX2 may prevent from PAVM development and subsequent related complications and avoid either surgery and/or percutaneous embolization and thus subsequent related complication.


Subject(s)
Cyclooxygenase 2/metabolism , Hepatic Veins/physiopathology , Hepatopulmonary Syndrome/blood , Hepatopulmonary Syndrome/physiopathology , Liver/blood supply , Liver/enzymology , Telangiectasia, Hereditary Hemorrhagic/enzymology , Animals , Arteriovenous Fistula/physiopathology , Disease Models, Animal , Embolization, Therapeutic , Humans , Hypoxia/physiopathology , Inflammation/physiopathology , Liver/physiopathology , Mice , Neovascularization, Pathologic , Pulmonary Artery/abnormalities , Pulmonary Artery/physiopathology , Pulmonary Veins/abnormalities , Pulmonary Veins/physiopathology , Vascular Endothelial Growth Factor A/metabolism
5.
Mol Cell Biochem ; 373(1-2): 247-57, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23124896

ABSTRACT

Hereditary haemorrhagic telangiectasia (HHT) is an autosomal dominant disease characterised by vascular dysplasia and increased bleeding that affect 1 in 5,000 people world-wide. Pathology is linked to mutations in genes encoding components of the heteromeric transforming growth factor-beta receptor (TGF-beta) and SMAD signalling pathway. Indeed HHT1 and HHT2 result from mutations in the genes encoding endoglin and activin-like kinase 1 (ALK1), TGF-beta receptor components. However, the fundamental cellular defects underlying HHT is poorly understood. Previously using confocal microscopy and N-glycosylation analysis, we found evidence that defective trafficking of endoglin from the endoplasmic reticulum (ER) to the plasma membrane is a mechanism underlying HHT1 in some patients. In this study, we used confocal microscopy to investigate whether a similar mechanism contributes to HHT2 pathology. To do this we expressed wild-type ALK1 and a number of HHT2 patient mutant variants as C-terminally tagged EGFP fusion proteins and tested their localisation in HeLa cells. We found that wild-type ALK1-EGFP was targeted predominantly to the plasma membrane, as evidenced by its colocalisation with the co-expressed HA-tagged endoglin. However, we found that in the majority of cases analysed the HHT2 patient mutant protein was retained within the ER as indicated by their colocalisation with the ER resident marker (calnexin) and lack of colocalisation with cell surface associated HA-endoglin. We conclude that defective trafficking and retention in the ER of mutant ALK1 protein is a possible mechanism of HHT2 in some patients.


Subject(s)
Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Endoplasmic Reticulum/enzymology , Telangiectasia, Hereditary Hemorrhagic/genetics , Calnexin/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Mutation, Missense , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Telangiectasia, Hereditary Hemorrhagic/enzymology
6.
Arterioscler Thromb Vasc Biol ; 33(2): 305-10, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23241407

ABSTRACT

OBJECTIVE: Vessels in brain arteriovenous malformations are prone to rupture. The underlying pathogenesis is not clear. Hereditary hemorrhagic telangiectasia type 2 patients with activin receptor-like kinase 1 (Alk1) mutation have a higher incidence of brain arteriovenous malformation than the general population. We tested the hypothesis that vascular endothelial growth factor impairs vascular integrity in the Alk1-deficient brain through reduction of mural cell coverage. METHODS AND RESULTS: Adult Alk1(1f/2f) mice (loxP sites flanking exons 4-6) and wild-type mice were injected with 2×10(7) PFU adenovious-cre recombinase and 2×10(9) genome copies of adeno-associated virus-vascular endothelial growth factor to induce focal homozygous Alk1 deletion (in Alk1(1f/2f) mice) and angiogenesis. Brain vessels were analyzed 8 weeks later. Compared with wild-type mice, the Alk1-deficient brain had more fibrin (99±30×10(3) pixels/mm(2) versus 40±13×10(3); P=0.001), iron deposition (508±506 pixels/mm(2) versus 6±49; P=0.04), and Iba1(+) microglia/macrophage infiltration (888±420 Iba1(+) cells/mm(2) versus 240±104 Iba1(+); P=0.001) after vascular endothelial growth factor stimulation. In the angiogenic foci, the Alk1-deficient brain had more α-smooth muscle actin negative vessels (52±9% versus 12±7%, P<0.001), fewer vascular-associated pericytes (503±179/mm(2) versus 931±115, P<0.001), and reduced platelet-derived growth factor receptor-ß expression. CONCLUSIONS: Reduction of mural cell coverage in response to vascular endothelial growth factor stimulation is a potential mechanism for the impairment of vessel wall integrity in hereditary hemorrhagic telangiectasia type 2-associated brain arteriovenous malformation.


Subject(s)
Activin Receptors, Type I/deficiency , Blood Vessels/enzymology , Brain/blood supply , Neovascularization, Pathologic , Pericytes/enzymology , Telangiectasia, Hereditary Hemorrhagic/enzymology , Vascular Endothelial Growth Factor A/metabolism , Actins/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type II , Animals , Becaplermin , Blood Vessels/pathology , Dependovirus/genetics , Disease Models, Animal , Fibrin/metabolism , Gene Transfer Techniques , Genetic Vectors , Iron/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Pericytes/pathology , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Vascular Endothelial Growth Factor A/genetics
7.
PLoS One ; 6(10): e26431, 2011.
Article in English | MEDLINE | ID: mdl-22028876

ABSTRACT

Activin A receptor, type II-like kinase 1 (also called ALK1), is a serine-threonine kinase predominantly expressed on endothelial cells surface. Mutations in its ACVRL1 encoding gene (12q11-14) cause type 2 Hereditary Haemorrhagic Telangiectasia (HHT2), an autosomal dominant multisystem vascular dysplasia. The study of the structural effects of mutations is crucial to understand their pathogenic mechanism. However, while an X-ray structure of ALK1 intracellular domain has recently become available (PDB ID: 3MY0), structure determination of ALK1 ectodomain (ALK1(EC)) has been elusive so far. We here describe the building of a homology model for ALK1(EC), followed by an extensive bioinformatic analysis, based on a set of 38 methods, of the effect of missense mutations at the sequence and structural level. ALK1(EC) potential interaction mode with its ligand BMP9 was then predicted combining modelling and docking data. The calculated model of the ALK1(EC) allowed mapping and a preliminary characterization of HHT2 associated mutations. Major structural changes and loss of stability of the protein were predicted for several mutations, while others were found to interfere mainly with binding to BMP9 or other interactors, like Endoglin (CD105), whose encoding ENG gene (9q34) mutations are known to cause type 1 HHT. This study gives a preliminary insight into the potential structure of ALK1(EC) and into the structural effects of HHT2 associated mutations, which can be useful to predict the potential effect of each single mutation, to devise new biological experiments and to interpret the biological significance of new mutations, private mutations, or non-synonymous polymorphisms.


Subject(s)
Activin Receptors, Type II/chemistry , Activin Receptors, Type II/genetics , Computational Biology , Mutation, Missense , Activin Receptors, Type II/metabolism , Amino Acid Sequence , Conserved Sequence , DNA Mutational Analysis , Enzyme Stability , Growth Differentiation Factor 2 , Growth Differentiation Factors/chemistry , Growth Differentiation Factors/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Static Electricity , Telangiectasia, Hereditary Hemorrhagic/enzymology , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology
8.
Lab Invest ; 87(7): 670-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17530030

ABSTRACT

Activin receptor-like kinase 1 (ALK1) is involved in the pathogenesis of hereditary hemorrhagic telangiectasia type II (HHT2) and pulmonary arterial hypertension. We have previously shown that Alk1 is predominantly expressed in the arterial endothelium and plays a pivotal role in the formation of embryonic blood vessels. At present, however, little is known about the precise expression pattern and function of ALK1 during extra-embryonic vascular development. Using previously generated lacZ reporter lines, we sought to examine the expression pattern and role of Alk1 during placental development in mice. Alk1 expression was restricted to endothelial cells of fetal vessels from the emergence of chorioallantoic fusion to the late gestational period, and no detectable Alk1 expression was observed in syncytiotrophoblasts or spongiotrophoblasts. Predominant arterial expression was observed in the umbilical and fetal placental vessels as well as in embryonic vessels. Morphological analysis of Alk1-null embryos indicates that Alk1 is essential for the development of distinct umbilical arteries and veins. The invasion of chorioallantoic mesoderm into the forming labyrinth layer was largely unaffected in the Alk1-null placenta, but chorioallantoic vessels appeared to be severely dilated and fused. Results from this study provide valuable information regarding the role of ALK1 in the development of placental vasculature as well as insights into the pathogenesis of HHT.


Subject(s)
Activin Receptors, Type I/metabolism , Arteries/enzymology , Endothelium, Vascular/enzymology , Neovascularization, Physiologic , Placenta/blood supply , Placentation/physiology , Telangiectasia, Hereditary Hemorrhagic/enzymology , Animals , Arteries/embryology , Arteries/growth & development , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic/physiology , Genes, Reporter , Immunohistochemistry , Lac Operon , Male , Mice , Mice, Transgenic , Pregnancy , Telangiectasia, Hereditary Hemorrhagic/pathology
9.
J Hum Genet ; 52(3): 237-243, 2007.
Article in English | MEDLINE | ID: mdl-17219009

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) or Osler-Rendu-Weber disease is a systemic fibrovascular dysplasia with an autosomal dominant inheritance pattern. Mutations in two genes, endoglin and ALK-1, are known to cause HHT, both of which mediate signaling by transforming growth factor beta ligands in vascular endothelial cells. Ten patients were analyzed. Diagnosis of HHT was carried out by means of family history, recurrent bleeding, and the presence of multiple telangiectases lesions. Conformation-sensitive gel electrophoresis analyses with consistent abnormal migration patterns were cloned and sequenced using the MegaBace 1000 DNA automated analyzer. Three novel mutations were identified in the coding sequence of the ALK-1 gene in five patients and their families, which demonstrated clinical manifestations of HHT type 2. These mutations included a G insertion and a T deletion of single base pairs in exons 3 and 7, as well as missense mutations in exons 7 and 8 of the ALK-1 gene. These data indicate that loss-of-function mutations in a single allele of the ALK1 locus are sufficient to contribute to defects in maintaining endothelial integrity. We suggest the high rate of mutation detection and the small size of the ALK-1 gene make genomic sequencing a viable diagnostic test for HHT2.


Subject(s)
Activin Receptors, Type II/genetics , Mutation/genetics , Telangiectasia, Hereditary Hemorrhagic/enzymology , Telangiectasia, Hereditary Hemorrhagic/genetics , Adult , Aged , Aged, 80 and over , Brazil , DNA Mutational Analysis , Exons/genetics , Female , Humans , Male , Middle Aged , Pedigree
10.
Circ Res ; 96(6): 684-92, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15718503

ABSTRACT

Decreased endothelial NO synthase (eNOS)-derived NO bioavailability and impaired vasomotor control are crucial factors in cardiovascular disease pathogenesis. Hereditary hemorrhagic telangiectasia type 1 (HHT1) is a vascular disorder associated with ENDOGLIN (ENG) haploinsufficiency and characterized by venous dilatations, focal loss of capillaries, and arteriovenous malformations (AVMs). We report that resistance arteries from Eng+/- mice display an eNOS-dependent enhancement in endothelium-dependent dilatation and impairment in the myogenic response, despite reduced eNOS levels. We have found that eNOS is significantly reduced in endoglin-deficient endothelial cells because of decreased eNOS protein half-life. We demonstrate that endoglin can reside in caveolae and associate with eNOS, suggesting a stabilizing function of endoglin for eNOS. After Ca2+-induced activation, endoglin-deficient endothelial cells have reduced eNOS/Hsp90 association, produce less NO, and generate more eNOS-derived superoxide (O2-), indicating that endoglin also facilitates eNOS/Hsp90 interactions and is an important regulator in the coupling of eNOS activity. Treatment with an O2- scavenger reverses the vasomotor abnormalities in Eng(+/-) arteries, suggesting that uncoupled eNOS and resulting impaired myogenic response represent early events in HHT1 pathogenesis and that the use of antioxidants may provide a novel therapeutic modality.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Nitric Oxide Synthase/physiology , Vascular Resistance/physiology , Acetylcholine/pharmacology , Animals , Antigens, CD , Blood Pressure/drug effects , Caveolin 1 , Caveolins/analysis , Cells, Cultured/drug effects , Down-Regulation , Endoglin , Endothelium, Vascular/physiology , Enzyme Activation/physiology , HSP90 Heat-Shock Proteins/analysis , HSP90 Heat-Shock Proteins/physiology , Heterozygote , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Liver/enzymology , Membrane Microdomains/chemistry , Membrane Microdomains/enzymology , Mesenteric Arteries/drug effects , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Receptors, Cell Surface , Signal Transduction/physiology , Superoxide Dismutase/pharmacology , Telangiectasia, Hereditary Hemorrhagic/enzymology , Telangiectasia, Hereditary Hemorrhagic/genetics , Umbilical Veins/cytology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/physiology , Vascular Resistance/genetics , Vasodilation/drug effects
11.
Circ Res ; 93(7): 682-9, 2003 Oct 03.
Article in English | MEDLINE | ID: mdl-12970115

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant vascular disorder characterized by epistaxis, mucocutaneous telangiectases, and arteriovenous malformations (AVM). Two genes are linked to HHT: endoglin (ENG) in HHT1 and activin receptor-like kinase 1 (ACVRL1; ALK1) in HHT2. Although both genes are involved in the transforming growth factor beta signaling pathways, the pathogenetic mechanisms for HHT remain elusive. It was shown that mutations in the Alk1 gene in mice and zebrafish resulted in an embryonic lethal phenotype due to severe dilation of blood vessels. We created a novel null mutant mouse line for Alk1 (Alk1lacZ) by replacing its exons, including the one that encodes the transmembrane domain, with the beta-galactosidase gene. Using Alk1lacZ mice, we show that Alk1 is predominantly expressed in developing arterial endothelium. Alk1 expression is greatly diminished in adult arteries, but is induced in preexisting feeding arteries and newly forming arterial vessels during wound healing and tumor angiogenesis. We also show that hemodynamic changes, which require vascular remodeling, may regulate Alk1 expression. Our studies suggest the role of Alk1 signaling in arterialization and remodeling of arteries. Contrary to the current view of HHT as venous disease, our findings suggest that the arterioles rather than the venules are the primary vessels affected by the loss of an Alk1 allele, and that blood vessels with reduction in Alk1 expression may harbor defects in responding to demands for vascular remodeling.


Subject(s)
Activin Receptors, Type I/metabolism , Arteries/enzymology , Endothelium, Vascular/enzymology , Activin Receptors, Type I/genetics , Activin Receptors, Type II , Animals , Arteries/embryology , Arteries/growth & development , Blood Flow Velocity , Embryo, Mammalian/enzymology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Inbred Strains , Mice, Transgenic , Telangiectasia, Hereditary Hemorrhagic/enzymology , Telangiectasia, Hereditary Hemorrhagic/pathology , Veins/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...