101
|
Abstract
Brain arteriovenous malformations (bAVMs) represent a high risk of intracranial hemorrhages, which are substantial causes of morbidity and mortality of bAVMs, especially in children and young adults. Although a variety of factors leading to hemorrhages of bAVMs are investigated extensively, their pathogenesis is still not well elucidated. The author has reviewed the updated data of genetic aspects of bAVMs, especially focusing on clinical and experimental knowledge from hereditary hemorrhagic telangiectasia, which is the representative genetic disease presenting with bAVMs caused by loss-of-function in one of the two genes: endoglin and activin receptor-like kinase 1. This knowledge may allow us to infer the pathogensis of sporadic bAVMs and in the development of new medical therapies for them.
Collapse
Affiliation(s)
- Masaki Komiyama
- Department of Neuro-Intervention, Osaka City General Hospital
| |
Collapse
|
102
|
Gkatzis K, Thalgott J, Dos-Santos-Luis D, Martin S, Lamandé N, Carette MF, Disch F, Snijder RJ, Westermann CJ, Mager JJ, Oh SP, Miquerol L, Arthur HM, Mummery CL, Lebrin F. Interaction Between ALK1 Signaling and Connexin40 in the Development of Arteriovenous Malformations. Arterioscler Thromb Vasc Biol 2016; 36:707-17. [PMID: 26821948 DOI: 10.1161/atvbaha.115.306719] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/20/2016] [Indexed: 01/01/2023]
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.
Collapse
MESH Headings
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/metabolism
- Activin Receptors, Type II/genetics
- Activin Receptors, Type II/metabolism
- Animals
- Arteriovenous Malformations/enzymology
- Arteriovenous Malformations/genetics
- Arteriovenous Malformations/pathology
- Cells, Cultured
- Connexins/genetics
- Connexins/metabolism
- Disease Models, Animal
- Endothelial Cells/enzymology
- Genetic Predisposition to Disease
- Haploinsufficiency
- Humans
- Mice, Mutant Strains
- Mice, Transgenic
- Neovascularization, Pathologic
- Phenotype
- RNA Interference
- Reactive Oxygen Species/metabolism
- Retinal Vessels/enzymology
- Retinal Vessels/pathology
- Signal Transduction
- Telangiectasia, Hereditary Hemorrhagic/enzymology
- Telangiectasia, Hereditary Hemorrhagic/genetics
- Telangiectasia, Hereditary Hemorrhagic/pathology
- Transfection
- Vascular Remodeling
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Konstantinos Gkatzis
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Jérémy Thalgott
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Damien Dos-Santos-Luis
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Sabrina Martin
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Noël Lamandé
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Marie France Carette
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Frans Disch
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Repke J Snijder
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Cornelius J Westermann
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Johannes J Mager
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - S Paul Oh
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Lucile Miquerol
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Helen M Arthur
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Christine L Mummery
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Franck Lebrin
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.).
| |
Collapse
|
103
|
Morrell NW, Bloch DB, ten Dijke P, Goumans MJTH, Hata A, Smith J, Yu PB, Bloch KD. Targeting BMP signalling in cardiovascular disease and anaemia. Nat Rev Cardiol 2016; 13:106-20. [PMID: 26461965 PMCID: PMC4886232 DOI: 10.1038/nrcardio.2015.156] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone morphogenetic proteins (BMPs) and their receptors, known to be essential regulators of embryonic patterning and organogenesis, are also critical for the regulation of cardiovascular structure and function. In addition to their contributions to syndromic disorders including heart and vascular development, BMP signalling is increasingly recognized for its influence on endocrine-like functions in postnatal cardiovascular and metabolic homeostasis. In this Review, we discuss several critical and novel aspects of BMP signalling in cardiovascular health and disease, which highlight the cell-specific and context-specific nature of BMP signalling. Based on advancing knowledge of the physiological roles and regulation of BMP signalling, we indicate opportunities for therapeutic intervention in a range of cardiovascular conditions including atherosclerosis and pulmonary arterial hypertension, as well as for anaemia of inflammation. Depending on the context and the repertoire of ligands and receptors involved in specific disease processes, the selective inhibition or enhancement of signalling via particular BMP ligands (such as in atherosclerosis and pulmonary arterial hypertension, respectively) might be beneficial. The development of selective small molecule antagonists of BMP receptors, and the identification of ligands selective for BMP receptor complexes expressed in the vasculature provide the most immediate opportunities for new therapies.
Collapse
Affiliation(s)
- Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Donald B Bloch
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Peter ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medicine Centre, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Marie-Jose T H Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medicine Centre, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Akiko Hata
- Cardiovascular Research Institute, University of California, 500 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Jim Smith
- MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Paul B Yu
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Kenneth D Bloch
- Anaesthesia Centre for Critical Care Research, Department of Anaesthesia, Critical Care and Pain Medicine, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
104
|
Corti P, Ieraci M, Tejero J. Characterization of zebrafish neuroglobin and cytoglobins 1 and 2: Zebrafish cytoglobins provide insights into the transition from six-coordinate to five-coordinate globins. Nitric Oxide 2015; 53:22-34. [PMID: 26721561 DOI: 10.1016/j.niox.2015.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/11/2015] [Accepted: 12/19/2015] [Indexed: 12/30/2022]
Abstract
Neuroglobin (Ngb) and cytoglobin (Cygb) are two six-coordinate heme proteins of unknown physiological function. Although studies on the mammalian proteins have elucidated aspects of Ngb and Cygb biophysics and indicated potential functions, the properties of non-mammalian Ngbs and Cygbs are largely uncharacterized. We have expressed the recombinant zebrafish proteins Ngb, Cygb1, and Cygb2 in Escherichia coli and characterized their nitrite reduction rates, spectral properties, autoxidation rate constants, redox potentials and lipid binding properties. The three zebrafish proteins can catalyze the reduction of nitrite to nitric oxide with a broad range of reaction rate constants. (Ngb, 0.68 ± 0.04 M(-1) s(-1); Cygb1, 28.6 ± 3.1 M(-1) s(-1); Cygb2, 0.94 ± 0.18 M(-1) s(-1)). We observe that zebrafish Ngb and Cygb2 have comparable spectral features to those of human Ngb and Cygb, consistent with a six-coordinate heme, whereas unexpectedly Cygb1 has a five-coordinate heme, a slower autoxidation and in general has properties more akin to oxygen transport proteins. In agreement with a possible oxygen carrier and nitrite reductase role, we detect mRNA transcript for Cygb1 but not Cygb2 or Ngb in zebrafish blood. Unlike human Cygb, neither of the zebrafish globins binds oleic acid with high affinity. This finding suggests that lipid binding may be a trait acquired later during evolution and not an ancestral property of cytoglobins. Altogether, our results uncover unexpected properties of zebrafish globins and reveal the pivotal role of cytoglobins in the transition of heme globins from six-coordinate to five-coordinate oxygen carriers and nitrite reductases.
Collapse
Affiliation(s)
- Paola Corti
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Matthew Ieraci
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jesús Tejero
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
105
|
García de Vinuesa A, Abdelilah-Seyfried S, Knaus P, Zwijsen A, Bailly S. BMP signaling in vascular biology and dysfunction. Cytokine Growth Factor Rev 2015; 27:65-79. [PMID: 26823333 DOI: 10.1016/j.cytogfr.2015.12.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The vascular system is critical for developmental growth, tissue homeostasis and repair but also for tumor development. Bone morphogenetic protein (BMP) signaling has recently emerged as a fundamental pathway of the endothelium by regulating cardiovascular and lymphatic development and by being causative for several vascular dysfunctions. Two vascular disorders have been directly linked to impaired BMP signaling: pulmonary arterial hypertension and hereditary hemorrhagic telangiectasia. Endothelial BMP signaling critically depends on the cellular context, which includes among others vascular heterogeneity, exposure to flow, and the intertwining with other signaling cascades (Notch, WNT, Hippo and hypoxia). The purpose of this review is to highlight the most recent findings illustrating the clear need for reconsidering the role of BMPs in vascular biology.
Collapse
Affiliation(s)
- Amaya García de Vinuesa
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, D-30625 Hannover, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - An Zwijsen
- VIB Center for the Biology of Disease, Leuven, Belgium; KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Médicale (INSERM, U1036), Grenoble F-38000, France; Commissariat à l'Énergie Atomique et aux Energies Alternatives, Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire Biologie du Cancer et de l'Infection, Grenoble F-38000, France; Université Grenoble-Alpes, Grenoble F-38000, France.
| |
Collapse
|
106
|
Ulrich F, Carretero-Ortega J, Menéndez J, Narvaez C, Sun B, Lancaster E, Pershad V, Trzaska S, Véliz E, Kamei M, Prendergast A, Kidd KR, Shaw KM, Castranova DA, Pham VN, Lo BD, Martin BL, Raible DW, Weinstein BM, Torres-Vázquez J. Reck enables cerebrovascular development by promoting canonical Wnt signaling. Development 2015; 143:147-59. [PMID: 26657775 DOI: 10.1242/dev.123059] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 11/25/2015] [Indexed: 01/03/2023]
Abstract
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.
Collapse
Affiliation(s)
- Florian Ulrich
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Jorge Carretero-Ortega
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Javier Menéndez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Carlos Narvaez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Belinda Sun
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Eva Lancaster
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Valerie Pershad
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Sean Trzaska
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Evelyn Véliz
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Makoto Kamei
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew Prendergast
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Kameha R Kidd
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenna M Shaw
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel A Castranova
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Van N Pham
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brigid D Lo
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - David W Raible
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Brant M Weinstein
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jesús Torres-Vázquez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
107
|
Peacock HM, Caolo V, Jones EAV. Arteriovenous malformations in hereditary haemorrhagic telangiectasia: looking beyond ALK1-NOTCH interactions. Cardiovasc Res 2015; 109:196-203. [PMID: 26645978 DOI: 10.1093/cvr/cvv264] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/29/2015] [Indexed: 12/20/2022] Open
Abstract
Hereditary haemorrhagic telangiectasia (HHT) is characterized by the development of arteriovenous malformations--enlarged shunts allowing arterial flow to bypass capillaries and enter directly into veins. HHT is caused by mutations in ALK1 or Endoglin; however, the majority of arteriovenous malformations are idiopathic and arise spontaneously. Idiopathic arteriovenous malformations differ from those due to loss of ALK1 in terms of both location and disease progression. Furthermore, while arteriovenous malformations in HHT and Alk1 knockout models have decreased NOTCH signalling, some idiopathic arteriovenous malformations have increased NOTCH signalling. The pathogenesis of these lesions also differs, with loss of ALK1 causing expansion of the shunt through proliferation, and NOTCH gain of function inducing initial shunt enlargement by cellular hypertrophy. Hence, we propose that idiopathic arteriovenous malformations are distinct from those of HHT. In this review, we explore the role of ALK1-NOTCH interactions in the development of arteriovenous malformations and examine a possible role of two signalling pathways downstream of ALK1, TMEM100 and IDs, in the development of arteriovenous malformations in HHT. A nuanced understanding of the precise molecular mechanisms underlying idiopathic and HHT-associated arteriovenous malformations will allow for development of targeted treatments for these lesions.
Collapse
Affiliation(s)
- Hanna M Peacock
- Department of Cardiovascular Science, Centre for Molecular and Vascular Biology, KU Leuven, UZ Herestraat 49-Box 911, 3000 Leuven, Belgium
| | - Vincenza Caolo
- Department of Cardiovascular Science, Centre for Molecular and Vascular Biology, KU Leuven, UZ Herestraat 49-Box 911, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Science, Centre for Molecular and Vascular Biology, KU Leuven, UZ Herestraat 49-Box 911, 3000 Leuven, Belgium
| |
Collapse
|
108
|
Animal Models in Studying Cerebral Arteriovenous Malformation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:178407. [PMID: 26649296 PMCID: PMC4663287 DOI: 10.1155/2015/178407] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/11/2015] [Accepted: 10/25/2015] [Indexed: 12/13/2022]
Abstract
Brain arteriovenous malformation (AVM) is an important cause of hemorrhagic stroke. The etiology is largely unknown and the therapeutics are controversial. A review of AVM-associated animal models may be helpful in order to understand the up-to-date knowledge and promote further research about the disease. We searched PubMed till December 31, 2014, with the term “arteriovenous malformation,” limiting results to animals and English language. Publications that described creations of AVM animal models or investigated AVM-related mechanisms and treatments using these models were reviewed. More than 100 articles fulfilling our inclusion criteria were identified, and from them eight different types of the original models were summarized. The backgrounds and procedures of these models, their applications, and research findings were demonstrated. Animal models are useful in studying the pathogenesis of AVM formation, growth, and rupture, as well as in developing and testing new treatments. Creations of preferable models are expected.
Collapse
|
109
|
Fish JE, Wythe JD. The molecular regulation of arteriovenous specification and maintenance. Dev Dyn 2015; 244:391-409. [PMID: 25641373 DOI: 10.1002/dvdy.24252] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/02/2015] [Accepted: 01/04/2015] [Indexed: 12/21/2022] Open
Abstract
The formation of a hierarchical vascular network, composed of arteries, veins, and capillaries, is essential for embryogenesis and is required for the production of new functional vasculature in the adult. Elucidating the molecular mechanisms that orchestrate the differentiation of vascular endothelial cells into arterial and venous cell fates is requisite for regenerative medicine, as the directed formation of perfused vessels is desirable in a myriad of pathological settings, such as in diabetes and following myocardial infarction. Additionally, this knowledge will enhance our understanding and treatment of vascular anomalies, such as arteriovenous malformations (AVMs). From studies in vertebrate model organisms, such as mouse, zebrafish, and chick, a number of key signaling pathways have been elucidated that are required for the establishment and maintenance of arterial and venous fates. These include the Hedgehog, Vascular Endothelial Growth Factor (VEGF), Transforming Growth Factor-β (TGF-β), Wnt, and Notch signaling pathways. In addition, a variety of transcription factor families acting downstream of, or in concert with, these signaling networks play vital roles in arteriovenous (AV) specification. These include Notch and Notch-regulated transcription factors (e.g., HEY and HES), SOX factors, Forkhead factors, β-Catenin, ETS factors, and COUP-TFII. It is becoming apparent that AV specification is a highly coordinated process that involves the intersection and carefully orchestrated activity of multiple signaling cascades and transcriptional networks. This review will summarize the molecular mechanisms that are involved in the acquisition and maintenance of AV fate, and will highlight some of the limitations in our current knowledge of the molecular machinery that directs AV morphogenesis.
Collapse
Affiliation(s)
- Jason E Fish
- Toronto General Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Canada
| | | |
Collapse
|
110
|
VEGF, Notch and TGFβ/BMPs in regulation of sprouting angiogenesis and vascular patterning. Biochem Soc Trans 2015; 42:1576-83. [PMID: 25399573 DOI: 10.1042/bst20140231] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The blood vasculature is constantly adapting to meet the demand from tissue. In so doing, branches may form, reorganize or regress. These complex processes employ integration of multiple signalling cascades, some of them being restricted to endothelial and mural cells and, hence, suitable for targeting of the vasculature. Both genetic and drug targeting experiments have demonstrated the requirement for the vascular endothelial growth factor (VEGF) system, the Delta-like-Notch system and the transforming growth factor β (TGFβ)/bone morphogenetic protein (BMP) cascades in vascular development. Although several of these signalling cascades in part converge into common downstream components, they differ in temporal and spatial regulation and expression. For example, the pro-angiogenic VEGFA is secreted by cells in need of oxygen, presented to the basal side of the endothelium, whereas BMP9 and BMP10 are supplied via the bloodstream in constant interaction with the apical side to suppress angiogenesis. Delta-like 4 (DLL4), on the other hand, is provided as an endothelial membrane bound ligand. In the present article, we discuss recent data on the integration of these pathways in the process of sprouting angiogenesis and vascular patterning and malformation.
Collapse
|
111
|
Rochon ER, Wright DS, Schubert MM, Roman BL. Context-specific interactions between Notch and ALK1 cannot explain ALK1-associated arteriovenous malformations. Cardiovasc Res 2015; 107:143-52. [PMID: 25969392 DOI: 10.1093/cvr/cvv148] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 05/07/2015] [Indexed: 01/17/2023] Open
Abstract
AIMS Notch and activin receptor-like kinase 1 (ALK1) have been implicated in arterial specification, angiogenic tip/stalk cell differentiation, and development of arteriovenous malformations (AVMs), and ALK1 can cooperate with Notch to up-regulate expression of Notch target genes in cultured endothelial cells. These findings suggest that Notch and ALK1 might collaboratively program arterial identity and prevent AVMs. We therefore sought to investigate the interaction between Notch and Alk1 signalling in the developing vertebrate vasculature. METHODS AND RESULTS We modulated Notch and Alk1 activities in zebrafish embryos and examined effects on Notch target gene expression and vascular morphology. Although Alk1 is not necessary for expression of Notch target genes in arterial endothelium, loss of Notch signalling unmasks a role for Alk1 in supporting hey2 and ephrinb2a expression in the dorsal aorta. In contrast, Notch and Alk1 play opposing roles in hey2 expression in cranial arteries and dll4 expression in all arterial endothelium, with Notch inducing and Alk1 repressing these genes. Although alk1 loss increases expression of dll4, AVMs in alk1 mutants could neither be phenocopied by Notch activation nor rescued by Dll4/Notch inhibition. CONCLUSION Control of Notch targets in arterial endothelium is context-dependent, with gene-specific and region-specific requirements for Notch and Alk1. Alk1 is not required for arterial identity, and perturbations in Notch signalling cannot account for alk1 mutant-associated AVMs. These data suggest that AVMs associated with ALK1 mutation are not caused by defective arterial specification or altered Notch signalling.
Collapse
Affiliation(s)
- Elizabeth R Rochon
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Daniel S Wright
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Max M Schubert
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Beth L Roman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto St, Pittsburgh, PA 15261, USA
| |
Collapse
|
112
|
Dynamic reciprocity revisited. J Theor Biol 2015; 370:205-8. [DOI: 10.1016/j.jtbi.2015.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 01/12/2015] [Accepted: 01/15/2015] [Indexed: 11/20/2022]
|
113
|
Letteboer TGW, Benzinou M, Merrick CB, Quigley DA, Zhau K, Kim IJ, To MD, Jablons DM, van Amstel JKP, Westermann CJJ, Giraud S, Dupuis-Girod S, Lesca G, Berg JH, Balmain A, Akhurst RJ. Genetic variation in the functional ENG allele inherited from the non-affected parent associates with presence of pulmonary arteriovenous malformation in hereditary hemorrhagic telangiectasia 1 (HHT1) and may influence expression of PTPN14. Front Genet 2015; 6:67. [PMID: 25815003 PMCID: PMC4357294 DOI: 10.3389/fgene.2015.00067] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 02/09/2015] [Indexed: 01/09/2023] Open
Abstract
HHT shows clinical variability within and between families. Organ site and prevalence of arteriovenous malformations (AVMs) depend on the HHT causative gene and on environmental and genetic modifiers. We tested whether variation in the functional ENG allele, inherited from the unaffected parent, alters risk for pulmonary AVM in HHT1 mutation carriers who are ENG haploinsufficient. Genetic association was found between rs10987746 of the wild type ENG allele and presence of pulmonary AVM [relative risk = 1.3 (1.0018-1.7424)]. The rs10987746-C at-risk allele associated with lower expression of ENG RNA in a panel of human lymphoblastoid cell lines (P = 0.004). Moreover, in angiogenically active human lung adenocarcinoma tissue, but not in uninvolved quiescent lung, rs10987746-C was correlated with expression of PTPN14 (P = 0.004), another modifier of HHT. Quantitative TAQMAN expression analysis in a panel of normal lung tissues from 69 genetically heterogeneous inter-specific backcross mice, demonstrated strong correlation between expression levels of Eng, Acvrl1, and Ptpn14 (r2 = 0.75-0.9, P < 1 × 10(-12)), further suggesting a direct or indirect interaction between these three genes in lung in vivo. Our data indicate that genetic variation within the single functional ENG gene influences quantitative and/or qualitative differences in ENG expression that contribute to risk of pulmonary AVM in HHT1, and provide correlative support for PTPN14 involvement in endoglin/ALK1 lung biology in vivo. PTPN14 has been shown to be a negative regulator of Yap/Taz signaling, which is implicated in mechanotransduction, providing a possible molecular link between endoglin/ALK1 signaling and mechanical stress. EMILIN2, which showed suggestive genetic association with pulmonary AVM, is also reported to interact with Taz in angiogenesis. Elucidation of the molecular mechanisms regulating these interactions in endothelial cells may ultimately provide more rational choices for HHT therapy.
Collapse
Affiliation(s)
- Tom G W Letteboer
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA ; Department of Medical Genetics, University Medical Centre Utrecht Utrecht, Netherlands
| | - Michael Benzinou
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA
| | - Christopher B Merrick
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA ; Department of Clinical Genetics, University of Dundee Dundee, UK
| | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA
| | - Kechen Zhau
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA
| | - Il-Jin Kim
- Department of Surgery, University of California, San Francisco San Francisco, CA, USA
| | - Minh D To
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA ; Department of Surgery, University of California, San Francisco San Francisco, CA, USA
| | - David M Jablons
- Department of Surgery, University of California, San Francisco San Francisco, CA, USA
| | | | | | - Sophie Giraud
- Department of Medical Genetics, Lyon University Hospital Lyon, France
| | | | - Gaetan Lesca
- Department of Medical Genetics, Lyon University Hospital Lyon, France
| | - Jonathan H Berg
- Department of Clinical Genetics, University of Dundee Dundee, UK
| | - Allan Balmain
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA ; Department of Biochemistry and Biophysics, University of California, San Francisco San Francisco, CA, USA ; Institute of Human Genetics, University of California, San Francisco San Francisco, CA, USA
| | - Rosemary J Akhurst
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco San Francisco, CA, USA ; Institute of Human Genetics, University of California, San Francisco San Francisco, CA, USA ; Department of Anatomy, University of California, San Francisco San Francisco, CA, USA
| |
Collapse
|
114
|
Asnani A, Peterson RT. The zebrafish as a tool to identify novel therapies for human cardiovascular disease. Dis Model Mech 2015; 7:763-7. [PMID: 24973746 PMCID: PMC4073266 DOI: 10.1242/dmm.016170] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Over the past decade, the zebrafish has become an increasingly popular animal model for the study of human cardiovascular disease. Because zebrafish embryos are transparent and their genetic manipulation is straightforward, the zebrafish has been used to recapitulate a number of cardiovascular disease processes ranging from congenital heart defects to arrhythmia to cardiomyopathy. The use of fluorescent reporters has been essential to identify two discrete phases of cardiomyocyte differentiation necessary for normal cardiac development in the zebrafish. These phases are analogous to the differentiation of the two progenitor heart cell populations in mammals, termed the first and second heart fields. The small size of zebrafish embryos has enabled high-throughput chemical screening to identify small-molecule suppressors of fundamental pathways in vasculogenesis, such as the BMP axis, as well as of common vascular defects, such as aortic coarctation. The optical clarity of zebrafish has facilitated studies of valvulogenesis as well as detailed electrophysiological mapping to characterize the early cardiac conduction system. One unique aspect of zebrafish larvae is their ability to oxygenate through diffusion alone, permitting the study of mutations that cause severe cardiomyopathy phenotypes such as silent heart and pickwickm171, which mimic titin mutations observed in human dilated cardiomyopathy. Above all, the regenerative capacity of zebrafish presents a particularly exciting opportunity to discover new therapies for cardiac injury, including scar formation following myocardial infarction. This Review will summarize the current state of the field and describe future directions to advance our understanding of human cardiovascular disease.
Collapse
Affiliation(s)
- Aarti Asnani
- Massachusetts General Hospital Cardiovascular Research Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Randall T Peterson
- Massachusetts General Hospital Cardiovascular Research Center, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
115
|
Tual-Chalot S, Oh SP, Arthur HM. Mouse models of hereditary hemorrhagic telangiectasia: recent advances and future challenges. Front Genet 2015; 6:25. [PMID: 25741358 PMCID: PMC4332371 DOI: 10.3389/fgene.2015.00025] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/19/2015] [Indexed: 12/15/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a genetic disorder characterized by a multi-systemic vascular dysplasia and hemorrhage. The precise factors leading to these vascular malformations are not yet understood and robust animal models of HHT are essential to gain a detailed understanding of the molecular and cellular events that lead to clinical symptoms, as well as to test new therapeutic modalities. Most cases of HHT are caused by mutations in either endoglin (ENG) or activin receptor-like kinase 1 (ACVRL1, also known as ALK1). Both genes are associated with TGFβ/BMP signaling, and loss of function mutations in the co-receptor ENG are causal in HHT1, while HHT2 is associated with mutations in the signaling receptor ACVRL1. Significant advances in mouse genetics have provided powerful ways to study the function of Eng and Acvrl1 in vivo, and to generate mouse models of HHT disease. Mice that are null for either Acvrl1 or Eng genes show embryonic lethality due to major defects in angiogenesis and heart development. However mice that are heterozygous for mutations in either of these genes develop to adulthood with no effect on survival. Although these heterozygous mice exhibit selected vascular phenotypes relevant to the clinical pathology of HHT, the phenotypes are variable and generally quite mild. An alternative approach using conditional knockout mice allows us to study the effects of specific inactivation of either Eng or Acvrl1 at different times in development and in different cell types. These conditional knockout mice provide robust and reproducible models of arteriovenous malformations, and they are currently being used to unravel the causal factors in HHT pathologies. In this review, we will summarize the strengths and limitations of current mouse models of HHT, discuss how knowledge obtained from these studies has already informed clinical care and explore the potential of these models for developing improved treatments for HHT patients in the future.
Collapse
Affiliation(s)
| | - S Paul Oh
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, FL, USA
| | - Helen M Arthur
- Institute of Genetic Medicine, Newcastle University , Newcastle, UK
| |
Collapse
|
116
|
Baeyens N, Nicoli S, Coon BG, Ross TD, Van den Dries K, Han J, Lauridsen HM, Mejean CO, Eichmann A, Thomas JL, Humphrey JD, Schwartz MA. Vascular remodeling is governed by a VEGFR3-dependent fluid shear stress set point. eLife 2015; 4. [PMID: 25643397 PMCID: PMC4337723 DOI: 10.7554/elife.04645] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 02/01/2015] [Indexed: 12/23/2022] Open
Abstract
Vascular remodeling under conditions of growth or exercise, or during recovery from arterial restriction or blockage is essential for health, but mechanisms are poorly understood. It has been proposed that endothelial cells have a preferred level of fluid shear stress, or ‘set point’, that determines remodeling. We show that human umbilical vein endothelial cells respond optimally within a range of fluid shear stress that approximate physiological shear. Lymphatic endothelial cells, which experience much lower flow in vivo, show similar effects but at lower value of shear stress. VEGFR3 levels, a component of a junctional mechanosensory complex, mediate these differences. Experiments in mice and zebrafish demonstrate that changing levels of VEGFR3/Flt4 modulates aortic lumen diameter consistent with flow-dependent remodeling. These data provide direct evidence for a fluid shear stress set point, identify a mechanism for varying the set point, and demonstrate its relevance to vessel remodeling in vivo. DOI:http://dx.doi.org/10.7554/eLife.04645.001 Blood and lymphatic vessels remodel their shape, diameter and connections during development, and throughout life in response to growth, exercise and disease. This process is called vascular remodeling. The endothelial cells that line the inside of blood and lymphatic vessels are constantly exposed to the frictional force from flowing blood, termed fluid shear stress. Changes in shear stress are sensed by the endothelial cells, which trigger vascular remodeling to return the stress to the original level. It has been proposed that remodeling is governed by a preferred level of fluid shear stress, or set point, against which deviations in the shear stress are compared. Thus, changing the fluid flow through a blood vessel increases or decreases shear stress, which results in the vessel remodeling to restore the original level of shear stress. Like all remodeling, this process involves inflammation to recruit white blood cells, which assist with the process. Baeyens et al. investigated whether such a shear stress set point exists and what its biological basis might be using cultured endothelial cells from human umbilical veins. These cells remained stable and in a resting state when a particular level of shear stress was applied to them; above or below this shear stress level, the cells produced an inflammatory response like that seen during vascular remodeling. This suggests that these cells do indeed have a set point for shear stress. The same response occurred in human lymphatic endothelial cells, although in these cells the shear stress set point was much lower, correlating with the low flow in lymphatic vessels. Baeyens et al. then discovered that the shear stress set point is related to the level of a protein called VEGFR3 in the cells, which was recently found to participate in shear stress sensing. Endothelial cells from lymphatic vessels normally produce much greater quantities of VEGFR3 than those from blood vessels. Reducing the amount of VEGFR3 in lymphatic endothelial cells increased the set point shear stress, while increasing the levels in blood vessel cells decreased the set point. This suggests that the levels of this protein account for the difference in the response of these two cell types. Baeyens et al. then tested this pathway by reducing the levels of VEGFR3 in zebrafish embryos and in adult mice. In both animals, this caused arteries to narrow, showing that VEGFR3 levels also control sensitivity to shear stress—and hence vascular remodeling—inside living creatures. Understanding in detail how vascular remodeling is regulated could help improve treatments for a wide range of cardiovascular conditions. To do so, further work will be needed to develop methods to control the sensitivity of endothelial cells to shear stress and to identify other proteins that might specifically control the narrowing or the expansion of vessels in human patients. DOI:http://dx.doi.org/10.7554/eLife.04645.002
Collapse
Affiliation(s)
- Nicolas Baeyens
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Stefania Nicoli
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Brian G Coon
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Tyler D Ross
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Koen Van den Dries
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Jinah Han
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Holly M Lauridsen
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, United States
| | - Cecile O Mejean
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Anne Eichmann
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, United States
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, United States
| | - Martin A Schwartz
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States
| |
Collapse
|
117
|
Tillet E, Bailly S. Emerging roles of BMP9 and BMP10 in hereditary hemorrhagic telangiectasia. Front Genet 2015; 5:456. [PMID: 25620979 PMCID: PMC4288046 DOI: 10.3389/fgene.2014.00456] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/12/2014] [Indexed: 12/21/2022] Open
Abstract
Rendu-Osler-Weber syndrome, also known as hereditary hemorrhagic telangiectasia (HHT), is an autosomal dominant vascular disorder. Three genes are causally related to HHT: the ENG gene encoding endoglin, a co-receptor of the TGFβ family (HHT1), the ACVRL1 gene encoding ALK1 (activin receptor-like kinase 1), a type I receptor of the TGFβ family (HHT2), and the SMAD4 gene, encoding a transcription factor critical for this signaling pathway. Bone morphogenetic proteins (BMPs) are growth factors of the TGFβ family. Among them, BMP9 and BMP10 have been shown to bind directly with high affinity to ALK1 and endoglin, and BMP9 mutations have recently been linked to a vascular anomaly syndrome that has phenotypic overlap with HHT. BMP9 and BMP10 are both circulating cytokines in blood, and the current working model is that BMP9 and BMP10 maintain a quiescent endothelial state that is dependent on the level of ALK1/endoglin activation in endothelial cells. In accordance with this model, to explain the etiology of HHT we hypothesize that a deficient BMP9/BMP10/ALK1/endoglin pathway may lead to re-activation of angiogenesis or a greater sensitivity to an angiogenic stimulus. Resulting endothelial hyperproliferation and hypermigration may lead to vasodilatation and generation of an arteriovenous malformation (AVM). HHT would thus result from a defect in the angiogenic balance. This review will focus on the emerging role played by BMP9 and BMP10 in the development of this disease and the therapeutic approaches that this opens.
Collapse
Affiliation(s)
- Emmanuelle Tillet
- Inserm, U1036 , Grenoble, France ; Laboratoire Biologie du Cancer et de l'Infection, Institut de Recherches en Technologies et Sciences pour le Vivant, Commissariat à l'énergie atomique et aux énergies alternatives , Grenoble, France ; Université Grenoble-Alpes , Grenoble, France
| | - Sabine Bailly
- Inserm, U1036 , Grenoble, France ; Laboratoire Biologie du Cancer et de l'Infection, Institut de Recherches en Technologies et Sciences pour le Vivant, Commissariat à l'énergie atomique et aux énergies alternatives , Grenoble, France ; Université Grenoble-Alpes , Grenoble, France
| |
Collapse
|
118
|
Goktas S, Chen CY, Kowalski WJ, Pekkan K. Hemodynamic flow visualization of early embryonic great vessels using μPIV. Methods Mol Biol 2015; 1189:17-30. [PMID: 25245684 DOI: 10.1007/978-1-4939-1164-6_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Microparticle image velocimetry (μPIV) is an evolving quantitative methodology to closely and accurately monitor the cardiac flow dynamics and mechanotransduction during vascular morphogenesis. While PIV technique has a long history, contemporary developments in advanced microscopy have significantly expanded its power. This chapter includes three new methods for μPIV acquisition in selected embryonic structures achieved through advanced optical imaging: (1) high-speed confocal scanning of transgenic zebrafish embryos, where the transgenic erythrocytes act as the tracing particles; (2) microinjection of artificial seeding particles in chick embryos visualized with stereomicroscopy; and (3) real-time, time-resolved optical coherence tomography acquisition of vitelline vessel flow profiles in chick embryos, tracking the erythrocytes.
Collapse
Affiliation(s)
- Selda Goktas
- Department of Mechanical Engineering, Koc University, Istanbul, Turkey
| | | | | | | |
Collapse
|
119
|
Roman BL, Finegold DN. Genetic and Molecular Basis for Hereditary Hemorrhagic Telangiectasia. CURRENT GENETIC MEDICINE REPORTS 2014. [DOI: 10.1007/s40142-014-0061-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
120
|
Kowalski WJ, Teslovich NC, Menon PG, Tinney JP, Keller BB, Pekkan K. Left atrial ligation alters intracardiac flow patterns and the biomechanical landscape in the chick embryo. Dev Dyn 2014; 243:652-62. [PMID: 24868595 DOI: 10.1002/dvdy.24107] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypoplastic left heart syndrome (HLHS) is a major human congenital heart defect that results in single ventricle physiology and high mortality. Clinical data indicate that intracardiac blood flow patterns during cardiac morphogenesis are a significant etiology. We used the left atrial ligation (LAL) model in the chick embryo to test the hypothesis that LAL immediately alters intracardiac flow streams and the biomechanical environment, preceding morphologic and structural defects observed in HLHS. RESULTS Using fluorescent dye injections, we found that intracardiac flow patterns from the right common cardinal vein, right vitelline vein, and left vitelline vein were altered immediately following LAL. Furthermore, we quantified a significant ventral shift of the right common cardinal and right vitelline vein flow streams. We developed an in silico model of LAL, which revealed that wall shear stress was reduced at the left atrioventricular canal and left side of the common ventricle. CONCLUSIONS Our results demonstrate that intracardiac flow patterns change immediately following LAL, supporting the role of hemodynamics in the progression of HLHS. Sites of reduced WSS revealed by computational modeling are commonly affected in HLHS, suggesting that changes in the biomechanical environment may lead to abnormal growth and remodeling of left heart structures.
Collapse
|
121
|
Shoemaker LD, Fuentes LF, Santiago SM, Allen BM, Cook DJ, Steinberg GK, Chang SD. Human brain arteriovenous malformations express lymphatic-associated genes. Ann Clin Transl Neurol 2014; 1:982-95. [PMID: 25574473 PMCID: PMC4284124 DOI: 10.1002/acn3.142] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/08/2014] [Accepted: 10/13/2014] [Indexed: 12/19/2022] Open
Abstract
Objective Brain arteriovenous malformations (AVMs) are devastating, hemorrhage-prone, cerebrovascular lesions characterized by well-defined feeding arteries, draining vein(s) and the absence of a capillary bed. The endothelial cells (ECs) that comprise AVMs exhibit a loss of arterial and venous specification. Given the role of the transcription factor COUP-TFII in vascular development, EC specification, and pathological angiogenesis, we examined human AVM tissue to determine if COUP-FTII may have a role in AVM disease biology. Methods We examined 40 human brain AVMs by immunohistochemistry (IHC) and qRT-PCR for the expression of COUP-TFII as well as other genes involved in venous and lymphatic development, maintenance, and signaling. We also examined proliferation and EC tube formation with human umbilical ECs (HUVEC) following COUP-TFII overexpression. Results We report that AVMs expressed COUP-TFII, SOX18, PROX1, NFATC1, FOXC2, TBX1, LYVE1, Podoplanin, and vascular endothelial growth factor (VEGF)-C, contained Ki67-positive cells and heterogeneously expressed genes involved in Hedgehog, Notch, Wnt, and VEGF signaling pathways. Overexpression of COUP-TFII alone in vitro resulted in increased EC proliferation and dilated tubes in an EC tube formation assay in HUVEC. Interpretation This suggests AVM ECs are further losing their arterial/venous specificity and acquiring a partial lymphatic molecular phenotype. There was significant correlation of gene expression with presence of clinical edema and acute hemorrhage. While the precise role of these genes in the formation, stabilization, growth and risk of hemorrhage of AVMs remains unclear, these findings have potentially important implications for patient management and treatment choice, and opens new avenues for future work on AVM disease mechanisms.
Collapse
Affiliation(s)
- Lorelei D Shoemaker
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Laurel F Fuentes
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Shauna M Santiago
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Breanna M Allen
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Douglas J Cook
- Centre for Neuroscience Studies and the Department of Surgery, Queen's University Kingston, Ontario, Canada
| | - Gary K Steinberg
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Steven D Chang
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| |
Collapse
|
122
|
Kowalski WJ, Pekkan K, Tinney JP, Keller BB. Investigating developmental cardiovascular biomechanics and the origins of congenital heart defects. Front Physiol 2014; 5:408. [PMID: 25374544 PMCID: PMC4204442 DOI: 10.3389/fphys.2014.00408] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 10/02/2014] [Indexed: 11/24/2022] Open
Abstract
Innovative research on the interactions between biomechanical load and cardiovascular (CV) morphogenesis by multiple investigators over the past 3 decades, including the application of bioengineering approaches, has shown that the embryonic heart adapts both structure and function in order to maintain cardiac output to the rapidly growing embryo. Acute adaptive hemodynamic mechanisms in the embryo include the redistribution of blood flow within the heart, dynamic adjustments in heart rate and developed pressure, and beat to beat variations in blood flow and vascular resistance. These biomechanically relevant events occur coincident with adaptive changes in gene expression and trigger adaptive mechanisms that include alterations in myocardial cell growth and death, regional and global changes in myocardial architecture, and alterations in central vascular morphogenesis and remodeling. These adaptive mechanisms allow the embryo to survive these biomechanical stresses (environmental, maternal) and to compensate for developmental errors (genetic). Recent work from numerous laboratories shows that a subset of these adaptive mechanisms is present in every developing multicellular organism with a “heart” equivalent structure. This chapter will provide the reader with an overview of some of the approaches used to quantify embryonic CV functional maturation and performance, provide several illustrations of experimental interventions that explore the role of biomechanics in the regulation of CV morphogenesis including the role of computational modeling, and identify several critical areas for future investigation as available experimental models and methods expand.
Collapse
Affiliation(s)
- William J Kowalski
- Cardiovascular Innovation Institute, University of Louisville Louisville, KY, USA ; Department of Pediatrics, University of Louisville Louisville, KY, USA
| | - Kerem Pekkan
- Department of Biomedical Engineering, Carnegie Mellon University Pittsburgh, PA, USA
| | - Joseph P Tinney
- Cardiovascular Innovation Institute, University of Louisville Louisville, KY, USA ; Department of Pediatrics, University of Louisville Louisville, KY, USA
| | - Bradley B Keller
- Cardiovascular Innovation Institute, University of Louisville Louisville, KY, USA ; Department of Pediatrics, University of Louisville Louisville, KY, USA ; Department of Biomedical Engineering, Carnegie Mellon University Pittsburgh, PA, USA
| |
Collapse
|
123
|
BMP signaling modulation attenuates cerebral arteriovenous malformation formation in a vertebrate model. J Cereb Blood Flow Metab 2014; 34:1688-94. [PMID: 25052553 PMCID: PMC4269730 DOI: 10.1038/jcbfm.2014.134] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 06/02/2014] [Accepted: 06/30/2014] [Indexed: 12/22/2022]
Abstract
Cerebral arteriovenous malformations (AVMs) are vascular anomalies that carry a high risk of stroke and death. To test potential AVM therapies, a reverse genetics approach was used to model AVMs in zebrafish. Antisense morpholino oligonucleotides were used to knockdown activin receptor-like kinase I (alk1), which encodes a transforming growth factor (TGF)-beta family type I receptor implicated in a subset of human AVMs. Knockdown of alk1 caused a spectrum of morphologic, functional, and molecular defects that resemble those seen in humans with AVMs. It was found that losartan, an angiotensin II receptor antagonist, attenuated abnormal blood vessel morphology and systemic manifestations of high-output arteriovenous shunting in vivo. SMAD1 phosphorylation was significantly decreased in alk1 morphants compared with uninjected organisms (0.189±0.0201, 0.429±0.0164, P=0.0002). After treatment, morphant SMAD1 levels approached uninjected levels (0.326±0.0360, P=0.0355) and were significantly higher than those seen in the morphant-control group (P=0.0294). These data suggest that modulating the BMP signaling pathway with losartan, a drug in widespread clinical use in humans as an antihypertensive, may have the potential to be further evaluated as a therapeutic strategy for patients with AVMs.
Collapse
|
124
|
Chou CW, Zhuo YL, Jiang ZY, Liu YW. The hemodynamically-regulated vascular microenvironment promotes migration of the steroidogenic tissue during its interaction with chromaffin cells in the zebrafish embryo. PLoS One 2014; 9:e107997. [PMID: 25248158 PMCID: PMC4172588 DOI: 10.1371/journal.pone.0107997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 08/24/2014] [Indexed: 11/18/2022] Open
Abstract
Background While the endothelium-organ interaction is critical for regulating cellular behaviors during development and disease, the role of blood flow in these processes is only partially understood. The dorsal aorta performs paracrine functions for the timely migration and differentiation of the sympatho-adrenal system. However, it is unclear how the adrenal cortex and medulla achieve and maintain specific integration and whether hemodynamic forces play a role. Methodology and Principal Findings In this study, the possible modulation of steroidogenic and chromaffin cell integration by blood flow was investigated in the teleostean counterpart of the adrenal gland, the interrenal gland, in the zebrafish (Danio rerio). Steroidogenic tissue migration and angiogenesis were suppressed by genetic or pharmacologic inhibition of blood flow, and enhanced by acceleration of blood flow upon norepinephrine treatment. Repressed steroidogenic tissue migration and angiogenesis due to flow deficiency were recoverable following restoration of flow. The regulation of interrenal morphogenesis by blood flow was found to be mediated through the vascular microenvironment and the Fibronectin-phosphorylated Focal Adhesion Kinase (Fn-pFak) signaling. Moreover, the knockdown of krüppel-like factor 2a (klf2a) or matrix metalloproteinase 2 (mmp2), two genes regulated by the hemodynamic force, phenocopied the defects in migration, angiogenesis, the vascular microenvironment, and pFak signaling of the steroidogenic tissue observed in flow-deficient embryos, indicating a direct requirement of mechanotransduction in these processes. Interestingly, epithelial-type steroidogenic cells assumed a mesenchymal-like character and downregulated β-Catenin at cell-cell junctions during interaction with chromaffin cells, which was reversed by inhibiting blood flow or Fn-pFak signaling. Blood flow obstruction also affected the migration of chromaffin cells, but not through mechanosensitive or Fn-pFak dependent mechanisms. Conclusions and Significance These results demonstrate that hemodynamically regulated Fn-pFak signaling promotes the migration of steroidogenic cells, ensuring their interaction with chromaffin cells along both sides of the midline during interrenal gland development.
Collapse
Affiliation(s)
- Chih-Wei Chou
- Department of Life Science, Tunghai University, Taichung, Taiwan
| | - You-Lin Zhuo
- Department of Life Science, Tunghai University, Taichung, Taiwan
| | - Zhe-Yu Jiang
- Department of Life Science, Tunghai University, Taichung, Taiwan
| | - Yi-Wen Liu
- Department of Life Science, Tunghai University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
125
|
Nielsen CM, Cuervo H, Ding VW, Kong Y, Huang EJ, Wang RA. Deletion of Rbpj from postnatal endothelium leads to abnormal arteriovenous shunting in mice. Development 2014; 141:3782-92. [PMID: 25209249 DOI: 10.1242/dev.108951] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Arteriovenous malformations (AVMs) are tortuous vessels characterized by arteriovenous (AV) shunts, which displace capillaries and shunt blood directly from artery to vein. Notch signaling regulates embryonic AV specification by promoting arterial, as opposed to venous, endothelial cell (EC) fate. To understand the essential role of endothelial Notch signaling in postnatal AV organization, we used inducible Cre-loxP recombination to delete Rbpj, a mediator of canonical Notch signaling, from postnatal ECs in mice. Deletion of endothelial Rbpj from birth resulted in features of AVMs by P14, including abnormal AV shunting and tortuous vessels in the brain, intestine and heart. We further analyzed brain AVMs, as they pose particular health risks. Consistent with AVM pathology, we found cerebral hemorrhage, hypoxia and necrosis, and neurological deficits. AV shunts originated from capillaries (and possibly venules), with the earliest detectable morphological abnormalities in AV connections by P8. Prior to AV shunt formation, alterations in EC gene expression were detected, including decreased Efnb2 and increased Pai1, which encodes a downstream effector of TGFβ signaling. After AV shunts had formed, whole-mount immunostaining showed decreased Efnb2 and increased Ephb4 expression within AV shunts, suggesting that ECs were reprogrammed from arterial to venous identity. Deletion of Rbpj from adult ECs led to tortuosities in gastrointestinal, uterine and skin vascular beds, but had mild effects in the brain. Our results demonstrate a temporal requirement for Rbpj in postnatal ECs to maintain proper artery, capillary and vein organization and to prevent abnormal AV shunting and AVM pathogenesis.
Collapse
Affiliation(s)
- Corinne M Nielsen
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Henar Cuervo
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Vivianne W Ding
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Yupeng Kong
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Rong A Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
126
|
Han C, Choe SW, Kim YH, Acharya AP, Keselowsky BG, Sorg BS, Lee YJ, Oh SP. VEGF neutralization can prevent and normalize arteriovenous malformations in an animal model for hereditary hemorrhagic telangiectasia 2. Angiogenesis 2014; 17:823-830. [PMID: 24957885 DOI: 10.1007/s10456-014-9436-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 06/05/2014] [Indexed: 12/18/2022]
Abstract
Arteriovenous malformation (AVM) refers to a vascular anomaly where arteries and veins are directly connected through a complex, tangled web of abnormal AV fistulae without a normal capillary network. Hereditary hemorrhagic telangiectasia (HHT) types 1 and 2 arise from heterozygous mutations in endoglin (ENG) and activin receptor-like kinase 1 (ALK1), respectively. HHT patients possess AVMs in various organs, and telangiectases (small AVMs) along the mucocutaneous surface. Understanding why and how AVMs develop is crucial for developing therapies to inhibit the formation, growth, or maintenance of AVMs in HHT patients. Previously, we have shown that secondary factors such as wounding are required for Alk1-deficient vessels to develop skin AVMs. Here, we present evidences that AVMs establish from nascent arteries and veins rather than from remodeling of a preexistent capillary network in the wound-induced skin AVM model. We also show that VEGF can mimic the wound effect on skin AVM formation, and VEGF-neutralizing antibody can prevent skin AVM formation and ameliorate internal bleeding in Alk1-deficient adult mice. With topical applications at different stages of AVM development, we demonstrate that the VEGF blockade can prevent the formation of AVM and cease the progression of AVM development. Taken together, the presented experimental model is an invaluable system for precise molecular mechanism of action of VEGF blockades as well as for preclinical screening of drug candidates for epistaxis and gastrointestinal bleedings.
Collapse
Affiliation(s)
- Chul Han
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Se-Woon Choe
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610.,Department of Biomedical Engineering, Tongmyong University, Busan, Republic of Korea
| | - Yong Hwan Kim
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Abhinav P Acharya
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL 32611
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL 32611
| | - Brian S Sorg
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL 32611
| | - Young-Jae Lee
- World Class University program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Incheon, Republic of Korea
| | - S Paul Oh
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610.,World Class University program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Incheon, Republic of Korea
| |
Collapse
|
127
|
Tual-Chalot S, Mahmoud M, Allinson KR, Redgrave RE, Zhai Z, Oh SP, Fruttiger M, Arthur HM. Endothelial depletion of Acvrl1 in mice leads to arteriovenous malformations associated with reduced endoglin expression. PLoS One 2014; 9:e98646. [PMID: 24896812 PMCID: PMC4045906 DOI: 10.1371/journal.pone.0098646] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/06/2014] [Indexed: 01/28/2023] Open
Abstract
Rare inherited cardiovascular diseases are frequently caused by mutations in genes that are essential for the formation and/or function of the cardiovasculature. Hereditary Haemorrhagic Telangiectasia is a familial disease of this type. The majority of patients carry mutations in either Endoglin (ENG) or ACVRL1 (also known as ALK1) genes, and the disease is characterized by arteriovenous malformations and persistent haemorrhage. ENG and ACVRL1 encode receptors for the TGFβ superfamily of ligands, that are essential for angiogenesis in early development but their roles are not fully understood. Our goal was to examine the role of Acvrl1 in vascular endothelial cells during vascular development and to determine whether loss of endothelial Acvrl1 leads to arteriovenous malformations. Acvrl1 was depleted in endothelial cells either in early postnatal life or in adult mice. Using the neonatal retinal plexus to examine angiogenesis, we observed that loss of endothelial Acvrl1 led to venous enlargement, vascular hyperbranching and arteriovenous malformations. These phenotypes were associated with loss of arterial Jag1 expression, decreased pSmad1/5/8 activity and increased endothelial cell proliferation. We found that Endoglin was markedly down-regulated in Acvrl1-depleted ECs showing endoglin expression to be downstream of Acvrl1 signalling in vivo. Endothelial-specific depletion of Acvrl1 in pups also led to pulmonary haemorrhage, but in adult mice resulted in caecal haemorrhage and fatal anaemia. We conclude that during development, endothelial Acvrl1 plays an essential role to regulate endothelial cell proliferation and arterial identity during angiogenesis, whilst in adult life endothelial Acvrl1 is required to maintain vascular integrity.
Collapse
Affiliation(s)
- Simon Tual-Chalot
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | - Marwa Mahmoud
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | | | - Rachael E. Redgrave
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | - Zhenhua Zhai
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | - S. Paul Oh
- Department of Physiology, University of Florida, Gainesville, Florida, United States of America
| | | | - Helen M. Arthur
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
- * E-mail:
| |
Collapse
|
128
|
Chen CY, Cheng CM. Microfluidics expands the zebrafish potentials in pharmaceutically relevant screening. Adv Healthc Mater 2014; 3:940-5. [PMID: 24459083 DOI: 10.1002/adhm.201300546] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 12/06/2013] [Indexed: 01/15/2023]
Abstract
The objective of this study is to enlarge the impact of microfluidics on the pharmaceutical industry by highlighting the reported scientific work on the synergistic relationship between zebrafish and microfluidics, and furthering that effort to shed light on how microfluidics can facilitate the use of zebrafish as a gene screening tool. Zebrafish is ranked the third most important animal model after rats and mice, according to a National Institutes of Health (NIH) announcement in 2003. It has become a staple for scientists to examine and subsequently begin to unravel the mystery of human diseases, and is increasingly used in toxicological studies for new drug development. The unique characteristics that this tiny fish possesses, including rapid growth rate, prodigious numbers of offspring, and eggs that develop outside the body, make it an invaluable genetic tool. Evidently, these advantages can be broadened with the addition of a properly designed microfluidic circuit. By means of the presented illustrations and demonstrated applications, the goal is to spark interest in the development of more novel microfluidic platform designs that can leverage the attributes of zebrafish and quickly come to commercial fruition.
Collapse
Affiliation(s)
- Chia-Yuan Chen
- Department of Mechanical Engineering; National Taiwan University of Science and Technology; Taipei 106 Taiwan
| | - Chao-Min Cheng
- Institute of Nanoengineering and Microsystems; National Tsing Hua University; Hsinchu 300 Taiwan
- Institute of Cellular and Organismic Biology; Academia Sinica; Taipei 115 Taiwan
| |
Collapse
|
129
|
Malformaciones arteriovenosas: un reto diagnóstico y terapéutico. ACTAS DERMO-SIFILIOGRAFICAS 2014; 105:347-58. [DOI: 10.1016/j.ad.2013.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 04/03/2013] [Accepted: 04/05/2013] [Indexed: 12/18/2022] Open
|
130
|
de Miguel R, López-Gutierrez J, Boixeda P. Arteriovenous Malformations: A Diagnostic and Therapeutic Challenge. ACTAS DERMO-SIFILIOGRAFICAS 2014. [DOI: 10.1016/j.adengl.2013.04.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
131
|
Walcott BP, Peterson RT. Zebrafish models of cerebrovascular disease. J Cereb Blood Flow Metab 2014; 34:571-7. [PMID: 24517974 PMCID: PMC3982096 DOI: 10.1038/jcbfm.2014.27] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 12/27/2013] [Accepted: 01/07/2014] [Indexed: 12/18/2022]
Abstract
Perturbations in cerebral blood flow and abnormalities in blood vessel structure are the hallmarks of cerebrovascular disease. While there are many genetic and environmental factors that affect these entities through a heterogeneous group of disease processes, the ultimate final pathologic insult in humans is defined as a stroke, or damage to brain parenchyma. In the case of ischemic stroke, blood fails to reach its target destination whereas in hemorrhagic stroke, extravasation of blood occurs outside of the blood vessel lumen, resulting in direct damage to brain parenchyma. As these acute events can be neurologically devastating, if not fatal, development of novel therapeutics are urgently needed. The zebrafish (Danio rerio) is an attractive model for the study of cerebrovascular disease because of its morphological and physiological similarity to human cerebral vasculature, its ability to be genetically manipulated, and its fecundity allowing for large-scale, phenotype-based screens.
Collapse
Affiliation(s)
- Brian P Walcott
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Randall T Peterson
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
132
|
Goetz JG, Steed E, Ferreira RR, Roth S, Ramspacher C, Boselli F, Charvin G, Liebling M, Wyart C, Schwab Y, Vermot J. Endothelial cilia mediate low flow sensing during zebrafish vascular development. Cell Rep 2014; 6:799-808. [PMID: 24561257 DOI: 10.1016/j.celrep.2014.01.032] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 12/20/2013] [Accepted: 01/23/2014] [Indexed: 11/16/2022] Open
Abstract
VIDEO ABSTRACT The pattern of blood flow has long been thought to play a significant role in vascular morphogenesis, yet the flow-sensing mechanism that is involved at early embryonic stages, when flow forces are low, remains unclear. It has been proposed that endothelial cells use primary cilia to sense flow, but this has never been tested in vivo. Here we show, by noninvasive, high-resolution imaging of live zebrafish embryos, that endothelial cilia progressively deflect at the onset of blood flow and that the deflection angle correlates with calcium levels in endothelial cells. We demonstrate that alterations in shear stress, ciliogenesis, or expression of the calcium channel PKD2 impair the endothelial calcium level and both increase and perturb vascular morphogenesis. Altogether, these results demonstrate that endothelial cilia constitute a highly sensitive structure that permits the detection of low shear forces during vascular morphogenesis.
Collapse
Affiliation(s)
- Jacky G Goetz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Emily Steed
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Rita R Ferreira
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Stéphane Roth
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Caroline Ramspacher
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Francesco Boselli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Gilles Charvin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Michael Liebling
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Claire Wyart
- Institut du Cerveau et de la Moelle Épinière (ICM), Hôpital de la Pitié-Salpêtrière, 75013 Paris, France; Inserm UMRS 1127, 75013 Paris, France; CNRS UMR 7225, 75013 Paris, France; UPMC University of Paris 06, 75005 Paris, France
| | - Yannick Schwab
- Electron Microscopy Core Facility, Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France.
| |
Collapse
|
133
|
Wilkinson RN, van Eeden FJ. The Zebrafish as a Model of Vascular Development and Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 124:93-122. [DOI: 10.1016/b978-0-12-386930-2.00005-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
134
|
Samson SC, Ferrer T, Jou CJ, Sachse FB, Shankaran SS, Shaw RM, Chi NC, Tristani-Firouzi M, Yost HJ. 3-OST-7 regulates BMP-dependent cardiac contraction. PLoS Biol 2013; 11:e1001727. [PMID: 24311987 PMCID: PMC3849020 DOI: 10.1371/journal.pbio.1001727] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 10/23/2013] [Indexed: 11/29/2022] Open
Abstract
During zebrafish cardiac development, 3-OST-7 constrains BMP signaling to the atrioventricular junction and precludes it from contractile myocardium, allowing tropomyosin-dependent sarcomere assembly and contraction. The 3-O-sulfotransferase (3-OST) family catalyzes rare modifications of glycosaminoglycan chains on heparan sulfate proteoglycans, yet their biological functions are largely unknown. Knockdown of 3-OST-7 in zebrafish uncouples cardiac ventricular contraction from normal calcium cycling and electrophysiology by reducing tropomyosin4 (tpm4) expression. Normal 3-OST-7 activity prevents the expansion of BMP signaling into ventricular myocytes, and ectopic activation of BMP mimics the ventricular noncontraction phenotype seen in 3-OST-7 depleted embryos. In 3-OST-7 morphants, ventricular contraction can be rescued by overexpression of tropomyosin tpm4 but not by troponin tnnt2, indicating that tpm4 serves as a lynchpin for ventricular sarcomere organization downstream of 3-OST-7. Contraction can be rescued by expression of 3-OST-7 in endocardium, or by genetic loss of bmp4. Strikingly, BMP misregulation seen in 3-OST-7 morphants also occurs in multiple cardiac noncontraction models, including potassium voltage-gated channel gene, kcnh2, affected in Romano-Ward syndrome and long-QT syndrome, and cardiac troponin T gene, tnnt2, affected in human cardiomyopathies. Together these results reveal 3-OST-7 as a key component of a novel pathway that constrains BMP signaling from ventricular myocytes, coordinates sarcomere assembly, and promotes cardiac contractile function. A highly complex environment at the cell surface and in the space between cells is thought to modulate cell behavior. Heparan sulfate proteoglycans are cell surface and extracellular matrix molecules that are covalently linked to long chains of repeating sugar units called glycosaminoglycan chains. These chains can be subjected to rare modifications and they are believed to influence specific cell signaling events in a lineage specific fashion in what is called the “glycocode.” Here we explore the functions of one member of a family of enzymes, 3-O-sulfotransferases (3-OSTs) that catalyzes a rare modification (3-O-sulfation) of glycosaminoglycans in zebrafish. We show that knockdown of 3-OST-7 results in a very specific phenotype, including loss of cardiac ventricle contraction. Knockdown of other 3-OST family members did not result in the same phenotype, suggesting that distinct 3-OST family members have distinct functions in vertebrates and lending in vivo evidence for the glycocode hypothesis. Mechanistically, we found that cardiac contraction can be rescued by reducing the amount of endogenous BMP4, and can be blocked by increasing BMP signaling, suggesting that the glycocode generated by 3-OST-7 is necessary to constrain BMP signaling in the heart for normal cardiac contraction. Furthermore, we show that tropomyosin4 (tpm4) is downstream of 3-OST-7 function, indicating that Tpm4 is key in this pathway to building the sarcomere, the functional contraction unit of the cardiomyocyte.
Collapse
Affiliation(s)
- Shiela C. Samson
- Department of Neurobiology & Anatomy, University of Utah Molecular Medicine Program, Salt Lake City, Utah, United States of America
| | - Tania Ferrer
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Chuanchau J. Jou
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Frank B. Sachse
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Sunita S. Shankaran
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Robin M. Shaw
- Department of Medicine, Cedars-Sinai Heart Institute, University of California, Los Angeles, California, United States of America
| | - Neil C. Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, California, United States of America
| | - Martin Tristani-Firouzi
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - H. Joseph Yost
- Department of Neurobiology & Anatomy, University of Utah Molecular Medicine Program, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
135
|
Anton H, Harlepp S, Ramspacher C, Wu D, Monduc F, Bhat S, Liebling M, Paoletti C, Charvin G, Freund JB, Vermot J. Pulse propagation by a capacitive mechanism drives embryonic blood flow. Development 2013; 140:4426-34. [DOI: 10.1242/dev.096768] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pulsatile flow is a universal feature of the blood circulatory system in vertebrates and can lead to diseases when abnormal. In the embryo, blood flow forces stimulate vessel remodeling and stem cell proliferation. At these early stages, when vessels lack muscle cells, the heart is valveless and the Reynolds number (Re) is low, few details are available regarding the mechanisms controlling pulses propagation in the developing vascular network. Making use of the recent advances in optical-tweezing flow probing approaches, fast imaging and elastic-network viscous flow modeling, we investigated the blood-flow mechanics in the zebrafish main artery and show how it modifies the heart pumping input to the network. The movement of blood cells in the embryonic artery suggests that elasticity of the network is an essential factor mediating the flow. Based on these observations, we propose a model for embryonic blood flow where arteries act like a capacitor in a way that reduces heart effort. These results demonstrate that biomechanics is key in controlling early flow propagation and argue that intravascular elasticity has a role in determining embryonic vascular function.
Collapse
Affiliation(s)
- Halina Anton
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Sebastien Harlepp
- Institut de Physique et de Chimie des Matériaux de Strasbourg, Université de Strasbourg, UMR 7504, 23 rue du Loess, 67034 Strasbourg, France
| | - Caroline Ramspacher
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Dave Wu
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Fabien Monduc
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Sandeep Bhat
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Michael Liebling
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Camille Paoletti
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | - Gilles Charvin
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| | | | - Julien Vermot
- Institut de Génétique Moleculaire et Cellulaire, CNRS/INSERM/UdS, 1 rue Laurent Fries, BP10142, 67404 Illkirch, France
| |
Collapse
|
136
|
Kochhan E, Lenard A, Ellertsdottir E, Herwig L, Affolter M, Belting HG, Siekmann AF. Blood flow changes coincide with cellular rearrangements during blood vessel pruning in zebrafish embryos. PLoS One 2013; 8:e75060. [PMID: 24146748 PMCID: PMC3795766 DOI: 10.1371/journal.pone.0075060] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 08/08/2013] [Indexed: 12/26/2022] Open
Abstract
After the initial formation of a highly branched vascular plexus, blood vessel pruning generates a hierarchically structured network with improved flow characteristics. We report here on the cellular events that occur during the pruning of a defined blood vessel in the eye of developing zebrafish embryos. Time-lapse imaging reveals that the connection of a new blood vessel sprout with a previously perfused multicellular endothelial tube leads to the formation of a branched, Y-shaped structure. Subsequently, endothelial cells in parts of the previously perfused branch rearrange from a multicellular into a unicellular tube, followed by blood vessel detachment. This process is accompanied by endothelial cell death. Finally, we show that differences in blood flow between neighboring vessels are important for the completion of the pruning process. Our data suggest that flow induced changes in tubular architecture ensure proper blood vessel pruning.
Collapse
Affiliation(s)
- Eva Kochhan
- Max Planck Institute for Molecular Biomedicine, Laboratory for Cardiovascular Patterning, Muenster, Germany
| | - Anna Lenard
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
| | - Elin Ellertsdottir
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
| | - Lukas Herwig
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
| | - Markus Affolter
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
| | - Heinz-Georg Belting
- Biozentrum der Universität Basel, Abteilung Zellbiologie, Basel, Switzerland
| | - Arndt F. Siekmann
- Max Planck Institute for Molecular Biomedicine, Laboratory for Cardiovascular Patterning, Muenster, Germany
- * E-mail:
| |
Collapse
|
137
|
Atri D, Larrivée B, Eichmann A, Simons M. Endothelial signaling and the molecular basis of arteriovenous malformation. Cell Mol Life Sci 2013; 71:10.1007/s00018-013-1475-1. [PMID: 24077895 PMCID: PMC3969452 DOI: 10.1007/s00018-013-1475-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/08/2013] [Accepted: 09/10/2013] [Indexed: 12/21/2022]
Abstract
Arteriovenous malformations occur when abnormalities of vascular patterning result in the flow of blood from arteries to veins without an intervening capillary bed. Recent work has revealed the importance of the Notch and TGF-β signaling pathways in vascular patterning. Specifically, Notch signaling has an increasingly apparent role in arterial specification and suppression of branching, whereas TGF-β is implicated in vascular smooth muscle development and remodeling under angiogenic stimuli. These physiologic roles, consequently, have implicated both pathways in the pathogenesis of arteriovenous malformation. In this review, we summarize the studies of endothelial signaling that contribute to arteriovenous malformation and the roles of genes implicated in their pathogenesis. We further discuss how endothelial signaling may contribute to vascular smooth muscle development and how knowledge of signaling pathways may provide us targets for medical therapy in these vascular lesions.
Collapse
Affiliation(s)
- Deepak Atri
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, United States
| | - Bruno Larrivée
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, United States
- Department of Ophthalmology, Hôpital Maisonneuve-Rosemont Research Centre, University of Montreal, Montreal, Canada
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, United States
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Paris, France
| | - Michael Simons
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| |
Collapse
|
138
|
Hawinkels LJ, Garcia de Vinuesa A, Ten Dijke P. Activin receptor-like kinase 1 as a target for anti-angiogenesis therapy. Expert Opin Investig Drugs 2013; 22:1371-83. [PMID: 24053899 DOI: 10.1517/13543784.2013.837884] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Formation of blood vessels from pre-existing ones, also termed angiogenesis, is of crucial importance for the outgrowth of tumours beyond 1 - 2 mm³. Therefore, anti-angiogenic therapies, mainly focussing on inhibition of vascular endothelial growth factor (VEGF) are used in clinical therapy. However, although initially reducing tumour size, therapy resistance occurs frequently and new targets are needed. A possible target is activin receptor-like kinase (ALK)-1, a transforming growth factor (TGF)-β type-I receptor, which binds bone morphogenetic protein (BMP)-9 and -10 with high affinity and has an important role in regulating angiogenesis. AREAS COVERED Several approaches to interfere with ALK1 signalling have been developed, that is, ALK1 neutralising antibodies and a soluble ALK1 extracellular domain/Fc fusion protein (ALK1-Fc), acting as a ligand trap. In this review, we discuss the involvement of ALK1 in angiogenesis, in a variety of diseases and the current status of the development of ALK1 inhibitors for cancer therapy. EXPERT OPINION Based on current, mainly preclinical studies on inhibition of ALK1 signalling by ligand traps and neutralising antibodies, targeting ALK1 seems very promising. Both ALK1-Fc and neutralising antibodies strongly inhibit angiogenesis in vitro and in vivo. The results from the first Phase I clinical trials are to be reported soon and multiple Phase II studies are ongoing.
Collapse
Affiliation(s)
- Lukas Jac Hawinkels
- Leiden University Medical Centre, Cancer Genomics Centre Netherlands and Centre for BioMedical Genetics, Department of Molecular Cell Biology , Building-2, S1-P, PO box 9600, 2300 RC Leiden , The Netherlands +31 71 526 9272 ; +31 71 526 8270 ;
| | | | | |
Collapse
|
139
|
Wooderchak-Donahue W, McDonald J, O’Fallon B, Upton P, Li W, Roman B, Young S, Plant P, Fülöp G, Langa C, Morrell N, Botella L, Bernabeu C, Stevenson D, Runo J, Bayrak-Toydemir P. BMP9 mutations cause a vascular-anomaly syndrome with phenotypic overlap with hereditary hemorrhagic telangiectasia. Am J Hum Genet 2013; 93:530-7. [PMID: 23972370 PMCID: PMC3769931 DOI: 10.1016/j.ajhg.2013.07.004] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/24/2013] [Accepted: 07/01/2013] [Indexed: 12/29/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT), the most common inherited vascular disorder, is caused by mutations in genes involved in the transforming growth factor beta (TGF-β) signaling pathway (ENG, ACVRL1, and SMAD4). Yet, approximately 15% of individuals with clinical features of HHT do not have mutations in these genes, suggesting that there are undiscovered mutations in other genes for HHT and possibly vascular disorders with overlapping phenotypes. The genetic etiology for 191 unrelated individuals clinically suspected to have HHT was investigated with the use of exome and Sanger sequencing; these individuals had no mutations in ENG, ACVRL1, and SMAD4. Mutations in BMP9 (also known as GDF2) were identified in three unrelated probands. These three individuals had epistaxis and dermal lesions that were described as telangiectases but whose location and appearance resembled lesions described in some individuals with RASA1-related disorders (capillary malformation-arteriovenous malformation syndrome). Analyses of the variant proteins suggested that mutations negatively affect protein processing and/or function, and a bmp9-deficient zebrafish model demonstrated that BMP9 is involved in angiogenesis. These data confirm a genetic cause of a vascular-anomaly syndrome that has phenotypic overlap with HHT.
Collapse
Affiliation(s)
- Whitney L. Wooderchak-Donahue
- Associated Regional and University Pathologists Institute for Clinical and Experimental Pathology, Salt Lake City, UT 84108, USA
| | - Jamie McDonald
- Department of Pathology, University of Utah, Salt Lake City, UT 84108, USA
| | - Brendan O’Fallon
- Associated Regional and University Pathologists Institute for Clinical and Experimental Pathology, Salt Lake City, UT 84108, USA
| | - Paul D. Upton
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Wei Li
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Beth L. Roman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Sarah Young
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Parker Plant
- Associated Regional and University Pathologists Institute for Clinical and Experimental Pathology, Salt Lake City, UT 84108, USA
| | - Gyula T. Fülöp
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28040, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid 28040, Spain
| | - Carmen Langa
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28040, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid 28040, Spain
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Luisa M. Botella
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28040, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid 28040, Spain
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28040, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid 28040, Spain
| | - David A. Stevenson
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT 84108, USA
| | - James R. Runo
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Pinar Bayrak-Toydemir
- Associated Regional and University Pathologists Institute for Clinical and Experimental Pathology, Salt Lake City, UT 84108, USA
- Department of Pathology, University of Utah, Salt Lake City, UT 84108, USA
| |
Collapse
|
140
|
Abstract
During development, mechanical forces cause changes in size, shape, number, position, and gene expression of cells. They are therefore integral to any morphogenetic processes. Force generation by actin-myosin networks and force transmission through adhesive complexes are two self-organizing phenomena driving tissue morphogenesis. Coordination and integration of forces by long-range force transmission and mechanosensing of cells within tissues produce large-scale tissue shape changes. Extrinsic mechanical forces also control tissue patterning by modulating cell fate specification and differentiation. Thus, the interplay between tissue mechanics and biochemical signaling orchestrates tissue morphogenesis and patterning in development.
Collapse
|
141
|
Laux DW, Young S, Donovan JP, Mansfield CJ, Upton PD, Roman BL. Circulating Bmp10 acts through endothelial Alk1 to mediate flow-dependent arterial quiescence. Development 2013; 140:3403-12. [PMID: 23863480 DOI: 10.1242/dev.095307] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Blood flow plays crucial roles in vascular development, remodeling and homeostasis, but the molecular pathways required for transducing flow signals are not well understood. In zebrafish embryos, arterial expression of activin receptor-like kinase 1 (alk1), which encodes a TGFβ family type I receptor, is dependent on blood flow, and loss of alk1 mimics lack of blood flow in terms of dysregulation of a subset of flow-responsive arterial genes and increased arterial endothelial cell number. These data suggest that blood flow activates Alk1 signaling to promote a flow-responsive gene expression program that limits nascent arterial caliber. Here, we demonstrate that restoration of endothelial alk1 expression to flow-deprived arteries fails to rescue Alk1 activity or normalize arterial endothelial cell gene expression or number, implying that blood flow may play an additional role in Alk1 signaling independent of alk1 induction. To this end, we define cardiac-derived Bmp10 as the crucial ligand for endothelial Alk1 in embryonic vascular development, and provide evidence that circulating Bmp10 acts through endothelial Alk1 to limit endothelial cell number in and thereby stabilize the caliber of nascent arteries. Thus, blood flow promotes Alk1 activity by concomitantly inducing alk1 expression and distributing Bmp10, thereby reinforcing this signaling pathway, which functions to limit arterial caliber at the onset of flow. Because mutations in ALK1 cause arteriovenous malformations (AVMs), our findings suggest that an impaired flow response initiates AVM development.
Collapse
Affiliation(s)
- Derek W Laux
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | | | | | | |
Collapse
|
142
|
Bone morphogenetic protein 9 (BMP9) controls lymphatic vessel maturation and valve formation. Blood 2013; 122:598-607. [PMID: 23741013 DOI: 10.1182/blood-2012-12-472142] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lymphatic vessels are critical for the maintenance of tissue fluid homeostasis and their dysfunction contributes to several human diseases. The activin receptor-like kinase 1 (ALK1) is a transforming growth factor-β family type 1 receptor that is expressed on both blood and lymphatic endothelial cells (LECs). Its high-affinity ligand, bone morphogenetic protein 9 (BMP9), has been shown to be critical for retinal angiogenesis. The aim of this work was to investigate whether BMP9 could play a role in lymphatic development. We found that Bmp9 deficiency in mice causes abnormal lymphatic development. Bmp9-knockout (KO) pups presented hyperplastic mesenteric collecting vessels that maintained LYVE-1 expression. In accordance with this result, we found that BMP9 inhibited LYVE-1 expression in LECs in an ALK1-dependent manner. Bmp9-KO pups also presented a significant reduction in the number and in the maturation of mesenteric lymphatic valves at embryonic day 18.5 and at postnatal days 0 and 4. Interestingly, the expression of several genes known to be involved in valve formation (Foxc2, Connexin37, EphrinB2, and Neuropilin1) was upregulated by BMP9 in LECS. Finally, we demonstrated that Bmp9-KO neonates and adult mice had decreased lymphatic draining efficiency. These data identify BMP9 as an important extracellular regulator in the maturation of the lymphatic vascular network affecting valve development and lymphatic vessel function.
Collapse
|
143
|
Maleki S, Björck HM, Paloschi V, Kjellqvist S, Folkersen L, Jackson V, Franco-Cereceda A, Eriksson P. Aneurysm Development in Patients With Bicuspid Aortic Valve (BAV): Possible Connection to Repair Deficiency? AORTA : OFFICIAL JOURNAL OF THE AORTIC INSTITUTE AT YALE-NEW HAVEN HOSPITAL 2013; 1:13-22. [PMID: 26798668 DOI: 10.12945/j.aorta.2013.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/15/2013] [Indexed: 01/25/2023]
Affiliation(s)
- Shohreh Maleki
- Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hanna M Björck
- Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Valentina Paloschi
- Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sanela Kjellqvist
- Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lasse Folkersen
- Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Veronica Jackson
- Cardiothoracic Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Anders Franco-Cereceda
- Cardiothoracic Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Per Eriksson
- Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
144
|
The ALK-1/Smad1 pathway in cardiovascular physiopathology. A new target for therapy? Biochim Biophys Acta Mol Basis Dis 2013; 1832:1492-510. [PMID: 23707512 DOI: 10.1016/j.bbadis.2013.05.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/04/2013] [Accepted: 05/13/2013] [Indexed: 01/04/2023]
Abstract
Activin receptor-like kinase-1 or ALK-1 is a type I cell surface receptor for the transforming growth factor-β (TGF-β) family of proteins. The role of ALK-1 in endothelial cells biology and in angiogenesis has been thoroughly studied by many authors. However, it has been recently suggested a possible role of ALK-1 in cardiovascular homeostasis. ALK-1 is not only expressed in endothelial cells but also in smooth muscle cells, myofibroblast, hepatic stellate cells, chondrocytes, monocytes, myoblasts, macrophages or fibroblasts, but its role in these cells have not been deeply analyzed. Due to the function of ALK-1 in these cells, this receptor plays a role in several cardiovascular diseases. Animals with ALK-1 haploinsufficiency and patients with mutations in Acvrl1 (the gene that codifies for ALK-1) develop type-2 Hereditary Hemorrhagic Telangiectasia. Moreover, ALK-1 heterozygous mice develop pulmonary hypertension. Higher levels of ALK-1 have been observed in atherosclerotic plaques, suggesting a possible protector role of this receptor. ALK-1 deficiency is also related to the development of arteriovenous malformations (AVMs). Besides, due to the ability of ALK-1 to regulate cell proliferation and migration, and to modulate extracellular matrix (ECM) protein expression in several cell types, ALK-1 has been now demonstrated to play an important role in cardiovascular remodeling. In this review, we would like to offer a complete vision of the role of ALK-1 in many process related to cardiovascular homeostasis, and the involvement of this protein in the development of cardiovascular diseases, suggesting the possibility of using the ALK-1/smad-1 pathway as a powerful therapeutic target.
Collapse
|
145
|
Choi EJ, Kim YH, Choe SW, Tak YG, Garrido-Martin EM, Chang M, Lee YJ, Oh SP. Enhanced responses to angiogenic cues underlie the pathogenesis of hereditary hemorrhagic telangiectasia 2. PLoS One 2013; 8:e63138. [PMID: 23675457 PMCID: PMC3651154 DOI: 10.1371/journal.pone.0063138] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/28/2013] [Indexed: 12/11/2022] Open
Abstract
Hereditary Hemorrhagic Telangiectasia (HHT) is a genetic vascular disease in which arteriovenous malformations (AVMs) manifest in skin and multiple visceral organs. HHT is caused by heterozygous mutations in endoglin (ENG), activin receptor-like kinase 1 (ALK1), or SMAD4. ALK1 regulates angiogenesis, but the precise function of ALK1 in endothelial cells (ECs) remains elusive. Since most blood vessels of HHT patients do not produce pathological vascular lesions, ALK1 heterozygous ECs may be normal unless additional genetic or environmental stresses are imposed. To investigate the cellular and biochemical phenotypes of Alk1-null versus Alk1-heterozygous ECs, we have generated pulmonary EC lines in which a genotype switch from the Alk1-conditional allele (Alk1 (2f)) to the Alk1-null allele (Alk1 (1f)) can be induced by tamoxifen treatment. Alk1-null (1 f/1 f) ECs displayed increased migratory properties in vitro in response to bFGF compared with Alk1-het (2 f/1 f) ECs. The 1 f/1 f-ECs formed a denser and more persistent tubular network as compared with their parental 2 f/1 f-ECs. Interestingly, the response to BMP-9 on SMAD1/5 phosphorylation was impaired in both 2 f/1 f- and 1 f/1 f-ECs at a comparable manner, suggesting that other factors in addition to SMADs may play a crucial role for enhanced angiogenic activity in 1 f/1 f-ECs. We also demonstrated in vivo that Alk1-deficient ECs exhibited high migratory and invasive properties. Taken together, these data suggest that enhanced responses to angiogenic cues in ALK1-deficient ECs underlie the pathogenesis of HHT2.
Collapse
Affiliation(s)
- Eun-Jung Choi
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Yong Hwan Kim
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Se-woon Choe
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Yu Gyoung Tak
- World Class University Program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Eva M. Garrido-Martin
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Myron Chang
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States of America
| | - Young Jae Lee
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- World Class University Program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - S. Paul Oh
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- World Class University Program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- * E-mail:
| |
Collapse
|
146
|
Seghers L, de Vries MR, Pardali E, Hoefer IE, Hierck BP, ten Dijke P, Goumans MJ, Quax PHA. Shear induced collateral artery growth modulated by endoglin but not by ALK1. J Cell Mol Med 2013; 16:2440-50. [PMID: 22436015 PMCID: PMC3823438 DOI: 10.1111/j.1582-4934.2012.01561.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) stimulates both ischaemia induced angiogenesis and shear stress induced arteriogenesis by signalling through different receptors. How these receptors are involved in both these processes of blood flow recovery is not entirely clear. In this study the role of TGF-β receptors 1 and endoglin is assessed in neovascularization in mice. Unilateral femoral artery ligation was performed in mice heterozygous for either endoglin or ALK1 and in littermate controls. Compared with littermate controls, blood flow recovery, monitored by laser Doppler perfusion imaging, was significantly hampered by maximal 40% in endoglin heterozygous mice and by maximal 49% in ALK1 heterozygous mice. Collateral artery size was significantly reduced in endoglin heterozygous mice compared with controls but not in ALK1 heterozygous mice. Capillary density in ischaemic calf muscles was unaffected, but capillaries from endoglin and ALK1 heterozygous mice were significantly larger when compared with controls. To provide mechanistic evidence for the differential role of endoglin and ALK1 in shear induced or ischaemia induced neovascularization, murine endothelial cells were exposed to shear stress in vitro. This induced increased levels of endoglin mRNA but not ALK1. In this study it is demonstrated that both endoglin and ALK1 facilitate blood flow recovery. Importantly, endoglin contributes to both shear induced collateral artery growth and to ischaemia induced angiogenesis, whereas ALK1 is only involved in ischaemia induced angiogenesis.
Collapse
Affiliation(s)
- Leonard Seghers
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Kowalski WJ, Dur O, Wang Y, Patrick MJ, Tinney JP, Keller BB, Pekkan K. Critical transitions in early embryonic aortic arch patterning and hemodynamics. PLoS One 2013; 8:e60271. [PMID: 23555940 PMCID: PMC3605337 DOI: 10.1371/journal.pone.0060271] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 02/25/2013] [Indexed: 02/01/2023] Open
Abstract
Transformation from the bilaterally symmetric embryonic aortic arches to the mature great vessels is a complex morphogenetic process, requiring both vasculogenic and angiogenic mechanisms. Early aortic arch development occurs simultaneously with rapid changes in pulsatile blood flow, ventricular function, and downstream impedance in both invertebrate and vertebrate species. These dynamic biomechanical environmental landscapes provide critical epigenetic cues for vascular growth and remodeling. In our previous work, we examined hemodynamic loading and aortic arch growth in the chick embryo at Hamburger-Hamilton stages 18 and 24. We provided the first quantitative correlation between wall shear stress (WSS) and aortic arch diameter in the developing embryo, and observed that these two stages contained different aortic arch patterns with no inter-embryo variation. In the present study, we investigate these biomechanical events in the intermediate stage 21 to determine insights into this critical transition. We performed fluorescent dye microinjections to identify aortic arch patterns and measured diameters using both injection recordings and high-resolution optical coherence tomography. Flow and WSS were quantified with 3D computational fluid dynamics (CFD). Dye injections revealed that the transition in aortic arch pattern is not a uniform process and multiple configurations were documented at stage 21. CFD analysis showed that WSS is substantially elevated compared to both the previous (stage 18) and subsequent (stage 24) developmental time-points. These results demonstrate that acute increases in WSS are followed by a period of vascular remodeling to restore normative hemodynamic loading. Fluctuations in blood flow are one possible mechanism that impacts the timing of events such as aortic arch regression and generation, leading to the variable configurations at stage 21. Aortic arch variations noted during normal rapid vascular remodeling at stage 21 identify a temporal window of increased vulnerability to aberrant aortic arch morphogenesis with the potential for profound effects on subsequent cardiovascular morphogenesis.
Collapse
Affiliation(s)
- William J. Kowalski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Onur Dur
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Yajuan Wang
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Michael J. Patrick
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Joseph P. Tinney
- Department of Pediatrics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
| | - Bradley B. Keller
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- Department of Pediatrics, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, United States of America
| | - Kerem Pekkan
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
148
|
Beets K, Huylebroeck D, Moya IM, Umans L, Zwijsen A. Robustness in angiogenesis: notch and BMP shaping waves. Trends Genet 2012; 29:140-9. [PMID: 23279848 DOI: 10.1016/j.tig.2012.11.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 10/31/2012] [Accepted: 11/16/2012] [Indexed: 12/20/2022]
Abstract
Vascular patterning involves sprouting of blood vessels, which is governed by orchestrated communication between cells in the surrounding tissue and endothelial cells (ECs) lining the blood vessels. Single ECs are selected for sprouting by hypoxia-induced stimuli and become the 'tip' or leader cell that guides new sprouts. The 'stalk' or trailing ECs proliferate for tube extension and lumenize the nascent vessel. Stalk and tip cells can dynamically switch their identities during this process in a Notch-dependent manner. Here, we review recent studies showing that bone morphogenetic protein (BMP) signaling coregulates Notch target genes in ECs. In particular, we focus on how Delta-like ligand 4 (DLL4)-Notch and BMP effector interplay may drive nonsynchronized oscillatory gene expression in ECs essential for setting sharp tip-stalk cell boundaries while sustaining a dynamic pool of nonsprouting ECs. Deeper knowledge about the coregulation of vessel plasticity in different vascular beds may result in refinement of anti-angiogenesis and vessel normalization therapies.
Collapse
Affiliation(s)
- Karen Beets
- Laboratory of Developmental Signaling, VIB Center for the Biology of Disease, VIB, 3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|
149
|
Abstract
BMP9 signaling has been implicated in hereditary hemorrhagic telangiectasia (HHT) and vascular remodeling, acting via the HHT target genes, endoglin and ALK1. This study sought to identify endothelial BMP9-regulated proteins that could affect the HHT phenotype. Gene ontology analysis of cDNA microarray data obtained after BMP9 treatment of primary human endothelial cells indicated regulation of chemokine, adhesion, and inflammation pathways. These responses included the up-regulation of the chemokine CXCL12/SDF1 and down-regulation of its receptor CXCR4. Quantitative mass spectrometry identified additional secreted proteins, including the chemokine CXCL10/IP10. RNA knockdown of endoglin and ALK1 impaired SDF1/CXCR4 regulation by BMP9. Because of the association of SDF1 with ischemia, we analyzed its expression under hypoxia in response to BMP9 in vitro, and during the response to hindlimb ischemia, in endoglin-deficient mice. BMP9 and hypoxia were additive inducers of SDF1 expression. Moreover, the data suggest that endoglin deficiency impaired SDF1 expression in endothelial cells in vivo. Our data implicate BMP9 in regulation of the SDF1/CXCR4 chemokine axis in endothelial cells and point to a role for BMP9 signaling via endoglin in a switch from an SDF1-responsive autocrine phenotype to an SDF1 nonresponsive paracrine state that represses endothelial cell migration and may promote vessel maturation.
Collapse
|
150
|
Watkins SC, Maniar S, Mosher M, Roman BL, Tsang M, St Croix CM. High resolution imaging of vascular function in zebrafish. PLoS One 2012; 7:e44018. [PMID: 22952858 PMCID: PMC3431338 DOI: 10.1371/journal.pone.0044018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 07/30/2012] [Indexed: 12/19/2022] Open
Abstract
Rationale The role of the endothelium in the pathogenesis of cardiovascular disease is an emerging field of study, necessitating the development of appropriate model systems and methodologies to investigate the multifaceted nature of endothelial dysfunction including disturbed barrier function and impaired vascular reactivity. Objective We aimed to develop and test an optimized high-speed imaging platform to obtain quantitative real-time measures of blood flow, vessel diameter and endothelial barrier function in order to assess vascular function in live vertebrate models. Methods and Results We used a combination of cutting-edge optical imaging techniques, including high-speed, camera-based imaging (up to 1000 frames/second), and 3D confocal methods to collect real time metrics of vascular performance and assess the dynamic response to the thromboxane A2 (TXA2) analogue, U-46619 (1 µM), in transgenic zebrafish larvae. Data obtained in 3 and 5 day post-fertilization larvae show that these methods are capable of imaging blood flow in a large (1 mm) segment of the vessel of interest over many cardiac cycles, with sufficient speed and sensitivity such that the trajectories of individual erythrocytes can be resolved in real time. Further, we are able to map changes in the three dimensional sizes of vessels and assess barrier function by visualizing the continuity of the endothelial layer combined with measurements of extravasation of fluorescent microspheres. Conclusions We propose that this system-based microscopic approach can be used to combine measures of physiologic function with molecular behavior in zebrafish models of human vascular disease.
Collapse
Affiliation(s)
- Simon C. Watkins
- Department of Cell Biology, The University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Salony Maniar
- Department of Environmental and Occupational Health, The University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mackenzie Mosher
- Department of Environmental and Occupational Health, The University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Beth L. Roman
- Department of Biological Sciences, The University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michael Tsang
- Department of Developmental Biology, The University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Claudette M. St Croix
- Department of Cell Biology, The University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Environmental and Occupational Health, The University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|