Notch Signaling in Vascular Smooth Muscle Cells.

[1]  M. Irving,et al.  End‐Stage Renal Disease in an Infant With Hajdu‐Cheney Syndrome , 2016, Therapeutic apheresis and dialysis : official peer-reviewed journal of the International Society for Apheresis, the Japanese Society for Apheresis, the Japanese Society for Dialysis Therapy.

[2]  T. Gridley,et al.  Notch signal reception is required in vascular smooth muscle cells for ductus arteriosus closure , 2016, Genesis.

[3]  J. T. Baeten,et al.  Loss of Notch2 and Notch3 in vascular smooth muscle causes patent ductus arteriosus , 2015, Genesis.

[4]  J. Kitajewski,et al.  Combined deficiency of Notch1 and Notch3 causes pericyte dysfunction, models CADASIL, and results in arteriovenous malformations , 2015, Scientific Reports.

[5]  I. Weissman,et al.  Pericytes are progenitors for coronary artery smooth muscle , 2015, eLife.

[6]  J. Roach,et al.  Heterozygous Loss-of-Function Mutations in DLL4 Cause Adams-Oliver Syndrome. , 2015, American journal of human genetics.

[7]  F. Gao,et al.  The Notch pathway mediates the angiotensin II-induced synthesis of extracellular matrix components in podocytes. , 2015, International journal of molecular medicine.

[8]  J. T. Baeten,et al.  Differential Regulation of NOTCH2 and NOTCH3 Contribute to Their Unique Functions in Vascular Smooth Muscle Cells* , 2015, The Journal of Biological Chemistry.

[9]  R. Lodi,et al.  Homozygous NOTCH3 null mutation and impaired NOTCH3 signaling in recessive early-onset arteriopathy and cavitating leukoencephalopathy , 2015, EMBO molecular medicine.

[10]  U. Lendahl,et al.  Notch3 Is Necessary for Blood Vessel Integrity in the Central Nervous System , 2015, Arteriosclerosis, thrombosis, and vascular biology.

[11]  J. Kitajewski,et al.  NOTCH decoys that selectively block DLL/NOTCH or JAG/NOTCH disrupt angiogenesis by unique mechanisms to inhibit tumor growth. , 2015, Cancer discovery.

[12]  E. Zackai,et al.  Truncating mutations in the last exon of NOTCH3 cause lateral meningocele syndrome , 2015, American journal of medical genetics. Part A.

[13]  B. Lilly,et al.  MicroRNA miR145 Regulates TGFBR2 Expression and Matrix Synthesis in Vascular Smooth Muscle Cells , 2015, Circulation research.

[14]  Zhenyi Liu,et al.  The intracellular domains of Notch1 and Notch2 are functionally equivalent during development and carcinogenesis , 2015, Development.

[15]  B. Osborne,et al.  Non-Canonical Notch Signaling in Cancer and Immunity , 2014, Front. Oncol..

[16]  Sarah M. Peterson,et al.  Notch signal integration in the vasculature during remodeling. , 2014, Vascular pharmacology.

[17]  B. Lilly,et al.  Notch signaling governs phenotypic modulation of smooth muscle cells. , 2014, Vascular pharmacology.

[18]  B. Lilly,et al.  Endothelial cells direct mesenchymal stem cells toward a smooth muscle cell fate. , 2014, Stem cells and development.

[19]  Michael B Elowitz,et al.  Fringe proteins modulate Notch-ligand cis and trans interactions to specify signaling states , 2014, eLife.

[20]  J. Roach,et al.  Mutations in NOTCH1 cause Adams-Oliver syndrome. , 2014, American journal of human genetics.

[21]  M. Graham,et al.  Notch signaling functions in retinal pericyte survival. , 2014, Investigative ophthalmology & visual science.

[22]  Zhongjie Sun,et al.  Molecular Mechanisms of Pulmonary Arterial Remodeling , 2014, Molecular medicine.

[23]  Hiroki Sato,et al.  Mutations of NOTCH3 in childhood pulmonary arterial hypertension , 2014, Molecular genetics & genomic medicine.

[24]  C. Moens,et al.  Notch3 establishes brain vascular integrity by regulating pericyte number , 2014, Development.

[25]  Wei Wang,et al.  Soluble Jagged1 Inhibits Pulmonary Hypertension by Attenuating Notch Signaling , 2013, Arteriosclerosis, thrombosis, and vascular biology.

[26]  J. Dauwerse,et al.  Hypomorphic NOTCH3 Alleles Do Not Cause CADASIL in Humans , 2013, Human mutation.

[27]  Jw Lee Mutations in PDGFRB and NOTCH3 are the first genetic causes identified for autosomal dominant infantile myofibromatosis , 2013, Clinical genetics.

[28]  Michael M. Wang,et al.  Von Willebrand Factor Inhibits Mature Smooth Muscle Gene Expression through Impairment of Notch Signaling , 2013, PloS one.

[29]  L. Liaw,et al.  A Receptor-Specific Function for Notch2 in Mediating Vascular Smooth Muscle Cell Growth Arrest Through Cyclin-dependent Kinase Inhibitor 1B , 2013, Circulation research.

[30]  Yiran Guo,et al.  Mutations in PDGFRB cause autosomal-dominant infantile myofibromatosis. , 2013, American journal of human genetics.

[31]  F. Alkuraya,et al.  Mutations in EOGT confirm the genetic heterogeneity of autosomal-recessive Adams-Oliver syndrome. , 2013, American journal of human genetics.

[32]  M. Lampugnani,et al.  Dll4 and PDGF-BB convert committed skeletal myoblasts to pericytes without erasing their myogenic memory. , 2013, Developmental cell.

[33]  M. Krasnow,et al.  Radial construction of an arterial wall. , 2012, Developmental cell.

[34]  P. Gaffney,et al.  RBPJ mutations identified in two families affected by Adams-Oliver syndrome. , 2012, American journal of human genetics.

[35]  G. Owens,et al.  Smooth muscle cell phenotypic switching in atherosclerosis. , 2012, Cardiovascular research.

[36]  A. Joutel,et al.  Notch signalling in smooth muscle cells during development and disease. , 2012, Cardiovascular research.

[37]  S. George,et al.  Wnt signalling in smooth muscle cells and its role in cardiovascular disorders. , 2012, Cardiovascular research.

[38]  B. Lilly,et al.  Notch2 and Notch3 Function Together to Regulate Vascular Smooth Muscle Development , 2012, PloS one.

[39]  Hideki Uosaki,et al.  Non-canonical Notch signaling: emerging role and mechanism. , 2012, Trends in cell biology.

[40]  Shi-You Chen,et al.  Transforming growth factor-β and smooth muscle differentiation. , 2012, World journal of biological chemistry.

[41]  M. Iruela-Arispe,et al.  Notch promotes vascular maturation by inducing integrin-mediated smooth muscle cell adhesion to the endothelial basement membrane. , 2012, Blood.

[42]  G. Owens,et al.  Epigenetic control of smooth muscle cell differentiation and phenotypic switching in vascular development and disease. , 2012, Annual review of physiology.

[43]  M. Majesky,et al.  Patterning the artery wall by lateral induction of Notch signaling. , 2012, Circulation.

[44]  J. Epstein,et al.  Notch Activation of Jagged1 Contributes to the Assembly of the Arterial Wall , 2012, Circulation.

[45]  E. Boscolo,et al.  JAGGED1 Signaling Regulates Hemangioma Stem Cell–to–Pericyte/Vascular Smooth Muscle Cell Differentiation , 2011, Arteriosclerosis, thrombosis, and vascular biology.

[46]  C. Betsholtz,et al.  Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. , 2011, Developmental cell.

[47]  Chulan Kwon,et al.  Notch Post-Translationally Regulates β-Catenin Protein in Stem and Progenitor Cells , 2011, Nature Cell Biology.

[48]  S. Patel-Hett,et al.  Signal transduction in vasculogenesis and developmental angiogenesis. , 2011, The International journal of developmental biology.

[49]  Chunxiang Zhang,et al.  The miR-143/145 Cluster Is a Novel Transcriptional Target of Jagged-1/Notch Signaling in Vascular Smooth Muscle Cells* , 2011, The Journal of Biological Chemistry.

[50]  A. Kispert,et al.  Notch Signaling Regulates Smooth Muscle Differentiation of Epicardium-Derived Cells , 2011, Circulation research.

[51]  S. Robertson,et al.  Mutations in NOTCH2 cause Hajdu-Cheney syndrome, a disorder of severe and progressive bone loss , 2011, Nature Genetics.

[52]  Oliver Distler,et al.  Notch signalling regulates fibroblast activation and collagen release in systemic sclerosis , 2011, Annals of the rheumatic diseases.

[53]  Youliang Wang,et al.  Endothelial Smad4 maintains cerebrovascular integrity by activating N-cadherin through cooperation with Notch. , 2011, Developmental cell.

[54]  T. Gridley,et al.  Patent ductus arteriosus in mice with smooth muscle-specific Jag1 deletion , 2010, Development.

[55]  B. Lilly,et al.  Notch3 Is Critical for Proper Angiogenesis and Mural Cell Investment , 2010, Circulation Research.

[56]  Rajendran Sanalkumar,et al.  Non-canonical activation of Notch signaling/target genes in vertebrates , 2010, Cellular and Molecular Life Sciences.

[57]  L. Liaw,et al.  Notch and Transforming Growth Factor-β (TGFβ) Signaling Pathways Cooperatively Regulate Vascular Smooth Muscle Cell Differentiation* , 2010, The Journal of Biological Chemistry.

[58]  D. Schaffer,et al.  Transforming Growth Factor‐β and Notch Signaling Mediate Stem Cell Differentiation into Smooth Muscle Cells , 2010, Stem cells.

[59]  A. Schedl,et al.  Cerebrovascular dysfunction and microcirculation rarefaction precede white matter lesions in a mouse genetic model of cerebral ischemic small vessel disease. , 2010, The Journal of clinical investigation.

[60]  Cuihua Zhang,et al.  Neointimal hyperplasia, vein graft remodeling, and long-term patency. , 2009, American journal of physiology. Heart and circulatory physiology.

[61]  R. Mecham,et al.  Vascular extracellular matrix and arterial mechanics. , 2009, Physiological reviews.

[62]  A. Gilmore,et al.  Notch activation induces Akt signaling via an autocrine loop to prevent apoptosis in breast epithelial cells. , 2009, Cancer research.

[63]  Marcus Fruttiger,et al.  The Notch Ligands Dll4 and Jagged1 Have Opposing Effects on Angiogenesis , 2009, Cell.

[64]  J. Liao,et al.  Smooth Muscle Notch1 Mediates Neointimal Formation After Vascular Injury , 2009, Circulation.

[65]  C. Betsholtz,et al.  Endothelial-mural cell signaling in vascular development and angiogenesis. , 2009, Arteriosclerosis, thrombosis, and vascular biology.

[66]  S. Artavanis-Tsakonas,et al.  Notch and Wnt signals cooperatively control cell proliferation and tumorigenesis in the intestine , 2009, Proceedings of the National Academy of Sciences.

[67]  M. Hirashima Regulation of endothelial cell differentiation and arterial specification by VEGF and Notch signaling , 2009, Anatomical science international.

[68]  B. Lilly,et al.  NOTCH3 Expression Is Induced in Mural Cells Through an Autoregulatory Loop That Requires Endothelial-Expressed JAGGED1 , 2009, Circulation research.

[69]  M. Majesky,et al.  Building a vessel wall with notch signaling. , 2009, Circulation research.

[70]  Holger Gerhardt,et al.  Angiogenesis: a team effort coordinated by notch. , 2009, Developmental cell.

[71]  M. Shin,et al.  Notch mediates Wnt and BMP signals in the early separation of smooth muscle progenitors and blood/endothelial common progenitors , 2009, Development.

[72]  I. Screpanti,et al.  Constitutively activated Notch signaling is involved in survival and apoptosis resistance of B-CLL cells. , 2009, Blood.

[73]  F. Oswald,et al.  The Notch signaling pathway: Transcriptional regulation at Notch target genes , 2009, Cellular and Molecular Life Sciences.

[74]  Rong A. Wang,et al.  Artery and vein size is balanced by Notch and ephrin B2/EphB4 during angiogenesis , 2008, Development.

[75]  Fredrik Lanner,et al.  Notch Signaling Regulates Platelet-Derived Growth Factor Receptor-β Expression in Vascular Smooth Muscle Cells , 2008, Circulation research.

[76]  G. Béréziat,et al.  The benefit of docosahexanoic acid on the migration of vascular smooth muscle cells is partially dependent on Notch regulation of MMP-2/-9. , 2008, The American journal of pathology.

[77]  D. Despres,et al.  Notch2 is required for the proliferation of cardiac neural crest‐derived smooth muscle cells , 2008, Developmental dynamics : an official publication of the American Association of Anatomists.

[78]  YuefengTang,et al.  Hairy-Related Transcription Factors Inhibit Notch-Induced Smooth Muscle α-Actin Expression by Interfering With Notch Intracellular Domain/CBF-1 Complex Interaction With the CBF-1–Binding Site , 2008 .

[79]  K. Kaestner,et al.  Endothelial expression of the Notch ligand Jagged1 is required for vascular smooth muscle development , 2008, Proceedings of the National Academy of Sciences.

[80]  J. Epstein,et al.  Myocardin regulates expression of contractile genes in smooth muscle cells and is required for closure of the ductus arteriosus in mice. , 2008, The Journal of clinical investigation.

[81]  B. Lilly,et al.  Transforming Growth Factor-β (TGF-β1) Down-regulates Notch3 in Fibroblasts to Promote Smooth Muscle Gene Expression* , 2008, Journal of Biological Chemistry.

[82]  Timothy B Sackton,et al.  Mutations in smooth muscle α-actin (ACTA2) lead to thoracic aortic aneurysms and dissections , 2007, Nature Genetics.

[83]  R. Adams,et al.  Regulation of vascular morphogenesis by Notch signaling. , 2007, Genes & development.

[84]  James D. Johnson,et al.  Notch signalling suppresses apoptosis in adult human and mouse pancreatic islet cells , 2007, Diabetologia.

[85]  C. Haslett,et al.  Mammalian NOTCH-1 Activates β1 Integrins via the Small GTPase R-Ras* , 2007, Journal of Biological Chemistry.

[86]  T. Gridley Notch signaling in vascular development and physiology , 2007, Development.

[87]  J. Epstein,et al.  An essential role for Notch in neural crest during cardiovascular development and smooth muscle differentiation. , 2007, The Journal of clinical investigation.

[88]  Alfonso Martinez Arias,et al.  Notch, a Universal Arbiter of Cell Fate Decisions , 2006, Science.

[89]  Hiroki Matsui,et al.  Jagged1-selective Notch Signaling Induces Smooth Muscle Differentiation via a RBP-Jκ-dependent Pathway* , 2006, Journal of Biological Chemistry.

[90]  L. Liaw,et al.  A novel mechanism of transcriptional repression of p27kip1 through Notch/HRT2 signaling in vascular smooth muscle cells , 2006, Thrombosis and Haemostasis.

[91]  I. Krantz,et al.  NOTCH2 mutations cause Alagille syndrome, a heterogeneous disorder of the notch signaling pathway. , 2006, American journal of human genetics.

[92]  A. Karsan,et al.  Smooth Muscle &agr;-Actin Is a Direct Target of Notch/CSL , 2006 .

[93]  D. Srivastava,et al.  Hrt and Hes negatively regulate Notch signaling through interactions with RBP-Jkappa. , 2006, Biochemical and biophysical research communications.

[94]  G. Stormo,et al.  Target Selectivity of Vertebrate Notch Proteins , 2006, Journal of Biological Chemistry.

[95]  A. Lalande,et al.  Mutations in myosin heavy chain 11 cause a syndrome associating thoracic aortic aneurysm/aortic dissection and patent ductus arteriosus , 2006, Nature Genetics.

[96]  L. Kedes,et al.  HERP1 Inhibits Myocardin-Induced Vascular Smooth Muscle Cell Differentiation by Interfering With SRF Binding to CArG Box , 2005, Arteriosclerosis, thrombosis, and vascular biology.

[97]  D. Srivastava,et al.  Mutations in NOTCH1 cause aortic valve disease , 2005, Nature.

[98]  Tan Xiao-mei,et al.  CADASIL , 2005 .

[99]  N. Perrimon,et al.  Notch modulates Wnt signalling by associating with Armadillo/β-catenin and regulating its transcriptional activity , 2005, Development.

[100]  W. Pear,et al.  Notch Signaling Represses Myocardin-induced Smooth Muscle Cell Differentiation* , 2005, Journal of Biological Chemistry.

[101]  Tannishtha Reya,et al.  Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance , 2005, Nature Immunology.

[102]  Cameron S. Osborne,et al.  Molecular Determinants of NOTCH4 Transcription in Vascular Endothelium , 2005, Molecular and Cellular Biology.

[103]  M. Viitanen,et al.  Mice carrying a R142C Notch 3 knock–in mutation do not develop a CADASIL‐like phenotype , 2005, Genesis.

[104]  B. Klonjkowski,et al.  Notch3 is required for arterial identity and maturation of vascular smooth muscle cells. , 2004, Genes & development.

[105]  D. Simon,et al.  Transcription Factor CHF1/Hey2 Regulates Neointimal Formation In Vivo and Vascular Smooth Muscle Proliferation and Migration In Vitro , 2004, Arteriosclerosis, thrombosis, and vascular biology.

[106]  P. Cahill,et al.  Notch 1 and 3 receptors modulate vascular smooth muscle cell growth, apoptosis and migration via a CBF‐1/RBP‐Jk dependent pathway , 2004, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[107]  G. Owens,et al.  Molecular regulation of vascular smooth muscle cell differentiation in development and disease. , 2004, Physiological reviews.

[108]  U. Lendahl,et al.  Cross-talk between the Notch and TGF-β signaling pathways mediated by interaction of the Notch intracellular domain with Smad3 , 2003, The Journal of cell biology.

[109]  U. Lendahl,et al.  Characterization of Notch3‐deficient mice: Normal embryonic development and absence of genetic interactions with a Notch1 mutation , 2003, Genesis.

[110]  L. Espinosa,et al.  Phosphorylation by Glycogen Synthase Kinase-3β Down-regulates Notch Activity, a Link for Notch and Wnt Pathways* , 2003, Journal of Biological Chemistry.

[111]  Da-Zhi Wang,et al.  Myocardin Is a Key Regulator of CArG-Dependent Transcription of Multiple Smooth Muscle Marker Genes , 2003, Circulation research.

[112]  L. Kedes,et al.  Notch Signaling in Vascular Development , 2003, Arteriosclerosis, thrombosis, and vascular biology.

[113]  Wenli Wang,et al.  Determinants of Notch-3 Receptor Expression and Signaling in Vascular Smooth Muscle Cells: Implications in Cell-Cycle Regulation , 2002, Circulation research.

[114]  I. Krantz,et al.  Analysis of Cardiovascular Phenotype and Genotype-Phenotype Correlation in Individuals With a JAG1 Mutation and/or Alagille Syndrome , 2002, Circulation.

[115]  Keith Brennan,et al.  CSL-independent Notch signalling: a checkpoint in cell fate decisions during development? , 2002, Current opinion in genetics & development.

[116]  Wenli Wang,et al.  Coordinate Notch3-Hairy-related Transcription Factor Pathway Regulation in Response to Arterial Injury , 2002, The Journal of Biological Chemistry.

[117]  Wenli Wang,et al.  Notch3 Signaling in Vascular Smooth Muscle Cells Induces c-FLIP Expression via ERK/MAPK Activation , 2002, The Journal of Biological Chemistry.

[118]  E. Lai Protein Degradation: Four E3s For The Notch Pathway , 2002, Current Biology.

[119]  J. Campos-Ortega,et al.  Notch signaling is required for arterial-venous differentiation during embryonic vascular development. , 2001, Development.

[120]  G. Weinmaster,et al.  Vascular expression of Notch pathway receptors and ligands is restricted to arterial vessels , 2001, Mechanisms of Development.

[121]  L. Liaw,et al.  Members of the Jagged/Notch gene families are expressed in injured arteries and regulate cell phenotype via alterations in cell matrix and cell-cell interaction. , 2001, The American journal of pathology.

[122]  C. Giannakopoulou,et al.  Infantile Myofibromatosis with Visceral Involvement and Complete Spontaneous Regression , 2001, The Journal of dermatology.

[123]  M. Cobb,et al.  Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. , 2001, Endocrine reviews.

[124]  N. Voelkel,et al.  Gene Expression Patterns in the Lungs of Patients With Primary Pulmonary Hypertension: A Gene Microarray Analysis , 2001, Circulation research.

[125]  G. Weinmaster,et al.  Defects in development of the kidney, heart and eye vasculature in mice homozygous for a hypomorphic Notch2 mutation. , 2001, Development.

[126]  A. Hamosh,et al.  Familial Tetralogy of Fallot caused by mutation in the jagged1 gene. , 2001, Human molecular genetics.

[127]  D. Srivastava,et al.  Members of the HRT family of basic helix-loop-helix proteins act as transcriptional repressors downstream of Notch signaling. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[128]  G. Weinmaster,et al.  Fringe differentially modulates Jagged1 and Delta1 signalling through Notch1 and Notch2 , 2000, Nature Cell Biology.

[129]  Yang Wang,et al.  Fringe is a glycosyltransferase that modifies Notch , 2000, Nature.

[130]  W. Richards,et al.  Dll4, a novel Notch ligand expressed in arterial endothelium. , 2000, Genes & development.

[131]  F. Chapon,et al.  The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. , 2000, The Journal of clinical investigation.

[132]  C. Betsholtz,et al.  Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. , 1999, Development.

[133]  M. H. Angelis,et al.  Expression of the mouse Delta1 gene during organogenesis and fetal development , 1999, Mechanisms of Development.

[134]  S. Artavanis-Tsakonas,et al.  Notch signaling: cell fate control and signal integration in development. , 1999, Science.

[135]  W. Pear,et al.  Cutting edge: protective effects of notch-1 on TCR-induced apoptosis. , 1999, Journal of immunology.

[136]  P. Underwood,et al.  Inhibition of endothelial cell adhesion and proliferation by extracellular matrix from vascular smooth muscle cells: role of type V collagen. , 1998, Atherosclerosis.

[137]  David J. Anderson,et al.  Molecular Distinction and Angiogenic Interaction between Embryonic Arteries and Veins Revealed by ephrin-B2 and Its Receptor Eph-B4 , 1998, Cell.

[138]  F. Kuo,et al.  Isolation and functional analysis of a cDNA for human Jagged2, a gene encoding a ligand for the Notch1 receptor , 1997, Molecular and cellular biology.

[139]  J. Weissenbach,et al.  Notch3 Mutations in Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), a Mendelian Condition Causing Stroke and Vascular Dementia , 1997, Annals of the New York Academy of Sciences.

[140]  Colin C. Collins,et al.  Alagille syndrome is caused by mutations in human Jagged1, which encodes a ligand for Notch1 , 1997, Nature Genetics.

[141]  J. Weissenbach,et al.  Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia , 1996, Nature.

[142]  R. Kageyama,et al.  Structure, chromosomal locus, and promoter analysis of the gene encoding the mouse helix-loop-helix factor HES-1. Negative autoregulation through the multiple N box elements. , 1994, The Journal of biological chemistry.

[143]  P. Simpson,et al.  The choice of cell fate in the epidermis of Drosophila , 1991, Cell.

[144]  R. Ross,et al.  A significant part of macrophage-derived growth factor consists of at least two forms of PDGF , 1985, Cell.

[145]  J. P. Wallace,et al.  References 1 , 1961 .

[146]  R. Punnett,et al.  The Theory of the Gene , 1926, Nature.

[147]  G. E. Twente,et al.  Patent ductus arteriosus , 2013 .

[148]  D. Abraham,et al.  Platelet-derived growth factor signaling in mesenchymal cells. , 2013, Frontiers in bioscience.

[149]  Hollis G. Potter,et al.  Author Manuscript , 2013 .

[150]  L. Liaw,et al.  muscle cells , 2012 .

[151]  T. Dang Notch, apoptosis and cancer. , 2012, Advances in experimental medicine and biology.

[152]  L. Liaw,et al.  Notch and Transforming Growth Factor-( TGF ) Signaling Pathways Cooperatively Regulate Vascular Smooth Muscle Cell Differentiation * , 2010 .

[153]  T. Gridley Notch signaling in the vasculature. , 2010, Current topics in developmental biology.

[154]  P. Stanley,et al.  Roles of glycosylation in Notch signaling. , 2010, Current topics in developmental biology.

[155]  B. Lilly,et al.  Differential gene expression in a coculture model of angiogenesis reveals modulation of select pathways and a role for Notch signaling. , 2009, Physiological genomics.

[156]  A. Karsan,et al.  Smooth Muscle alpha-actin is a direct target of Notch/CSL. , 2006, Circulation research.

[157]  I. Krantz,et al.  Jagged1 mutations in Alagille syndrome , 2001, Human mutation.