101
|
Koenig SN, Bosse K, Majumdar U, Bonachea EM, Radtke F, Garg V. Endothelial Notch1 Is Required for Proper Development of the Semilunar Valves and Cardiac Outflow Tract. J Am Heart Assoc 2016; 5:e003075. [PMID: 27107132 PMCID: PMC4843530 DOI: 10.1161/jaha.115.003075] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/16/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Congenital heart disease is the most common type of birth defect, affecting ≈2% of the population. Malformations involving the cardiac outflow tract and semilunar valves account for >50% of these cases predominantly because of a bicuspid aortic valve, which has an estimated prevalence of 1% in the population. We previously reported that mutations in NOTCH1 were a cause of bicuspid aortic valve in nonsyndromic autosomal-dominant human pedigrees. Subsequently, we described a highly penetrant mouse model of aortic valve disease, consisting of a bicuspid aortic valve with thickened cusps and associated stenosis and regurgitation, in Notch1-haploinsufficient adult mice backcrossed into a Nos3-null background. METHODS AND RESULTS Here, we described the congenital cardiac abnormalities in Notch1(+/-);Nos3(-/-) embryos that led to ≈65% lethality by postnatal day 10. Although expected Mendelian ratios of Notch1(+/-);Nos3(-/-) embryos were found at embryonic day 18.5, histological examination revealed thickened, malformed semilunar valve leaflets accompanied by additional anomalies of the cardiac outflow tract including ventricular septal defects and overriding aorta. The aortic valve leaflets of Notch1(+/-);Nos3(-/-) embryos at embryonic day 15.5 were significantly thicker than controls, consistent with a defect in remodeling of the semilunar valve cushions. In addition, we generated mice haploinsufficient for Notch1 specifically in endothelial and endothelial-derived cells in a Nos3-null background and found that Notch1(fl/+);Tie2-Cre(+/-);Nos3(-/-) mice recapitulate the congenital cardiac phenotype of Notch1(+/-);Nos3(-/-) embryos. CONCLUSIONS Our data demonstrate the role of endothelial Notch1 in the proper development of the semilunar valves and cardiac outflow tract.
Collapse
Affiliation(s)
- Sara N Koenig
- Center for Cardiovascular and Pulmonary Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH
| | - Kevin Bosse
- Center for Cardiovascular and Pulmonary Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH
| | - Uddalak Majumdar
- Center for Cardiovascular and Pulmonary Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH
| | | | - Freddy Radtke
- Ecole Polytechnique Fédérale de Lausanne, Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Vidu Garg
- Center for Cardiovascular and Pulmonary Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH Department of Pediatrics, The Ohio State University, Columbus, OH Department of Molecular Genetics, The Ohio State University, Columbus, OH
| |
Collapse
|
102
|
Kanz D, Konantz M, Alghisi E, North TE, Lengerke C. Endothelial-to-hematopoietic transition: Notch-ing vessels into blood. Ann N Y Acad Sci 2016; 1370:97-108. [DOI: 10.1111/nyas.13030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Dirk Kanz
- Department of Stem Cell and Regenerative Biology; Harvard University; Boston Massachusetts
| | - Martina Konantz
- Department of Biomedicine; University Hospital Basel; Basel Switzerland
| | - Elisa Alghisi
- Department of Biomedicine; University Hospital Basel; Basel Switzerland
| | - Trista E. North
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston Massachusetts
- Harvard Stem Cell Institute; Cambridge Massachusetts
| | - Claudia Lengerke
- Department of Biomedicine; University Hospital Basel; Basel Switzerland
- Division of Hematology; University Hospital Basel; Basel Switzerland
| |
Collapse
|
103
|
Valve Interstitial Cells Act in a Pericyte Manner Promoting Angiogensis and Invasion by Valve Endothelial Cells. Ann Biomed Eng 2016; 44:2707-23. [PMID: 26905695 PMCID: PMC4983529 DOI: 10.1007/s10439-016-1567-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/09/2016] [Indexed: 12/24/2022]
Abstract
Neovascularization is an understudied aspect of calcific aortic valve disease (CAVD). Within diseased valves, cells along the neovessels' periphery stain for pericyte markers, but it is unclear whether valvular interstitial cells (VICs) can demonstrate a pericyte-like phenotype. This investigation examined the perivascular potential of VICs to regulate valve endothelial cell (VEC) organization and explored the role of Angiopoeitin1-Tie2 signaling in this process. Porcine VECs and VICs were fluorescently tracked and co-cultured in Matrigel over 7 days. VICs regulated early VEC network organization in a ROCK-dependent manner, then guided later VEC network contraction through chemoattraction. Unlike vascular control cells, the valve cell cultures ultimately formed invasive spheroids with 3D angiogenic-like sprouts. VECs co-cultured with VICs displayed significantly more invasion than VECs alone; with VICs generally leading and wrapping around VEC invasive sprouts. Lastly, Angiopoietin1-Tie2 signaling was found to regulate valve cell organization during VEC/VIC spheroid formation and invasion. VICs demonstrated pericyte-like behaviors toward VECs throughout sustained co-culture. The change from a vasculogenic network to an invasive sprouting spheroid suggests that both cell types undergo phenotypic changes during long-term culture in the model angiogenic environment. Valve cells organizing into spheroids and undergoing 3D invasion of Matrigel demonstrated several typical angiogenic-like phenotypes dependent on basal levels of Angiopoeitin1-Tie2 signaling and ROCK activation. These results suggest that the ectopic sustained angiogenic environment during the early stages of valve disease promotes organized activity by both VECs and VICs, contributing to neovessel formation and the progression of CAVD.
Collapse
|
104
|
Huk DJ, Austin BF, Horne TE, Hinton RB, Ray WC, Heistad DD, Lincoln J. Valve Endothelial Cell-Derived Tgfβ1 Signaling Promotes Nuclear Localization of Sox9 in Interstitial Cells Associated With Attenuated Calcification. Arterioscler Thromb Vasc Biol 2015; 36:328-38. [PMID: 26634652 DOI: 10.1161/atvbaha.115.306091] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 11/18/2015] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Aortic valve disease, including calcification, affects >2% of the human population and is caused by complex interactions between multiple risk factors, including genetic mutations, the environment, and biomechanics. At present, there are no effective treatments other than surgery, and this is because of the limited understanding of the mechanisms that underlie the condition. Previous work has shown that valve interstitial cells within the aortic valve cusps differentiate toward an osteoblast-like cell and deposit bone-like matrix that leads to leaflet stiffening and calcific aortic valve stenosis. However, the mechanisms that promote pathological phenotypes in valve interstitial cells are unknown. APPROACH AND RESULTS Using a combination of in vitro and in vivo tools with mouse, porcine, and human tissue, we show that in valve interstitial cells, reduced Sox9 expression and nuclear localization precedes the onset of calcification. In vitro, Sox9 nuclear export and calcific nodule formation is prevented by valve endothelial cells. However, in vivo, loss of Tgfβ1 in the endothelium leads to reduced Sox9 expression and calcific aortic valve disease. CONCLUSIONS Together, these findings suggest that reduced nuclear localization of Sox9 in valve interstitial cells is an early indicator of calcification, and therefore, pharmacological targeting to prevent nuclear export could serve as a novel therapeutic tool in the prevention of calcification and stenosis.
Collapse
Affiliation(s)
- Danielle J Huk
- From the Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, Miami, FL (D.J.H.); Center for Cardiovascular Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, OH (D.J.H., B.F.A., T.E.H., J.L.); Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (R.B.H.); Battelle Center for Mathematical Medicine, Nationwide Children's Hospital Research Institute, Columbus, OH (W.C.R.); The Ohio State University Interdisciplinary Graduate Program in Biophysics, Columbus, OH (W.C.R.); Department of Pediatrics, The Ohio State University, Columbus, OH (W.C.R., J.L.); and Division of Cardiovascular Medicine and Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA (D.D.H.)
| | - Blair F Austin
- From the Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, Miami, FL (D.J.H.); Center for Cardiovascular Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, OH (D.J.H., B.F.A., T.E.H., J.L.); Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (R.B.H.); Battelle Center for Mathematical Medicine, Nationwide Children's Hospital Research Institute, Columbus, OH (W.C.R.); The Ohio State University Interdisciplinary Graduate Program in Biophysics, Columbus, OH (W.C.R.); Department of Pediatrics, The Ohio State University, Columbus, OH (W.C.R., J.L.); and Division of Cardiovascular Medicine and Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA (D.D.H.)
| | - Tori E Horne
- From the Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, Miami, FL (D.J.H.); Center for Cardiovascular Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, OH (D.J.H., B.F.A., T.E.H., J.L.); Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (R.B.H.); Battelle Center for Mathematical Medicine, Nationwide Children's Hospital Research Institute, Columbus, OH (W.C.R.); The Ohio State University Interdisciplinary Graduate Program in Biophysics, Columbus, OH (W.C.R.); Department of Pediatrics, The Ohio State University, Columbus, OH (W.C.R., J.L.); and Division of Cardiovascular Medicine and Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA (D.D.H.)
| | - Robert B Hinton
- From the Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, Miami, FL (D.J.H.); Center for Cardiovascular Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, OH (D.J.H., B.F.A., T.E.H., J.L.); Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (R.B.H.); Battelle Center for Mathematical Medicine, Nationwide Children's Hospital Research Institute, Columbus, OH (W.C.R.); The Ohio State University Interdisciplinary Graduate Program in Biophysics, Columbus, OH (W.C.R.); Department of Pediatrics, The Ohio State University, Columbus, OH (W.C.R., J.L.); and Division of Cardiovascular Medicine and Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA (D.D.H.)
| | - William C Ray
- From the Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, Miami, FL (D.J.H.); Center for Cardiovascular Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, OH (D.J.H., B.F.A., T.E.H., J.L.); Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (R.B.H.); Battelle Center for Mathematical Medicine, Nationwide Children's Hospital Research Institute, Columbus, OH (W.C.R.); The Ohio State University Interdisciplinary Graduate Program in Biophysics, Columbus, OH (W.C.R.); Department of Pediatrics, The Ohio State University, Columbus, OH (W.C.R., J.L.); and Division of Cardiovascular Medicine and Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA (D.D.H.)
| | - Donald D Heistad
- From the Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, Miami, FL (D.J.H.); Center for Cardiovascular Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, OH (D.J.H., B.F.A., T.E.H., J.L.); Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (R.B.H.); Battelle Center for Mathematical Medicine, Nationwide Children's Hospital Research Institute, Columbus, OH (W.C.R.); The Ohio State University Interdisciplinary Graduate Program in Biophysics, Columbus, OH (W.C.R.); Department of Pediatrics, The Ohio State University, Columbus, OH (W.C.R., J.L.); and Division of Cardiovascular Medicine and Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA (D.D.H.)
| | - Joy Lincoln
- From the Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, Miami, FL (D.J.H.); Center for Cardiovascular Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, OH (D.J.H., B.F.A., T.E.H., J.L.); Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (R.B.H.); Battelle Center for Mathematical Medicine, Nationwide Children's Hospital Research Institute, Columbus, OH (W.C.R.); The Ohio State University Interdisciplinary Graduate Program in Biophysics, Columbus, OH (W.C.R.); Department of Pediatrics, The Ohio State University, Columbus, OH (W.C.R., J.L.); and Division of Cardiovascular Medicine and Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA (D.D.H.).
| |
Collapse
|
105
|
Martin PS, Kloesel B, Norris RA, Lindsay M, Milan D, Body SC. Embryonic Development of the Bicuspid Aortic Valve. J Cardiovasc Dev Dis 2015; 2:248-272. [PMID: 28529942 PMCID: PMC5438177 DOI: 10.3390/jcdd2040248] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bicuspid aortic valve (BAV) is the most common congenital valvular heart defect with an overall frequency of 0.5%–1.2%. BAVs result from abnormal aortic cusp formation during valvulogenesis, whereby adjacent cusps fuse into a single large cusp resulting in two, instead of the normal three, aortic cusps. Individuals with BAV are at increased risk for ascending aortic disease, aortic stenosis and coarctation of the aorta. The frequent occurrence of BAV and its anatomically discrete but frequent co-existing diseases leads us to suspect a common cellular origin. Although autosomal-dominant transmission of BAV has been observed in a few pedigrees, notably involving the gene NOTCH1, no single-gene model clearly explains BAV inheritance, implying a complex genetic model involving interacting genes. Several sequencing studies in patients with BAV have identified rare and uncommon mutations in genes of cardiac embryogenesis. But the extensive cell-cell signaling and multiple cellular origins involved in cardiac embryogenesis preclude simplistic explanations of this disease. In this review, we examine the series of events from cellular and transcriptional embryogenesis of the heart, to development of the aortic valve.
Collapse
Affiliation(s)
- Peter S. Martin
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
| | - Benjamin Kloesel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell Biology, Children’s Research Institute, Medical University of South Carolina, 173 Ashley St, Charleston, SC 29403, USA; E-Mail:
| | - Mark Lindsay
- Cardiovascular Research Center, Richard B. Simches Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; E-Mails: (M.L.); (D.M.)
| | - David Milan
- Cardiovascular Research Center, Richard B. Simches Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; E-Mails: (M.L.); (D.M.)
| | - Simon C. Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
- Author to whom correspondence should be addressed: E-Mail: ; Tel.: +1-617-732-7330; Fax: +1-617-730-2813
| |
Collapse
|
106
|
Amarillo IE, O'Connor S, Lee CK, Willing M, Wambach JA. De novo 9q gain in an infant with tetralogy of Fallot with absent pulmonary valve: Patient report and review of congenital heart disease in 9q duplication syndrome. Am J Med Genet A 2015; 167A:2966-74. [PMID: 26768185 DOI: 10.1002/ajmg.a.37296] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 08/05/2015] [Indexed: 12/14/2022]
Abstract
Genomic disruptions, altered epigenetic mechanisms, and environmental factors contribute to the heterogeneity of congenital heart defects (CHD). In recent years, chromosomal microarray analysis (CMA) has led to the identification of numerous copy number variations (CNV) in patients with CHD. Genes disrupted by and within these CNVs thus represent excellent candidate genes for CHD. Microduplications of 9q (9q+) have been described in patients with CHD, however, the critical gene locus remains undetermined. Here we discuss an infant with tetralogy of Fallot with absent pulmonary valve, fetal hydrops, and a 3.76 Mb de novo contiguous gain of 9q34.2-q34.3 detected by CMA, and confirmed by karyotype and FISH studies. This duplicated interval disrupted RXRA (retinoid X receptor alpha; OMIM #180245) at intron 1. We also review CHD findings among previously reported patients with 9q (9q+) duplication syndrome. This is the first report implicating RXRA in CHD with 9q duplication, providing additional data in understanding the genetic etiology of tetralogy of Fallot, CHD, and disorders linked to 9q microduplication syndrome. This report also highlights the significance of CMA in the clinical diagnosis and genetic counseling of patients and families with complex CHD.
Collapse
Affiliation(s)
- Ina E Amarillo
- Department of Pathology and Immunology, Cytogenomics Laboratory, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Shawn O'Connor
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Caroline K Lee
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Marcia Willing
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Jennifer A Wambach
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| |
Collapse
|
107
|
Horne TE, VandeKopple M, Sauls K, Koenig SN, Anstine LJ, Garg V, Norris RA, Lincoln J. Dynamic Heterogeneity of the Heart Valve Interstitial Cell Population in Mitral Valve Health and Disease. J Cardiovasc Dev Dis 2015; 2:214-232. [PMID: 26527432 PMCID: PMC4625550 DOI: 10.3390/jcdd2030214] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The heart valve interstitial cell (VIC) population is dynamic and thought to mediate lay down and maintenance of the tri-laminar extracellular matrix (ECM) structure within the developing and mature valve throughout life. Disturbances in the contribution and distribution of valve ECM components are detrimental to biomechanical function and associated with disease. This pathological process is associated with activation of resident VICs that in the absence of disease reside as quiescent cells. While these paradigms have been long standing, characterization of this abundant and ever-changing valve cell population is incomplete. Here we examine the expression pattern of Smooth muscle α-actin, Periostin, Twist1 and Vimentin in cultured VICs, heart valves from healthy embryonic, postnatal and adult mice, as well as mature valves from human patients and established mouse models of disease. We show that the VIC population is highly heterogeneous and phenotypes are dependent on age, species, location, and disease state. Furthermore, we identify phenotypic diversity across common models of mitral valve disease. These studies significantly contribute to characterizing the VIC population in health and disease and provide insights into the cellular dynamics that maintain valve structure in healthy adults and mediate pathologic remodeling in disease states.
Collapse
Affiliation(s)
- Tori E Horne
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, 575 Children's Drive, Research Building III, WB4239, Columbus, OH 43215, USA; (T.E.H.); (M.V.); (S.N.K.); (L.J.A.); (V.G.)
| | - Matthew VandeKopple
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, 575 Children's Drive, Research Building III, WB4239, Columbus, OH 43215, USA; (T.E.H.); (M.V.); (S.N.K.); (L.J.A.); (V.G.)
| | - Kimberly Sauls
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.); (R.A.N.)
| | - Sara N Koenig
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, 575 Children's Drive, Research Building III, WB4239, Columbus, OH 43215, USA; (T.E.H.); (M.V.); (S.N.K.); (L.J.A.); (V.G.)
| | - Lindsey J Anstine
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, 575 Children's Drive, Research Building III, WB4239, Columbus, OH 43215, USA; (T.E.H.); (M.V.); (S.N.K.); (L.J.A.); (V.G.)
| | - Vidu Garg
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, 575 Children's Drive, Research Building III, WB4239, Columbus, OH 43215, USA; (T.E.H.); (M.V.); (S.N.K.); (L.J.A.); (V.G.)
- Department of Pediatrics, The Ohio State University, Columbus, OH 43215, USA
| | - Russell A Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.); (R.A.N.)
| | - Joy Lincoln
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, 575 Children's Drive, Research Building III, WB4239, Columbus, OH 43215, USA; (T.E.H.); (M.V.); (S.N.K.); (L.J.A.); (V.G.)
- Department of Pediatrics, The Ohio State University, Columbus, OH 43215, USA
| |
Collapse
|
108
|
Hjortnaes J, Shapero K, Goettsch C, Hutcheson JD, Keegan J, Kluin J, Mayer JE, Bischoff J, Aikawa E. Valvular interstitial cells suppress calcification of valvular endothelial cells. Atherosclerosis 2015; 242:251-260. [PMID: 26232165 DOI: 10.1016/j.atherosclerosis.2015.07.008] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 07/02/2015] [Accepted: 07/02/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is the most common heart valve disease in the Western world. We previously proposed that valvular endothelial cells (VECs) replenish injured adult valve leaflets via endothelial-to-mesenchymal transformation (EndMT); however, whether EndMT contributes to valvular calcification is unknown. We hypothesized that aortic VECs undergo osteogenic differentiation via an EndMT process that can be inhibited by valvular interstitial cells (VICs). APPROACH AND RESULTS VEC clones underwent TGF-β1-mediated EndMT, shown by significantly increased mRNA expression of the EndMT markers α-SMA (5.3 ± 1.2), MMP-2 (13.5 ± 0.6) and Slug (12 ± 2.1) (p < 0.05), (compared to unstimulated controls). To study the effects of VIC on VEC EndMT, clonal populations of VICs were derived from the same valve leaflets, placed in co-culture with VECs, and grown in control/TGF-β1 supplemented media. In the presence of VICs, EndMT was inhibited, shown by decreased mRNA expression of α-SMA (0.1 ± 0.5), MMP-2 (0.1 ± 0.1), and Slug (0.2 ± 0.2) (p < 0.05). When cultured in osteogenic media, VECs demonstrated osteogenic changes confirmed by increase in mRNA expression of osteocalcin (8.6 ± 1.3), osteopontin (3.7 ± 0.3), and Runx2 (5.5 ± 1.5). The VIC presence inhibited VEC osteogenesis, demonstrated by decreased expression of osteocalcin (0.4 ± 0.1) and osteopontin (0.2 ± 0.1) (p < 0.05). Time course analysis suggested that EndMT precedes osteogenesis, shown by an initial increase of α-SMA and MMP-2 (day 7), followed by an increase of osteopontin and osteocalcin (day 14). CONCLUSIONS The data indicate that EndMT may precede VEC osteogenesis. This study shows that VICs inhibit VEC EndMT and osteogenesis, indicating the importance of VEC-VIC interactions in valve homeostasis.
Collapse
Affiliation(s)
- Jesper Hjortnaes
- Center of Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston.,Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kayle Shapero
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston
| | - Claudia Goettsch
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston
| | - Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston
| | - Joshua Keegan
- Center of Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston
| | - Jolanda Kluin
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - John E Mayer
- Department of Thoracic Surgery, Boston Children's Hospital, Harvard Medical School, Boston
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston
| | - Elena Aikawa
- Center of Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston.,Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston
| |
Collapse
|
109
|
Endocardial Brg1 disruption illustrates the developmental origins of semilunar valve disease. Dev Biol 2015; 407:158-72. [PMID: 26100917 DOI: 10.1016/j.ydbio.2015.06.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/12/2015] [Accepted: 06/13/2015] [Indexed: 11/24/2022]
Abstract
The formation of intricately organized aortic and pulmonic valves from primitive endocardial cushions of the outflow tract is a remarkable accomplishment of embryonic development. While not always initially pathologic, developmental semilunar valve (SLV) defects, including bicuspid aortic valve, frequently progress to a disease state in adults requiring valve replacement surgery. Disrupted embryonic growth, differentiation, and patterning events that "trigger" SLV disease are coordinated by gene expression changes in endocardial, myocardial, and cushion mesenchymal cells. We explored roles of chromatin regulation in valve gene regulatory networks by conditional inactivation of the Brg1-associated factor (BAF) chromatin remodeling complex in the endocardial lineage. Endocardial Brg1-deficient mouse embryos develop thickened and disorganized SLV cusps that frequently become bicuspid and myxomatous, including in surviving adults. These SLV disease-like phenotypes originate from deficient endocardial-to-mesenchymal transformation (EMT) in the proximal outflow tract (pOFT) cushions. The missing cells are replaced by compensating neural crest or other non-EMT-derived mesenchyme. However, these cells are incompetent to fully pattern the valve interstitium into distinct regions with specialized extracellular matrices. Transcriptomics reveal genes that may promote growth and patterning of SLVs and/or serve as disease-state biomarkers. Mechanistic studies of SLV disease genes should distinguish between disease origins and progression; the latter may reflect secondary responses to a disrupted developmental system.
Collapse
|
110
|
Chen J, Ryzhova LM, Sewell-Loftin MK, Brown CB, Huppert SS, Baldwin HS, Merryman WD. Notch1 Mutation Leads to Valvular Calcification Through Enhanced Myofibroblast Mechanotransduction. Arterioscler Thromb Vasc Biol 2015; 35:1597-605. [PMID: 26023079 DOI: 10.1161/atvbaha.114.305095] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/19/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Calcific aortic valve disease (CAVD) is a significant cardiovascular disorder, and controversy exists as to whether it is primarily a dystrophic or osteogenic process in vivo. In this study, we sought to clarify the mechanism of CAVD by assessing a genetic mutation, Notch1 heterozygosity, which leads to CAVD with 100% penetrance in humans. APPROACH AND RESULTS Murine immortalized Notch1(+/-) aortic valve interstitial cells (AVICs) were isolated and expanded in vitro. Molecular signaling of wild-type and Notch1(+/-) AVICs were compared to identify changes in pathways that have been linked to CAVD-transforming growth factor-β1/bone morphogenetic protein, mitogen-activated protein kinase, and phosphoinositide 3-kinase/protein kinase B-and assessed for calcification potential. Additionally, AVIC mechanobiology was studied in a physiologically relevant, dynamic mechanical environment (10% cyclic strain) to investigate differences in responses between the cell types. We found that Notch1(+/-) AVICs resembled a myofibroblast-like phenotype expressing higher amounts of cadherin-11, a known mediator of dystrophic calcification, and decreased Runx2, a known osteogenic marker. We determined that cadherin-11 expression is regulated by Akt activity, and inhibition of Akt phosphorylation significantly reduced cadherin-11 expression. Moreover, in the presence of cyclic strain, Notch1(+/-) AVICs exhibited significantly upregulated phosphorylation of Akt at Ser473 and smooth muscle α-actin expression, indicative of a fully activated myofibroblast. Finally, these Notch1-mediated alterations led to enhanced dystrophic calcific nodule formation. CONCLUSIONS This study presents novel insights in our understanding of Notch1-mediated CAVD by demonstrating that the mutation leads to AVICs that are fully activated myofibroblasts, resulting in dystrophic, but not osteogenic, calcification.
Collapse
Affiliation(s)
- Joseph Chen
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - Larisa M Ryzhova
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - M K Sewell-Loftin
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - Christopher B Brown
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - Stacey S Huppert
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - H Scott Baldwin
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.)
| | - W David Merryman
- From the Department of Biomedical Engineering (J.C., L.M.R., M.K.S.-L., W.D.M.) and Divison of Cardiology, Department of Pediatrics (C.B.B., H.S.B.), Vanderbilt University, Nashville, TN; and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital, OH (S.S.H.).
| |
Collapse
|
111
|
Farrar EJ, Huntley GD, Butcher J. Endothelial-derived oxidative stress drives myofibroblastic activation and calcification of the aortic valve. PLoS One 2015; 10:e0123257. [PMID: 25874717 PMCID: PMC4395382 DOI: 10.1371/journal.pone.0123257] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 02/20/2015] [Indexed: 01/14/2023] Open
Abstract
Aims Oxidative stress is present in and contributes to calcification of the aortic valve, but the driving factors behind the initiation of valve oxidative stress are not well understood. We tested whether the valve endothelium acts as an initiator and propagator of oxidative stress in aortic valve disease. Methods and Results Calcified human aortic valves showed side-specific elevation of superoxide in the endothelium, co-localized with high VCAM1 expression, linking oxidative stress, inflammation, and valve degeneration. Treatment with inflammatory cytokine TNFα increased superoxide and oxidative stress and decreased eNOS and VE-cadherin acutely over 48 hours in aortic valve endothelial cells (VEC) and chronically over 21 days in ex vivo AV leaflets. Co-treatment of VEC with tetrahydrobiopterin (BH4) but not apocynin mitigated TNFα-driven VEC oxidative stress. Co-treatment of ex vivo AV leaflets with TNFα+BH4 or TNFα+peg-SOD rescued endothelial function and mitigated inflammatory responses. Both BH4 and peg-SOD rescued valve leaflets from the pro-osteogenic effects of TNFα treatment, but only peg-SOD was able to mitigate the fibrogenic effects, including increased collagen and αSMA expression. Conclusions Aortic valve endothelial cells are a novel source of oxidative stress in aortic valve disease. TNFα-driven VEC oxidative stress causes loss of endothelial protective function, chronic inflammation, and fibrogenic and osteogenic activation, mitigated differentially by BH4 and peg-SOD. These mechanisms identify new targets for tailored antioxidant therapy focused on mitigation of oxidative stress and restoration of endothelial protection.
Collapse
Affiliation(s)
- Emily J. Farrar
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Geoffrey D. Huntley
- Mayo Medical School, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jonathan Butcher
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
112
|
Koenig SN, Bosse KM, Nadorlik HA, Lilly B, Garg V. Evidence of Aortopathy in Mice with Haploinsufficiency of Notch1 in Nos3-Null Background. J Cardiovasc Dev Dis 2015; 2:17-30. [PMID: 25914885 PMCID: PMC4407710 DOI: 10.3390/jcdd2010017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Thoracic aortic aneurysms (TAA) are a significant cause of morbidity and mortality in humans. While the exact etiology is unknown, genetic factors play an important role. Mutations in NOTCH1 have been linked to bicuspid aortic valve (BAV) and aortopathy in humans. The aim of this study was to determine if haploinsufficiency of Notch1 contributes to aortopathy using Notch1+/−; Nos3−/− mice. Echocardiographic analysis of Notch1+/−; Nos3−/− mice reveals effacement of the sinotubular junction and a trend toward dilation of the aortic sinus. Furthermore, examination of the proximal aorta of Notch1+/−; Nos3−/− mice reveals elastic fiber degradation, a trend toward increased matrix metalloproteinase 2 expression, and increased smooth muscle cell apoptosis, features characteristic of aneurysmal disease. Although at a lower penetrance, we also found features consistent with aortopathic changes in Notch1 heterozygote mice and in Nos3-null mice. Our findings implicate a novel role for Notch1 in aortopathy of the proximal aorta.
Collapse
Affiliation(s)
- Sara N. Koenig
- The Center for Cardiovascular and Pulmonary Research and Heart Center, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; E-Mails: (S.N.K.); (K.M.B.); (H.A.N.); (B.L.)
- Department of Pediatrics, The Ohio State University, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Kevin M. Bosse
- The Center for Cardiovascular and Pulmonary Research and Heart Center, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; E-Mails: (S.N.K.); (K.M.B.); (H.A.N.); (B.L.)
| | - Holly A. Nadorlik
- The Center for Cardiovascular and Pulmonary Research and Heart Center, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; E-Mails: (S.N.K.); (K.M.B.); (H.A.N.); (B.L.)
- Department of Pediatrics, The Ohio State University, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Brenda Lilly
- The Center for Cardiovascular and Pulmonary Research and Heart Center, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; E-Mails: (S.N.K.); (K.M.B.); (H.A.N.); (B.L.)
- Department of Pediatrics, The Ohio State University, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Vidu Garg
- The Center for Cardiovascular and Pulmonary Research and Heart Center, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; E-Mails: (S.N.K.); (K.M.B.); (H.A.N.); (B.L.)
- Department of Pediatrics, The Ohio State University, 700 Children’s Drive, Columbus, OH 43205, USA
- Department of Molecular Genetics, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-614-355-5740; Fax: +1-614-355-5725
| |
Collapse
|
113
|
Lincoln J. To 'cell' and back! J Mol Cell Cardiol 2015; 81:94-5. [PMID: 25681583 DOI: 10.1016/j.yjmcc.2015.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 02/01/2015] [Indexed: 10/24/2022]
Affiliation(s)
- Joy Lincoln
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
114
|
Shapero K, Wylie-Sears J, Levine RA, Mayer JE, Bischoff J. Reciprocal interactions between mitral valve endothelial and interstitial cells reduce endothelial-to-mesenchymal transition and myofibroblastic activation. J Mol Cell Cardiol 2015; 80:175-85. [PMID: 25633835 PMCID: PMC4346432 DOI: 10.1016/j.yjmcc.2015.01.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 01/14/2015] [Accepted: 01/18/2015] [Indexed: 11/26/2022]
Abstract
Thickening of mitral leaflets, endothelial-to-mesenchymal transition (EndMT), and activated myofibroblast-like interstitial cells have been observed in ischemic mitral valve regurgitation. We set out to determine if interactions between mitral valve endothelial cells (VEC) and interstitial cells (VIC) might affect these alterations. We used in vitro co-culture in Transwell™ inserts to test the hypothesis that VIC secrete factors that inhibit EndMT and conversely, that VEC secrete factors that mitigate the activation of VIC to a myofibroblast-like, activated phenotype. Primary cultures and clonal populations of ovine mitral VIC and VEC were used. Western blot, quantitative reverse transcriptase PCR (qPCR) and functional assays were used to assess changes in cell phenotype and behavior. VIC or conditioned media from VIC inhibited transforming growth factorβ (TGFβ)-induced EndMT in VEC, as indicated by reduced expression of EndMT markers α-smooth muscle actin (α-SMA), Slug, Snai1 and MMP-2 and maintained ability of VEC to mediate leukocyte adhesion, an important endothelial function. VEC or conditioned media from VEC reversed the spontaneous cell culture-induced change in VIC to an activated phenotype, as indicated by reduced expression of α-SMA and type I collagen, increased expression chondromodulin-1 (Chm1), and reduced contractile activity. These results demonstrate that mitral VEC and VIC secrete soluble factors that can reduce VIC activation and inhibit TGFβ-driven EndMT, respectively. These findings suggest that the endothelium of the mitral valve is critical for the maintenance of a quiescent VIC phenotype and that, in turn, VIC prevent EndMT. We speculate that disturbance of the ongoing reciprocal interactions between VEC and VICs in vivo may contribute to the thickened and fibrotic leaflets observed in ischemic mitral regurgitation, and in other types of valve disease.
Collapse
Affiliation(s)
- Kayle Shapero
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, USA; Biomedical Engineering Department, Boston University, Boston, MA, USA; Department of Cardiothoracic Surgery, Boston Children's Hospital, Harvard Medical School, USA
| | - Jill Wylie-Sears
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, USA
| | - Robert A Levine
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - John E Mayer
- Department of Cardiothoracic Surgery, Boston Children's Hospital, Harvard Medical School, USA
| | - Joyce Bischoff
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, USA.
| |
Collapse
|
115
|
Abstract
Valvular heart disease is associated with significant morbidity and mortality and often the result of congenital malformations. However, the prevalence is increasing in adults not only because of the growing aging population, but also because of improvements in the medical and surgical care of children with congenital heart valve defects. The success of the Human Genome Project and major advances in genetic technologies, in combination with our increased understanding of heart valve development, has led to the discovery of numerous genetic contributors to heart valve disease. These have been uncovered using a variety of approaches including the examination of familial valve disease and genome-wide association studies to investigate sporadic cases. This review will discuss these findings and their implications in the treatment of valvular heart disease.
Collapse
Affiliation(s)
- Stephanie LaHaye
- Center for Cardiovascular and Pulmonary Research and The Heart Center, Room WB4221, Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | | | | |
Collapse
|
116
|
Godby RC, Munjal C, Opoka AM, Smith JM, Yutzey KE, Narmoneva DA, Hinton RB. Cross Talk between NOTCH Signaling and Biomechanics in Human Aortic Valve Disease Pathogenesis. J Cardiovasc Dev Dis 2014; 1:237-256. [PMID: 29552567 PMCID: PMC5856658 DOI: 10.3390/jcdd1030237] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aortic valve disease is a burgeoning public health problem associated with significant mortality. Loss of function mutations in NOTCH1 cause bicuspid aortic valve (BAV) and calcific aortic valve disease. Because calcific nodules manifest on the fibrosa side of the cusp in low fluidic oscillatory shear stress (OSS), elucidating pathogenesis requires approaches that consider both molecular and mechanical factors. Therefore, we examined the relationship between NOTCH loss of function (LOF) and biomechanical indices in healthy and diseased human aortic valve interstitial cells (AVICs). An orbital shaker system was used to apply cyclic OSS, which mimics the cardiac cycle and hemodynamics experienced by AVICs in vivo. NOTCH LOF blocked OSS-induced cell alignment in human umbilical vein endothelial cells (HUVECs), whereas AVICs did not align when subjected to OSS under any conditions. In healthy AVICs, OSS resulted in decreased elastin (ELN) and α-SMA (ACTA2). NOTCH LOF was associated with similar changes, but in diseased AVICs, NOTCH LOF combined with OSS was associated with increased α-SMA expression. Interestingly, AVICs showed relatively higher expression of NOTCH2 compared to NOTCH1. Biomechanical interactions between endothelial and interstitial cells involve complex NOTCH signaling that contributes to matrix homeostasis in health and disorganization in disease.
Collapse
Affiliation(s)
- Richard C. Godby
- Division of Cardiology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Charu Munjal
- Division of Cardiology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Amy M. Opoka
- Division of Cardiology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - J. Michael Smith
- TriHealth Heart Institute, Cardio-Thoracic Surgery, Good Samaritan Hospital, Cincinnati, OH 45242, USA
| | - Katherine E. Yutzey
- Molecular Cardiovascular Biology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Daria A. Narmoneva
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Robert B. Hinton
- Division of Cardiology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-513-636-0389; Fax: +1-513-636-5958
| |
Collapse
|
117
|
Review of Molecular and Mechanical Interactions in the Aortic Valve and Aorta: Implications for the Shared Pathogenesis of Aortic Valve Disease and Aortopathy. J Cardiovasc Transl Res 2014; 7:823-46. [DOI: 10.1007/s12265-014-9602-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 10/30/2014] [Indexed: 01/08/2023]
|
118
|
Cheng J, Koenig SN, Kuivaniemi HS, Garg V, Hans CP. Pharmacological inhibitor of notch signaling stabilizes the progression of small abdominal aortic aneurysm in a mouse model. J Am Heart Assoc 2014; 3:e001064. [PMID: 25349182 PMCID: PMC4338693 DOI: 10.1161/jaha.114.001064] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background The progression of abdominal aortic aneurysm (AAA) involves a sustained influx of proinflammatory macrophages, which exacerbate tissue injury by releasing cytokines, chemokines, and matrix metalloproteinases. Previously, we showed that Notch deficiency reduces the development of AAA in the angiotensin II–induced mouse model by preventing infiltration of macrophages. Here, we examined whether Notch inhibition in this mouse model prevents progression of small AAA and whether these effects are associated with altered macrophage differentiation. Methods and Results Treatment with pharmacological Notch inhibitor (DAPT [N‐(N‐[3,5‐difluorophenacetyl]‐L‐alanyl)‐S‐phenylglycine t‐butyl ester]) at day 3 or 8 of angiotensin II infusion arrested the progression of AAA in Apoe−/− mice, as demonstrated by a decreased luminal diameter and aortic width. The abdominal aortas of Apoe−/− mice treated with DAPT showed decreased expression of matrix metalloproteinases and presence of elastin precursors including tropoelastin and hyaluronic acid. Marginal adventitial thickening observed in the aorta of DAPT‐treated Apoe−/− mice was not associated with increased macrophage content, as observed in the mice treated with angiotensin II alone. Instead, DAPT‐treated abdominal aortas showed increased expression of Cd206‐positive M2 macrophages and decreased expression of Il12‐positive M1 macrophages. Notch1 deficiency promoted M2 differentiation of macrophages by upregulating transforming growth factor β2 in bone marrow–derived macrophages at basal levels and in response to IL4. Protein expression of transforming growth factor β2 and its downstream effector pSmad2 also increased in DAPT‐treated Apoe−/− mice, indicating a potential link between Notch and transforming growth factor β2 signaling in the M2 differentiation of macrophages. Conclusions Pharmacological inhibitor of Notch signaling prevents the progression of AAA by macrophage differentiation–dependent mechanisms. The study also provides insights for novel therapeutic strategies to prevent the progression of small AAA.
Collapse
Affiliation(s)
- Jeeyun Cheng
- Center for Cardiovascular and Pulmonary Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH (J.C., S.N.K., V.G., C.P.H.)
| | - Sara N Koenig
- Center for Cardiovascular and Pulmonary Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH (J.C., S.N.K., V.G., C.P.H.)
| | - Helena S Kuivaniemi
- The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA (H.S.K.)
| | - Vidu Garg
- Center for Cardiovascular and Pulmonary Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH (J.C., S.N.K., V.G., C.P.H.) Department of Pediatrics, The Ohio State University, Columbus, OH (V.G., C.P.H.) Department of Molecular Genetics, The Ohio State University, Columbus, OH (V.G.)
| | - Chetan P Hans
- Center for Cardiovascular and Pulmonary Research and The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH (J.C., S.N.K., V.G., C.P.H.) Department of Pediatrics, The Ohio State University, Columbus, OH (V.G., C.P.H.)
| |
Collapse
|
119
|
Abstract
PURPOSE OF REVIEW Aortic valve disease (AVD) is a growing public health problem, and the pathogenesis underlying AVD is complex. The lack of durable bioprostheses and pharmacologic therapies remain central needs in care. The purpose of this review is to highlight recent clinical studies that impact the care of children with AVD and is to explore ongoing translational research efforts. RECENT FINDINGS Clinical studies have evaluated the durability of bioprosthetics and surgical strategies, tested statins during early disease, and identified new predictive biomarkers. Large animal models have demonstrated the effectiveness of a novel bioprosthetic scaffold. Mouse models of latent AVD have advanced our ability to elucidate natural history and perform preclinical studies that test new treatments in the context of early disease. SUMMARY Current priorities for AVD patients include identifying new pharmacologic treatments and developing durable bioprostheses. Multidisciplinary efforts are needed that bridge pediatric and adult programs, and bring together different types of expertise and leverage network and consortium resources. As our understanding of the underlying complex genetics is better defined, companion diagnostics may transform future clinical trials and ultimately improve the care of patients with AVD by promoting personalized medicine and early intervention.
Collapse
|
120
|
Sox9- and Scleraxis-Cre Lineage Fate Mapping in Aortic and Mitral Valve Structures. J Cardiovasc Dev Dis 2014. [DOI: 10.3390/jcdd1020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
121
|
Abstract
Normal valve structures consist of stratified layers of specialized extracellular matrix (ECM) interspersed with valve interstitial cells (VICs) and surrounded by a monolayer of valve endothelial cells (VECs). VECs play essential roles in establishing the valve structures during embryonic development, and are important for maintaining life-long valve integrity and function. In contrast to a continuous endothelium over the surface of healthy valve leaflets, VEC disruption is commonly observed in malfunctioning valves and is associated with pathological processes that promote valve disease and dysfunction. Despite the clinical relevance, focused studies determining the contribution of VECs to development and disease processes are limited. The isolation of VECs from animal models would allow for cell-specific experimentation. VECs have been isolated from large animal adult models but due to their small population size, fragileness, and lack of specific markers, no reports of VEC isolations in embryos or adult small animal models have been reported. Here we describe a novel method that allows for the direct isolation of VECs from mice at embryonic and adult stages. Utilizing the Tie2-GFP reporter model that labels all endothelial cells with Green Fluorescent Protein (GFP), we have been successful in isolating GFP-positive (and negative) cells from the semilunar and atrioventricular valve regions using fluorescence activated cell sorting (FACS). Isolated GFP-positive VECs are enriched for endothelial markers, including CD31 and von Willebrand Factor (vWF), and retain endothelial cell expression when cultured; while, GFP-negative cells exhibit molecular profiles and cell shapes consistent with VIC phenotypes. The ability to isolate embryonic and adult murine VECs allows for previously unattainable molecular and functional studies to be carried out on a specific valve cell population, which will greatly improve our understanding of valve development and disease mechanisms.
Collapse
Affiliation(s)
- Lindsey J Miller
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University; Center for Cardiovascular and Pulmonary Research, The Heart Center, The Research Institute at Nationwide Children's Hospital
| | - Joy Lincoln
- Center for Cardiovascular and Pulmonary Research, The Heart Center, The Research Institute at Nationwide Children's Hospital; Department of Pediatrics, The Ohio State University;
| |
Collapse
|
122
|
Abstract
Valvular heart disease occurs as either a congenital or acquired condition and advances in medical care have resulted in valve disease becoming increasingly prevalent. Unfortunately, treatments remain inadequate because of our limited understanding of the genetic and molecular etiology of diseases affecting the heart valves. Therefore, surgical repair or replacement remains the most effective option, which comes with additional complications and no guarantee of life-long success. Over the past decade, there have been significant advances in our understanding of cardiac valve development and, not surprisingly, mutations in these developmental genes have been identified in humans with congenital valve malformations. Concurrently, there has been a greater realization that acquired valve disease is not simply a degenerative process. Molecular investigation of acquired valve disease has identified that numerous signaling pathways critical for normal valve development are re-expressed in diseased valves. This review will discuss recent advances in our understanding of the development of the heart valves, as well as the implications of these findings on the genetics of congenital and acquired valvular heart disease.
Collapse
Affiliation(s)
- Joy Lincoln
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital
| | | |
Collapse
|
123
|
Barnette DN, VandeKopple M, Wu Y, Willoughby DA, Lincoln J. RNA-seq analysis to identify novel roles of scleraxis during embryonic mouse heart valve remodeling. PLoS One 2014; 9:e101425. [PMID: 24983472 PMCID: PMC4077804 DOI: 10.1371/journal.pone.0101425] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 06/06/2014] [Indexed: 11/19/2022] Open
Abstract
Heart valve disease affects up to 30% of the population and has been shown to have origins during embryonic development. Valvulogenesis begins with formation of endocardial cushions in the atrioventricular canal and outflow tract regions. Subsequently, endocardial cushions remodel, elongate and progressively form mature valve structures composed of a highly organized connective tissue that provides the necessary biomechanical function throughout life. While endocardial cushion formation has been well studied, the processes required for valve remodeling are less well understood. The transcription factor Scleraxis (Scx) is detected in mouse valves from E15.5 during initial stages of remodeling, and expression remains high until birth when formation of the highly organized mature structure is complete. Heart valves from Scx-/- mice are abnormally thick and develop fibrotic phenotypes similar to human disease by juvenile stages. These phenotypes begin around E15.5 and are associated with defects in connective tissue organization and valve interstitial cell differentiation. In order to understand the etiology of this phenotype, we analyzed the transcriptome of remodeling valves isolated from E15.5 Scx-/- embryos using RNA-seq. From this, we have identified a profile of protein and non-protein mRNAs that are dependent on Scx function and using bioinformatics we can predict the molecular functions and biological processes affected by these genes. These include processes and functions associated with gene regulation (methyltransferase activity, DNA binding, Notch signaling), vitamin A metabolism (retinoic acid biosynthesis) and cellular development (cell morphology, cell assembly and organization). In addition, several mRNAs are affected by alternative splicing events in the absence of Scx, suggesting additional roles in post-transcriptional modification. In summary, our findings have identified transcriptome profiles from abnormal heart valves isolated from E15.5 Scx-/- embryos that could be used in the future to understand mechanisms of heart valve disease in the human population.
Collapse
Affiliation(s)
- Damien N. Barnette
- Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, Miami, Florida, United States of America
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, Ohio, United States of America
| | - Matthew VandeKopple
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, Ohio, United States of America
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Yonggan Wu
- Ocean Ridge Biosciences LLC, Palm Beach Gardens, Florida, United States of America
| | - David A. Willoughby
- Ocean Ridge Biosciences LLC, Palm Beach Gardens, Florida, United States of America
| | - Joy Lincoln
- Center for Cardiovascular and Pulmonary Research and The Heart Center at Nationwide Children's Hospital Research Institute, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
124
|
Abstract
Calcific aortic valve disease (CAVD) is a major contributor to cardiovascular morbidity and mortality and, given its association with age, the prevalence of CAVD is expected to continue to rise as global life expectancy increases. No drug strategies currently exist to prevent or treat CAVD. Given that valve replacement is the only available clinical option, patients often cope with a deteriorating quality of life until diminished valve function demands intervention. The recognition that CAVD results from active cellular mechanisms suggests that the underlying pathways might be targeted to treat the condition. However, no such therapeutic strategy has been successfully developed to date. One hope was that drugs already used to treat vascular complications might also improve CAVD outcomes, but the mechanisms of CAVD progression and the desired therapeutic outcomes are often different from those of vascular diseases. Therefore, we discuss the benchmarks that must be met by a CAVD treatment approach, and highlight advances in the understanding of CAVD mechanisms to identify potential novel therapeutic targets.
Collapse
Affiliation(s)
- Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, 3 Blackfan Circle, 17th Floor, Center for Life Sciences Boston, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, 3 Blackfan Circle, 17th Floor, Center for Life Sciences Boston, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - W David Merryman
- Department of Biomedical Engineering, 2213 Garland Avenue, Vanderbilt University, Nashville, TN 37212, USA
| |
Collapse
|
125
|
Mathieu P, Boulanger MC. Basic mechanisms of calcific aortic valve disease. Can J Cardiol 2014; 30:982-93. [PMID: 25085215 DOI: 10.1016/j.cjca.2014.03.029] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/03/2014] [Accepted: 03/19/2014] [Indexed: 02/06/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common heart valve disorder. There is no medical treatment to prevent and/or promote the regression of CAVD. Hence, it is of foremost importance to delineate and understand the key basic underlying mechanisms involved in CAVD. In the past decade our comprehension of the underpinning processes leading to CAVD has expanded at a fast pace. Hence, our understanding of the basic pathobiological processes implicated in CAVD might lead eventually to the development of novel pharmaceutical therapies for CAVD. In this review, we discuss molecular processes that are implicated in fibrosis and mineralization of the aortic valve. Specifically, we address the role of lipid retention, inflammation, phosphate signalling and osteogenic transition in the development of CAVD. Interplays between these different processes and the key regulation pathways are discussed along with their clinical relevance.
Collapse
Affiliation(s)
- Patrick Mathieu
- Laboratoire d'Études Moléculaires des Valvulopathies (LEMV), Groupe de Recherche en Valvulopathies (GRV), Québec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, Québec, Québec, Canada.
| | - Marie-Chloé Boulanger
- Laboratoire d'Études Moléculaires des Valvulopathies (LEMV), Groupe de Recherche en Valvulopathies (GRV), Québec Heart and Lung Institute/Research Center, Department of Surgery, Laval University, Québec, Québec, Canada
| |
Collapse
|
126
|
Gould ST, Matherly EE, Smith JN, Heistad DD, Anseth KS. The role of valvular endothelial cell paracrine signaling and matrix elasticity on valvular interstitial cell activation. Biomaterials 2014; 35:3596-606. [PMID: 24462357 DOI: 10.1016/j.biomaterials.2014.01.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 01/07/2014] [Indexed: 12/23/2022]
Abstract
The effects of valvular endothelial cell (VlvEC) paracrine signaling on VIC phenotype and nodule formation were tested using a co-culture platform with physiologically relevant matrix elasticities and diffusion distance. 100 μm thin poly(ethylene glycol) (PEG) hydrogels of 3-27 kPa Young's moduli were fabricated in transwell inserts. VICs were cultured on the gels, as VIC phenotype is known to change significantly within this range, while VlvECs lined the underside of the membrane. Co-culture with VlvECs significantly reduced VIC activation to the myofibroblast phenotype on all gels with the largest percent decrease on the 3 kPa gels (~70%), while stiffer gels resulted in approximately 20-30% decrease. Additionally, VlvECs significantly reduced αSMA protein expression (~2 fold lower) on both 3 and 27 kPa gels, as well as the number (~2 fold lower) of nodules formed on the 27 kPa gels. Effects of VlvECs were prevented when nitric oxide (NO) release was inhibited with l-NAME, suggesting that VlvEC produced NO inhibits VIC activation. Withdrawal of l-NAME after 3, 5, and 7 days with restoration of VlvEC NO production for 2 additional days led to a partial reversal of VIC activation (~25% decrease). A potential mechanism by which VlvEC produced NO reduced VIC activation was studied by inhibiting initial and mid-stage cGMP pathway molecules. Inhibition of soluble guanylyl cyclase (sGC) with ODQ or protein kinase G (PKG) with RBrcGMP or stimulation of Rho kinase (ROCK) with LPA, abolished VlvEC effects on VIC activation. This work contributes substantially to the understanding of the valve endothelium's role in preventing VIC functions associated with aortic valve stenosis initiation and progression.
Collapse
Affiliation(s)
- Sarah T Gould
- Department of Chemical and Biological Engineering, The BioFrontiers Institute, Boulder, CO 80303, USA
| | - Emily E Matherly
- Department of Chemical and Biological Engineering, The BioFrontiers Institute, Boulder, CO 80303, USA
| | - Jennifer N Smith
- Department of Chemical and Biological Engineering, The BioFrontiers Institute, Boulder, CO 80303, USA
| | - Donald D Heistad
- Departments of Internal Medicine and Pharmacology, University of Iowa Health Care, Iowa City, IA 52242, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, The BioFrontiers Institute, Boulder, CO 80303, USA; Howard Hughes Medical Institute University of Colorado, Boulder, CO 80303, USA.
| |
Collapse
|
127
|
Notch γ-secretase inhibitor dibenzazepine attenuates angiotensin II-induced abdominal aortic aneurysm in ApoE knockout mice by multiple mechanisms. PLoS One 2013; 8:e83310. [PMID: 24358274 PMCID: PMC3865307 DOI: 10.1371/journal.pone.0083310] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 11/02/2013] [Indexed: 01/01/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening aortic disease in the elderly. Activation of Notch1 pathway plays a critical role in the development of AAA, but the underlying mechanisms remain poorly understood. In the present study, we explored the mechanisms by which Notch1 activation regulates angiotensin II (Ang II)-induced AAA formation and evaluated the therapeutic potential of a new Notch γ-secretase inhibitor, dibenzazepine (DBZ), for the treatment of AAA. Apolipoprotein E knockout (Apo E(-/-)) mice infused for 4 weeks with Ang II (1000 ng/kg/min, IP) using osmotic mini-pumps were received an intraperitoneal injection of either vehicle or 1 mg/kg/d DBZ. Notch1 signaling was activated in AAA tissue from both Ang II-infused Apo E(-/-) mice and human undergoing AAA repair in vivo, with increased expression of Notch intracellular domain (NICD) and its target gene Hes1, and this effect was effectively blocked by DBZ. Moreover, infusion of Ang II markedly increased the incidence and severity of AAA in Apo E(-/-) mice. In contrast, inhibition of Notch activation by DBZ prevented AAA formation in vivo. Furthermore, DBZ markedly prevented Ang II-stimulated accumulation of macrophages and CD4(+) T cells, and ERK-mediated angiogenesis, simultaneously reversed Th2 response, in vivo. In conclusion, these findings provide new insight into the multiple mechanisms of Notch signaling involved in AAA formation and suggest that γ-secretase inhibitor DBZ might be a novel therapeutic drug for treating AAAS.
Collapse
|
128
|
Barnette DN, Hulin A, Ahmed ASI, Colige AC, Azhar M, Lincoln J. Tgfβ-Smad and MAPK signaling mediate scleraxis and proteoglycan expression in heart valves. J Mol Cell Cardiol 2013; 65:137-46. [PMID: 24157418 DOI: 10.1016/j.yjmcc.2013.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/10/2013] [Accepted: 10/09/2013] [Indexed: 11/17/2022]
Abstract
Mature heart valves are complex structures consisting of three highly organized extracellular matrix layers primarily composed of collagens, proteoglycans and elastin. Collectively, these diverse matrix components provide all the necessary biomechanical properties for valve function throughout life. In contrast to healthy valves, myxomatous valve disease is the most common cause of mitral valve prolapse in the human population and is characterized by an abnormal abundance of proteoglycans within the valve tri-laminar structure. Despite the clinical significance, the etiology of this phenotype is not known. Scleraxis (Scx) is a basic-helix-loop-helix transcription factor that we previously showed to be required for establishing heart valve structure during remodeling stages of valvulogenesis. In this study, we report that remodeling heart valves from Scx null mice express decreased levels of proteoglycans, particularly chondroitin sulfate proteoglycans (CSPGs), while overexpression in embryonic avian valve precursor cells and adult porcine valve interstitial cells increases CSPGs. Using these systems we further identify that Scx is positively regulated by canonical Tgfβ2 signaling during this process and this is attenuated by MAPK activity. Finally, we show that Scx is increased in myxomatous valves from human patients and mouse models, and overexpression in human mitral valve interstitial cells modestly increases proteoglycan expression consistent with myxomatous mitral valve phenotypes. Together, these studies identify an important role for Scx in regulating proteoglycans in embryonic and mature valve cells and suggest that imbalanced regulation could influence myxomatous pathogenesis.
Collapse
Affiliation(s)
- Damien N Barnette
- Molecular and Cellular Pharmacology Graduate Program, Leonard M. Miller School of Medicine, P.O. Box 016189 (R-189), Miami, FL, USA; Center for Cardiovascular and Pulmonary Research at Nationwide Children's Hospital Research Institute, 700 Children's Drive, Columbus, OH 43205, USA; The Heart Center at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | | | | | | | | | | |
Collapse
|
129
|
MacGrogan D, Luxán G, de la Pompa JL. Genetic and functional genomics approaches targeting the Notch pathway in cardiac development and congenital heart disease. Brief Funct Genomics 2013; 13:15-27. [PMID: 24106100 DOI: 10.1093/bfgp/elt036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The Notch signalling pathway plays crucial roles in cardiac development and postnatal cardiac homoeostasis. Gain- and loss-of-function approaches indicate that Notch promotes or inhibits cardiogenesis in a stage-dependent manner. However, the molecular mechanisms are poorly defined because many downstream effectors remain to be identified. Genome-scale analyses are shedding light on the genes that are regulated by Notch signalling and the mechanisms underlying this regulation. We review the functional data that implicates Notch in cardiac morphogenetic processes and expression profiling studies that enlighten the regulatory networks behind them. A recurring theme is that Notch cross-talks reiteratively with other key signalling pathways including Wnt and Bmp to coordinate cell and tissue interactions during cardiogenesis.
Collapse
Affiliation(s)
- Donal MacGrogan
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain. Tel.: +34-620-936633; Fax: +34-91-4531304;
| | | | | |
Collapse
|
130
|
Andersen TA, Troelsen KDLL, Larsen LA. Of mice and men: molecular genetics of congenital heart disease. Cell Mol Life Sci 2013; 71:1327-52. [PMID: 23934094 PMCID: PMC3958813 DOI: 10.1007/s00018-013-1430-1] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/16/2013] [Accepted: 07/18/2013] [Indexed: 12/21/2022]
Abstract
Congenital heart disease (CHD) affects nearly 1 % of the population. It is a complex disease, which may be caused by multiple genetic and environmental factors. Studies in human genetics have led to the identification of more than 50 human genes, involved in isolated CHD or genetic syndromes, where CHD is part of the phenotype. Furthermore, mapping of genomic copy number variants and exome sequencing of CHD patients have led to the identification of a large number of candidate disease genes. Experiments in animal models, particularly in mice, have been used to verify human disease genes and to gain further insight into the molecular pathology behind CHD. The picture emerging from these studies suggest that genetic lesions associated with CHD affect a broad range of cellular signaling components, from ligands and receptors, across down-stream effector molecules to transcription factors and co-factors, including chromatin modifiers.
Collapse
Affiliation(s)
- Troels Askhøj Andersen
- Wilhelm Johannsen Centre for Functional Genome Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | | | | |
Collapse
|