39 results on '"Morrisey, E"'
Search Results
2. Multiple dose-dependent roles for Sox2 in the patterning and differentiation of anterior foregut endoderm
- Author
-
Hogan, B. L. M., Taranova, O., Okubo, T., Morrisey, E. E., Nam, K.-T., Goldenring, J. R., Kurotani, R., Que, J., and Pevny, L. H.
- Subjects
stomatognathic system ,embryonic structures ,respiratory system - Abstract
Sox2 is expressed in developing foregut endoderm, with highest levels in the future esophagus and anterior stomach. By contrast, Nkx2.1 (Titf1) is expressed ventrally, in the future trachea. In humans, heterozygosity for SOX2 is associated with anopthalmiaesophageal-genital syndrome (OMIM 600992), a condition including esophageal atresia (EA) and tracheoesophageal fistula (TEF), in which the trachea and esophagus fail to separate. Mouse embryos heterozygous for the null allele, Sox2EGFP, appear normal. However, further reductions in Sox2, using Sox2LP and Sox2COND hypomorphic alleles, result in multiple abnormalities. Approximately 60% of Sox2EGFP/COND embryos have EA with distal TEF in which Sox2 is undetectable by immunohistochemistry or western blot. The mutant esophagus morphologically resembles the trachea, with ectopic expression of Nkx2.1, a columnar, ciliated epithelium, and very few p63+ basal cells. By contrast, the abnormal foregut of Nkx2.1-null embryos expresses elevated Sox2 and p63, suggesting reciprocal regulation of Sox2 and Nkx2.1 during early dorsal/ventral foregut patterning. Organ culture experiments further suggest that FGF signaling from the ventral mesenchyme regulates Sox2 expression in the endoderm. In the 40% Sox2EGFP/COND embryos in which Sox2 levels are ~18% of wild type there is no TEF. However, the esophagus is still abnormal, with luminal mucus-producing cells, fewer p63+ cells, and ectopic expression of genes normally expressed in glandular stomach and intestine. In all hypomorphic embryos the forestomach has an abnormal phenotype, with reduced keratinization, ectopic mucus cells and columnar epithelium. These findings suggest that Sox2 plays a second role in establishing the boundary between the keratinized, squamous esophagus/forestomach and glandular hindstomach.
- Published
- 2007
- Full Text
- View/download PDF
3. BRD4 regulates Nanog expression in mouse embryonic stem cells and preimplantation embryos
- Author
-
Liu, W, primary, Stein, P, additional, Cheng, X, additional, Yang, W, additional, Shao, N-Y, additional, Morrisey, E E, additional, Schultz, R M, additional, and You, J, additional
- Published
- 2014
- Full Text
- View/download PDF
4. WIKI4, a Novel Inhibitor of Tankyrase and Wnt/beta-Catenin Signaling
- Author
-
Morrisey, E, James, RG, Davidson, KC, Bosch, KA, Biechele, TL, Robin, NC, Taylor, RJ, Major, MB, Camp, ND, Fowler, K, Martins, TJ, Moon, RT, Morrisey, E, James, RG, Davidson, KC, Bosch, KA, Biechele, TL, Robin, NC, Taylor, RJ, Major, MB, Camp, ND, Fowler, K, Martins, TJ, and Moon, RT
- Abstract
The Wnt/ß-catenin signaling pathway controls important cellular events during development and often contributes to disease when dysregulated. Using high throughput screening we have identified a new small molecule inhibitor of Wnt/ß-catenin signaling, WIKI4. WIKI4 inhibits expression of ß-catenin target genes and cellular responses to Wnt/ß-catenin signaling in cancer cell lines as well as in human embryonic stem cells. Furthermore, we demonstrate that WIKI4 mediates its effects on Wnt/ß-catenin signaling by inhibiting the enzymatic activity of TNKS2, a regulator of AXIN ubiquitylation and degradation. While TNKS has previously been shown to be the target of small molecule inhibitors of Wnt/ß-catenin signaling, WIKI4 is structurally distinct from previously identified TNKS inhibitors.
- Published
- 2012
5. A house with many rooms: how the heart got its chambers with foxn4
- Author
-
Cohen, E. D., primary and Morrisey, E. E., additional
- Published
- 2008
- Full Text
- View/download PDF
6. GATA4 transcription factor is required for ventral morphogenesis and heart tube formation.
- Author
-
Kuo, C T, primary, Morrisey, E E, additional, Anandappa, R, additional, Sigrist, K, additional, Lu, M M, additional, Parmacek, M S, additional, Soudais, C, additional, and Leiden, J M, additional
- Published
- 1997
- Full Text
- View/download PDF
7. Characterization of Wnt gene expression in developing and postnatal hair follicles and identification of Wnt5a as a target of Sonic hedgehog in hair follicle morphogenesis
- Author
-
Reddy, S., Andl, T., Bagasra, A., Lu, M. M., Epstein, D. J., Morrisey, E. E., and Millar, S. E.
- Published
- 2001
- Full Text
- View/download PDF
8. GATA-6 activates transcription of surfactant protein A.
- Author
-
Bruno, M D, Korfhagen, T R, Liu, C, Morrisey, E E, and Whitsett, J A
- Abstract
Surfactant protein A (SP-A) is a member of the collectin family of innate host defense molecules expressed primarily in respiratory epithelial cells of the lung. SP-A concentrations are influenced by both cell-specific and ubiquitous nuclear proteins that regulate SP-A gene transcription in a cell-selective and temporally regulated manner. In this work, a consensus GATA-binding site (GBS) was identified at positions -69 to -64 of the mouse SP-A gene. The transcriptional activity of wild-type SP-A reporter constructs in HeLa cells was increased 5-10-fold when cotransfected with a GATA-6 expression plasmid. Deletion of the GBS completely blocked transactivation by GATA-6. Transfection of a construct expressing GATA-6-engrailed fusion protein inhibited basal expression of the SP-A/chloramphenicol acetyltransferase construct in MLE-15 cells. Nuclear extract proteins from MLE-15 cells bound to the GBS in the mouse SP-A gene, and a supershifted band was detected with a GATA-6-specific antibody. Transactivation of the wild-type SP-A constructs by GATA-6 increased transcriptional activity 7-10-fold, whereas thyroid transcription factor-1 (TTF-1) increased the activity of these constructs 12-18-fold. The effects of cotransactivating with both GATA-6 and TTF-1 expression constructs were additive. However, mutation of the TTF-1-binding sites alone or in combination decreased GATA-6 transactivation. Likewise, mutation of the GBS blocked TTF-1 activation of the SP-A promoter. In situ hybridization demonstrated GATA-6 mRNA in the peripheral epithelial cells of fetal mouse lung, consistent with the sites of SP-A expression. GATA-6 is expressed in respiratory epithelial cells and binds to a cis-acting element in the SP-A gene promoter, activating the transcriptional activity of the gene.
- Published
- 2000
9. GATA-4 activates transcription via two novel domains that are conserved within the GATA-4/5/6 subfamily.
- Author
-
Morrisey, E E, Ip, H S, Tang, Z, and Parmacek, M S
- Abstract
GATA-4 is one of the earliest developmental markers of the precardiac mesoderm, heart, and gut and has been shown to activate regulatory elements controlling transcription of genes encoding cardiac-specific proteins. To elucidate the molecular mechanisms underlying the transcriptional activity of the GATA-4 protein, structure-function analyses were performed. These analyses revealed that the C-terminal zinc finger and adjacent basic domain of GATA-4 is bifunctional, modulating both DNA-binding and nuclear localization activities. The N terminus of the protein encodes two independent transcriptional Activation Domains (amino acids 1-74 and amino acids 130-177). Amino acid residues were identified within each domain that are required for transcriptional activation. Finally, we have shown that regions of Xenopus GATA-5 and -6 corresponding to Activation Domains I and II, respectively, function as potent transcriptional activators. The identification and functional characterization of two evolutionarily conserved transcriptional Activation Domains within the GATA-4/5/6 subfamily suggests that each of these domains modulates critical functions in the transcriptional regulatory program(s) encoded by GATA-4, -5, and -6 during vertebrate development. As such these data provide novel insights into the molecular mechanisms that control development of the heart.
- Published
- 1997
10. GATA6 regulates HNF4 and is required for differentiation of visceral endoderm in the mouse embryo.
- Author
-
Morrisey, E E, Tang, Z, Sigrist, K, Lu, M M, Jiang, F, Ip, H S, and Parmacek, M S
- Abstract
GATA6 belongs to a family of zinc finger transcription factors that play important roles in transducing nuclear events that regulate cellular differentiation and embryonic morphogenesis in vertebrate species. To examine the function of GATA6 during embryonic development, gene targeting was used to generate GATA6-deficient (GATA6(-/-)) ES cells and mice harboring a null mutation in GATA6. Differentiated embryoid bodies derived from GATA6(-/-) ES cells lack a covering layer of visceral endoderm and severely attenuate, or fail to express, genes encoding early and late endodermal markers, including HNF4, GATA4, alpha-fetoprotein (AFP), and HNF3beta. Homozygous GATA6(-/-) mice died between embryonic day (E) 6.5 and E7. 5 and exhibited a specific defect in endoderm differentiation including severely down-regulated expression of GATA4 and absence of HNF4 gene expression. Moreover, widespread programmed cell death was observed within the embryonic ectoderm of GATA6-deficient embryos, a finding also observed in HNF4-deficient embryos. Consistent with these data, forced expression of GATA6 activated the HNF4 promoter in nonendodermal cells. Finally, to examine the function of GATA6 during later embryonic development, GATA6(-/-)-C57BL/6 chimeric mice were generated. lacZ-tagged GATA6(-/-) ES cells contributed to all embryonic tissues with the exception of the endodermally derived bronchial epithelium. Taken together, these data suggest a model in which GATA6 lies upstream of HNF4 in a transcriptional cascade that regulates differentiation of the visceral endoderm. In addition, these data demonstrate that GATA6 is required for establishment of the endodermally derived bronchial epithelium.
- Published
- 1998
11. Developmental pattern of expression and genomic organization of the calponin-h1 gene. A contractile smooth muscle cell marker.
- Author
-
Samaha, F F, Ip, H S, Morrisey, E E, Seltzer, J, Tang, Z, Solway, J, and Parmacek, M S
- Abstract
Calponin-h1 is a 34-kDa myofibrillar thin filament, actin-binding protein that is expressed exclusively in smooth muscle cells (SMCs) in adult animals. To examine the molecular mechanisms that regulate SMC-specific gene expression, we have examined the temporal, spatial, and cell cycle-regulated patterns of expression of calponin-h1 gene expression and isolated and structurally characterized the murine calponin-h1 gene. Calponin-h1 mRNA is expressed exclusively in SMC-containing tissues in adult animals. During murine embryonic development, calponin-h1 gene expression is (i) detectable in E9.5 embryos in the dorsal aorta, cardiac outflow tract, and tubular heart, (ii) sequentially up-regulated in SMC-containing tissues, and (iii) down-regulated to non-detectable levels in the heart during late fetal development. In addition, the gene is expressed in resting rat aortic SMCs, but its expression is rapidly down-regulated when growth-arrested cells reenter phase G1 of the cell cycle and proliferate. Calponin-h1 is encoded by a 10.7-kilobase single copy gene composed of seven exons, which is part of a multigene family. Transient transfection analyses demonstrated that 1.5 kilobases of calponin-h1 5'-flanking sequence is sufficient to program high level transcription of a luciferase reporter gene in cultured primary rat aortic SMCs and the smooth muscle cell line, A7r5. Taken together, these data suggest that the calponin-h1 gene will serve as an excellent model system with which to examine the molecular mechanisms that regulate SMC lineage specification, differentiation, and phenotypic modulation.
- Published
- 1996
12. Structure and expression of a smooth muscle cell-specific gene, SM22 alpha.
- Author
-
Solway, J, Seltzer, J, Samaha, F F, Kim, S, Alger, L E, Niu, Q, Morrisey, E E, Ip, H S, and Parmacek, M S
- Abstract
SM22 alpha is expressed exclusively in smooth muscle-containing tissues of adult animals and is one of the earliest markers of differentiated smooth muscle cells (SMCs). To examine the molecular mechanisms that regulate SMC-specific gene expression, we have isolated and structurally characterized the murine SM22 alpha gene. SM22 alpha is a 6.2-kilobase single copy gene composed of five exons. SM22 alpha mRNA is expressed at high levels in the aorta, uterus, lung, and intestine, and in primary cultures of rat aortic SMCs, and the SMC line, A7r5. In contrast to genes encoding SMC contractile proteins, SM22 alpha gene expression is not decreased in proliferating SMCs. Transient transfection experiments demonstrated that 441 base pairs of SM22 alpha 5'-flanking sequence was necessary and sufficient to program high level transcription of a luciferase reporter gene in both primary rat aortic SMCs and A7r5 cells. DNA sequence analyses revealed that the 441-base pair promoter contains two CArG/SRF boxes, a CACC box, and one potential MEF-2 binding site, cis-acting elements which are each important regulators of striated muscle transcription. Taken together, these studies have identified the murine SM22 alpha promoter as an excellent model system for studies of developmentally regulated, lineage-specific gene expression in SMCs.
- Published
- 1995
13. Compartmented, Filament Wound, One-Piece Aircraft Fuel Tanks.
- Author
-
DEPARTMENT OF THE AIR FORCE WASHINGTON DC, Morrisey,E J, DEPARTMENT OF THE AIR FORCE WASHINGTON DC, and Morrisey,E J
- Abstract
An external aircraft fuel tank is provided which comprises a filament-reinforced fuel cell assembled with aerodynamic end shapes having a plurality of layers of adhesive resin-impregnated filament wrappings over the assembly. Also provided is a method for making the fuel tank. (Author), Supersedes PAT-APP-582 514-84.
- Published
- 1985
14. Method of Making Compartmented, Filament Wound, One-Piece Aircraft Fuel Tanks.
- Author
-
DEPARTMENT OF THE AIR FORCE WASHINGTON DC, Morrisey,E J, DEPARTMENT OF THE AIR FORCE WASHINGTON DC, and Morrisey,E J
- Abstract
An external aircraft fuel tank is provided which comprises a filament-reinforced fuel cell assembled with aerodynamic end shapes having a plurality of layers of adhesive resin-impregnated filament wrappings over the assembly. Also provided is a method for making the fuel tank. (Author), Supersedes PAT-APPL-339 258-82, AD-D009 351.
- Published
- 1984
15. Investigation of Perforated Plastic Spheres Concept for Fuel Tank Fire Suppression.
- Author
-
MONSANTO RESEARCH CORP DAYTON OH, Botteri, B. P., Gandee, G. W., Morrisey, E. J., MONSANTO RESEARCH CORP DAYTON OH, Botteri, B. P., Gandee, G. W., and Morrisey, E. J.
- Abstract
An investigation was conducted to establish the feasibility of using perforated hollow plastic spheres to pack aircraft fuel tanks to provide fire and explosion suppression capability. The program involved establishing several sphere configurations, producing test quantities, determining performance under electrical spark and incendiary gunfire conditions, and evaluating fuel system compatibility. These sphere configurations varying in diameter from 3/4 to 1 inch with perforations of 0.060 to 0.100 mils were evaluated. All configurations provided some explosion suppression, but the goal of 3 psi maximum peak pressure rise required for fuel tank applications was not achieved. Fuel system compatibility was slightly inferior to that experienced with polyurethane foam., Sponsored in part by the Bureau of Mines, Explosives Research Center, Pittsburgh, Pa.
- Published
- 1969
16. Reciprocal requirements for EDA/EDAR/NF-kappaB and Wnt/beta- catenin signaling pathways in hair follicle induction
- Author
-
Zhang, Y., Tomann, P., Andl, T., Gallant, NM., Huelsken, J., Jerchow, B., Birchmeier, W., Paus, R., Piccolo, S., Mikkola, M. L., Morrisey, E. E., Overbeek, P. A., Scheidereit, C., Millar, S. E., and Schmidt-Ullrich, R.
- Subjects
integumentary system - Abstract
Wnt/beta-catenin and NF-kappaB signaling mechanisms provide central controls in development and disease, but how these pathways intersect is unclear. Using hair follicle induction as a model system, we show that patterning of dermal Wnt/beta-catenin signaling requires epithelial beta- catenin activity. We find that Wnt/beta-catenin signaling is absolutely required for NF-kappaB activation, and that Edar is a direct Wnt target gene. Wnt/beta-catenin signaling is initially activated independently of EDA/EDAR/NF-kappaB activity in primary hair follicle primordia. However, Eda/Edar/NF-kappaB signaling is required to refine the pattern of Wnt/beta-catenin activity, and to maintain this activity at later stages of placode development. We show that maintenance of localized expression of Wnt10b and Wnt10a requires NF-kappaB signaling, providing a molecular explanation for the latter observation, and identify Wnt10b as a direct NF-kappaB target. These data reveal a complex interplay and interdependence of Wnt/beta-catenin and EDA/EDAR/NF-kappaB signaling pathways in initiation and maintenance of primary hair follicle placodes.
17. beta-Catenin is required for specification of proximal/distal cell fate during lung morphogenesis
- Author
-
Mucenski, M. L., Wert, S. E., Nation, J. M., Loudy, D. E., Huelsken, J., Birchmeier, W., Morrisey, E. E., and Whitsett, J. A.
- Subjects
respiratory system ,respiratory tract diseases - Abstract
The lungs are divided, both structurally and functionally, into two distinct components, the proximal airways, which conduct air, and the peripheral airways, which mediate gas exchange. The mechanisms that control the specification of these two structures during lung development are currently unknown. Here we show that beta-catenin signaling is required for the formation of the distal, but not the proximal, airways. When the gene for beta-catenin was conditionally excised in epithelial cells of the developing mouse lung prior to embryonic day 14.5, the proximal lung tubules grew and differentiated appropriately. The mice, however, died at birth because of respiratory failure. Analysis of the lungs by in situ hybridization and immunohistochemistry, using molecular markers of the epithelial and mesenchymal components of both proximal and peripheral airways, showed that the lungs were composed primarily of proximal airways. These observations establish, for the first time, both the sites and timing of specification of the proximal and peripheral airways in the developing lung, and that beta-catenin is one of the essential components of this specification.
18. Airway-derived emphysema-specific alveolar type II cells exhibit impaired regenerative potential in COPD.
- Author
-
Hu Y, Hu Q, Ansari M, Riemondy K, Pineda R, Sembrat J, Leme AS, Ngo K, Morgenthaler O, Ha K, Gao B, Janssen WJ, Basil MC, Kliment CR, Morrisey E, Lehmann M, Evans CM, Schiller HB, and Königshoff M
- Subjects
- Animals, Humans, Mice, Male, Disease Models, Animal, Pulmonary Surfactant-Associated Protein C metabolism, Female, Organoids, Uteroglobin metabolism, Uteroglobin genetics, Pulmonary Alveoli cytology, Pulmonary Alveoli pathology, Stem Cells cytology, Stem Cells metabolism, Middle Aged, Pulmonary Disease, Chronic Obstructive physiopathology, Pulmonary Disease, Chronic Obstructive metabolism, Alveolar Epithelial Cells metabolism, Regeneration, Pulmonary Emphysema physiopathology, Pulmonary Emphysema metabolism
- Abstract
Emphysema, the progressive destruction of gas exchange surfaces in the lungs, is a hallmark of COPD that is presently incurable. This therapeutic gap is largely due to a poor understanding of potential drivers of impaired tissue regeneration, such as abnormal lung epithelial progenitor cells, including alveolar type II (ATII) and airway club cells. We discovered an emphysema-specific subpopulation of ATII cells located in enlarged distal alveolar sacs, termed asATII cells. Single-cell RNA sequencing and in situ localisation revealed that asATII cells co-express the alveolar marker surfactant protein C and the club cell marker secretaglobin-3A2 (SCGB3A2). A similar ATII subpopulation derived from club cells was also identified in mouse COPD models using lineage labelling. Human and mouse ATII subpopulations formed 80-90% fewer alveolar organoids than healthy controls, indicating reduced progenitor function. Targeting asATII cells or their progenitor club cells could reveal novel COPD treatment strategies., Competing Interests: Conflict of interest: C.M. Evans reports grants from Cystic Fibrosis Foundation, Department of Defense, and Enterprise Therapeutics, outside the submitted work, and royalties from Eleven P15 consulting. C.R. Kliment serves on an advisory board of Verona Pharmaceuticals. The remaining authors have no conflicts of interest., (Copyright ©The authors 2024.)
- Published
- 2024
- Full Text
- View/download PDF
19. Author Correction: SARS-CoV-2 disrupts host epigenetic regulation via histone mimicry.
- Author
-
Kee J, Thudium S, Renner DM, Glastad K, Palozola K, Zhang Z, Li Y, Lan Y, Cesare J, Poleshko A, Kiseleva AA, Truitt R, Cardenas-Diaz FL, Zhang X, Xie X, Kotton DN, Alysandratos KD, Epstein JA, Shi PY, Yang W, Morrisey E, Garcia BA, Berger SL, Weiss SR, and Korb E
- Published
- 2023
- Full Text
- View/download PDF
20. Publisher Correction: SARS-CoV-2 disrupts host epigenetic regulation via histone mimicry.
- Author
-
Kee J, Thudium S, Renner DM, Glastad K, Palozola K, Zhang Z, Li Y, Lan Y, Cesare J, Poleshko A, Kiseleva AA, Truitt R, Cardenas-Diaz FL, Zhang X, Xie X, Kotton DN, Alysandratos KD, Epstein JA, Shi PY, Yang W, Morrisey E, Garcia BA, Berger SL, Weiss SR, and Korb E
- Published
- 2023
- Full Text
- View/download PDF
21. SARS-CoV-2 disrupts host epigenetic regulation via histone mimicry.
- Author
-
Kee J, Thudium S, Renner DM, Glastad K, Palozola K, Zhang Z, Li Y, Lan Y, Cesare J, Poleshko A, Kiseleva AA, Truitt R, Cardenas-Diaz FL, Zhang X, Xie X, Kotton DN, Alysandratos KD, Epstein JA, Shi PY, Yang W, Morrisey E, Garcia BA, Berger SL, Weiss SR, and Korb E
- Subjects
- Chromatin genetics, Chromatin metabolism, Chromatin Assembly and Disassembly, Epigenome genetics, Humans, COVID-19 genetics, COVID-19 metabolism, COVID-19 virology, Epigenesis, Genetic, Histones chemistry, Histones metabolism, Host Microbial Interactions, Molecular Mimicry, SARS-CoV-2 genetics, SARS-CoV-2 metabolism, SARS-CoV-2 pathogenicity, Viral Proteins chemistry, Viral Proteins genetics, Viral Proteins metabolism
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and caused the devastating global pandemic of coronavirus disease 2019 (COVID-19), in part because of its ability to effectively suppress host cell responses
1-3 . In rare cases, viral proteins dampen antiviral responses by mimicking critical regions of human histone proteins4-8 , particularly those containing post-translational modifications required for transcriptional regulation9-11 . Recent work has demonstrated that SARS-CoV-2 markedly disrupts host cell epigenetic regulation12-14 . However, how SARS-CoV-2 controls the host cell epigenome and whether it uses histone mimicry to do so remain unclear. Here we show that the SARS-CoV-2 protein encoded by ORF8 (ORF8) functions as a histone mimic of the ARKS motifs in histone H3 to disrupt host cell epigenetic regulation. ORF8 is associated with chromatin, disrupts regulation of critical histone post-translational modifications and promotes chromatin compaction. Deletion of either the ORF8 gene or the histone mimic site attenuates the ability of SARS-CoV-2 to disrupt host cell chromatin, affects the transcriptional response to infection and attenuates viral genome copy number. These findings demonstrate a new function of ORF8 and a mechanism through which SARS-CoV-2 disrupts host cell epigenetic regulation. Further, this work provides a molecular basis for the finding that SARS-CoV-2 lacking ORF8 is associated with decreased severity of COVID-19., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
22. Endothelial Foxp1 Regulates Neointimal Hyperplasia Via Matrix Metalloproteinase-9/Cyclin Dependent Kinase Inhibitor 1B Signal Pathway.
- Author
-
Chen X, Xu J, Bao W, Li H, Wu W, Liu J, Pi J, Tomlinson B, Chan P, Ruan C, Zhang Q, Zhang L, Fan H, Morrisey E, Liu Z, Zhang Y, Lin L, Liu J, and Zhuang T
- Subjects
- Animals, Cell Movement, Cell Proliferation, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Endothelium metabolism, Forkhead Transcription Factors, Hyperplasia pathology, Mice, Myocytes, Smooth Muscle metabolism, Repressor Proteins metabolism, Signal Transduction, Transcription Factors metabolism, Matrix Metalloproteinase 9 metabolism, Neointima pathology
- Abstract
Background The endothelium is essential for maintaining vascular physiological homeostasis and the endothelial injury leads to the neointimal hyperplasia because of the excessive proliferation of vascular smooth muscle cells. Endothelial Foxp1 (forkhead box P1) has been shown to control endothelial cell (EC) proliferation and migration in vitro. However, whether EC-Foxp1 participates in neointimal formation in vivo is not clear. Our study aimed to investigate the roles and mechanisms of EC-Foxp1 in neointimal hyperplasia. Methods and Results The wire injury femoral artery neointimal hyperplasia model was performed in Foxp1 EC-specific loss-of-function and gain-of-function mice. EC-Foxp1 deletion mice displayed the increased neointimal formation through elevation of vascular smooth muscle cell proliferation and migration, and the reduction of EC proliferation hence reendothelialization after injury. In contrast, EC-Foxp1 overexpression inhibited the neointimal formation. EC-Foxp1 paracrine regulated vascular smooth muscle cell proliferation and migration via targeting matrix metalloproteinase-9. Also, EC-Foxp1 deletion impaired EC repair through reduction of EC proliferation via increasing cyclin dependent kinase inhibitor 1B expression. Delivery of cyclin dependent kinase inhibitor 1B-siRNA to ECs using RGD (Arg-Gly-Asp)-peptide magnetic nanoparticle normalized the EC-Foxp1 deletion-mediated impaired EC repair and attenuated the neointimal formation. EC-Foxp1 regulates matrix metalloproteinase-9/cyclin dependent kinase inhibitor 1B signaling pathway to control injury induced neointimal formation. Conclusions Our study reveals that targeting EC-Foxp1-matrix metalloproteinase-9/cyclin dependent kinase inhibitor 1B pathway might provide future novel therapeutic interventions for restenosis.
- Published
- 2022
- Full Text
- View/download PDF
23. National Heart, Lung, and Blood Institute and Building Respiratory Epithelium and Tissue for Health (BREATH) Consortium Workshop Report: Moving Forward in Lung Regeneration.
- Author
-
Hynds RE, Zacharias WJ, Nikolić MZ, Königshoff M, Eickelberg O, Gosens R, de Coppi P, Janes SM, Morrisey E, Clevers H, Ryan AL, Stripp BR, Sun X, Kim CF, and Lin QS
- Subjects
- Animals, Cell- and Tissue-Based Therapy, Congresses as Topic, Education, Humans, National Heart, Lung, and Blood Institute (U.S.), United States, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells transplantation, Lung physiology, Lung Diseases metabolism, Lung Diseases therapy, Regeneration, Respiratory Mucosa physiology
- Abstract
The National Heart, Lung, and Blood Institute of the National Institutes of Health, together with the Longfonds BREATH consortium, convened a working group to review the field of lung regeneration and suggest avenues for future research. The meeting took place on May 22, 2019, at the American Thoracic Society 2019 conference in Dallas, Texas, United States, and brought together investigators studying lung development, adult stem-cell biology, induced pluripotent stem cells, biomaterials, and respiratory disease. The purpose of the working group was 1 ) to examine the present status of basic science approaches to tackling lung disease and promoting lung regeneration in patients and 2 ) to determine priorities for future research in the field.
- Published
- 2021
- Full Text
- View/download PDF
24. SARS-CoV-2 induces double-stranded RNA-mediated innate immune responses in respiratory epithelial derived cells and cardiomyocytes.
- Author
-
Li Y, Renner DM, Comar CE, Whelan JN, Reyes HM, Cardenas-Diaz FL, Truitt R, Tan LH, Dong B, Alysandratos KD, Huang J, Palmer JN, Adappa ND, Kohanski MA, Kotton DN, Silverman RH, Yang W, Morrisey E, Cohen NA, and Weiss SR
- Abstract
Coronaviruses are adept at evading host antiviral pathways induced by viral double-stranded RNA, including interferon (IFN) signaling, oligoadenylate synthetase-ribonuclease L (OAS-RNase L), and protein kinase R (PKR). While dysregulated or inadequate IFN responses have been associated with severe coronavirus infection, the extent to which the recently emerged SARS-CoV-2 activates or antagonizes these pathways is relatively unknown. We found that SARS-CoV-2 infects patient-derived nasal epithelial cells, present at the initial site of infection, induced pluripotent stem cell-derived alveolar type 2 cells (iAT2), the major cell type infected in the lung, and cardiomyocytes (iCM), consistent with cardiovascular consequences of COVID-19 disease. Robust activation of IFN or OAS-RNase L is not observed in these cell types, while PKR activation is evident in iAT2 and iCM. In SARS-CoV-2 infected Calu-3 and A549
ACE2 lung-derived cell lines, IFN induction remains relatively weak; however activation of OAS-RNase L and PKR is observed. This is in contrast to MERS-CoV, which effectively inhibits IFN signaling as well as OAS-RNase L and PKR pathways, but similar to mutant MERS-CoV lacking innate immune antagonists. Remarkably, both OAS-RNase L and PKR are activated in MAVS knockout A549ACE2 cells, demonstrating that SARS-CoV-2 can induce these host antiviral pathways despite minimal IFN production. Moreover, increased replication and cytopathic effect in RNASEL knockout A549ACE2 cells implicates OAS-RNase L in restricting SARS-CoV-2. Finally, while SARS-CoV-2 fails to antagonize these host defense pathways, which contrasts with other coronaviruses, the IFN signaling response is generally weak. These host-virus interactions may contribute to the unique pathogenesis of SARS-CoV-2., Significance: SARS-CoV-2 emergence in late 2019 led to the COVID-19 pandemic that has had devastating effects on human health and the economy. Early innate immune responses are essential for protection against virus invasion. While inadequate innate immune responses are associated with severe COVID-19 diseases, understanding of the interaction of SARS-CoV-2 with host antiviral pathways is minimal. We have characterized the innate immune response to SARS-CoV-2 infections in relevant respiratory tract derived cells and cardiomyocytes and found that SARS-CoV-2 activates two antiviral pathways, oligoadenylate synthetase-ribonuclease L (OAS-RNase L), and protein kinase R (PKR), while inducing minimal levels of interferon. This in contrast to MERS-CoV which inhibits all three pathways. Activation of these pathways may contribute to the distinctive pathogenesis of SARS-CoV-2.- Published
- 2020
- Full Text
- View/download PDF
25. Endothelial Foxp1 Suppresses Atherosclerosis via Modulation of Nlrp3 Inflammasome Activation.
- Author
-
Zhuang T, Liu J, Chen X, Zhang L, Pi J, Sun H, Li L, Bauer R, Wang H, Yu Z, Zhang Q, Tomlinson B, Chan P, Zheng X, Morrisey E, Liu Z, Reilly M, and Zhang Y
- Subjects
- Animals, Atherosclerosis genetics, Atherosclerosis pathology, Endothelial Cells pathology, Forkhead Transcription Factors genetics, Human Umbilical Vein Endothelial Cells pathology, Humans, Inflammasomes genetics, Inflammasomes metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, NLR Family, Pyrin Domain-Containing 3 Protein genetics, Repressor Proteins genetics, Atherosclerosis metabolism, Endothelial Cells metabolism, Forkhead Transcription Factors metabolism, Human Umbilical Vein Endothelial Cells metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Repressor Proteins metabolism
- Abstract
Rationale: Endothelial dysfunction results in sustained and chronic vascular inflammation, which is central to atherosclerotic diseases. However, transcriptional regulation of vascular endothelial inflammation has not been well clarified., Objective: This study aims to explore Foxp (forkhead box P) transcription factor 1 in regulation of endothelial homeostasis, atherogenesis, and its mechanisms., Methods and Results: To assess the importance of Foxp1 in atherosclerosis, Foxp1 expression was analyzed in human coronary artery and mouse artery, and we observed significant downregulation of Foxp1 in atherosclerotic and atherosusceptible endothelium. Endothelial-specific Foxp1 knockout mice (Foxp1
ECKO ) were bred onto ApoeKO mice to generate endothelial Foxp1-deletion hyperlipidemic model Foxp1ECKO ;ApoeKO , which displayed significant increases in atherosclerotic lesion formation in aortas and aortic roots with enhanced monocyte adhesion, migration, and infiltration into the vascular wall and formation of inflammatory lipid-laden macrophages. In contrast, endothelial-specific Foxp1 overexpression mice Foxp1ECTg ;ApoeKO exhibited reduced atherosclerotic lesion formation with less monocyte infiltration. Foxp1 was further identified as a gatekeeper of vessel inflammation by direct regulation of endothelial inflammasome components, including Nlrp3 (NLR [nucleotide-binding and leucine-rich repeat immune receptors] family pyrin domain containing 3), caspase-1, and IL (interleukin)-1β. Moreover, endothelial Foxp1 was found to be regulated by Klf2 (Kruppel-like factor 2). Oscillatory shear stress downregulated Foxp1 expression via repressing Klf2 expression in endothelium, and, therefore, promoted endothelial inflammasome activation, leading to atherosclerotic lesion formation. Simvastatin upregulated the reduced expression of Klf2 and Foxp1 in atherosusceptible vascular endothelium and alleviated vascular inflammation contributing to its inhibitory effect in atherosclerosis., Conclusions: These data are the first in vivo experimental validation of an atheroprotective role of endothelial Klf2 and Foxp1, which reveals a Klf2-Foxp1 transcriptional network in endothelial cells as a novel regulator of endothelial inflammasome activation for atherogenesis, therefore, provides opportunities for therapeutic intervention of atherosclerotic diseases and uncovers a novel atheroprotective mechanism for simvastatin.- Published
- 2019
- Full Text
- View/download PDF
26. Endothelial Forkhead Box Transcription Factor P1 Regulates Pathological Cardiac Remodeling Through Transforming Growth Factor-β1-Endothelin-1 Signal Pathway.
- Author
-
Liu J, Zhuang T, Pi J, Chen X, Zhang Q, Li Y, Wang H, Shen Y, Tomlinson B, Chan P, Yu Z, Cheng Y, Zheng X, Reilly M, Morrisey E, Zhang L, Liu Z, and Zhang Y
- Subjects
- Angiotensin II metabolism, Animals, Aorta surgery, Disease Models, Animal, Endothelin-1 metabolism, Fibrosis, Forkhead Transcription Factors genetics, Heart Failure genetics, Humans, Mice, Mice, Knockout, Nanotubes, Peptide, RNA, Small Interfering genetics, Repressor Proteins genetics, Signal Transduction, Transforming Growth Factor beta1 genetics, Ventricular Remodeling, Endothelium, Vascular physiology, Forkhead Transcription Factors metabolism, Heart Failure metabolism, Myocardium pathology, Repressor Proteins metabolism, Transforming Growth Factor beta1 metabolism
- Abstract
Background: Pathological cardiac fibrosis and hypertrophy, the common features of left ventricular remodeling, often progress to heart failure. Forkhead box transcription factor P1 (Foxp1) in endothelial cells (ECs) has been shown to play an important role in heart development. However, the effect of EC-Foxp1 on pathological cardiac remodeling has not been well clarified. This study aims to determine the role of EC-Foxp1 in pathological cardiac remodeling and the underlying mechanisms., Methods: Foxp1 EC-specific loss-of-function and gain-of-function mice were generated, and an angiotensin II infusion or a transverse aortic constriction operation mouse model was used to study the cardiac remodeling mechanisms. Foxp1 downstream target gene transforming growth factor-β1 (TGF-β1) was confirmed by chromatin immunoprecipitation and luciferase assays. Finally, the effects of TGF-β1 blockade on EC-Foxp1 deletion-mediated profibrotic and prohypertrophic phenotypic changes were further confirmed by pharmacological inhibition, more specifically by RGD-peptide magnetic nanoparticle target delivery of TGF-β1-siRNA to ECs., Results: Foxp1 expression is significantly downregulated in cardiac ECs during angiotensin II-induced cardiac remodeling. EC-Foxp1 deletion results in severe cardiac remodeling, including more cardiac fibrosis with myofibroblast formation and extracellular matrix protein production, as well as decompensated cardiac hypertrophy and further exacerbation of cardiac dysfunction on angiotensin II infusion or transverse aortic constriction operation. In contrast, EC-Foxp1 gain of function protects against pathological cardiac remodeling and improves cardiac dysfunction. TGF-β1 signals are identified as Foxp1 direct target genes, and EC-Foxp1 deletion upregulates TGF-β1 signals to promote myofibroblast formation through fibroblast proliferation and transformation, resulting in severe cardiac fibrosis. Moreover, EC-Foxp1 deletion enhances TGF-β1-promoted endothelin-1 expression, which significantly increases cardiomyocyte size and reactivates cardiac fetal genes, leading to pathological cardiac hypertrophy. Correspondingly, these EC-Foxp1 deletion-mediated profibrotic and prohypertrophic phenotypic changes and cardiac dysfunction are normalized by the blockade of TGF-β1 signals through pharmacological inhibition and RGD-peptide magnetic nanoparticle target delivery of TGF-β1-siRNA to ECs., Conclusions: EC-Foxp1 regulates the TGF-β1-endothelin-1 pathway to control pathological cardiac fibrosis and hypertrophy, resulting in cardiac dysfunction. Therefore, targeting the EC-Foxp1-TGF-β1-endothelin-1 pathway might provide a future novel therapy for heart failure.
- Published
- 2019
- Full Text
- View/download PDF
27. Cell-Specific Effects of GATA (GATA Zinc Finger Transcription Factor Family)-6 in Vascular Smooth Muscle and Endothelial Cells on Vascular Injury Neointimal Formation.
- Author
-
Zhuang T, Liu J, Chen X, Pi J, Kuang Y, Wang Y, Tomlinson B, Chan P, Zhang Q, Li Y, Yu Z, Zheng X, Reilly M, Morrisey E, Zhang L, Liu Z, and Zhang Y
- Subjects
- Animals, Cell Movement genetics, Cell Proliferation genetics, Disease Models, Animal, Female, GATA6 Transcription Factor genetics, Hyperplasia pathology, Male, Mice, Mice, Inbred C57BL, Random Allocation, Sensitivity and Specificity, Transcription Factors metabolism, Gene Expression Regulation, Muscle, Smooth, Vascular metabolism, Neointima pathology, Vascular System Injuries pathology, Zinc Fingers genetics
- Abstract
Objective- Transcription factor GATA (GATA zinc finger transcription factor family)-6 is highly expressed in vessels and rapidly downregulated in balloon-injured carotid arteries and viral delivery of GATA-6 to the vessels limited the neointimal formation, however, little is known about its cell-specific regulation of in vivo vascular smooth muscle cell (VSMC) phenotypic state contributing to neointimal formation. This study aims to determine the role of vascular cell-specific GATA-6 in ligation- or injury-induced neointimal hyperplasia in vivo. Approach and Results- Endothelial cell and VSMC-specific GATA-6 deletion mice are generated, and the results indicate that endothelial cell-specific GATA-6 deletion mice exhibit significant decrease of VSMC proliferation and attenuation of neointimal formation after artery ligation and injury compared with the wild-type littermate control mice. PDGF (platelet-derived growth factor)-B is identified as a direct target gene, and endothelial cell-GATA-6-PDGF-B pathway regulates VSMC proliferation and migration in a paracrine manner which controls the neointimal formation. In contrast, VSMC-specific GATA-6 deletion promotes injury-induced VSMC transformation from contractile to proliferative synthetic phenotype leading to increased neointimal formation. CCN (cysteine-rich 61/connective tissue growth factor/nephroblastoma overexpressed family)-5 is identified as a novel target gene, and VSMC-specific CCN-5 overexpression in mice reverses the VSMC-GATA-6 deletion-mediated increased cell proliferation and migration and finally attenuates the neointimal formation. Conclusions- This study gives us a direct in vivo evidence of GATA-6 cell lineage-specific regulation of PDGF-B and CCN-5 on VSMC phenotypic state, proliferation and migration contributing to neointimal formation, which advances our understanding of in vivo neointimal hyperplasia, meanwhile also provides opportunities for future therapeutic interventions.
- Published
- 2019
- Full Text
- View/download PDF
28. Protein kinase R-like endoplasmatic reticulum kinase is a mediator of stretch in ventilator-induced lung injury.
- Author
-
Dolinay T, Aonbangkhen C, Zacharias W, Cantu E, Pogoriler J, Stablow A, Lawrence GG, Suzuki Y, Chenoweth DM, Morrisey E, Christie JD, Beers MF, and Margulies SS
- Subjects
- Adult, Aged, Animals, Female, Humans, Lung pathology, Male, Middle Aged, Pulmonary Stretch Receptors pathology, Rats, Rats, Sprague-Dawley, Respiratory Mucosa metabolism, Respiratory Mucosa pathology, Swine, Ventilator-Induced Lung Injury pathology, Endoplasmic Reticulum Stress physiology, Lung metabolism, Pulmonary Stretch Receptors metabolism, Ventilator-Induced Lung Injury metabolism, eIF-2 Kinase physiology
- Abstract
Background: Acute respiratory distress syndrome (ARDS) is a severe form of lung injury characterized by damage to the epithelial barrier with subsequent pulmonary edema and hypoxic respiratory failure. ARDS is a significant medical problem in intensive care units with associated high care costs. There are many potential causes of ARDS; however, alveolar injury associated with mechanical ventilation, termed ventilator-induced lung injury (VILI), remains a well-recognized contributor. It is thus critical to understand the mechanism of VILI. Based on our published preliminary data, we hypothesized that the endoplasmic reticulum (ER) stress response molecule Protein Kinase R-like Endoplasmic Reticulum Kinase (PERK) plays a role in transmitting mechanosensory signals the alveolar epithelium., Methods: ER stress signal responses to mechanical stretch were studied in ex-vivo ventilated pig lungs. To explore the effect of PERK inhibition on VILI, we ventilated live rats and compared lung injury parameters to non-ventilated controls. The effect of stretch-induced epithelial ER Ca
2+ signaling on PERK was studied in stretched alveolar epithelial monolayers. To confirm the activation of PERK in human disease, ER stress signaling was compared between ARDS and non-ARDS lungs., Results: Our studies revealed increased PERK-specific ER stress signaling in response to overstretch. PERK inhibition resulted in dose-dependent improvement of alveolar inflammation and permeability. Our data indicate that stretch-induced epithelial ER Ca2+ release is an activator of PERK. Experiments with human lung tissue confirmed PERK activation by ARDS., Conclusion: Our study provides evidences that PERK is a mediator stretch signals in the alveolar epithelium.- Published
- 2018
- Full Text
- View/download PDF
29. Elevated Expression of miR302-367 in Endothelial Cells Inhibits Developmental Angiogenesis via CDC42/CCND1 Mediated Signaling Pathways.
- Author
-
Pi J, Liu J, Zhuang T, Zhang L, Sun H, Chen X, Zhao Q, Kuang Y, Peng S, Zhou X, Yu Z, Tao T, Tomlinson B, Chan P, Tian Y, Fan H, Liu Z, Zheng X, Morrisey E, and Zhang Y
- Subjects
- Animals, Antagomirs genetics, Antagomirs metabolism, Base Sequence, Binding Sites, Cell Movement, Cell Proliferation, Cyclin D1 metabolism, Embryo, Mammalian, Endothelial Cells cytology, Female, Mice, Mice, Transgenic, MicroRNAs agonists, MicroRNAs antagonists & inhibitors, MicroRNAs metabolism, Oligoribonucleotides genetics, Oligoribonucleotides metabolism, Pregnancy, Primary Cell Culture, Rhombencephalon blood supply, Rhombencephalon growth & development, Signal Transduction, cdc42 GTP-Binding Protein metabolism, Cyclin D1 genetics, Endothelial Cells metabolism, Gene Expression Regulation, Developmental, MicroRNAs genetics, Neovascularization, Physiologic genetics, Rhombencephalon metabolism, cdc42 GTP-Binding Protein genetics
- Abstract
Rationale: Angiogenesis is critical for embryonic development and microRNAs fine-tune this process, but the underlying mechanisms remain incompletely understood. Methods: Endothelial cell (EC) specific miR302-367 line was used as gain-of-function and anti-miRs as loss-of-function models to investigate the effects of miR302-367 on developmental angiogenesis with embryonic hindbrain vasculature as an in vivo model and fibrin gel beads and tube formation assay as in vitro models. Cell migration was evaluated by Boyden chamber and scratch wound healing assay and cell proliferation by cell count, MTT assay, Ki67 immunostaining and PI cell cycle analysis. RNA high-throughput sequencing identified miR-target genes confirmed by chromatin immunoprecipitation and 3'-UTR luciferase reporter assay, and finally target site blocker determined the pathway contributing significantly to the phenotype observed upon microRNA expression. Results: Elevated EC miR302-367 expression reduced developmental angiogenesis, whereas it was enhanced by inhibition of miR302-367, possibly due to the intrinsic inhibitory effects on EC migration and proliferation. We identified Cdc42 as a direct target gene and elevated EC miR302-367 decreased total and active Cdc42, and further inhibited F-actin formation via the WASP and Klf2/Grb2/Pak1/LIM-kinase/Cofilin pathways. MiR302-367-mediated-Klf2 regulation of Grb2 for fine-tuning Pak1 activation contributing to the inhibited F-actin formation, and then the attenuation of EC migration. Moreover, miR302-367 directly down-regulated EC Ccnd1 and impaired cell proliferation via the Rb/E2F pathway. Conclusion: miR302-367 regulation of endothelial Cdc42 and Ccnd1 signal pathways for EC migration and proliferation advances our understanding of developmental angiogenesis, and meanwhile provides a rationale for future interventions of pathological angiogenesis that shares many common features of physiological angiogenesis., Competing Interests: Competing interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
30. A MicroRNA302-367-Erk1/2-Klf2-S1pr1 Pathway Prevents Tumor Growth via Restricting Angiogenesis and Improving Vascular Stability.
- Author
-
Pi J, Tao T, Zhuang T, Sun H, Chen X, Liu J, Cheng Y, Yu Z, Zhu HH, Gao WQ, Suo Y, Wei X, Chan P, Zheng X, Tian Y, Morrisey E, Zhang L, and Zhang Y
- Subjects
- Angiogenesis Inhibitors biosynthesis, Animals, Carcinoma, Lewis Lung, Coculture Techniques, Human Umbilical Vein Endothelial Cells metabolism, Humans, Melanoma, Experimental, Mice, Mice, Transgenic, Neoplasms pathology, Neoplasms prevention & control, Neovascularization, Pathologic pathology, Neovascularization, Pathologic prevention & control, Sphingosine-1-Phosphate Receptors, Xenograft Model Antitumor Assays methods, Kruppel-Like Transcription Factors biosynthesis, MAP Kinase Signaling System physiology, MicroRNAs biosynthesis, Neoplasms metabolism, Neovascularization, Pathologic metabolism, Receptors, Lysosphingolipid biosynthesis
- Abstract
Rationale: Angiogenic hypersprouting and leaky vessels are essential for tumor growth. MicroRNAs have unique therapeutic advantages by targeting multiple pathways of tumor-associated angiogenesis, but the function of individual miRNAs of miR302-367 cluster in angiogenesis and tumors has not yet been fully evaluated., Objective: To investigate the functions of miR302-367 in developmental angiogenesis and tumor angiogenesis and explore the molecular mechanisms of microRNA for the treatment of pathological neovascularization-related diseases., Methods and Results: Here, we show that miR302-367 elevation in endothelial cells reduces retinal sprouting angiogenesis and promotes vascular stability in vivo, ex vivo, and in vitro. Erk1/2 is identified as direct target of miR302-367, and downregulation of Erk1/2 on miR302-367 elevation in endothelial cells increases the expression of Klf2 and in turn S1pr1 and its downstream target VE-cadherin, suppressing angiogenesis and improving vascular stability. Conversely, both pharmacological blockade and genetic deletion of S1pr1 in endothelial cells reverse the antiangiogenic and vascular stabilizing effect of miR302-367 in mice. Tumor angiogenesis shares features of developmental angiogenesis, and endothelial specific elevation of miR302-367 reduces tumor growth by restricting sprout angiogenesis and decreasing vascular permeability via the same Erk1/2-Klf2-S1pr1 pathways., Conclusions: MiR302-367 regulation of an Erk1/2-Klf2-S1pr1 pathway in the endothelium advances our understanding of angiogenesis, meanwhile also provides opportunities for therapeutic intervention of tumor growth., (© 2016 American Heart Association, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
31. Regulation of cardiomyocyte proliferation by Foxp1.
- Author
-
Wang Y and Morrisey E
- Subjects
- Animals, Cell Proliferation, Chromatin Immunoprecipitation, Fibroblast Growth Factors metabolism, Forkhead Transcription Factors genetics, HMGB Proteins metabolism, Mice, Models, Biological, Mutation, Repressor Proteins genetics, SOXF Transcription Factors metabolism, Wnt Proteins metabolism, beta Catenin metabolism, Forkhead Transcription Factors metabolism, Myocytes, Cardiac cytology, Repressor Proteins metabolism
- Published
- 2010
- Full Text
- View/download PDF
32. Altered ultrasonic vocalization in mice with a disruption in the Foxp2 gene.
- Author
-
Shu W, Cho JY, Jiang Y, Zhang M, Weisz D, Elder GA, Schmeidler J, De Gasperi R, Sosa MA, Rabidou D, Santucci AC, Perl D, Morrisey E, and Buxbaum JD
- Subjects
- Animals, Blotting, Southern, Cerebellar Cortex pathology, Forkhead Transcription Factors, Immunohistochemistry, In Situ Nick-End Labeling, Mice physiology, Purkinje Cells pathology, Cerebellar Cortex abnormalities, Mice genetics, Repressor Proteins genetics, Ultrasonics, Vocalization, Animal
- Abstract
Neurobiology of speech and language has previously been studied in the KE family, in which half of the members have severe impairment in both speech and language. The gene responsible for the phenotype was mapped to chromosome 7q31 and identified as the FOXP2 gene, coding for a transcription factor containing a polyglutamine tract and a forkhead DNA-binding domain. Because of linkage studies implicating 7q31 in autism, where language impairment is a component of the disorder, and in specific language impairment, FOXP2 has also been considered as a potential susceptibility locus for the language deficits in autism and/or specific language impairment. In this study, we characterized mice with a disruption in the murine Foxp2 gene. Disruption of both copies of the Foxp2 gene caused severe motor impairment, premature death, and an absence of ultrasonic vocalizations that are elicited when pups are removed from their mothers. Disruption of a single copy of the gene led to modest developmental delay but a significant alteration in ultrasonic vocalization in response to such separation. Learning and memory appear normal in the heterozygous animals. Cerebellar abnormalities were observed in mice with disruptions in Foxp2, with Purkinje cells particularly affected. Our findings support a role for Foxp2 in cerebellar development and in a developmental process that subsumes social communication functions in diverse organisms.
- Published
- 2005
- Full Text
- View/download PDF
33. When your patients try to sell you....
- Author
-
Morrisey E
- Subjects
- Humans, Dentist-Patient Relations, Marketing, Practice Management, Dental organization & administration
- Published
- 2002
34. The bone morphogenic protein antagonist gremlin regulates proximal-distal patterning of the lung.
- Author
-
Lu MM, Yang H, Zhang L, Shu W, Blair DG, and Morrisey EE
- Subjects
- Animals, Cytokines, Embryo, Mammalian metabolism, Embryo, Mammalian physiology, Embryonic and Fetal Development, Humans, Mice embryology, Mice, Transgenic genetics, Muscle, Smooth embryology, Proteins genetics, Proteins metabolism, Proteolipids genetics, Pulmonary Surfactants genetics, Pulmonary Surfactants metabolism, Transcription, Genetic physiology, Body Patterning physiology, Bone Morphogenetic Proteins antagonists & inhibitors, Intercellular Signaling Peptides and Proteins, Lung embryology, Proteins physiology
- Abstract
The proximal-distal patterning of lung epithelium involves a complex series of signaling and transcriptional events resulting in the programmed differentiation of highly specialized cells for gas exchange and surfactant protein expression essential for postnatal lung function. The BMP signaling pathway has been shown to regulate cellular differentiation in the lung as well as other tissues. In this report, we show that the can family of related BMP antagonists, including gremlin, cer-1, PRDC, and Dan are expressed in the lung during embryonic development with gremlin expression observed in the proximal airway epithelium. The role of gremlin in lung development was explored by overexpressing it in the distal lung epithelium of transgenic mice using the human SP-C promoter. SP-C/gremlin transgenic mice exhibited a disruption of the proximal-distal patterning found in the airways of the mammalian lung. Expanded expression of the proximal epithelial cell markers CC10 and HFH-4 (Foxj1) was observed in the distal regions of transgenic lungs. Furthermore, smooth muscle alpha-actin expression was observed surrounding the distal airways of SP-C/gremlin mice, indicating a proximalization of distal lung tubules. These data suggest that gremlin plays an important role in lung morphogenesis by regulating the proximal-distal patterning of the lung during development., (Copyright 2001 Wiley-Liss, Inc.)
- Published
- 2001
- Full Text
- View/download PDF
35. Characterization of a new subfamily of winged-helix/forkhead (Fox) genes that are expressed in the lung and act as transcriptional repressors.
- Author
-
Shu W, Yang H, Zhang L, Lu MM, and Morrisey EE
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cloning, Molecular, DNA, Complementary, Epithelium metabolism, Forkhead Transcription Factors, Lung embryology, Mice, Molecular Sequence Data, Repressor Proteins chemistry, Repressor Proteins pharmacology, Sequence Homology, Amino Acid, Gene Expression Regulation, Developmental, Lung metabolism, Repressor Proteins genetics, Repressor Proteins physiology, Transcription Factors
- Abstract
Epithelial gene expression in the lung is thought to be regulated by the coordinate activity of several different families of transcription factors including the Fox family of winged-helix/forkhead DNA-binding proteins. In this report, we have identified and characterized two members of this Fox gene family, Foxp1 and Foxp2, and show that they comprise a new subfamily of Fox genes expressed in the lung. Foxp1 and Foxp2 are expressed at high levels in the lung as early as E12.5 of mouse development with Foxp2 expression restricted to the airway epithelium. In addition, Foxp1 and Foxp2 are expressed at lower levels in neural, intestinal, and cardiovascular tissues during development. Upon differentiation of the airway epithelium along the proximal-distal axis, Foxp2 expression becomes restricted to the distal alveolar epithelium whereas Foxp1 expression is observed in the distal epithelium and mesenchyme. Foxp1 and Foxp2 can regulate epithelial lung gene transcription as was demonstrated by their ability to dramatically repress the mouse CC10 promoter and, to a lesser extent, the human surfactant protein C promoter. In addition, GAL4 fusion proteins encoding subdomains of Foxp1 and Foxp2 demonstrate that an independent and homologous transcriptional repression domain lies within the N-terminal end of the proteins. Together, these studies suggest that Foxp1 and Foxp2 are important regulators of lung epithelial gene transcription.
- Published
- 2001
- Full Text
- View/download PDF
36. GATA-6: the proliferation stops here: cell proliferation in glomerular mesangial and vascular smooth muscle cells.
- Author
-
Morrisey EE
- Subjects
- Cell Cycle, Cell Division drug effects, Cell Division physiology, Cyclin-Dependent Kinase Inhibitor p21, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclins metabolism, Cyclins pharmacology, DNA-Binding Proteins pharmacology, E2F Transcription Factors, Enhancer Elements, Genetic, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacology, GATA6 Transcription Factor, Glomerular Mesangium cytology, Glomerulonephritis metabolism, Glomerulonephritis pathology, Muscle, Smooth, Vascular cytology, Retinoblastoma-Binding Protein 1, Transcription Factors pharmacology, Carrier Proteins, Cell Cycle Proteins, DNA-Binding Proteins metabolism, Glomerular Mesangium metabolism, Muscle, Smooth, Vascular metabolism, Transcription Factors metabolism
- Published
- 2000
- Full Text
- View/download PDF
37. The gene encoding the mitogen-responsive phosphoprotein Dab2 is differentially regulated by GATA-6 and GATA-4 in the visceral endoderm.
- Author
-
Morrisey EE, Musco S, Chen MY, Lu MM, Leiden JM, and Parmacek MS
- Subjects
- 3T3 Cells, Adaptor Proteins, Signal Transducing, Animals, Apoptosis Regulatory Proteins, Base Sequence, Cell Differentiation, DNA, Complementary metabolism, DNA-Binding Proteins physiology, GATA4 Transcription Factor, GATA6 Transcription Factor, Genes, Tumor Suppressor, Humans, In Situ Hybridization, Mice, Molecular Sequence Data, Phosphoproteins genetics, Plasmids, Promoter Regions, Genetic, Protein Structure, Tertiary, Recombinant Fusion Proteins metabolism, Stem Cells cytology, Transcription Factors physiology, Transcription, Genetic, Transcriptional Activation, Tumor Suppressor Proteins, Adaptor Proteins, Vesicular Transport, DNA-Binding Proteins metabolism, Endoderm metabolism, Gene Expression Regulation, Developmental, Phosphoproteins metabolism, Proteins, Transcription Factors metabolism
- Abstract
Gene targeting studies have demonstrated that the zinc finger transcription factor GATA-6 lies upstream in a transcriptional cascade that controls differentiation of the visceral endoderm. To understand the function of GATA-6 in the visceral endoderm and to identify genes regulated by GATA-6 in this tissue, subtractive hybridization was performed using template cDNAs derived from differentiated wild-type embryonic stem (ES) cells and GATA-6(-/-) ES cells, respectively. These analyses revealed that the gene encoding Dab2, a mitogen-responsive phosphoprotein, is differentially expressed in wild-type and GATA-6-deficient ES cells. Consistent with these findings, Dab2 is expressed in the visceral endoderm of wild-type embryos but not in the visceral endoderm of GATA-6-deficient embryos. Cotransfection experiments demonstrate that the human Dab2 promoter can be transactivated by forced expression of GATA-6 in NIH-3T3 cells. In contrast, forced expression of GATA-4 does not transactivate the human Dab2 promoter and Dab2 is expressed in the visceral endoderm of GATA-4 null embryos. Surprisingly, the specificity of GATA-6-induced transactivation of the Dab2 promoter is not mediated through its zinc finger DNA-binding domain. Taken together, these data demonstrate that the mitogen-responsive phosphoprotein Dab2 is a downstream target of GATA-6 in the visceral endoderm. Moreover, these data demonstrate that molecular mechanisms have evolved that direct, and distinguish, the functional specificity of GATA family members when they are developmentally coexpressed.
- Published
- 2000
- Full Text
- View/download PDF
38. GATA-5: a transcriptional activator expressed in a novel temporally and spatially-restricted pattern during embryonic development.
- Author
-
Morrisey EE, Ip HS, Tang Z, Lu MM, and Parmacek MS
- Subjects
- 3T3 Cells, Amino Acid Sequence, Animals, Base Sequence, COS Cells, Cloning, Molecular, DNA, Complementary, Embryonic and Fetal Development, GATA5 Transcription Factor, Gastrula chemistry, Heart embryology, Lung embryology, Mesoderm chemistry, Mice, Molecular Sequence Data, Muscle, Smooth chemistry, Muscle, Smooth cytology, Organ Specificity, RNA, Messenger analysis, Sequence Homology, Amino Acid, DNA-Binding Proteins genetics, Gene Expression Regulation, Developmental physiology, Trans-Activators genetics, Transcription Factors genetics
- Abstract
Members of the GATA family of zinc finger transcription factors regulate critical steps of cellular differentiation during vertebrate development. In the studies described in this report, we have isolated and functionally characterized the murine GATA-5 cDNA and protein and defined the temporal and spatial pattern of GATA-5 gene expression during mammalian development. The amino terminus of the mouse GATA-5 protein shares high level amino acid sequence identity with the murine GATA-4 and -6 proteins, but not with other members of the GATA family. GATA-5 binds to the functionally important CEF-1 nuclear protein binding site in the cardiac-specific slow/cardiac troponin C (cTnC) transcriptional enhancer and overexpression of GATA-5 transactivates the cTnC enhancer in noncardiac muscle cell lines. During embryonic and postnatal development, the pattern of GATA-5 gene expression differs significantly from that of other GATA family members. In the primitive streak embryo, GATA-5 mRNA is detectable in the precardiac mesoderm. Within the embryonic heart, the GATA-5 gene is expressed within the atrial and ventricular chambers (ED 9.5), becomes restricted to the atrial endocardium (ED 12.5), and is subsequently not expressed in the heart during late fetal and postnatal development. Moreover, coincident with the earliest steps in lung development, only the GATA-5 gene is expressed within the pulmonary mesenchyme. Finally, the GATA-5 gene is expressed in tissue-restricted subsets of smooth muscle cells (SMCs), including bronchial SMCs and SMCs in the bladder wall. These data are consistent with a model in which GATA-5 performs a unique temporally and spatially restricted function in the embryonic heart and lung. Moreover, these data suggest that GATA-5 may play an important role in the transcriptional program(s) that underlies smooth muscle cell diversity.
- Published
- 1997
- Full Text
- View/download PDF
39. GATA-6: a zinc finger transcription factor that is expressed in multiple cell lineages derived from lateral mesoderm.
- Author
-
Morrisey EE, Ip HS, Lu MM, and Parmacek MS
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Binding Sites, Cardiovascular System embryology, Cell Lineage genetics, Clone Cells, DNA, Complementary isolation & purification, Digestive System embryology, GATA6 Transcription Factor, Lung embryology, Mice, Molecular Sequence Data, Muscle, Skeletal embryology, Muscle, Smooth, Vascular embryology, RNA analysis, Rats, Troponin genetics, Troponin C, Urogenital System embryology, DNA-Binding Proteins genetics, Gene Expression Regulation, Developmental, Mesoderm physiology, Transcription Factors genetics, Zinc Fingers genetics
- Abstract
Members of the GATA family of zinc finger transcription factors play important roles in the development of several mesodermally derived cell lineages. In the studies described in this report, we have isolated and functionally characterized the murine GATA-6 cDNA and protein and defined the temporal and spatial patterns of GATA-6 gene expression during mammalian development. The GATA-6 and -4 proteins share high-level amino acid sequence identity over a proline-rich region at the amino terminus of the protein that is not conserved in other GATA family members. GATA-6 binds to a functionally important nuclear protein binding site within the cardiac-specific cardiac troponin C (cTnC) transcriptional enhancer. Moreover, the cTnC promoter enhancer can be transactivated by overexpression of GATA-6 in noncardiac muscle cells. During early murine embryonic development, the patterns of GATA-6 and -4 gene expression are similar, with expression of GATA-6 restricted to the precardiac mesoderm, the embryonic heart tube, and the primitive gut. However, coincident with the onset of vasculogenesis and development of the respiratory and urogenital tracts, only the GATA-6 gene is expressed in arterial smooth muscle cells, the developing bronchi, and the urogenital ridge and bladder. These data are consistent with a model in which GATA-6 functions in concert with GATA-4 to direct tissue-specific gene expression during formation of the mammalian heart and gastrointestinal tract, but performs a unique function in programming lineage-restricted gene expression in the arterial system, the bladder, and the embryonic lung.
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.