1
|
Stefanovic S, Etchevers HC, Zaffran S. Outflow Tract Formation-Embryonic Origins of Conotruncal Congenital Heart Disease. J Cardiovasc Dev Dis 2021; 8:jcdd8040042. [PMID: 33918884 PMCID: PMC8069607 DOI: 10.3390/jcdd8040042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 12/13/2022] Open
Abstract
Anomalies in the cardiac outflow tract (OFT) are among the most frequent congenital heart defects (CHDs). During embryogenesis, the cardiac OFT is a dynamic structure at the arterial pole of the heart. Heart tube elongation occurs by addition of cells from pharyngeal, splanchnic mesoderm to both ends. These progenitor cells, termed the second heart field (SHF), were first identified twenty years ago as essential to the growth of the forming heart tube and major contributors to the OFT. Perturbation of SHF development results in common forms of CHDs, including anomalies of the great arteries. OFT development also depends on paracrine interactions between multiple cell types, including myocardial, endocardial and neural crest lineages. In this publication, dedicated to Professor Andriana Gittenberger-De Groot and her contributions to the field of cardiac development and CHDs, we review some of her pioneering studies of OFT development with particular interest in the diverse origins of the many cell types that contribute to the OFT. We also discuss the clinical implications of selected key findings for our understanding of the etiology of CHDs and particularly OFT malformations.
Collapse
|
2
|
Dinsmore CJ, Soriano P. MAPK and PI3K signaling: At the crossroads of neural crest development. Dev Biol 2018; 444 Suppl 1:S79-S97. [PMID: 29453943 PMCID: PMC6092260 DOI: 10.1016/j.ydbio.2018.02.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 02/08/2023]
Abstract
Receptor tyrosine kinase-mediated growth factor signaling is essential for proper formation and development of the neural crest. The many ligands and receptors implicated in these processes signal through relatively few downstream pathways, frequently converging on the MAPK and PI3K pathways. Despite decades of study, there is still considerable uncertainty about where and when these signaling pathways are required and how they elicit particular responses. This review summarizes our current understanding of growth factor-induced MAPK and PI3K signaling in the neural crest.
Collapse
Affiliation(s)
- Colin J Dinsmore
- Department of Cell, Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, USA.
| |
Collapse
|
3
|
Keller KC, Ding H, Tieu R, Sparks NRL, Ehnes DD, Zur Nieden NI. Wnt5a Supports Osteogenic Lineage Decisions in Embryonic Stem Cells. Stem Cells Dev 2016; 25:1020-32. [PMID: 26956615 DOI: 10.1089/scd.2015.0367] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The specification of pluripotent stem cells into the bone-forming osteoblasts has been explored in a number of studies. However, the current body of literature has yet to adequately address the role of Wnt glycoproteins in the differentiation of pluripotent stem cells along the osteogenic lineage. During mouse embryonic stem cell (ESC) in vitro osteogenesis, the noncanonical WNT5a is expressed early on. Cells either sorted by their positive WNT5a expression or when supplemented with recombinant WNT5a (rWNT5a) during a 2-day window showed significantly enhanced osteogenic yield. Mechanistically, rWNT5a supplementation upregulated protein kinase C (PKC), calcium/calmodulin-dependent kinase II (CamKII) and c-Jun N-terminal kinase (JNK) activity while antagonizing the key effector of canonical Wnt signaling: β-catenin. Conversely, when recombinant WNT3a (rWNT3a) or other positive regulators of β-catenin were employed during this same time window there was a decrease in osteogenic marker expression. However, if rWNT3a was supplemented during a time window following rWNT5a treatment, osteogenic differentiation was enhanced both in murine and human ESCs. Elucidating the role of these WNT ligands in directing the early stages of osteogenesis has the potential to considerably improve tissue engineering protocols and applications for regenerative medicine.
Collapse
Affiliation(s)
- Kevin C Keller
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California
| | - Huawen Ding
- 2 Applied Stem Cell Technologies Unit, Department for Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology , Leipzig, Germany
| | - Rudy Tieu
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California
| | - Nicole R L Sparks
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California
| | - Devon D Ehnes
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California
| | - Nicole I Zur Nieden
- 1 Department of Cell Biology & Neuroscience, Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside , Riverside, California.,2 Applied Stem Cell Technologies Unit, Department for Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology , Leipzig, Germany
| |
Collapse
|
4
|
Methyl viologen induces neural differentiation on murine P19 cells. In Vitro Cell Dev Biol Anim 2016; 52:466-72. [PMID: 26831493 DOI: 10.1007/s11626-016-0001-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 01/05/2016] [Indexed: 10/22/2022]
Abstract
Methyl viologen is a highly effective contact herbicide, a neurotoxic compound and an inducer of reactive oxygen species, which generate oxidative stress in cells. Reactive oxygen species has been known to function as an important messenger in cell differentiation. In this study, we investigated the effect of methyl viologen on neural cell differentiation using pluripotent mouse embryonal carcinoma P19 cells, which reportedly differentiate into neural cells upon exposure to retinoic acid. Methyl viologen, an inducer of intracellular reactive oxygen species, induced the differentiation of P19 cells into neural cells in the presence of neurofilament. Reduced glutathione, an eliminator of reactive oxygen species, also induced neural differentiation in P19 cells. These results suggest that P19 cells differentiate into neural cells, conceivably independent of intracellular reactive oxygen species.
Collapse
|
5
|
Kovalevich J, Yen W, Ozdemir A, Langford D. Cocaine induces nuclear export and degradation of neuronal retinoid X receptor-γ via a TNF-α/JNK- mediated mechanism. J Neuroimmune Pharmacol 2015; 10:55-73. [PMID: 25586717 PMCID: PMC4336643 DOI: 10.1007/s11481-014-9573-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 11/26/2014] [Indexed: 12/14/2022]
Abstract
Cocaine abuse represents an immense societal health and economic burden for which no effective treatment currently exists. Among the numerous intracellular signaling cascades impacted by exposure to cocaine, increased and aberrant production of pro-inflammatory cytokines in the CNS has been observed. Additionally, we have previously reported a decrease in retinoid-X-receptor-gamma (RXR-γ) in brains of mice chronically exposed to cocaine. Through obligate heterodimerization with a number of nuclear receptors, RXRs serve as master regulatory transcription factors, which can potentiate or suppress expression of a wide spectrum of genes. Little is known about the regulation of RXR levels, but previous studies indicate cellular stressors such as cytokines negatively regulate levels of RXRs in vitro. To evaluate the mechanism underlying the cocaine-induced decreases in RXR-γ levels observed in vivo, we exposed neurons to cocaine in vitro and examined pathways which may contribute to disruption in RXR signaling, including activation of stress pathways by cytokine induction. In these studies, we provide the first evidence that cocaine exposure disrupts neuronal RXR-γ signaling in vitro by promoting its nuclear export and degradation. Furthermore, we demonstrate this effect may be mediated, at least in part, by cocaine-induced production of TNF-α and its downstream effector c-Jun-NH-terminal kinase (JNK). Findings from this study are therefore applicable to both cocaine abuse and to pathological conditions characterized by neuroinflammatory factors, such as neurodegenerative disease.
Collapse
Affiliation(s)
- Jane Kovalevich
- Department of Neuroscience, Temple University School of Medicine, Medical Education Research Building, 3500 North Broad Street, Philadelphia, PA, 19140, USA
| | | | | | | |
Collapse
|
6
|
Yang H, Keen CL, Lanoue L. Influence of intracellular zinc on cultures of rat cardiac neural crest cells. ACTA ACUST UNITED AC 2015; 104:11-22. [PMID: 25689142 DOI: 10.1002/bdrb.21135] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 01/08/2015] [Indexed: 11/06/2022]
Abstract
BACKGROUND Developmental zinc (Zn) deficiency increases the incidence of heart anomalies in rat fetuses, in regions and structures derived from the outflow tract. Given that the development of the outflow tract requires the presence of cardiac neural crest cells (cNCC), we speculated that Zn deficiency selectively kills cNCC and could lead to heart malformations. METHODS Cardiac NCC were isolated from E10.5 rat embryos and cultured in control media (CTRL), media containing 3 μM of the cell permeable metal chelator N, N, N', N'-tetrakis (2-pyridylmethyl) ethylene diamine (TPEN), or in TPEN-treated media supplemented with 3 μM Zn (TPEN + Zn). Cardiac NCC were collected after 6, 8, and 24 h of treatment to assess cell viability, proliferation, and apoptosis. RESULTS The addition of TPEN to the culture media reduced free intracellular Zn pools and cell viability as assessed by low ATP production, compared to cells grown in control or Zn-supplemented media. There was an accumulation of reactive oxygen species, a release of mitochondrial cytochrome c into the cytoplasm, and an increased cellular expression of active caspase-3 in TPEN-treated cNCC compared to cNCC cultured in CTRL or TPEN + Zn media. CONCLUSION Zn deficiency can result in oxidative stress in cNCC, and subsequent decreases in their population and metabolic activity. These data support the concept that Zn deficiency associated developmental heart defects may arise in part as a consequence of altered cNCC metabolism.
Collapse
Affiliation(s)
- Hsunhui Yang
- Department of Nutrition, University of California, Davis, California
| | | | | |
Collapse
|
7
|
|
8
|
Rappolee DA, Awonuga AO, Puscheck EE, Zhou S, Xie Y. Benzopyrene and experimental stressors cause compensatory differentiation in placental trophoblast stem cells. Syst Biol Reprod Med 2010; 56:168-83. [PMID: 20377314 DOI: 10.3109/19396360903431638] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Stress causes decreased cell accumulation in early periimplantation embryos and the placental trophoblast stem cells derived from them. Benzopyrene and many other stressors activate stress enzymes that lead to suppressed stem cell accumulation through diminished proliferation and increased apoptosis. Trophoblast stem cells proliferate and a subpopulation of early postimplantation trophoblast cells differentiate to produce the first placental hormones that arise in the implanting conceptus. These hormones mediate antiluteolytic effects that enable the continuation of a successful implantation. The normal determination and differentiation of placental trophoblast stem cells is dependent upon a series of transcription factors. But, these transcription factors can also be modulated by stress through the activity of stress enzymes. This review enumerates and analyzes recent reports on the effects of benzopyrene on placental function in terms of the emerging paradigm that placental differentiation from stem cells can be regulated when insufficient production of stem cells is caused by stress. In addition, we review the other effects caused by benzopyrene throughout placental development.
Collapse
Affiliation(s)
- Daniel A Rappolee
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | | | | | | | | |
Collapse
|
9
|
Zile MH. Vitamin A-not for your eyes only: requirement for heart formation begins early in embryogenesis. Nutrients 2010; 2:532-50. [PMID: 22254040 PMCID: PMC3257662 DOI: 10.3390/nu2050532] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/07/2010] [Accepted: 05/18/2010] [Indexed: 12/25/2022] Open
Abstract
Vitamin A insufficiency has profound adverse effects on embryonic development. Major advances in understanding the role of vitamin A in vertebrate heart formation have been made since the discovery that the vitamin A active form, all-trans-retinoic acid, regulates many genes, including developmental genes. Among the experimental models used, the vitamin A-deficient avian embryo has been an important tool to study the function of vitamin A during early heart formation. A cluster of retinoic acid-regulated developmental genes have been identified that participate in building the heart. In the absence of retinoic acid the embryonic heart develops abnormally leading to embryolethality.
Collapse
Affiliation(s)
- Maija H Zile
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
10
|
|
11
|
Sanbe A, Mizutani R, Miyauchi N, Yamauchi J, Nagase T, Yamamura KI, Tanoue A. Inhibitory effects of cigarette smoke extract on neural crest migration occur through suppression of R-spondin1 expression via aryl hydrocarbon receptor. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2009; 380:569-76. [PMID: 19768455 DOI: 10.1007/s00210-009-0455-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Accepted: 09/05/2009] [Indexed: 12/21/2022]
Abstract
Although it is known that smoking during pregnancy induces fetal malformations, few basic studies at the molecular level are currently available. Since it is known that neural crest cells (NCC) play an important role in tissue development and differentiation, we investigated the influence of cigarette smoke extract (CSE) on NCC migration. CSE treatment reduced the migration index of NCC in dose- and tar-content-dependent manners without induction of apoptosis or decrease in proliferation of NCC. alpha-Naphthoflavone, an antagonist of aryl hydrocarbon receptor (AhR), prevented the reduction in NCC migration that was otherwise induced by CSE treatment. Overexpression of AhR caused a significant decrease in NCC migration index, implying that CSE can attenuate NCC migration through AhR signaling. Transcriptome analysis revealed that overexpression of AhR led to decreased expression of R-spondin1 in NCC. Furthermore, overexpression of R-spondin1 prevented the inhibitory effect of CSE on NCC. These results suggest that CSE causes suppressed expression of R-spondin1 by activating signals via the AhR, which leads to impaired neural crest cell migration.
Collapse
Affiliation(s)
- Atsushi Sanbe
- Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
12
|
Genetic approaches for changing the heart and dissecting complex syndromes. J Mol Cell Cardiol 2008; 45:148-55. [PMID: 18601931 DOI: 10.1016/j.yjmcc.2008.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 05/21/2008] [Accepted: 06/03/2008] [Indexed: 01/18/2023]
Abstract
The genetic, biochemical and molecular bases of human cardiac disease have been the focus of extensive research efforts for many years. Early animal models of cardiovascular disease used pharmacologic or surgical interventions, or took advantage of naturally occurring genetic abnormalities and the data obtained were largely correlative. The inability to directly alter an organism's genetic makeup and cellular protein content and accurately measure the results of that manipulation precluded rigorous examination of true cause-effect and structure-function relationships. Directed genetic manipulation in the mouse gave researchers the ability to modify and control the mammalian heart's protein content, resulting in the rational design of models that could provide critical links between the mutated or absent protein and disease. Two techniques that have proven particularly useful are transgenesis, which involves the random insertion of ectopic genetic material of interest into a "host" genome, and gene targeting, which utilizes homologous recombination at a pre-selected locus. Initially, transgenesis and gene targeting were used to examine systemic loss-of-function and gain-of-function, respectively, but further refinements in both techniques have allowed for investigations of organ-specific, cell type-specific, developmental stage-sensitive and dose-dependent effects. Genetically engineered animal models of pediatric and adult cardiac disease have proven that, when used appropriately, these tools have the power to extend mere observation to the establishment of true causative proof. We illustrate the power of the general approach by showing how genetically engineered mouse models can define the precise signaling pathways that are affected by the gain-of-function mutation that underlies Noonan syndrome. Increasingly precise and modifiable animal models of human cardiac disease will allow researchers to determine not only pathogenesis, but also guide treatment and the development of novel therapies.
Collapse
|
13
|
Lopez V, Keen CL, Lanoue L. Prenatal zinc deficiency: influence on heart morphology and distribution of key heart proteins in a rat model. Biol Trace Elem Res 2008; 122:238-55. [PMID: 18224284 DOI: 10.1007/s12011-007-8079-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Revised: 10/25/2007] [Accepted: 11/23/2007] [Indexed: 12/22/2022]
Abstract
The etiology of congenital heart disease is multifactorial, with genetics and nutritional deficiencies recognized as causative agents. Maternal zinc (Zn) deficiency is associated with an increased risk for fetal heart malformations; however, the contributing mechanisms have yet to be identified. In this study, we fed pregnant rats a Zn-adequate diet (ZnA), a Zn-deficient (ZnD), or a restricted amount of Zn adequate diet (RF) beginning on gestation day (GD) 4.5, to examine whether increased cell death and changes in cardiac neural crest cells (NCC) play a role in Zn deficiency-induced heart defects. Fetuses were collected on GD 13.5, 15.5, and 18.5 and processed for GATA-4, FOG-2, connexin-43 (Cx43), HNK-1, smooth muscle alpha-actin (SMA) and cleaved caspase-3 protein expression. Fetuses from ZnA-fed dams showed normal heart development, whereas fetuses from dams fed with the ZnD diet exhibited a variety of heart anomalies, particularly in the region of the outflow tract. HNK-1 expression was lower than normal in the hearts of GD13.5 and 15.5 ZnD fetuses, particularly in the right atrium and in the distal tip of the interventricular septum. Conversely, Cx43 immunoreactivity was increased throughout the heart in fetuses from ZnD dams compared to fetuses from control dams. The distribution and intensity of expression of SMA, GATA-4, FOG-2, and markers of apoptosis were similar among the three groups. We propose that Zn deficiency induced alterations in the distribution of Cx43 and HNK-1 in fetal hearts contribute to the occurrence of the developmental heart anomalies.
Collapse
Affiliation(s)
- Veronica Lopez
- Department of Nutrition, University of California, Davis, One Shields Ave., Meyer Hall, Davis, CA 95616, USA
| | | | | |
Collapse
|
14
|
Hoover LL, Burton EG, Brooks BA, Kubalak SW. The expanding role for retinoid signaling in heart development. ScientificWorldJournal 2008; 8:194-211. [PMID: 18661045 PMCID: PMC2559957 DOI: 10.1100/tsw.2008.39] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The importance of retinoid signaling during cardiac development has long been appreciated, but recently has become a rapidly expanding field of research. Experiments performed over 50 years ago showed that too much or too little maternal intake of vitamin A proved detrimental for embryos, resulting in a cadre of predictable cardiac developmental defects. Germline and conditional knockout mice have revealed which molecular players in the vitamin A signaling cascade are potentially responsible for regulating specific developmental events, and many of these molecules have been temporally and spatially characterized. It is evident that intact and controlled retinoid signaling is necessary for each stage of cardiac development to proceed normally, including cardiac lineage determination, heart tube formation, looping, epicardium formation, ventricular maturation, chamber and outflow tract septation, and coronary arteriogenesis. This review summarizes many of the significant milestones in this field and particular attention is given to recently uncovered cross-talk between retinoid signaling and other developmentally significant pathways. It is our hope that this review of the role of retinoid signaling during formation, remodeling, and maturation of the developing heart will serve as a tool for future discoveries.
Collapse
Affiliation(s)
- Loretta L Hoover
- Department of Cell Biology and Anatomy, Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, SC, USA.
| | | | | | | |
Collapse
|
15
|
Han J, Li L, Zhang Z, Xiao Y, Lin J, Zheng L, Li Y. Platelet-derived growth factor C plays a role in the branchial arch malformations induced by retinoic acid. ACTA ACUST UNITED AC 2007; 79:221-30. [PMID: 17183585 DOI: 10.1002/bdra.20329] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND All-trans-retinoic acid (RA) can produce branchial arch abnormalities in postimplantation rodent embryos cultured in vitro. Platelet-derived growth factor C (PDGF-C) was recently identified as a member of the PDGF ligand family. Many members of the PDGF family are essential for branchial arch morphogenesis and can be regulated by RA. The roles of PDGF-C in branchial arch malformations induced by RA and possible mechanisms were investigated. METHODS In whole embryo culture (WEC), mouse embryos were exposed to RA at 0, 0.1, 0.4, 1.0, or 10.0 microM, PDGF-C at 25, 50, or 75 ng/mL, or PDGF-C at 25, 50, or 75 ng/mL containing 0.4 microM RA. After 48 h of culture, mouse embryos were examined for dysmorphogenesis, and whole-mount immunohistochemistry was applied to PDGF-C. In explant cultures, explants were exposed to the same doses of RA and PDGF-C as WEC. Semiquantitative RT-PCR, zymography, and reverse zymography were used to evaluate the expressions and activities of matrix metalloproteinase (MMP)-2, MMP-14, and tissue inhibitor of metalloproteinase (TIMP)-2. RESULTS PDGF-C was reduced by RA, and exogenous PDGF-C rescued the branchial arch malformations induced by RA. Moreover, PDGF-C prevented RA-induced inhibition of the migratory ability of mesenchymal cells in the first branchial arch, by regulating the expressions of MMP-2, MMP-14, and TIPM-2. CONCLUSIONS Our results suggest that RA exposure reduces the expression of PDGF-C. The branchial arch malformations resulting from fetal RA exposure are caused at least partially by loss of PDGF-C and subsequent misregulations of the expressions of MMP-2, MMP-14, and TIMP-2.
Collapse
Affiliation(s)
- Jing Han
- Department of Food Science and Nutrition, School of Public Health, Peking University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
16
|
Cipollone D, Amati F, Carsetti R, Placidi S, Biancolella M, D'Amati G, Novelli G, Siracusa G, Marino B. A multiple retinoic acid antagonist induces conotruncal anomalies, including transposition of the great arteries, in mice. Cardiovasc Pathol 2007; 15:194-202. [PMID: 16844550 DOI: 10.1016/j.carpath.2006.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2005] [Revised: 02/08/2006] [Accepted: 04/10/2006] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The morphogenetic mechanisms that are responsible for the transposition of the great arteries are still largely unknown, mainly because this malformation is very difficult to experimentally reproduce. The aim of the present study was to test the effect of BMS-189453, a retinoic acid antagonist, on murine heart morphogenesis. METHODS We administered this drug at 5 mg/kg body weight (twice, at a 12-h interval) to pregnant mice on 6.25/6.75 days postcoitum (dpc) (Group A), 6.75/7.25 dpc (Group B), 7.25/7.75 dpc (Group C), 7.75/8.25 dpc (Group D), or 8.25/8.75 dpc (Group E). At birth, the anatomical features of fetuses were evaluated by stereomicroscopic examination. RESULTS In Group A (18 fetuses), cardiovascular anatomy was normal in 10 (56%) cases, and 8 (44%) fetuses presented with transposition of the great arteries. In Group B, no fetuses were obtained. In Group C (78 fetuses), cardiovascular anatomy was normal in 19 (24%) cases, while 59 (76%) mice presented with various types of cardiac defects, including 48 transpositions of the great arteries (61%). In Group D (80 fetuses), cardiac defects were seen in 22 (27%) mice: 14 of these (17%) were transpositions of the great arteries. In Group E (72 fetuses), cardiovascular anatomy was normal in all cases. Of 248 fetuses analyzed, 87% presented with thymic aplasia or hypoplasia, and 20% presented with meroanencephalia and/or rachischisis. CONCLUSIONS Transposition of the great arteries can be consistently reproduced in mice by administration of a retinoic acid competitive antagonist on 7.5 dpc.
Collapse
Affiliation(s)
- Daria Cipollone
- Department of Public Health and Cell Biology, University of Rome Tor Vergata, 00173 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Han J, Li L, Zhang Z, Xiao Y, Lin J, Li Y. PDGF-C participates in branchial arch morphogenesis and is down-regulated by retinoic acid. Toxicol Lett 2006; 166:248-54. [PMID: 16956736 DOI: 10.1016/j.toxlet.2006.07.308] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Revised: 07/18/2006] [Accepted: 07/20/2006] [Indexed: 12/26/2022]
Abstract
Retinoic acid (RA) is a teratogen that induces a variety of craniofacial abnormalities, including branchial arch deformities and cleft palate. Platelet-derived growth factor C (PDGF-C) is a recently identified member of the PDGF family. PDGF-C contributes to normal development of the heart, central nervous system, kidney and palatogenesis. But the roles of PDGF-C in branchial arches development and the relationship between PDGF-C and RA-induced branchial arches abnormalities are poorly understood. We examined the effects of RA on PDGF-C and its receptor PDGFR-alpha expressions. We demonstrated that administration of RA to mouse embryos resulted in dramatic losses of PDGF-C and its receptor PDGFR-alpha. Furthermore, we confirmed that blocking PDGF-C signaling by anti-PDGF-C neutralization antibody led to branchial arch malformations similar to that of RA induced, both hypoplastic branchial arches and FBA. These findings suggest the down-regulation of PDGF-C may be one of mechanisms of branchial arch abnormalities induced by RA and PDGF-C signaling is required for branchial arch morphogenesis.
Collapse
Affiliation(s)
- Jing Han
- Department of Food Science and Nutrition, School of Public Health, Peking University, Beijing, China
| | | | | | | | | | | |
Collapse
|
18
|
Nakamura T, Colbert MC, Robbins J. Neural crest cells retain multipotential characteristics in the developing valves and label the cardiac conduction system. Circ Res 2006; 98:1547-54. [PMID: 16709902 DOI: 10.1161/01.res.0000227505.19472.69] [Citation(s) in RCA: 262] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multipotent neural crest cells (NCCs) are a major extracardiac component of cardiovascular development. Although recognized as contributing cells to the arterial valves at early developmental stages, NCC persistence in the valves at later times or in the adult heart is controversial. We analyzed NCC persistence and contributions to both semilunar and atrioventricular (AV) valves in the mature heart. Two NCC-specific promoters driving Cre recombinase, Wnt1-Cre and P0-Cre, were mated with floxed reporter mice, R26R or CAG-CAT-EGFP, to map NCC fate. Hearts were analyzed before aorticopulmonary (AP) septation through adult stages. As previously demonstrated, strong NCC labeling was detected in ventral and dorsal outflow cushions before AP septation. In contrast to previous reports, we found that substantial numbers of labeled cells persisted in the semilunar valves in late fetal, neonatal, and adult hearts. Furthermore, NCCs were also found in the AV valves, almost exclusively in the septal leaflets. NCCs in the AV valves expressed melanocytic and neurogenic markers. However, cells labeled in the proximal cardiac conduction system exhibited neurogenic and gliagenic markers, whereas some NCCs expressed no differentiation specific markers. These results suggest that cardiac NCCs contribute to the mature valves and the cardiac conduction system and retain multipotent characteristics late in development.
Collapse
Affiliation(s)
- Tomoki Nakamura
- Department of Pediatrics, Division of Molecular Cardiovascular Biology, Children's Hospital Research Foundation, Cincinnati, Ohio 45229-3039, USA
| | | | | |
Collapse
|
19
|
Lee GS, Liao X, Cantor RM, Collins MD. Interactive effects of cadmium and all-trans-retinoic acid on the induction of forelimb ectrodactyly in C57BL/6 mice. ACTA ACUST UNITED AC 2006; 76:19-28. [PMID: 16369952 DOI: 10.1002/bdra.20201] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Most toxicological studies have tested single chemical agents at relatively high doses, and fewer studies have addressed the toxic effects of chemical interactions. It is important to understand the toxicity of chemical mixtures in order to assess the more realistic risks of environmental and occupational exposures. A number of chemicals are known to induce a predominantly postaxial forelimb ectrodactyly in C57BL/6 mice, including acetazolamide, ethanol, cadmium, valproic acid, carbon dioxide, dimethadione, phenytoin, and 13-cis-retinoic acid and all-trans-retinoic acid (RA). In the present study, the interactive effects of coadministration of cadmium and RA on developing limbs were investigated. METHODS Pregnant C57BL/6 mice were treated with different intraperitoneal (IP) doses of cadmium chloride (CdCl2) and/or RA on gestational day (GD) 9.5, and fetuses were collected on GD 18 and double stained for examination of skeletal defects. RESULTS When RA was given simultaneously with cadmium, a significant increase in the incidence and severity of forelimb ectrodactyly (predominantly postaxial) was observed compared to the results with corresponding doses of cadmium or RA alone. When mice were exposed to subthreshold doses of both cadmium (0.5 mg/kg) and RA (1 mg/kg), the combined treatment exceeded the threshold, resulting in forelimb ectrodactyly in 19% of the fetuses. Moreover, coadministration of cadmium and RA at doses exceeding the respective thresholds showed a synergistic effect, that is, 92% of fetuses were found with the forelimb defect as opposed to 10% if the response were additive. CONCLUSIONS The findings demonstrate that concurrent exposure to these teratogens can have a synergistic effect and that subteratogenic doses may combine to exceed a threshold.
Collapse
Affiliation(s)
- Grace S Lee
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles School of Public Health, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
20
|
Abstract
The neural crest is a transient, migratory cell population found in all vertebrate embryos that generate a diverse range of cell and tissue derivatives including, but not limited, to the neurons and glia of the peripheral nervous system, smooth muscle, connective tissue, melanocytes, craniofacial cartilage, and bone. Over the past few years, many studies have provided tremendous insights into understanding the mechanisms regulating the induction and migration of neural crest cell development. This review highlights the surprising and perhaps unexpected roles for morphogens in these distinct processes. A comparison of studies performed in several different vertebrates emphasizes the requirement for coordination between multiple signaling pathways in the induction and migration of neural crest cells in the developing embryo.
Collapse
Affiliation(s)
- Natalie C Jones
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, Missouri 64110, USA
| | | |
Collapse
|
21
|
Mizuno N, Kokubu H, Sato M, Nishimura A, Yamauchi J, Kurose H, Itoh H. G protein-coupled receptor signaling through Gq and JNK negatively regulates neural progenitor cell migration. Proc Natl Acad Sci U S A 2005; 102:12365-70. [PMID: 16116085 PMCID: PMC1194958 DOI: 10.1073/pnas.0506101102] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
In the early development of the central nervous system, neural progenitor cells divide in an asymmetric manner and migrate along the radial glia cells. The radial migration is an important process for the proper lamination of the cerebral cortex. Recently, a new mode of the radial migration was found at the intermediate zone where the neural progenitor cells become multipolar and reduce the migration rate. However, the regulatory signals for the radial migration are unknown. Using the migration assay in vitro, we examined how neural progenitor cell migration is regulated. Neural progenitor cells derived from embryonic mouse telencephalon migrated on laminin-coated dishes. Endothelin (ET)-1 inhibited the neural progenitor cell migration. This ET-1 effect was blocked by BQ788, a specific inhibitor of the ETB receptor, and by the expression of a carboxyl-terminal peptide of Galpha q but not Galpha i. The expression of constitutively active mutant of Galpha q, Galpha qR183C, inhibited the migration of neural progenitor cells. Moreover, the inhibitory effect of ET-1 was suppressed by the c-Jun N-terminal kinase (JNK) inhibitor SP600125 and the expression of the JNK-binding domain of JNK-interacting protein-1, a specific inhibitor of the JNK pathway. Using the slice culture system of embryonic brain, we demonstrated that ET-1 and the constitutively active mutant of Galpha q caused the retention of the neural progenitor cells in the intermediate zone and JNK-binding domain of JNK-interacting protein-1 abrogated the effect of ET-1. These results indicated that G protein-coupled receptor signaling negatively regulates neural progenitor cell migration through Gq and JNK.
Collapse
Affiliation(s)
- Norikazu Mizuno
- Department of Cell Biology, Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | | | | | | | | | | | | |
Collapse
|
22
|
Williams SS, Mear JP, Liang HC, Potter SS, Aronow BJ, Colbert MC. Large-scale reprogramming of cranial neural crest gene expression by retinoic acid exposure. Physiol Genomics 2005; 19:184-97. [PMID: 15466718 DOI: 10.1152/physiolgenomics.00136.2004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Although retinoic acid (RA), the active form of vitamin A, is required for normal embryonic growth and development, it is also a powerful teratogen. Infants born to mothers exposed to retinoids during pregnancy have a 25-fold increased risk for malformations, nearly exclusively of cranial neural crest-derived tissues. To characterize neural crest cell responses to RA, we exposed murine crest cultures to teratogenic levels of RA and subjected their RNA to microarray-based gene expression profile analysis using Affymetrix MG-U74Av2 GeneChips. RNAs were isolated from independent cultures treated with 10(-6) M RA for 6, 12, 24, or 48 h. Statistical analyses of gene expression profile data facilitated identification of the 205 top-ranked differentially regulated genes whose expression was reproducibly changed by RA over time. Cluster analyses of these genes across the independently treated sample series revealed distinctive kinetic patterns of altered gene expression. The largest group was transiently affected within the first 6 h of exposure, representing early responding genes. Group 2 showed sustained induction by RA over all times, whereas group 3 was characterized by the suppression of a time-dependent expression increase normally seen in untreated cells. Additional patterns demonstrated time-dependent increased or decreased expression among genes not normally regulated to a significant extent. Gene function analysis revealed that more than one-third of all RA-regulated genes were associated with developmental regulation, including both canonical and noncanonical Wnt signaling pathways. Multiple genes associated with cell adhesion and cell cycle regulation, recognized targets for the biological effects of RA, were also affected. Taken together, these results support the hypothesis that the teratogenic effects of RA derive from reprogramming gene expression of a host of genes, which play critical roles during embryonic development regulating pathways that determine subsequent differentiation of cranial neural crest cells.
Collapse
Affiliation(s)
- Sarah S Williams
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | |
Collapse
|
23
|
Wang L, Mear JP, Kuan CY, Colbert MC. Retinoic acid induces CDK inhibitors and growth arrest specific (Gas) genes in neural crest cells. Dev Growth Differ 2005; 47:119-30. [PMID: 15839997 DOI: 10.1111/j.1440-169x.2005.00788.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Retinoic acid (RA), the active metabolite of vitamin A, regulates cellular growth and differentiation during embryonic development. In excess, this vitamin is also highly teratogenic to animals and humans. The neural crest is particularly sensitive to RA, and high levels adversely affect migration, proliferation and cell death. We investigated potential gene targets of RA associated with neural crest proliferation by determining RA-mediated changes in gene expression over time, using microarrays. Statistical analysis of the top ranked RA-regulated genes identified modest changes in multiple genes previously associated with cell cycle control and proliferation including the cyclin-dependent kinase inhibitors Cdkn1a (p21), Cdkn2b (p15(INK4b)), and Gas3/PMP22. The expression of p21 and p15(INK4b) contribute to decreased proliferation by blocking cell cycle progression at G1-S. This checkpoint is pivotal to decisions regulating proliferation, apoptosis, or differentiation. We have also confirmed the overexpression of Gas3/PMP22 in RA-treated neural crests, which is associated with cytoskeletal changes and increased apoptosis. Our results suggest that increases in multiple components of diverse regulatory pathways have an overall cumulative effect on cellular decisions. This heterogeneity contributes to the pleiotropic effects of RA, specifically those affecting proliferation and cell death.
Collapse
Affiliation(s)
- Linping Wang
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|
24
|
Abstract
Of all the model organisms used to study human development, rodents such as mice most accurately reflect human craniofacial development. Collective advances in mouse embryology and mouse genetics continue to shape our understanding of neural crest cell development and by extrapolation the etiology of human congenital head and facial birth defects. The aim of this review is to highlight the considerable progress being made in our understanding of cranial neural crest cell patterning in mouse embryos.
Collapse
Affiliation(s)
- Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.
| |
Collapse
|
25
|
Mehrotra M, Krane SM, Walters K, Pilbeam C. Differential regulation of platelet-derived growth factor stimulated migration and proliferation in osteoblastic cells. J Cell Biochem 2005; 93:741-52. [PMID: 15660418 DOI: 10.1002/jcb.20138] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Osteoblastic migration and proliferation in response to growth factors are essential for skeletal development, bone remodeling, and fracture repair, as well as pathologic processes, such as metastasis. We studied migration in response to platelet-derived growth factor (PDGF, 10 ng/ml) in a wounding model. PDGF stimulated a twofold increase in migration of osteoblastic MC3T3-E1 cells and murine calvarial osteoblasts over 24-48 h. PDGF also stimulated a tenfold increase in 3H-thymidine (3H-TdR) incorporation in MC3T3-E1 cells. Migration and DNA replication, as measured by BrdU incorporation, could be stimulated in the same cell. Blocking DNA replication with aphidicolin did not reduce the distance migrated. To examine the role of mitogen-activated protein (MAP) kinases in migration and proliferation, we used specific inhibitors of p38 MAP kinase, extracellular signal regulated kinase (ERK), and c-Jun N-terminal kinase (JNK). For these signaling studies, proliferation was measured by carboxyfluorescein diacetate succinimidyl ester (CFSE) using flow cytometry. Inhibition of the p38 MAP kinase pathway by SB203580 and SB202190 blocked PDGF-stimulated migration but had no effect on proliferation. Inhibition of the ERK pathway by PD98059 and U0126 inhibited proliferation but did not inhibit migration. Inhibition of JNK activity by SP600125 inhibited both migration and proliferation. Hence, the stimulation of migration and proliferation by PDGF occurred by both overlapping and independent pathways. The JNK pathway was involved in both migration and proliferation, whereas the p38 pathway was predominantly involved in migration and the ERK pathway predominantly involved in proliferation.
Collapse
Affiliation(s)
- Meenal Mehrotra
- University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | | | | | | |
Collapse
|
26
|
Lefebvre P, Martin PJ, Flajollet S, Dedieu S, Billaut X, Lefebvre B. Transcriptional activities of retinoic acid receptors. VITAMINS AND HORMONES 2005; 70:199-264. [PMID: 15727806 DOI: 10.1016/s0083-6729(05)70007-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Vitamin A derivatives plays a crucial role in embryonic development, as demonstrated by the teratogenic effect of either an excess or a deficiency in vitamin A. Retinoid effects extend however beyond embryonic development, and tissue homeostasis, lipid metabolism, cellular differentiation and proliferation are in part controlled through the retinoid signaling pathway. Retinoids are also therapeutically effective in the treatment of skin diseases (acne, psoriasis and photoaging) and of some cancers. Most of these effects are the consequences of retinoic acid receptors activation, which triggers transcriptional events leading either to transcriptional activation or repression of retinoid-controlled genes. Synthetic molecules are able to mimic part of the biological effects of the natural retinoic acid receptors, all-trans retinoic acid. Therefore, retinoic acid receptors are considered as highly valuable therapeutic targets and limiting unwanted secondary effects due to retinoid treatment requires a molecular knowledge of retinoic acid receptors biology. In this review, we will examine experimental evidence which provide a molecular basis for the pleiotropic effects of retinoids, and emphasize the crucial roles of coregulators of retinoic acid receptors, providing a conceptual framework to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Philippe Lefebvre
- INSERM U459 and Ligue Nationale Contre le Cancer, Faculté de Médecine de Lille, 59045 Lille cedex, France
| | | | | | | | | | | |
Collapse
|
27
|
Selmin O, Thorne PA, Caldwell PT, Johnson PD, Runyan RB. Effects of trichloroethylene and its metabolite trichloroacetic acid on the expression of vimentin in the rat H9c2 cell line. Cell Biol Toxicol 2005; 21:83-95. [PMID: 16142583 DOI: 10.1007/s10565-005-0124-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2004] [Accepted: 05/03/2005] [Indexed: 12/21/2022]
Abstract
Trichloroethylene (TCE) and its metabolite trichloroacetic acid (TCAA) are environmental contaminants with specific toxicity for the embryonic heart. In an effort to identify the cellular pathways disrupted by TCE and TCAA during heart development, we investigated their effects on expression of vimentin, a marker of cardiac differentiation. Previous studies had shown that the level of vimentin transcript was inhibited in rat embryonic heart after maternal exposure to TCE via drinking water. In the same study, maternal exposure to TCAA produced the opposite effect, inducing an increased level of vimentin mRNA. In this study, we selected an in vitro system, the rat cardiac myoblast cell line H9c2, to further characterize the molecular mechanisms used by TCE and TCAA to disrupt normal heart development. In particular, we investigated the effects of both toxicants on vimentin, at both the RNA and protein levels, using dose-response and time course curves. Our experimental findings indicate that vimentin expression is affected by TCE and TCAA in H9c2 cells similarly as in vivo. The work is significant because it provides a suitable in vitro model for studies looking at toxicant effects on myocardiac cells, and it suggests that vimentin is a good marker of TCE exposure in the embryonic heart.
Collapse
Affiliation(s)
- O Selmin
- Department of Veterinary Sciences and Microbiology, University of Arizona, Tucson, Arizona 85721-0090, USA.
| | | | | | | | | |
Collapse
|
28
|
White SR, Tse R, Marroquin BA. Stress-activated protein kinases mediate cell migration in human airway epithelial cells. Am J Respir Cell Mol Biol 2005; 32:301-10. [PMID: 15668325 DOI: 10.1165/rcmb.2004-0118oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Airway epithelial cell (AEC) repair immediately after injury requires coordinated cell spreading and migration at the site of injury. Stress-activated protein kinases such as p38 MAPK and c-Jun N-terminal Protein Kinase (JNK) modulate several responses to cell stress and injury, but their role in AEC migration is not clear. We examined migration in confluent 16HBE14o(-) human AEC lines and in primary AEC grown on collagen-IV. Wounds were created by mechanical abrasion and followed to closure using digital microscopy. Inhibitors of either p38 extracellular signal-regulated kinase (ERK)1/2 (PD98059), mitogen-activated protein kinase (MAPK) (SB203580), or JNK (SP600125) could block cell migration substantially. Inhibiting JNK but not p38 MAPK or ERK1/2 blocked extension of cells into the wound region from the original line of injury. Initial migration was associated with phosphorylation of ERK, p38 MAPK, and JNK within 5-15 min. The downstream effector of p38, heat shock protein 27, also was phosphorylated rapidly after injury; phosphorylation could be blocked by prior treatment with SB203580 but not SP600125. The downstream effector of JNK, c-Jun, likewise was phosphorylated rapidly after injury and could be blocked by inhibiting JNK. Our data demonstrate that p38 MAPK, JNK, and ERK1/2 participate in the early stages of AEC migration.
Collapse
Affiliation(s)
- Steven R White
- University of Chicago, Section of Pulmonary and Critical Care Medicine, 5841 S. Maryland Ave., MC 6076, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
29
|
Altan ZM, Fenteany G. c-Jun N-terminal kinase regulates lamellipodial protrusion and cell sheet migration during epithelial wound closure by a gene expression-independent mechanism. Biochem Biophys Res Commun 2004; 322:56-67. [PMID: 15313173 DOI: 10.1016/j.bbrc.2004.07.079] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2004] [Indexed: 02/05/2023]
Abstract
c-Jun N-terminal kinase (JNK) is emerging as an important regulator of cell migration. Perturbing the JNK signaling pathway with three structurally and mechanistically distinct inhibitors that selectively target either JNKs themselves or the upstream mixed-lineage kinases, we found dramatic inhibition of membrane protrusion and cell sheet migration during wound closure in Madin-Darby canine kidney (MDCK) epithelial cell monolayers. Extension of lamellipodia is blocked from the earliest times after wounding in the presence of JNK pathway inhibitors, whereas assembly of non-protrusive actin bundles at the wound margin is unaffected. Inhibitors of the other mitogen-activated protein kinase (MAPK) pathways, the extracellular signal-regulated kinase and p38 MAPK pathways, only have comparatively weak or marginal inhibitory effects on wound closure. Multiple splice variants of both JNK1 and JNK2 are expressed in MDCK cells, and JNK1 and JNK2 are rapidly and transiently activated upon wounding. Phosphorylation of c-Jun does not appear relevant to MDCK wound closure, and membrane protrusion directly after wounding is not affected by inhibitors of RNA or protein synthesis. While most known substrates of JNK are transcription factors or proteins regulating apoptosis, our data indicate that JNK regulates protrusion and migration in a gene expression-independent manner and suggest an important cytoplasmic role for JNK in the control of cell motility.
Collapse
Affiliation(s)
- Z Melis Altan
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
| | | |
Collapse
|
30
|
Abstract
Recent advances in genetic manipulation have greatly expanded our understanding of cellular responses to platelet-derived growth factors (PDGFs) during animal development. In addition to driving mesenchymal proliferation, PDGFs have been shown to direct the migration, differentiation and function of a variety of specialized mesenchymal and migratory cell types, both during development and in the adult animal. Furthermore, the availability of genomic sequence data has facilitated the identification of novel PDGF and PDGF receptor (PDGFR) family members in C. elegans, Drosophila, Xenopus, zebrafish and mouse. Early data from these different systems suggest that some functions of PDGFs have been evolutionarily conserved.
Collapse
Affiliation(s)
- Renée V Hoch
- Program in Developmental Biology and Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | | |
Collapse
|
31
|
Yanfeng W, Saint-Jeannet JP, Klein PS. Wnt-frizzled signaling in the induction and differentiation of the neural crest. Bioessays 2003; 25:317-25. [PMID: 12655639 DOI: 10.1002/bies.10255] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The neural crest is a transient population of multipotent progenitors arising at the lateral edge of the neural plate in vertebrate embryos. After delamination and migration from the neuroepithelium, these cells contribute to a diverse array of tissues including neurons, smooth muscle, craniofacial cartilage, bone cells, endocrine cells and pigment cells. Considerable progress in recent years has furthered our understanding at a molecular level of how this important group of cells is generated and how they are assigned to specific lineages. Here we review a number of recent studies supporting a role for Wnt signaling in neural crest induction, differentiation, and apoptosis. We also summarize the timing of expression of a number of Wnt ligands and receptors with respect to neural crest induction.
Collapse
Affiliation(s)
- Wang Yanfeng
- Cell and Molecular Biology Graduate Group, Department of Medicine and Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
32
|
Ioroi T, Yamamori M, Yagi K, Hirai M, Zhan Y, Kim S, Iwao H. Dominant negative c-Jun inhibits platelet-derived growth factor-directed migration by vascular smooth muscle cells. J Pharmacol Sci 2003; 91:145-8. [PMID: 12686758 DOI: 10.1254/jphs.91.145] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The mitogen-activated protein (MAP) kinase pathways has been shown to be necessary for mitogen-stimulated proliferation, but its role in cell migration has not been fully understood. In this study, we investigated the possible contribution of signaling pathways through c-Jun in platelet-derived growth factor (PDGF)-BB directed cell migration in rat aortic vascular smooth muscle cells (VSMCs) infected with a recombinant adenovirus containing the dominant-negative c-Jun (Ad-DN-c-Jun). DN-c-Jun protein was expressed dose-dependently in VSMCs infected with Ad-DN-c-Jun. Expression of DN-c-Jun significantly inhibited VSMC migration induced by PDGF-BB. Our results provide the first evidence that signaling pathways through c-Jun participates in cell migration induced by PDGF-BB in addition to other MAP kinase pathways in VSMCs.
Collapse
Affiliation(s)
- Takeshi Ioroi
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Braz JC, Bueno OF, De Windt LJ, Molkentin JD. PKC alpha regulates the hypertrophic growth of cardiomyocytes through extracellular signal-regulated kinase1/2 (ERK1/2). J Cell Biol 2002; 156:905-19. [PMID: 11864993 PMCID: PMC2173307 DOI: 10.1083/jcb.200108062] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Members of the protein kinase C (PKC) isozyme family are important signal transducers in virtually every mammalian cell type. Within the heart, PKC isozymes are thought to participate in a signaling network that programs developmental and pathological cardiomyocyte hypertrophic growth. To investigate the function of PKC signaling in regulating cardiomyocyte growth, adenoviral-mediated gene transfer of wild-type and dominant negative mutants of PKC alpha, beta II, delta, and epsilon (only wild-type zeta) was performed in cultured neonatal rat cardiomyocytes. Overexpression of wild-type PKC alpha, beta II, delta, and epsilon revealed distinct subcellular localizations upon activation suggesting unique functions of each isozyme in cardiomyocytes. Indeed, overexpression of wild-type PKC alpha, but not betaI I, delta, epsilon, or zeta induced hypertrophic growth of cardiomyocytes characterized by increased cell surface area, increased [(3)H]-leucine incorporation, and increased expression of the hypertrophic marker gene atrial natriuretic factor. In contrast, expression of dominant negative PKC alpha, beta II, delta, and epsilon revealed a necessary role for PKC alpha as a mediator of agonist-induced cardiomyocyte hypertrophy, whereas dominant negative PKC epsilon reduced cellular viability. A mechanism whereby PKC alpha might regulate hypertrophy was suggested by the observations that wild-type PKC alpha induced extracellular signal-regulated kinase1/2 (ERK1/2), that dominant negative PKC alpha inhibited PMA-induced ERK1/2 activation, and that dominant negative MEK1 (up-stream of ERK1/2) inhibited wild-type PKC alpha-induced hypertrophic growth. These results implicate PKC alpha as a necessary mediator of cardiomyocyte hypertrophic growth, in part, through a ERK1/2-dependent signaling pathway.
Collapse
Affiliation(s)
- Julian C Braz
- Department of Pediatrics, University of Cincinnati, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|