201
|
Shih HP, Gross MK, Kioussi C. Muscle development: forming the head and trunk muscles. Acta Histochem 2007; 110:97-108. [PMID: 17945333 PMCID: PMC6317512 DOI: 10.1016/j.acthis.2007.08.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Revised: 08/16/2007] [Accepted: 08/21/2007] [Indexed: 01/26/2023]
Abstract
The morphological events forming the body's musculature are sensitive to genetic and environmental perturbations with high incidence of congenital myopathies, muscular dystrophies and degenerations. Pattern formation generates branching series of states in the genetic regulatory network. Different states of the network specify pre-myogenic progenitor cells in the head and trunk. These progenitors reveal their myogenic nature by the subsequent onset of expression of the master switch gene MyoD and/or Myf5. Once initiated, the myogenic progression that ultimately forms mature muscle appears to be quite similar in head and trunk skeletal muscle. Several genes that are essential in specifying pre-myogenic progenitors in the trunk are known. Pax3, Lbx1, and a number of other homeobox transcription factors are essential in specifying pre-myogenic progenitors in the dermomyotome, from which the epaxial and hypaxial myoblasts, which express myogenic regulatory factors (MRFs), emerge. The proteins involved in specifying pre-myogenic progenitors in the head are just beginning to be discovered and appear to be distinct from those in the trunk. The homeobox gene Pitx2, the T-box gene Tbx1, and the bHLH genes Tcf21 and Msc encode transcription factors that play roles in specifying progenitor cells that will give rise to branchiomeric muscles of the head. Pitx2 is expressed well before the onset of myogenic progression in the first branchial arch (BA) mesodermal core and is essential for the formation of first BA derived muscle groups. Anterior-posterior patterning events that occur during gastrulation appear to initiate the Pitx2 expression domain in the cephalic and BA mesoderm. Pitx2 therefore contributes to the establishment of network states, or kernels, that specify pre-myogenic progenitors for extraocular and mastication muscles. A detailed understanding of the molecular mechanisms that regulate head muscle specification and formation provides the foundation for understanding congenital myopathies. Current technology and mouse model systems help to elucidate the molecular basis on etiology and repair of muscular degenerative diseases.
Collapse
Affiliation(s)
- Hung Ping Shih
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331
| | - Michael K. Gross
- Department of Biochemistry and Biophysics, College of Sciences, Oregon State University, Corvallis, OR 97331
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331
- Corresponding Author, , T (541) 737-2179, F (541) 737-3999
| |
Collapse
|
202
|
Feldkamp ML, Carey JC, Sadler TW. Development of gastroschisis: review of hypotheses, a novel hypothesis, and implications for research. Am J Med Genet A 2007; 143A:639-52. [PMID: 17230493 DOI: 10.1002/ajmg.a.31578] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gastroschisis, a ventral body wall defect, is a continuing challenge and concern to researchers, clinicians, and epidemiologists seeking to identify its cause(s) and pathogenesis. Concern has been renewed in recent years because, unlike most other birth defects, rates of gastroschisis are reportedly increasing in many developed and developing countries. No tenable explanation or specific causes have been identified for this trend. Rates of gastroschisis are particularly high among pregnancies of very young women. Such an intriguing association, not observed to this degree with other birth defects, may afford clues to the defect's cause. Understanding the causes of gastroschisis may provide insight to the defect's origin. In pursuing such causal studies, it would be helpful to understand the embryogenesis of gastroschisis. To date, four main embryologic hypotheses have been proposed: (1) Failure of mesoderm to form in the body wall; (2) Rupture of the amnion around the umbilical ring with subsequent herniation of bowel; (3) Abnormal involution of the right umbilical vein leading to weakening of the body wall and gut herniation; and (4) Disruption of the right vitelline (yolk sac) artery with subsequent body wall damage and gut herniation. Although based on embryological phenomena, these hypotheses do not provide an adequate explanation for how gastroschisis would occur. Therefore, we propose an alternative hypothesis, based on well described embryonic events. Specifically, we propose that abnormal folding of the body wall results in a ventral body wall defect through which the gut herniates, leading to the clinical presentation of gastroschisis. This hypothesis potentially explains the origin of gastroschisis as well as that of other developmental defects of the ventral wall.
Collapse
Affiliation(s)
- Marcia L Feldkamp
- Department of Pediatrics, Division of Medical Genetics, University of Utah Health Sciences Center, Salt Lake City, UT 84132, USA.
| | | | | |
Collapse
|
203
|
Abstract
Anterior segment dysgenesis (ASD) is a failure of the normal development of the tissues of the anterior segment of the eye. It leads to anomalies in the structure of the mature anterior segment, associated with an increased risk of glaucoma and corneal opacity. Several different gene mutations have been identified underlying these anomalies with the majority of ASD genes encoding transcriptional regulators. In this review, the role of the ASD genes, PITX2 and FOXC1, is considered in relation to the embryology of the anterior segment, the biochemical function of these proteins, and their role in development and disease aetiology. The emerging view is that these genes act in concert to specify a population of mesenchymal progenitor cells, mainly of neural crest origin, as they migrate anteriorly around the embryonic optic cup. These same genes then regulate mesenchymal cell differentiation to give rise to distinct anterior segment tissues. Development appears critically sensitive to gene dosage, and variation in the normal level of transcription factor activity causes a range of anterior segment anomalies. Interplay between PITX2 and FOXC1 in the development of different anterior segment tissues may partly explain the phenotypic variability and the genetic heterogeneity characteristic of ASD.
Collapse
Affiliation(s)
- J C Sowden
- Developmental Biology Unit, University College London Institute of Child Health and Great Ormond Street Hospital for Children NHS Trust, London, UK.
| |
Collapse
|
204
|
Zhu X, Gleiberman AS, Rosenfeld MG. Molecular physiology of pituitary development: signaling and transcriptional networks. Physiol Rev 2007; 87:933-63. [PMID: 17615393 DOI: 10.1152/physrev.00006.2006] [Citation(s) in RCA: 240] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The pituitary gland is a central endocrine organ regulating basic physiological functions, including growth, the stress response, reproduction, metabolic homeostasis, and lactation. Distinct hormone-producing cell types in the anterior pituitary arise from a common ectodermal primordium during development by extrinsic and intrinsic mechanisms, providing a powerful model system for elucidating general principles in mammalian organogenesis. The central purpose of this review is to inspect the integrated signaling and transcriptional events that affect precursor proliferation, cell lineage commitment, terminal differentiation, and physiological regulation by hypothalamic tropic factors.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, La Jolla, California 92093, USA.
| | | | | |
Collapse
|
205
|
Kao WWY, Liu CY. The use of transgenic and knock-out mice in the investigation of ocular surface cell biology. Ocul Surf 2007; 1:5-19. [PMID: 17075625 DOI: 10.1016/s1542-0124(12)70003-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The transgenic and knock-out mice created by transgenesis and gene targeting techniques are very useful for elucidating the pathophysiology of human diseases caused by altered genetic functions. Many of the experimental mouse lines exhibit ocular surface disorders. However, embryonic lethality and congenital defects found in many of the transgenic and knock-out mice preclude their use for studying the consequences of altered genetic functions in adult animals. To circumvent these difficulties, we have established binary inducible mouse models, using the corneal keratocyte-specific keratocan promoter, and the tetracycline-inducible gene expression system (reverse tetracycline transcription activator--rtTA). In these models, the animals function normally until they are fed doxycycline, thus inducing the overexpression of inserted transgenes by keratocytes. We have also developed inserted rtTA and Cre reporter gene constructs to create genetically modified mouse lines that have tissue-specific gene alterations to study acquired conditions, e.g., wound healing and irregular hormone and cytokine signaling that offsets homeostasis in adults. Furthermore, the genes that are ubiquitously expressed in many tissues can be specifically ablated solely in ocular surface tissues to examine their function, since the loss of such a gene in ocular surface tissues will not be life-threatening. It is noteworthy that these altered mouse lines can also be used as models for the development of therapeutic treatment regimens of diseases using gene therapy and stem cell strategies.
Collapse
Affiliation(s)
- Winston W-Y Kao
- Department of Opthalmology, University of Cincinnati, Cincinnati, OH 45267-0527, USA.
| | | |
Collapse
|
206
|
Ai D, Wang J, Amen M, Lu MF, Amendt BA, Martin JF. Nuclear factor 1 and T-cell factor/LEF recognition elements regulate Pitx2 transcription in pituitary development. Mol Cell Biol 2007; 27:5765-75. [PMID: 17562863 PMCID: PMC1952127 DOI: 10.1128/mcb.01848-06] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 01/17/2007] [Accepted: 05/21/2007] [Indexed: 01/13/2023] Open
Abstract
Pitx2, a paired-related homeobox gene that is mutated in Rieger syndrome I, is the earliest known marker of oral ectoderm. Pitx2 was previously shown to be required for tooth, palate, and pituitary development in mice; however, the mechanisms regulating Pitx2 transcription in the oral ectoderm are poorly understood. Here we used an in vivo transgenic approach to investigate the mechanisms regulating Pitx2 transcription. We identified a 7-kb fragment that directs LacZ expression in oral ectoderm and in many of its derivatives. Deletion analysis of transgenic embryos reduced this fragment to a 520-bp region that directed LacZ activity to Rathke's pouch. A comparison of the mouse and human sequences revealed a conserved nuclear factor 1 (NF-1) recognition element near a consensus T-cell factor (TCF)/LEF binding site. The mutation of either site individually abolished LacZ activity in transgenic embryos, identifying Pitx2 as a direct target of Wnt signaling in pituitary development. These findings uncover a requirement for NF-1 and TCF factors in Pitx2 transcriptional regulation in the pituitary and provide insight into the mechanisms controlling region-specific transcription in the oral ectoderm and its derivatives.
Collapse
Affiliation(s)
- Di Ai
- Institute of Biosciences and Technology, Texas A&M System Health Science Center, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
207
|
Selamet Tierney ES, Marans Z, Rutkin MB, Chung WK. Variants of the CFC1 gene in patients with laterality defects associated with congenital cardiac disease. Cardiol Young 2007; 17:268-74. [PMID: 17445335 DOI: 10.1017/s1047951107000455] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/12/2006] [Indexed: 11/05/2022]
Abstract
OBJECTIVES This study was designed to assess the frequency and types of genetic variants in CFC1 in children with laterality disorders associated with cardiovascular involvement. BACKGROUND Laterality syndromes are estimated to comprise 3% of neonates with congenital cardiac disease. Genetic predisposition in some cases of laterality defects has been suggested by associated chromosomal anomalies and familial aggregation, often within consanguineous families, suggesting autosomal recessive inheritance. Mice with induced homozygous mutations in cfc1, and heterozygous CFC1 mutations in humans, have been associated with laterality defects. METHODS Direct sequence analysis of the coding sequence of CFC1 was performed in 42 subjects with laterality defects and congenital cardiac disease. RESULTS We identified 3 synonymous coding variants, 3 non-synonymous coding variants (N21H, R47Q, and R78W), and 2 intronic variants in CFC1. The N21H variant was observed in 3 of 19 affected Caucasians, and the R47Q variant in another 2. Neither polymorphism was observed in Caucasian controls. Furthermore, all subjects with the N21H polymorphism had double outlet right ventricle. Transmission of both the N21H and R47Q polymorphisms from unaffected parents was demonstrated, and all three non-synonymous variants had significant allele frequencies in unaffected African-American subjects, suggesting that other factors must also contribute to laterality defects. CONCLUSIONS Three non-synonymous variants in CFC1 were identified, the N21H variant being associated with laterality defects in Caucasians, but not fully penetrant. One or more of these non-synonymous missense variants may act as a susceptibility allele in conjunction with other genes, and/or environmental factors, to cause laterality defects.
Collapse
Affiliation(s)
- Elif Seda Selamet Tierney
- Division of Pediatric Cardiology, Morgan Stanley Children's Hospital of New York Presbyterian, Columbia University, College of Physicians & Surgeons, New York, NY, USA.
| | | | | | | |
Collapse
|
208
|
Chen CP. Syndromes and Disorders Associated with Omphalocele (III): Single Gene Disorders, Neural Tube Defects, Diaphragmatic Defects and Others. Taiwan J Obstet Gynecol 2007; 46:111-20. [PMID: 17638618 DOI: 10.1016/s1028-4559(07)60004-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Omphalocele can be associated with single gene disorders, neural tube defects, diaphragmatic defects, fetal valproate syndrome, and syndromes of unknown etiology. This article provides a comprehensive review of omphalocele-related disorders: otopalatodigital syndrome type II; Melnick-Needles syndrome; Rieger syndrome; neural tube defects; Meckel syndrome; Shprintzen-Goldberg omphalocele syndrome; lethal omphalocele-cleft palate syndrome; cerebro-costo-mandibular syndrome; fetal valproate syndrome; Marshall-Smith syndrome; fibrochondrogenesis; hydrolethalus syndrome; Fryns syndrome; omphalocele, diaphragmatic defects, radial anomalies and various internal malformations; diaphragmatic defects, limb deficiencies and ossification defects of skull; Donnai-Barrow syndrome; CHARGE syndrome; Goltz syndrome; Carpenter syndrome; Toriello-Carey syndrome; familial omphalocele; Cornelia de Lange syndrome; C syndrome; Elejalde syndrome; Malpuech syndrome; cervical ribs, Sprengel anomaly, anal atresia and urethral obstruction; hydrocephalus with associated malformations; Kennerknecht syndrome; lymphedema, atrial septal defect and facial changes; and craniosynostosismental retardation syndrome of Lin and Gettig. Perinatal identification of omphalocele should alert one to the possibility of omphalocele-related disorders and familial inheritance and prompt a thorough genetic counseling for these disorders.
Collapse
Affiliation(s)
- Chih-Ping Chen
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei, Taiwan.
| |
Collapse
|
209
|
Lin L, Cui L, Zhou W, Dufort D, Zhang X, Cai CL, Bu L, Yang L, Martin J, Kemler R, Rosenfeld MG, Chen J, Evans SM. Beta-catenin directly regulates Islet1 expression in cardiovascular progenitors and is required for multiple aspects of cardiogenesis. Proc Natl Acad Sci U S A 2007; 104:9313-8. [PMID: 17519333 PMCID: PMC1890491 DOI: 10.1073/pnas.0700923104] [Citation(s) in RCA: 210] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent studies have demonstrated that the LIM homeodomain transcription factor Islet1 (Isl1) marks pluripotent cardiovascular progenitor cells and is required for proliferation, survival, and migration of recently defined second heart field progenitors. Factors that are upstream of Isl1 in cardiovascular progenitors have not yet been defined. Here we demonstrate that beta-catenin is required for Isl1 expression in cardiac progenitors, directly regulating the Isl1 promoter. Ablation of beta-catenin in Isl1-expressing progenitors disrupts multiple aspects of cardiogenesis, resulting in embryonic lethality at E13. beta-Catenin is also required upstream of a number of genes required for pharyngeal arch, outflow tract, and/or atrial septal morphogenesis, including Tbx2, Tbx3, Wnt11, Shh, and Pitx2. Our findings demonstrate that beta-catenin signaling regulates proliferation and survival of cardiac progenitors.
Collapse
Affiliation(s)
- Lizhu Lin
- *Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Li Cui
- *Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Wenlai Zhou
- Howard Hughes Medical Institute and Department of Medicine, and
| | - Daniel Dufort
- Department of Obstetrics and Gynecology, Royal Victoria Hospital, 687 Pine Avenue West, Montreal, QC, Canada H3A 1A1; and
| | - Xiaoxue Zhang
- *Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Chen-Leng Cai
- *Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Lei Bu
- *Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Lei Yang
- *Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Jody Martin
- *Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Rolf Kemler
- Max-Planck-Institute of Immunobiology, Stubeweg 51, 79108 Freiburg, Germany
| | | | - Ju Chen
- School of Medicine, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | - Sylvia M. Evans
- *Skaggs School of Pharmacy and Pharmaceutical Sciences
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
210
|
Rulifson IC, Karnik SK, Heiser PW, ten Berge D, Chen H, Gu X, Taketo MM, Nusse R, Hebrok M, Kim SK. Wnt signaling regulates pancreatic beta cell proliferation. Proc Natl Acad Sci U S A 2007; 104:6247-52. [PMID: 17404238 PMCID: PMC1847455 DOI: 10.1073/pnas.0701509104] [Citation(s) in RCA: 274] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Indexed: 01/09/2023] Open
Abstract
There is widespread interest in defining factors and mechanisms that stimulate proliferation of pancreatic islet cells. Wnt signaling is an important regulator of organ growth and cell fates, and genes encoding Wnt-signaling factors are expressed in the pancreas. However, it is unclear whether Wnt signaling regulates pancreatic islet proliferation and differentiation. Here we provide evidence that Wnt signaling stimulates islet beta cell proliferation. The addition of purified Wnt3a protein to cultured beta cells or islets promoted expression of Pitx2, a direct target of Wnt signaling, and Cyclin D2, an essential regulator of beta cell cycle progression, and led to increased beta cell proliferation in vitro. Conditional pancreatic beta cell expression of activated beta-catenin, a crucial Wnt signal transduction protein, produced similar phenotypes in vivo, leading to beta cell expansion, increased insulin production and serum levels, and enhanced glucose handling. Conditional beta cell expression of Axin, a potent negative regulator of Wnt signaling, led to reduced Pitx2 and Cyclin D2 expression by beta cells, resulting in reduced neonatal beta cell expansion and mass and impaired glucose tolerance. Thus, Wnt signaling is both necessary and sufficient for islet beta cell proliferation, and our study provides previously unrecognized evidence of a mechanism governing endocrine pancreas growth and function.
Collapse
Affiliation(s)
| | | | - Patrick W. Heiser
- Diabetes Center, University of California, San Francisco, CA 94143-0573
| | - Derk ten Berge
- Departments of *Developmental Biology and
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305-5329; and
| | | | - Xueying Gu
- Departments of *Developmental Biology and
| | - Makoto M. Taketo
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Yoshida-Konoé-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Roel Nusse
- Departments of *Developmental Biology and
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305-5329; and
| | - Matthias Hebrok
- Diabetes Center, University of California, San Francisco, CA 94143-0573
| | - Seung K. Kim
- Departments of *Developmental Biology and
- Medicine, Oncology Division, Stanford University, Stanford, CA 94305-5329
| |
Collapse
|
211
|
Shih HP, Gross MK, Kioussi C. Cranial muscle defects of Pitx2 mutants result from specification defects in the first branchial arch. Proc Natl Acad Sci U S A 2007; 104:5907-12. [PMID: 17384148 PMCID: PMC1851590 DOI: 10.1073/pnas.0701122104] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Indexed: 12/11/2022] Open
Abstract
Pitx2 expression is observed during all states of the myogenic progression in embryonic muscle anlagen and persists in adult muscle. Pitx2 mutant mice form all but a few muscle anlagen. Loss or degeneration in muscle anlagen could generally be attributed to the loss of a muscle attachment site induced by some other aspect of the Pitx2 phenotype. Muscles derived from the first branchial arch were absent, whereas muscles derived from the second branchial arch were merely distorted in Pitx2 mutants at midgestation. Pitx2 was expressed well before, and was required for, initiation of the myogenic progression in the first, but not second, branchial arch mesoderm. Pitx2 was also required for expression of premyoblast specification markers Tbx1, Tcf21, and Msc in the first, but not second, branchial arch. First, but not second, arch mesoderm of Pitx2 mutants failed to enlarge after embryonic day 9.5, well before the onset of the myogenic progression. Thus, Pitx2 contributes to specification of first, but not second, arch mesoderm. The jaw of Pitx2 mutants was vestigial by midgestation, but significant size reductions were observed as early as embryonic day 10.5. The diminutive first branchial arch of mutants could not be explained by loss of mesoderm alone, suggesting that Pitx2 contributes to the earliest specification of jaw itself.
Collapse
Affiliation(s)
- Hung Ping Shih
- *Department of Pharmaceutical Sciences, College of Pharmacy, and
- Department of Biochemistry and Biophysics, College of Sciences, Oregon State University, Corvallis, OR 97331
| | - Michael K. Gross
- Department of Biochemistry and Biophysics, College of Sciences, Oregon State University, Corvallis, OR 97331
| | - Chrissa Kioussi
- *Department of Pharmaceutical Sciences, College of Pharmacy, and
| |
Collapse
|
212
|
Abstract
The vertebrate lung consists of multiple cell types that are derived primarily from endodermal and mesodermal compartments of the early embryo. The process of pulmonary organogenesis requires the generation of precise signaling centers that are linked to transcriptional programs that, in turn, regulate cell numbers, differentiation, and behavior, as branching morphogenesis and alveolarization proceed. This review summarizes knowledge regarding the expression and proposed roles of transcription factors influencing lung formation and function with particular focus on knowledge derived from the study of the mouse. A group of transcription factors active in the endodermally derived cells of the developing lung tubules, including thyroid transcription factor-1 (TTF-1), beta-catenin, Forkhead orthologs (FOX), GATA, SOX, and ETS family members are required for normal lung morphogenesis and function. In contrast, a group of distinct proteins, including FOXF1, POD1, GLI, and HOX family members, play important roles in the developing lung mesenchyme, from which pulmonary vessels and bronchial smooth muscle develop. Lung formation is dependent on reciprocal signaling among cells of both endodermal and mesenchymal compartments that instruct transcriptional processes mediating lung formation and adaptation to breathing after birth.
Collapse
Affiliation(s)
- Yutaka Maeda
- Division of Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | |
Collapse
|
213
|
Shih HP, Gross MK, Kioussi C. Expression pattern of the homeodomain transcription factor Pitx2 during muscle development. Gene Expr Patterns 2007; 7:441-51. [PMID: 17166778 DOI: 10.1016/j.modgep.2006.11.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2006] [Revised: 11/03/2006] [Accepted: 11/08/2006] [Indexed: 11/25/2022]
Abstract
Late-stage Pitx2(+/LacZ) mouse embryos stained with x-gal appeared to have blue muscles, suggesting that Pitx2 expression specifically marks some phase of the myogenic progression or muscle anlagen formation. Detailed temporal and spatial analyses were undertaken to determine the extent and onset of Pitx2 expression in muscle. Pitx2 was specifically expressed in the vast majority of muscles of the head and trunk in late embryos and adults. Early Pitx2 expression in the cephalic mesoderm, first branchial arch and somatopleure preceded specification of head muscle. In contrast, Pitx2 expression appeared to follow muscle specification events in the trunk. However, Pitx2 expression was rapidly upregulated in these myogenic structures by E10.5. Upregulation correlated tightly with the apposition of a non-myogenic, Pitx2-expressing, cell cluster lateral to the dermomyotome. This cluster first appeared at the forelimb level at E10.25, gradually elongated in the posterior direction, appeared to aggregate from delaminated cells emanating from the ventrally located somatopleure, and was named the dorsal somatopleure. Immunohistochemistry on appendicular sections after E10.5 demonstrated that Pitx2 neatly marked the areas of muscle anlagen, that Pax3, Lbx1, and the muscle regulatory factors (MRFs) stained only subsets of Pitx2(+) cells within these areas, and that virtually all Pitx2(+) cells in these areas express at least one of these known myogenic markers. Taken together, the results demonstrate that, within muscle anlagen, Pitx2 marks the muscle lineage more completely that any of the known markers, and are consistent with a role for Pitx2 in muscle anlagen formation or maintenance.
Collapse
Affiliation(s)
- Hung Ping Shih
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | | | | |
Collapse
|
214
|
|
215
|
Martínez-Fernandez S, Hernández-Torres F, Franco D, Lyons GE, Navarro F, Aránega AE. Pitx2c overexpression promotes cell proliferation and arrests differentiation in myoblasts. Dev Dyn 2006; 235:2930-9. [PMID: 16958127 DOI: 10.1002/dvdy.20924] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Pitx2 is a paired-related homeobox gene that has been shown to play a central role during development. In the mouse, there are three isoforms, Pitx2a, b, and c, which differ only in their amino terminal regions. Pitx2 is expressed in myotomes, myoblasts, and myofibers and may be involved in muscle patterning. However, the mechanism by which Pitx2 acts in muscle cell lineages as well as the distinct functions of the individual isoforms have not been investigated. In this study, we used Sol8 myoblasts to investigate the function of Pitx2 in skeletal myogenesis. We found that Pitx2c is the main Pitx2 isoform present in Sol8 myoblasts. Overexpression of Pitx2c in Sol8 myoblasts inhibited myocyte differentiation and myotube formation. Furthermore, Sol8 cells overexpressing Pitx2c maintained high proliferative capacity and a significant up-regulation of the cell cycle genes cyclin D1, cyclin D2, and c-myc. Gene expression analysis for Pax3 and the s MyoD and myogenin showed that Pitx2c-overexpression caused Sol8 cells to remain as myoblasts, in an undifferentiated myogenic state. Furthermore, down-regulation of the muscle-specific genes sTnI and MyHC3 demonstrated that Sol8-overexpressing Pitx2c myoblasts failed to reach terminal differentiation. This study sheds light on previously unknown functions of the Pitx2c isoform in balancing proliferation vs. differentiation in a myogenic cell line.
Collapse
|
216
|
Gerhart J, Elder J, Neely C, Schure J, Kvist T, Knudsen K, George-Weinstein M. MyoD-positive epiblast cells regulate skeletal muscle differentiation in the embryo. J Cell Biol 2006; 175:283-92. [PMID: 17060497 PMCID: PMC2064569 DOI: 10.1083/jcb.200605037] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2006] [Accepted: 09/15/2006] [Indexed: 11/22/2022] Open
Abstract
MyoD mRNA is expressed in a subpopulation of cells within the embryonic epiblast. Most of these cells are incorporated into somites and synthesize Noggin. Ablation of MyoD-positive cells in the epiblast subsequently results in the herniation of organs through the ventral body wall, a decrease in the expression of Noggin, MyoD, Myf5, and myosin in the somites and limbs, and an increase in Pax-3-positive myogenic precursors. The addition of Noggin lateral to the somites compensates for the loss of MyoD-positive epiblast cells. Skeletal muscle stem cells that arise in the epiblast are utilized in the somites to promote muscle differentiation by serving as a source of Noggin.
Collapse
Affiliation(s)
- Jacquelyn Gerhart
- Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, Philadelphia, PA 19131, USA
| | | | | | | | | | | | | |
Collapse
|
217
|
Hirano M, Kiyonari H, Inoue A, Furushima K, Murata T, Suda Y, Aizawa S. A new serine/threonine protein kinase, Omphk1, essential to ventral body wall formation. Dev Dyn 2006; 235:2229-37. [PMID: 16715502 DOI: 10.1002/dvdy.20823] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Here, we report a new serine/threonine protein kinase of the SNF1 subfamily Omphk1. Two Omphk homologues exist in each vertebrate species, and one homologue exists in Drosophila and Caenorhabditis elegans; the kinase domain is highly conserved among these homologues, and several domains are conserved among vertebrate Omphk. Omphk1 expression dynamically changes in the developing central nervous system, is found ubiquitously in epidermis, and is present uniquely in several other tissues. Its expression is also found in each tissue associated with the ventral body wall closure: the primary body wall composed of primitive ectoderm and each component of the secondary body wall. Concomitantly, its null mutant exhibits omphalocele with a failure in closure of the secondary body wall. There are no apparent gross morphological defects in brain, however, despite the unique Omphk1 expression in this tissue.
Collapse
Affiliation(s)
- Mariko Hirano
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology, RIKEN Kobe, Minami-machi, Chuo-ku, Kobe, Japan
| | | | | | | | | | | | | |
Collapse
|
218
|
Quentien MH, Barlier A, Franc JL, Pellegrini I, Brue T, Enjalbert A. Pituitary transcription factors: from congenital deficiencies to gene therapy. J Neuroendocrinol 2006; 18:633-42. [PMID: 16879162 DOI: 10.1111/j.1365-2826.2006.01461.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Despite the existence of interspecies phenotypic variability, animal models have yielded valuable insights into human pituitary diseases. Studies on Snell and Jackson mice known to have growth hormone, prolactin and thyroid-stimulating hormone deficiencies involving the hypoplastic pituitary gland have led to identifying alterations of the pituitary specific POU homeodomain Pit-1 transcription factor gene. The human phenotype associated with rare mutations in this gene was found to be similar to that of these mice mutants. Terminal differentiation of lactotroph cells and direct regulation of the prolactin gene both require interactions between Pit-1 and cell type specific partners, including panpituitary transcriptional regulators such as Pitx1 and Pitx2. Synergistic activation of the prolactin promoter by Pitx factors and Pit-1 is involved not only in basal condition, but also in responsiveness to forskolin, thyrotrophin-releasing-hormone and epidermal growth factor. In corticotroph cells, Pitx1 interacts with Tpit. Tpit mutations have turned out to be the main molecular cause of neonatal isolated adrenocorticotrophin deficiency. This finding supports the idea that Tpit plays an essential role in the differentiation of the pro-opiomelanocortin pituitary lineage. The effects of Pit-1 are not restricted to hormone gene regulation because this factor also contributes to cell division and protects the cell from programmed cell death. Lentiviral vectors expressing a Pit-1 dominant negative mutant induced time- and dose-dependent cell death in somatotroph and lactotroph adenomas in vitro. Gene transfer by lentiviral vectors should provide a promising step towards developing an efficient specific therapeutic approach by which a gene therapy programme for treating human pituitary adenomas could be based.
Collapse
Affiliation(s)
- M H Quentien
- ICNE-UMR6544-CNRS-Université de la Méditerranée, Institut Jean Roche, Marseille, France.
| | | | | | | | | | | |
Collapse
|
219
|
Ai D, Liu W, Ma L, Dong F, Lu MF, Wang D, Verzi MP, Cai C, Gage PJ, Evans S, Black BL, Brown NA, Martin JF. Pitx2 regulates cardiac left-right asymmetry by patterning second cardiac lineage-derived myocardium. Dev Biol 2006; 296:437-49. [PMID: 16836994 PMCID: PMC5851592 DOI: 10.1016/j.ydbio.2006.06.009] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2006] [Revised: 06/01/2006] [Accepted: 06/01/2006] [Indexed: 11/28/2022]
Abstract
Current models of left-right asymmetry hold that an early asymmetric signal is generated at the node and transduced to lateral plate mesoderm in a linear signal transduction cascade through the function of the Nodal signaling molecule. The Pitx2 homeobox gene functions at the final stages of this cascade to direct asymmetric morphogenesis of selected organs including the heart. We previously showed that Pitx2 regulated an asymmetric pathway that was independent of cardiac looping suggesting a second asymmetric cardiac pathway. It has been proposed that in the cardiac outflow tract Pitx2 functions in both cardiac neural crest, as a target of canonical Wnt-signaling, and in the mesoderm-derived cardiac second lineage. We used fate mapping, conditional loss of function, and chimera analysis in mice to investigate the role of Pitx2 in outflow tract morphogenesis. Our findings reveal that Pitx2 is dispensable in the cardiac neural crest but functions in second lineage myocardium revealing that this cardiac progenitor field is patterned asymmetrically.
Collapse
Affiliation(s)
- Di Ai
- Institute of Biosciences and Technology, Texas A and M System, Health Science Center, 2121 Holcombe Blvd, Houston, TX 77030, USA
| | - Wei Liu
- Institute of Biosciences and Technology, Texas A and M System, Health Science Center, 2121 Holcombe Blvd, Houston, TX 77030, USA
| | - Lijiang Ma
- Institute of Biosciences and Technology, Texas A and M System, Health Science Center, 2121 Holcombe Blvd, Houston, TX 77030, USA
| | - Feiyan Dong
- Institute of Biosciences and Technology, Texas A and M System, Health Science Center, 2121 Holcombe Blvd, Houston, TX 77030, USA
| | - Mei-Fang Lu
- Institute of Biosciences and Technology, Texas A and M System, Health Science Center, 2121 Holcombe Blvd, Houston, TX 77030, USA
| | - Degang Wang
- Institute of Biosciences and Technology, Texas A and M System, Health Science Center, 2121 Holcombe Blvd, Houston, TX 77030, USA
| | - Michael P. Verzi
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143-2240, USA
| | - Chenleng Cai
- Institute of Molecular Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Philip J. Gage
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sylvia Evans
- Institute of Molecular Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brian L. Black
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143-2240, USA
| | - Nigel A. Brown
- Division of Basic Medical Sciences, St. George’s Hospital Medical School, University of London, Cranmer Terrace, London SW17 ORE, UK
| | - James F. Martin
- Institute of Biosciences and Technology, Texas A and M System, Health Science Center, 2121 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
220
|
Bothe I, Dietrich S. The molecular setup of the avian head mesoderm and its implication for craniofacial myogenesis. Dev Dyn 2006; 235:2845-60. [PMID: 16894604 DOI: 10.1002/dvdy.20903] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The head mesoderm is the mesodermal tissue on either side of the brain, from forebrain to hindbrain levels, and gives rise to the genuine head muscles. Its relatedness to the more posterior paraxial mesoderm, the somites, which generate the muscles of the trunk, is conversely debated. To gain insight into the molecular setup of the head mesoderm, its similarity or dissimilarity to the somitic mesoderm, and the implications of its setup for the progress of muscle formation, we investigated the expression of markers (1) for mesoderm segmentation and boundary formation, (2) for regional specification and somitogenesis and (3) for the positive and negative control of myogenic differentiation. We show that the head mesoderm is molecularly distinct from somites. It is not segmented; even the boundary to the first somite is ill-defined. Importantly, the head mesoderm lacks the transcription factors driving muscle differentiation while genes suppressing differentiation and promoting cell proliferation are expressed. These factors show anteroposteriorly and dorsoventrally regionalised but overlapping expression. Notably, expression extends into the areas that actively contribute to the heart, overlapping with the expression of cardiac markers.
Collapse
Affiliation(s)
- Ingo Bothe
- King's College London, Department of Craniofacial Development, Guy's Hospital, London, United Kingdom
| | | |
Collapse
|
221
|
Sclafani AM, Skidmore JM, Ramaprakash H, Trumpp A, Gage PJ, Martin DM. Nestin-Cre mediated deletion of Pitx2 in the mouse. Genesis 2006; 44:336-44. [PMID: 16823861 DOI: 10.1002/dvg.20220] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Nestin-Cre mice are widely used to generate gene deletions in the developing brain. Surprisingly, fewNestin-Cre lines have been characterized for their temporal and brain region-specific recombination. In addition, some Nestin-Cre lines express Cre outside the central nervous system, making it difficult to choose appropriate lines for targeting genes with brain region-restricted expression. Here we describe the properties of a Nestin-Cre transgenic line and its use for conditional deletions of Pitx2, a paired-like homeodomain transcription factor. We report that Nestin-Cre conditional Pitx2 mutant mice have ocular and craniofacial defects consistent with the role of human PITX2 in Rieger syndrome. Conditional mutants exhibit defects in midbrain neuronal development similar to those in Pitx2 homozygous null embryos, but lack the abnormalities in subthalamic nucleus neurons that occur with complete loss of Pitx2 function. These data indicate that normal differentiation of midbrain neurons depends upon adequate Pitx2 function during the period of active neurogenesis.
Collapse
Affiliation(s)
- Anthony M Sclafani
- Molecular, Cellular and Developmental Biology Graduate Program, Yale College of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
222
|
Berry FB, Lines MA, Oas JM, Footz T, Underhill DA, Gage PJ, Walter MA. Functional interactions between FOXC1 and PITX2 underlie the sensitivity to FOXC1 gene dose in Axenfeld-Rieger syndrome and anterior segment dysgenesis. Hum Mol Genet 2006; 15:905-19. [PMID: 16449236 DOI: 10.1093/hmg/ddl008] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Axenfeld-Rieger ocular dysgenesis is associated with mutations of the human PITX2 and FOXC1 genes, which encode transcription factors of the homeodomain and forkhead types, respectively. We have identified a functional link between FOXC1 and PITX2 which we propose underpins the similar Axenfeld-Rieger phenotype caused by mutations of these genes. FOXC1 and PITX2A physically interact, and this interaction requires crucial functional domains on both proteins: the C-terminal activation domain of FOXC1 and the homeodomain of PITX2. Immunofluorescence further shows PITX2A and FOXC1 to be colocalized within a common nuclear subcompartment. Furthermore, PITX2A can function as a negative regulator of FOXC1 transactivity. This work ties both proteins into a common pathway and offers an explanation of why increased FOXC1 gene dosage produces a phenotype resembling that of PITX2 deletions and mutations. Ocular phenotypes arise despite the deregulated expression of FOXC1-target genes through mutations in FOXC1 or PITX2. Ultimately, PITX2 loss of function mutations have a compound effect: the reduced expression of PITX2-target genes coupled with the extensive activation of FOXC1-regulated targets. Our findings indicate that the functional interaction between FOXC1 and PITX2A underlies the sensitivity to FOXC1 gene dosage in Axenfeld-Rieger syndrome and related anterior segment dysgeneses.
Collapse
Affiliation(s)
- Fred B Berry
- Department of Ophthalmology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7.
| | | | | | | | | | | | | |
Collapse
|
223
|
Zhang HZ, Degar BA, Rogoulina S, Resor C, Booth CJ, Sinning J, Gage PJ, Forget BG. Hematopoiesis following disruption of the Pitx2 homeodomain gene. Exp Hematol 2006; 34:167-78. [PMID: 16459185 DOI: 10.1016/j.exphem.2005.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2005] [Revised: 10/14/2005] [Accepted: 11/03/2005] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Study the effect of loss of expression of Pitx2, a homeodomain gene preferentially expressed in murine hematopoietic stem/progenitor cells, on hematopoietic stem cells (HSCs). METHODS We examined the fetal livers of mouse embryos with homozygous disruption of the Pitx2 gene, using flow cytometry immunophenotyping analysis, as well as immunohistochemistry techniques. We further investigated the role of Pitx2 in HSCs using a chimeric mouse model system. Pitx2 null embryonic stem (ES) cell clones were generated from embryonic day 3.5 blastocysts of Pitx2 null embryos. The Pitx2 null donor ES cell contribution to the adult hematopoietic system was confirmed by identifying donor-specific glucose-phosphate isomerase isotype in the erythrocytes using cellulose acetate eletrophoresis, and by demonstrating donor-specific major histocompatibility complex antigen allotype on the granulocytes/monocytes and T and B lymphocytes of the chimeric mice using flow cytometry analysis. RESULTS Pitx2 homozygous null fetal livers are decreased in size and overall cellularity. The erythroid cell component of these livers is further reduced as compared to that of their wild-type and heterozygous littermates. Detailed quantitative analysis of the chimeric mice revealed contribution of Pitx2 null ES cells to erythroid, myeloid, lymphoid, and megakaryocytic lineages. The quantitative level of ES cell contribution to the peripheral hematopoietic cells was proportional to the level of general chimerism as determined by coat color. CONCLUSION Although the fetal livers of Pitx2 null embryos displayed signs of impaired erythropoiesis, Pitx2 gene disrupted HSCs can contribute to hematopoiesis under physiological conditions.
Collapse
Affiliation(s)
- Hui Z Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06520-8021, USA
| | | | | | | | | | | | | | | |
Collapse
|
224
|
Kieusseian A, Chagraoui J, Kerdudo C, Mangeot PE, Gage PJ, Navarro N, Izac B, Uzan G, Forget BG, Dubart-Kupperschmitt A. Expression of Pitx2 in stromal cells is required for normal hematopoiesis. Blood 2006; 107:492-500. [PMID: 16195330 PMCID: PMC1895608 DOI: 10.1182/blood-2005-02-0529] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Accepted: 07/20/2005] [Indexed: 11/20/2022] Open
Abstract
Although the expression of Pitx2, a bicoid family homeodomain transcription factor, is highly regulated during hematopoiesis, its function during this process was not documented; we thus studied hematopoiesis in Pitx2-null mice. We found that Pitx2(-/-) embryos display hypoplastic livers with reduced numbers of hematopoietic cells, but these cells had normal hematopoietic potential, as evidenced by colony-forming assays, immature progenitor cell assays, and long-term repopulation assays. Because the microenvironment is also crucial to the development of normal hematopoiesis, we established Pitx2(-/-) and Pitx2(+/+) stromas from fetal liver and studied their hematopoietic supportive capacity. We showed that the frequency of cobblestone area-forming cells was 4-fold decreased when using Pitx2(-/-) stromal cells compared with Pitx2(+/+) stromal cells, whatever the Pitx2 genotype of hematopoietic cells tested in this assay. This defect was rescued by expression of Pitx2 into Pitx2(-/-) fetal liver stromal cells, demonstrating a major and direct role of Pitx2 in the hematopoietic supportive capacity of fetal liver stroma. Finally, we showed a reduced capacity of MS5 stromal cells expressing Pitx2 RNAi to support human hematopoiesis. Altogether these data showed that Pitx2 has major functions in the hematopoietic supportive capacity of fetal liver and adult bone marrow stromal cells.
Collapse
|
225
|
Peeters H, Devriendt K. Human laterality disorders. Eur J Med Genet 2006; 49:349-62. [PMID: 16461029 DOI: 10.1016/j.ejmg.2005.12.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Accepted: 12/07/2005] [Indexed: 12/12/2022]
Abstract
Heterotaxia is a group of congenital disorders characterized by a misplacement of one or more organs according to the left-right axis. Bilateral asymmetry of internal organs is conserved among all vertebrate species. Analyses in animal models such as mouse, chicken, frog and zebrafish allowed for a remarkable progress of knowledge on the embryonic and genetic mechanisms underlying internal left-right asymmetry. In this review we focus on the insights from these model organisms that are useful for a better understanding of the etiology and pathogenesis of human heterotaxia. The known causes of human heterotaxia are reviewed and situated within the conceptual framework that originates from vertebrate model organisms. Furthermore, we attempt to apply the rapidly increasing insights gained from both animal models and human genetics to clinical practice in order to contribute to a more accurate conceptual classification, genetic diagnosis and counseling.
Collapse
Affiliation(s)
- Hilde Peeters
- Department of Human Genetics, University Hospital Gasthuisberg, University of Leuven, Herestraat 49, B-3000 Leuven, Belgium.
| | | |
Collapse
|
226
|
Abstract
Extraocular muscle is fundamentally distinct from other skeletal muscles. Here, we review the biological organization of the extraocular muscles with the intent of understanding this novel muscle group in the context of oculomotor system function. The specific objectives of this review are threefold. The first objective is to understand the anatomic arrangement of the extraocular muscles and their compartmental or layered organization in the context of a new concept of orbital mechanics, the active pulley hypothesis. The second objective is to present an integrated view of the morphologic, cellular, and molecular differences between extraocular and the more traditional skeletal muscles. The third objective is to relate recent data from functional and molecular biology studies to the established extraocular muscle fiber types. Developmental mechanisms that may be responsible for the divergence of the eye muscles from a skeletal muscle prototype also are considered. Taken together, a multidisciplinary understanding of extraocular muscle biology in health and disease provides insights into oculomotor system function and malfunction. Moreover, because the eye muscles are selectively involved or spared in a variety of neuromuscular diseases, knowledge of their biology may improve current pathogenic models of and treatments for devastating systemic diseases.
Collapse
Affiliation(s)
- Robert F Spencer
- Department of Anatomy, Medical College of Virginia, Richmond, VA 23298, USA
| | | |
Collapse
|
227
|
Abstract
Unraveling the complex tissue interactions necessary to generate the structural and functional diversity present among craniofacial muscles is challenging. These muscles initiate their development within a mesenchymal population bounded by the brain, pharyngeal endoderm, surface ectoderm, and neural crest cells. This set of spatial relations, and in particular the segmental properties of these adjacent tissues, are unique to the head. Additionally, the lack of early epithelialization in head mesoderm necessitates strategies for generating discrete myogenic foci that may differ from those operating in the trunk. Molecular data indeed indicate dissimilar methods of regulation, yet transplantation studies suggest that some head and trunk myogenic populations are interchangeable. The first goal of this review is to present key features of these diversities, identifying and comparing tissue and molecular interactions regulating myogenesis in the head and trunk. Our second focus is on the diverse morphogenetic movements exhibited by craniofacial muscles. Precursors of tongue muscles partly mimic migrations of appendicular myoblasts, whereas myoblasts destined to form extraocular muscles condense within paraxial mesoderm, then as large cohorts they cross the mesoderm:neural crest interface en route to periocular regions. Branchial muscle precursors exhibit yet another strategy, establishing contacts with neural crest populations before branchial arch formation and maintaining these relations through subsequent stages of morphogenesis. With many of the prerequisite stepping-stones in our knowledge of craniofacial myogenesis now in place, discovering the cellular and molecular interactions necessary to initiate and sustain the differentiation and morphogenesis of these neglected craniofacial muscles is now an attainable goal.
Collapse
Affiliation(s)
- Drew M Noden
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA.
| | | |
Collapse
|
228
|
Ramsdell AF. Left–right asymmetry and congenital cardiac defects: Getting to the heart of the matter in vertebrate left–right axis determination. Dev Biol 2005; 288:1-20. [PMID: 16289136 DOI: 10.1016/j.ydbio.2005.07.038] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2005] [Revised: 07/21/2005] [Accepted: 07/26/2005] [Indexed: 01/20/2023]
Abstract
Cellular and molecular left-right differences that are present in the mesodermal heart fields suggest that the heart is lateralized from its inception. Left-right asymmetry persists as the heart fields coalesce to form the primary heart tube, and overt, morphological asymmetry first becomes evident when the heart tube undergoes looping morphogenesis. Thereafter, chamber formation, differentiation of the inflow and outflow tracts, and position of the heart relative to the midline are additional features of heart development that exhibit left-right differences. Observations made in human clinical studies and in animal models of laterality disease suggest that all of these features of cardiac development are influenced by the embryonic left-right body axis. When errors in left-right axis determination happen, they almost always are associated with complex congenital heart malformations. The purpose of this review is to highlight what is presently known about cardiac development and upstream processes of left-right axis determination, and to consider how perturbation of the left-right body plan might ultimately result in particular types of congenital heart defects.
Collapse
Affiliation(s)
- Ann F Ramsdell
- Department of Cell and Developmental Biology and Anatomy, School of Medicine and Program in Women's Studies, College of Arts and Sciences, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
229
|
Porter JD, Israel S, Gong B, Merriam AP, Feuerman J, Khanna S, Kaminski HJ. Distinctive morphological and gene/protein expression signatures during myogenesis in novel cell lines from extraocular and hindlimb muscle. Physiol Genomics 2005; 24:264-75. [PMID: 16291736 DOI: 10.1152/physiolgenomics.00234.2004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscles are not created equal. The underutilized concept of muscle allotypes defines distinct muscle groups that differ in their intrinsic capacity to express novel traits when exposed to a facilitating extrinsic environment. Allotype-specific traits may have significance as determinants of the preferential involvement or sparing of muscle groups that is observed in a variety of neuromuscular diseases. Little is known, however, of the developmental mechanisms underlying the distinctive skeletal muscle allotypes. The lack of appropriate in vitro models, to dissociate the cell-autonomous and non-cell-autonomous mechanisms behind allotype diversity, has been a barrier to such studies. Here, we derived novel cell lines from the extraocular and hindlimb muscle allotypes and assessed their similarities and differences during early myogenesis using morphological and gene/protein expression profiling tools. Our data establish that there are fundamental differences in the transcriptional and cellular signaling pathways used by the two myoblast lineages. Taken together, these data show that myoblast lineage plays a significant role in the divergence of the distinctive muscle groups or allotypes.
Collapse
Affiliation(s)
- John D Porter
- Department of Neurology, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, Ohio, USA.
| | | | | | | | | | | | | |
Collapse
|
230
|
Evans AL, Gage PJ. Expression of the homeobox gene Pitx2 in neural crest is required for optic stalk and ocular anterior segment development. Hum Mol Genet 2005; 14:3347-59. [PMID: 16203745 DOI: 10.1093/hmg/ddi365] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Heterozygous mutations in the homeobox gene, PITX2, result in ocular anterior segment defects and a high incidence of early-onset glaucoma. Pitx2 is expressed in both the neural crest and the mesoderm-derived precursors of the periocular mesenchyme. Complete loss of function in mice results in agenesis or severe disruption of periocular mesenchyme structures and extrinsic defects in early optic nerve development. However, the specific requirements for Pitx2 in neural crest versus mesoderm could not be determined using these mice, and only roles in the initial stages of eye development could be assessed due to early embryonic lethality. To determine the specific roles of Pitx2 in the neural crest precursor pool, we generated neural crest-specific Pitx2 knockout mice (Pitx2-ncko). Because Pitx2-nkco mice are viable, we also analyzed gene function in later eye development. Pitx2 is intrinsically required in neural crest for specification of corneal endothelium, corneal stroma and the sclera. Pitx2 function in neural crest is also required for normal development of ocular blood vessels. Pitx2-ncko mice exhibit a unique optic nerve phenotype in which the eyes are progressively displaced towards the midline until they are directly attached to the ventral hypothalamus. As Pitx2 is not expressed in the optic stalk, an essential function of PITX2 protein in neural crest is to regulate an extrinsic factor(s) required for development of the optic nerve. We propose a revised model of optic nerve development and new mechanisms that may underlie the etiology of glaucoma in Axenfeld-Rieger patients.
Collapse
Affiliation(s)
- Amanda L Evans
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, 48109, USA
| | | |
Collapse
|
231
|
Wei Q. Pitx2a binds to human papillomavirus type 18 E6 protein and inhibits E6-mediated P53 degradation in HeLa cells. J Biol Chem 2005; 280:37790-7. [PMID: 16129685 PMCID: PMC1479768 DOI: 10.1074/jbc.m502974200] [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: 11/06/2022] Open
Abstract
Binding of high risk human papillomavirus (HPV) E6 protein to E6-associated protein (E6AP), a cellular ubiquitin-protein ligase, enables E6AP to ubiquitinate p53, leading to p53 degradation in cervical cancer cells such as HeLa cells. Here we report that Pitx2a, a bicoid-type homeodomain transcription factor, can bind to HPV E6 protein and inhibit E6/E6AP-mediated p53 degradation. Deletion of the Pitx2a homeodomain abrogates its ability to bind to HPV E6 protein and to induce p53 accumulation in HeLa cells, suggesting that the homeodomain of Pitx2a is essential for inhibition of E6/E6AP-mediated p53 degradation. Recombinant Pitx2a can also block E6/E6AP-mediated p53 degradation in vitro, indicating that this function of Pitx2a is independent of its transcription activity. Pitx2a does not regulate Hdm2-mediated p53 degradation, because Pitx2a does not affect p53 protein levels in HPV-negative cells, such as HCT116, U2OS, and C33A cells. In addition, Pitx2a-induced p53 is transcriptionally active and maintains its specific DNA binding activity in HeLa cells. Taken together, these findings suggest that, by binding to E6, Pitx2a interferes with E6/E6AP-mediated p53 degradation, leading to the accumulation of functional p53 protein in HeLa cells.
Collapse
Affiliation(s)
- Qize Wei
- Laboratory of Molecular Cardiology, NHLBI, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
232
|
Laughton KW, Fisher KRS, Halina WG, Partlow GD. Schistosomus Reflexus Syndrome: A Heritable Defect in Ruminants. Anat Histol Embryol 2005; 34:312-8. [PMID: 16159373 DOI: 10.1111/j.1439-0264.2005.00624.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Schistosomus reflexus (SR) is a rare and fatal congenital disorder. Primarily observed in ruminants, its defining features include spinal inversion, exposure of the abdominal viscera because of a fissure of the ventral abdominal wall, limb ankylosis, positioning of the limbs adjacent to the skull and, lung and diaphragm hypoplasia. Variable components of SR include scoliosis, cleft sternum, exposure of thoracic viscera, and abnormalities of the digestive and urogenital systems. This report presents the findings from an anatomical analysis of a female Holstein SR calf with thoracoschisis, scoliosis and anomalies of the appendicular skeleton, cardiovascular, respiratory, digestive and urogenital systems. Many of these malformations have not been previously reported. The reproductive tract of this case is particularly unique, displaying Muellerian duct abnormalities. These abnormalities suggest SR occurs as early as the post-gastrulation embryo and involves the intermediate mesoderm. Preliminary analysis of associated cases suggests that SR has a genetic aetiology.
Collapse
Affiliation(s)
- K W Laughton
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | | | | | | |
Collapse
|
233
|
Charles MA, Suh H, Hjalt TA, Drouin J, Camper SA, Gage PJ. PITX genes are required for cell survival and Lhx3 activation. Mol Endocrinol 2005; 19:1893-903. [PMID: 15761027 DOI: 10.1210/me.2005-0052] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The PITX family of transcription factors regulate the development of many organs. Pitx1 mutants have a mild pituitary phenotype, but Pitx2 is necessary for the development of Rathke's pouch, expression of essential transcription factors in gonadotropes, and expansion of the Pit1 lineage. We report that lack of Pitx2 causes the pouch to undergo excessive cell death, resulting in severe pituitary hypoplasia. Transgenic overexpression of PITX2 in the pituitary can increase the gonadotrope population, suggesting that the absolute concentration of PITX2 is important for normal pituitary cell lineage expansion. We show that PITX1 and PITX2 proteins are present in similar expression patterns throughout pituitary development and in the mature pituitary. Both transcription factors are preferentially expressed in adult gonadotropes and thyrotropes, suggesting the possibility of overlap in maintenance of adult pituitary functions within these cell types. Double knockouts of Pitx1 and Pitx2 exhibit severe pituitary hypoplasia and fail to express the transcription factor LHX3. This indicates that these PITX genes are upstream of Lhx3 and have compensatory roles during development. Thus, the combined dosage of these PITX family members is vital for pituitary development, and their persistent coexpression in the adult pituitary suggests a continued role in maintenance of pituitary function.
Collapse
Affiliation(s)
- Michael A Charles
- 4301 MSRB III, 1500 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, MI 48109-0638, USA
| | | | | | | | | | | |
Collapse
|
234
|
Zhu X, Lin CR, Prefontaine GG, Tollkuhn J, Rosenfeld MG. Genetic control of pituitary development and hypopituitarism. Curr Opin Genet Dev 2005; 15:332-40. [PMID: 15917210 DOI: 10.1016/j.gde.2005.04.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2005] [Accepted: 04/12/2005] [Indexed: 10/25/2022]
Abstract
The pituitary gland functions as a relay between the hypothalamus and peripheral target organs that regulate basic physiological functions, including growth, the stress response, reproduction, metabolism and lactation. The development of the pituitary gland has been studied extensively in mice, and has begun to be explored in zebrafish, an animal model system amenable to forward genetics. Multiple signaling molecules and transcription factors, expressed in overlapping but distinct spatial and temporal patterns, are required at various stages of pituitary development. Defects in this precisely regulated genetic program lead to diverse pituitary dysfunction. The animal models have greatly enhanced our understanding of molecular mechanisms underlying pituitary development in addition to congenital pituitary disorders in humans.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
235
|
Muncke N, Niesler B, Roeth R, Schön K, Rüdiger HJ, Goldmuntz E, Goodship J, Rappold G. Mutational analysis of the PITX2 coding region revealed no common cause for transposition of the great arteries (dTGA). BMC MEDICAL GENETICS 2005; 6:20. [PMID: 15890066 PMCID: PMC1142516 DOI: 10.1186/1471-2350-6-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Accepted: 05/12/2005] [Indexed: 11/25/2022]
Abstract
BACKGROUND PITX2 is a bicoid-related homeodomain transcription factor that plays an important role in asymmetric cardiogenesis. Loss of function experiments in mice cause severe heart malformations, including transposition of the great arteries (TGA). TGA accounts for 5-7% of all congenital heart diseases affecting 0.2 per 1000 live births, thereby representing the most frequent cyanotic heart defect diagnosed in the neonatal period. METHODS To address whether altered PITX2 function could also contribute to the formation of dTGA in humans, we screened 96 patients with dTGA by means of dHPLC and direct sequencing for mutations within the PITX2 gene. RESULTS Several SNPs could be detected, but no stop or frame shift mutation. In particular, we found seven intronic and UTR variants, two silent mutations and two polymorphisms within the coding region. CONCLUSION As most sequence variants were also found in controls we conclude that mutations in PITX2 are not a common cause of dTGA.
Collapse
Affiliation(s)
- Nadja Muncke
- Institut für Humangenetik, Universität Heidelberg, INF 366, 69120 Heidelberg, Germany
| | - Beate Niesler
- Institut für Humangenetik, Universität Heidelberg, INF 366, 69120 Heidelberg, Germany
| | - Ralph Roeth
- Institut für Humangenetik, Universität Heidelberg, INF 366, 69120 Heidelberg, Germany
| | - Karin Schön
- Institut für Humangenetik, Universität Heidelberg, INF 366, 69120 Heidelberg, Germany
| | - Heinz-Juergen Rüdiger
- Abteilung für Kardiologie, Kinderklinik Heidelberg, INF 153, 69120 Heidelberg, Germany
| | - Elizabeth Goldmuntz
- Division of Cardiology, Department of Pediatrics, University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Judith Goodship
- Institute of Human Genetics, International Center for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Gudrun Rappold
- Institut für Humangenetik, Universität Heidelberg, INF 366, 69120 Heidelberg, Germany
| |
Collapse
|
236
|
Philips ST, Albin RL, Martin DM. Genetics of subthalamic nucleus in development and disease. Exp Neurol 2005; 192:320-30. [PMID: 15755549 DOI: 10.1016/j.expneurol.2004.11.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Revised: 11/05/2004] [Accepted: 11/09/2004] [Indexed: 11/16/2022]
Abstract
The subthalamic nucleus (STN) is a crucial node in the basal ganglia. Clinical success in targeting the STN for deep brain stimulation in Parkinson's disease patients has prompted increased interest in understanding STN biology. In this report, we discuss recent evidence for transcription factor mediated regulation of STN development. We also review STN developmental neurobiology and known patterns of gene expression in the developing and mature STN.
Collapse
Affiliation(s)
- Steven T Philips
- Department of Pediatrics, The University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
237
|
Person AD, Garriock RJ, Krieg PA, Runyan RB, Klewer SE. Frzb modulates Wnt-9a-mediated beta-catenin signaling during avian atrioventricular cardiac cushion development. Dev Biol 2005; 278:35-48. [PMID: 15649459 DOI: 10.1016/j.ydbio.2004.10.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2004] [Revised: 10/08/2004] [Accepted: 10/21/2004] [Indexed: 10/26/2022]
Abstract
Normal development of the cardiac atrioventricular (AV) endocardial cushions is essential for proper ventricular septation and morphogenesis of the mature mitral and tricuspid valves. In this study, we demonstrate spatially restricted expression of both Wnt-9a (formerly Wnt-14) and the secreted Wnt antagonist Frzb in AV endocardial cushions of the developing chicken heart. Wnt-9a expression is detected only in AV canal endocardial cells, while Frzb expression is detected in both endocardial and transformed mesenchymal cells of the developing AV cardiac cushions. We present evidence that Wnt-9a promotes cell proliferation in the AV canal and overexpression of Wnt-9a in ovo results in enlarged endocardial cushions and AV inlet obstruction. Wnt-9a stimulates beta-catenin-responsive transcription in AV canal cells, duplicates the embryonic axis upon ventral injections in Xenopus embryos and appears to regulate cell proliferation by activating a Wnt/beta-catenin signaling pathway. Additional functional studies reveal that Frzb inhibits Wnt-9a-mediated cell proliferation in cardiac cushions. Together, these data argue that Wnt-9a and Frzb regulate mesenchymal cell proliferation leading to proper AV canal cushion outgrowth and remodeling in the developing avian heart.
Collapse
Affiliation(s)
- Anthony D Person
- Department of Cell Biology and Anatomy, University Medical Center, University of Arizona School of Medicine, 1501 N. Campbell Avenue, PO Box 245044, Tucson, AZ 85724, USA.
| | | | | | | | | |
Collapse
|
238
|
Zhu X, Rosenfeld MG. Transcriptional control of precursor proliferation in the early phases of pituitary development. Curr Opin Genet Dev 2005; 14:567-74. [PMID: 15380249 DOI: 10.1016/j.gde.2004.08.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The anterior pituitary is derived from Rathke's pouch arising from the oral ectoderm. The initial apparently uniform precursor cells proliferate and differentiate into six different cell types that are present in mature gland by integrative interactions between different signaling molecules and transcription factors. This system provides an opportunity to understand gene regulation in the cellular processes of precursor cell proliferation, determination, and differentiation events during organogenesis. Recent studies have made significant advances in our appreciation of the molecular mechanisms by which transcription factors regulate these cellular processes.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA.
| | | |
Collapse
|
239
|
Brewer S, Williams T. Finally, a sense of closure? Animal models of human ventral body wall defects. Bioessays 2005; 26:1307-21. [PMID: 15551266 DOI: 10.1002/bies.20137] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Malformations concerning the ventral body wall constitute one of the leading categories of human birth defects and are present in about one out of every 2000 live births. Although the occurrence of these defects is relatively common, few detailed experimental studies exist on the development and closure of the ventral body wall in mouse and human. This field is further complicated by the array of theories on the pathogenesis of body wall defects and the likelihood that there is no single cause for these abnormalities. In this review, we summarize what is known concerning the mechanisms of normal ventral body wall closure in humans and mice. We then outline the theories that have been proposed concerning human body wall closure abnormalities and examine the growing number of mouse mutations that impact normal ventral body wall closure. Finally, we speculate how studies in animal models such as mouse and Drosophila are beginning to provide a much-needed mechanistic framework with which to identify and characterize the genes and tissues required for this vital aspect of human embryogenesis.
Collapse
Affiliation(s)
- Stephanie Brewer
- Department of Craniofacial Biology and Cell and Developmental Biology, University of Colorado Health Sciences Center, 12801 East 17th Avenue, Denver, CO 80045, USA
| | | |
Collapse
|
240
|
Person AD, Klewer SE, Runyan RB. Cell Biology of Cardiac Cushion Development. INTERNATIONAL REVIEW OF CYTOLOGY 2005; 243:287-335. [PMID: 15797462 DOI: 10.1016/s0074-7696(05)43005-3] [Citation(s) in RCA: 269] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The valves of the heart develop in the embryo from precursor structures called endocardial cushions. After cardiac looping, endocardial cushion swellings form and become populated by valve precursor cells formed by an epithelial-mesenchymal transition (EMT). Endocardial cushions subsequently undergo directed growth and remodeling to form the valvular structures and the membranous septa of the mature heart. The developmental processes that mediate cushion formation include many prototypic cellular actions including adhesion, signaling, migration, secretion, replication, differentiation, and apoptosis. Cushion morphogenesis is unique in that these cellular possesses occur in a functioning organ where the cushions act as valves even while developing into definitive valvular structures. Cardiovascular defects are the most common congenital defects, and one of the most common causes of death during infancy. Thus, there is significant interest in understanding the mechanisms that underlie this complex developmental process. In this regard, substantial progress has been made by incorporating an understanding of cardiac morphology and cell biology with the rapidly expanding repertoire of molecular mechanisms gained through human genetics and research using animal models. This article reviews cardiac morphogenesis as it relates to heart valve formation and highlights selected growth factors, intracellular signaling mediators, and extracellular matrix components involved in the creation and remodeling of endocardial cushions into mature cardiac structures.
Collapse
Affiliation(s)
- Anthony D Person
- Department of Cell Biology and Anatomy, University of Arizona School of Medicine, Tucson, Arizona 85724, USA
| | | | | |
Collapse
|
241
|
Shi X, Bosenko DV, Zinkevich NS, Foley S, Hyde DR, Semina EV, Vihtelic TS. Zebrafish pitx3 is necessary for normal lens and retinal development. Mech Dev 2004; 122:513-27. [PMID: 15804565 DOI: 10.1016/j.mod.2004.11.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2004] [Revised: 11/11/2004] [Accepted: 11/19/2004] [Indexed: 01/23/2023]
Abstract
The human PITX3 gene encodes a bicoid-like homeodomain transcription factor associated with a variety of congenital ocular conditions, including anterior segment dysgenesis, Peter's anomaly, and cataracts. We identified a zebrafish pitx3 gene encoding a protein (Pitx3) that possesses 63% amino acid identity with human PITX3. The zebrafish pitx3 gene encompasses approximately 16.5kb on chromosome 13 and consists of four exons, which is similar to the genomic organization of other pitx genes. Expression of the zebrafish pitx3 gene was studied by in situ mRNA hybridization and RT-PCR. The pitx3 transcripts were detected throughout development with the greatest level of expression occurring in the developing lens and brain at 24hpf. In adults, the highest expression was detected in the eye. Morpholinos were used to knockdown expression of the Pitx3 protein and a control morpholino that contains five mismatched bases was used to confirm the specificity of the phenotypes. The morphants had small eyes, misshapen heads and reduced jaws and fins relative to controls. The morphants exhibited abnormalities in lens development and their retinas contained pyknotic nuclei accompanied by a reduction in the number of cells in different neuronal classes. This suggests the lens is required for retinal development or Pitx3 has an unexpected role in retinal cell differentiation or survival. These results demonstrate zebrafish pitx3 represents a true ortholog of the human PITX3 gene and the general function of the Pitx3 protein in lens development is conserved between mammals and the teleost fish.
Collapse
Affiliation(s)
- Xiaohai Shi
- Department of Biological Sciences, Center for Zebrafish Research, Galvin Life Sciences Center, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | | | | | | | |
Collapse
|
242
|
Kishigami S, Yoshikawa SI, Castranio T, Okazaki K, Furuta Y, Mishina Y. BMP signaling through ACVRI is required for left–right patterning in the early mouse embryo. Dev Biol 2004; 276:185-93. [PMID: 15531373 DOI: 10.1016/j.ydbio.2004.08.042] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2003] [Revised: 07/07/2004] [Accepted: 08/20/2004] [Indexed: 12/01/2022]
Abstract
Vertebrate organisms are characterized by dorsal-ventral and left-right asymmetry. The process that establishes left-right asymmetry during vertebrate development involves bone morphogenetic protein (BMP)-dependent signaling, but the molecular details of this signaling pathway remain poorly defined. This study tests the role of the BMP type I receptor ACVRI in establishing left-right asymmetry in chimeric mouse embryos. Mouse embryonic stem (ES) cells with a homozygous deletion at Acvr1 were used to generate chimeric embryos. Chimeric embryos were rescued from the gastrulation defect of Acvr1 null embryos but exhibited abnormal heart looping and embryonic turning. High mutant contribution chimeras expressed left-side markers such as nodal bilaterally in the lateral plate mesoderm (LPM), indicating that loss of ACVRI signaling leads to left isomerism. Expression of lefty1 was absent in the midline of chimeric embryos, but shh, a midline marker, was expressed normally, suggesting that, despite formation of midline, its barrier function was abolished. High-contribution chimeras also lacked asymmetric expression of nodal in the node. These data suggest that ACVRI signaling negatively regulates left-side determinants such as nodal and positively regulates lefty1. These functions maintain the midline, restrict expression of left-side markers, and are required for left-right pattern formation during embryogenesis in the mouse.
Collapse
Affiliation(s)
- Satoshi Kishigami
- Molecular Developmental Biology Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | |
Collapse
|
243
|
Golding JP, Partridge TA, Beauchamp JR, King T, Brown NA, Gassmann M, Zammit PS. Mouse myotomes pairs exhibit left-right asymmetric expression of MLC3F and alpha-skeletal actin. Dev Dyn 2004; 231:795-800. [PMID: 15499557 DOI: 10.1002/dvdy.20176] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Most muscle originates from the myotomal compartment of the somites, paired structures flanking the neural tube. Whereas vertebrate embryos show molecular and morphological asymmetry about the left-right body axis, somitic myogenesis is thought to occur symmetrically. Here, we provide the first evidence that myotome pairs are transiently left-right asymmetric, with higher expression of alpha-skeletal actin and myosin light chain 3F (MLC3F) on the left side between embryonic day 9.5-10.25. In iv mutants with situs inversus, the asymmetric expression of alpha-skeletal actin and MLC3F was inverted, showing that this process is regulated by global left-right axis cues, initiated before gastrulation. However, although left-sided identity is later maintained by Pitx2 genes, we found that Pitx2c null embryos have normal left-biased expression of alpha-skeletal actin and MLC3F. Myotome asymmetry, therefore, is downstream of the iv mutation but upstream of, or unrelated to, the Pitx2c pathway.
Collapse
Affiliation(s)
- Jon P Golding
- Muscle Cell Biology Group, Medical Research Council Clinical Sciences Centre, London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
244
|
Yamada R, Mizutani-Koseki Y, Koseki H, Takahashi N. Requirement for Mab21l2 during development of murine retina and ventral body wall. Dev Biol 2004; 274:295-307. [PMID: 15385160 DOI: 10.1016/j.ydbio.2004.07.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Revised: 07/16/2004] [Accepted: 07/20/2004] [Indexed: 10/26/2022]
Abstract
The mab-21 gene was first identified because of its requirement for ray identity specification in Caenorhabditis elegans. It is now known to constitute a family of genes that are highly conserved from vertebrates to invertebrates, and two homologues Mab21l1 and Mab21l2 have been identified in many species. Here we describe the generation of Mab21l2-deficient mice, which have defects in eye and body wall formation. The mutant mouse eye has a rudimentary retina, as a result of insufficient invagination of the optic vesicle due to deficient proliferation, causing the absence of lens. The defects in optic vesicle development correlate with reduced expression of Chx10, which is also required for retina development; Rx, Lhx2, and Pax6 expression is not significantly affected. We conclude that Mab21l2 expression is essential for optic vesicle growth and formation of the optic cup, its absence causing reduced expression of Chx10. Mutant mice also display abnormal extrusion of abdominal organs, defects in ventral body wall formation, resulting in death in utero at mid-gestational stage. Our results reveal that Mab21l2 plays crucial roles in retina and in ventral body wall formation.
Collapse
Affiliation(s)
- Ryuichi Yamada
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, 630-0101, Japan
| | | | | | | |
Collapse
|
245
|
Zhang M, Bolfing MF, Knowles HJ, Karnes H, Hackett BP. Foxj1 regulates asymmetric gene expression during left-right axis patterning in mice. Biochem Biophys Res Commun 2004; 324:1413-20. [PMID: 15504371 DOI: 10.1016/j.bbrc.2004.09.207] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Indexed: 10/26/2022]
Abstract
Mice with a targeted mutation of the foxj1 gene demonstrate either D- or L-looping of the embryonic cardiac tube. Foxj1 is expressed in ventral cells of the embryonic node prior to asymmetric, left-right expression of other genes. Despite an absence of 9+2 cilia in foxj1(-/-) mice, 9+0 cilia are present in the node of foxj1(-/-) embryos. In foxj1(-/-) embryos, the patterns of expression of the TGF-beta family member nodal and the homeobox family member pitx2 are randomized. No expression of the TGF-beta family member lefty-2 is observed in any foxj1(-/-) early somite stage embryos. Foxj1 thus acts early in left-right axis patterning and regulates asymmetric gene expression. This regulation does not appear to be the result of a direct interaction between Foxj1 and the genes examined.
Collapse
Affiliation(s)
- Min Zhang
- Developmental Biology Research Unit, The Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
246
|
Schubert SW, Kardash E, Khan MA, Cheusova T, Kilian K, Wegner M, Hashemolhosseini S. Interaction, cooperative promoter modulation, and renal colocalization of GCMa and Pitx2. J Biol Chem 2004; 279:50358-65. [PMID: 15385555 DOI: 10.1074/jbc.m404587200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The transcription factor GCMa is a member of a new small family of transcription factors with a conserved zinc-containing DNA-binding domain. All members of this transcription factor family play crucial roles as master regulators during development. GCMa is restricted to placenta during development and to kidney and thymus at postnatal stages. It is essential for the formation of the placental labyrinth and as a consequence for survival of the embryo from mid-embryogenesis onwards. Here, we identify Pitx transcription factors as GCMa-interacting proteins. We show that Pitx proteins interact via their conserved homeodomain with the DNA-binding domain of GCMa. As a consequence, Pitx proteins and GCMa exhibit cooperative DNA binding. Furthermore, Pitx proteins influence GCMa-dependent promoter activation in a cell-specific manner. One of the three Pitx paralogues in mice, Pitx2, is the predominant Pitx member present in the placenta and colocalizes on the cellular level with GCMa in the kidney. This is the first description of a regulatory cross-talk between a transcription factor of the GCM family and a homeodomain protein.
Collapse
Affiliation(s)
- Steffen W Schubert
- Institut für Biochemie, Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
247
|
Marques S, Borges AC, Silva AC, Freitas S, Cordenonsi M, Belo JA. The activity of the Nodal antagonist Cerl-2 in the mouse node is required for correct L/R body axis. Genes Dev 2004; 18:2342-7. [PMID: 15466485 PMCID: PMC522983 DOI: 10.1101/gad.306504] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Correct establishment of the left/right (L/R) body asymmetry in the mouse embryo requires asymmetric activation of the evolutionarily conserved Nodal signaling cascade in the left lateral plate mesoderm (L-LPM). Furthermore, the presence of Nodal in the node is essential for its own expression in the L-LPM. Here, we have characterized the function of cerl-2, a novel Nodal antagonist, which displays a unique asymmetric expression on the right side of the mouse node. cerl-2 knockout mice display multiple laterality defects including randomization of the L/R axis. These defects can be partially rescued by removing one nodal allele. Our results demonstrate that Cerl-2 plays a key role in restricting the Nodal signaling pathway toward the left side of the mouse embryo by preventing its activity in the right side.
Collapse
Affiliation(s)
- Sara Marques
- Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
| | | | | | | | | | | |
Collapse
|
248
|
Bamforth SD, Bragança J, Farthing CR, Schneider JE, Broadbent C, Michell AC, Clarke K, Neubauer S, Norris D, Brown NA, Anderson RH, Bhattacharya S. Cited2 controls left-right patterning and heart development through a Nodal-Pitx2c pathway. Nat Genet 2004; 36:1189-1196. [PMID: 15475956 DOI: 10.1038/ng1446] [Citation(s) in RCA: 161] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2004] [Accepted: 09/07/2004] [Indexed: 12/18/2022]
Abstract
Malformations of the septum, outflow tract and aortic arch are the most common congenital cardiovascular defects and occur in mice lacking Cited2, a transcriptional coactivator of TFAP2. Here we show that Cited2(-/-) mice also develop laterality defects, including right isomerism, abnormal cardiac looping and hyposplenia, which are suppressed on a mixed genetic background. Cited2(-/-) mice lack expression of the Nodal target genes Pitx2c, Nodal and Ebaf in the left lateral plate mesoderm, where they are required for establishing laterality and cardiovascular development. CITED2 and TFAP2 were detected at the Pitx2c promoter in embryonic hearts, and they activate Pitx2c transcription in transient transfection assays. We propose that an abnormal Nodal-Pitx2c pathway represents a unifying mechanism for the cardiovascular malformations observed in Cited2(-/-) mice, and that such malformations may be the sole manifestation of a laterality defect.
Collapse
Affiliation(s)
- Simon D Bamforth
- Department of Cardiovascular Medicine, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Vincent SD, Norris DP, Le Good JA, Constam DB, Robertson EJ. Asymmetric Nodal expression in the mouse is governed by the combinatorial activities of two distinct regulatory elements. Mech Dev 2004; 121:1403-15. [PMID: 15454269 DOI: 10.1016/j.mod.2004.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2004] [Revised: 05/18/2004] [Accepted: 06/02/2004] [Indexed: 11/15/2022]
Abstract
In all vertebrates, invariant left/right (L/R) positioning and organization of the internal viscera is controlled by a conserved pathway. Nodal, a member of the TGFbeta superfamily is a critical upstream component responsible for initiating L/R axis determination. Asymmetric Nodal expression in the node preceeds and foreshadows morphological L/R asymmetry. Here we address the mechanism of Nodal activation in the left LPM by studying the function of a novel enhancer element, the AIE. We show this element is exclusively active in cells of the left lateral plate mesoderm (LPM) and is not itself responding to Nodal asymmetry. To test the hypothesis that this element may initiate asymmetric Nodal expression in the LPM, we deleted it from the mouse germ line. Mice homozygous for the AIE deletion (Nodal(deltaaie/deltaaie)) show no defects. However, we find that the AIE contributes to regulating the level of asymmetric Nodal activity; analysis of transheterozygous embryos (Nodal(deltaaie/null)) shows reduced Nodal expression in the left LPM associated with a low penetrance of L/R defects. Our findings point to the existence of two independent pathways that control Nodal expression in the left LPM.
Collapse
Affiliation(s)
- Stéphane D Vincent
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA.
| | | | | | | | | |
Collapse
|
250
|
Bamforth SD, Bragança J, Farthing CR, Schneider JE, Broadbent C, Michell AC, Clarke K, Neubauer S, Norris D, Brown NA, Anderson RH, Bhattacharya S. Cited2 controls left-right patterning and heart development through a Nodal-Pitx2c pathway. Nat Genet 2004; 36:1189-1196. [DOI: https:/doi.org/10.1038/ng1446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|