Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34878091

ABSTRACT

A major feature of Saethre-Chotzen syndrome is coronal craniosynostosis, the fusion of the frontal and parietal bones at the coronal suture. It is caused by heterozygous loss-of-function mutations in either of the bHLH transcription factors TWIST1 and TCF12. Although compound heterozygous Tcf12; Twist1 mice display severe coronal synostosis, the individual role of Tcf12 had remained unexplored. Here, we show that Tcf12 controls several key processes in calvarial development, including the rate of frontal and parietal bone growth, and the boundary between sutural and osteogenic cells. Genetic analysis supports an embryonic requirement for Tcf12 in suture formation, as combined deletion of Tcf12 in embryonic neural crest and mesoderm, but not in postnatal suture mesenchyme, disrupts the coronal suture. We also detected asymmetric distribution of mesenchymal cells on opposing sides of the wild-type frontal and parietal bones, which prefigures later bone overlap at the sutures. In Tcf12 mutants, reduced asymmetry is associated with bones meeting end-on-end, possibly contributing to synostosis. Our results support embryonic requirements of Tcf12 in proper formation of the overlapping coronal suture.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Craniosynostoses/metabolism , Osteogenesis , Skull/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Craniosynostoses/embryology , Craniosynostoses/genetics , Mesenchymal Stem Cells/metabolism , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Neural Crest/metabolism , Skull/metabolism
2.
Cell ; 135(7): 1299-310, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19109898

ABSTRACT

Rats have important advantages over mice as an experimental system for physiological and pharmacological investigations. The lack of rat embryonic stem (ES) cells has restricted the availability of transgenic technologies to create genetic models in this species. Here, we show that rat ES cells can be efficiently derived, propagated, and genetically manipulated in the presence of small molecules that specifically inhibit GSK3, MEK, and FGF receptor tyrosine kinases. These rat ES cells express pluripotency markers and retain the capacity to differentiate into derivatives of all three germ layers. Most importantly, they can produce high rates of chimerism when reintroduced into early stage embryos and can transmit through the germline. Establishment of authentic rat ES cells will make possible sophisticated genetic manipulation to create models for the study of human diseases.


Subject(s)
Blastocyst/cytology , Embryonic Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Chimera , Epigenesis, Genetic , Female , Fibroblast Growth Factors/antagonists & inhibitors , Glycogen Synthase Kinases/antagonists & inhibitors , Male , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Rats , Rats, Inbred Strains , Signal Transduction
3.
Development ; 143(3): 504-15, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26718006

ABSTRACT

The role of the Hippo signaling pathway in cranial neural crest (CNC) development is poorly understood. We used the Wnt1(Cre) and Wnt1(Cre2SOR) drivers to conditionally ablate both Yap and Taz in the CNC of mice. When using either Cre driver, Yap and Taz deficiency in the CNC resulted in enlarged, hemorrhaging branchial arch blood vessels and hydrocephalus. However, Wnt1(Cre2SOR) mutants had an open cranial neural tube phenotype that was not evident in Wnt1(Cre) mutants. In O9-1 CNC cells, the loss of Yap impaired smooth muscle cell differentiation. RNA-sequencing data indicated that Yap and Taz regulate genes encoding Fox transcription factors, specifically Foxc1. Proliferation was reduced in the branchial arch mesenchyme of Yap and Taz CNC conditional knockout (CKO) embryos. Moreover, Yap and Taz CKO embryos had cerebellar aplasia similar to Dandy-Walker spectrum malformations observed in human patients and mouse embryos with mutations in Foxc1. In embryos and O9-1 cells deficient for Yap and Taz, Foxc1 expression was significantly reduced. Analysis of Foxc1 regulatory regions revealed a conserved recognition element for the Yap and Taz DNA binding co-factor Tead. ChIP-PCR experiments supported the conclusion that Foxc1 is directly regulated by the Yap-Tead complex. Our findings uncover important roles for Yap and Taz in CNC diversification and development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Face/embryology , Neural Crest/embryology , Phosphoproteins/metabolism , Skull/embryology , Animals , Apoptosis/genetics , Cell Cycle Proteins , Cell Differentiation , Cell Proliferation , Embryo Loss/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Gene Deletion , Gene Expression Regulation, Developmental , Hemorrhage/pathology , Hydrocephalus/embryology , Hydrocephalus/pathology , Mandible/pathology , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Neural Tube Defects/pathology , Phenotype , Sequence Analysis, RNA , Signal Transduction , Trans-Activators , YAP-Signaling Proteins
4.
Nat Commun ; 15(1): 6948, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138165

ABSTRACT

Cranial sutures separate neighboring skull bones and are sites of bone growth. A key question is how osteogenic activity is controlled to promote bone growth while preventing aberrant bone fusions during skull expansion. Using single-cell transcriptomics, lineage tracing, and mutant analysis in zebrafish, we uncover key developmental transitions regulating bone formation at sutures during skull expansion. In particular, we identify a subpopulation of mesenchyme cells in the mid-suture region that upregulate a suite of genes including BMP antagonists (e.g. grem1a) and pro-angiogenic factors. Lineage tracing with grem1a:nlsEOS reveals that this mid-suture subpopulation is largely non-osteogenic. Moreover, combinatorial mutation of BMP antagonists enriched in this mid-suture subpopulation results in increased BMP signaling in the suture, misregulated bone formation, and abnormal suture morphology. These data reveal establishment of a non-osteogenic mesenchyme population in the mid-suture region that restricts bone formation through local BMP antagonism, thus ensuring proper suture morphology.


Subject(s)
Bone Morphogenetic Proteins , Cranial Sutures , Mesoderm , Osteogenesis , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/embryology , Zebrafish/genetics , Cranial Sutures/metabolism , Cranial Sutures/embryology , Cranial Sutures/growth & development , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Mesoderm/metabolism , Mesoderm/embryology , Mesoderm/cytology , Gene Expression Regulation, Developmental , Signal Transduction , Skull/embryology , Single-Cell Analysis , Mutation
5.
JBMR Plus ; 7(4): e10716, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37065628

ABSTRACT

Craniosynostosis is a congenital anomaly characterized by the premature fusion of cranial sutures. Sutures are a critical connective tissue that regulates bone growth; their aberrant fusion results in abnormal shapes of the head and face. The molecular and cellular mechanisms have been investigated for a long time, but knowledge gaps remain between genetic mutations and mechanisms of pathogenesis for craniosynostosis. We previously demonstrated that the augmentation of bone morphogenetic protein (BMP) signaling through constitutively active BMP type 1A receptor (caBmpr1a) in neural crest cells (NCCs) caused the development of premature fusion of the anterior frontal suture, leading to craniosynostosis in mice. In this study, we demonstrated that ectopic cartilage forms in sutures prior to premature fusion in caBmpr1a mice. The ectopic cartilage is subsequently replaced by bone nodules leading to premature fusion with similar but unique fusion patterns between two neural crest-specific transgenic Cre mouse lines, P0-Cre and Wnt1-Cre mice, which coincides with patterns of premature fusion in each line. Histologic and molecular analyses suggest that endochondral ossification in the affected sutures. Both in vitro and in vivo observations suggest a greater chondrogenic capacity and reduced osteogenic capability of neural crest progenitor cells in mutant lines. These results suggest that the augmentation of BMP signaling alters the cell fate of cranial NCCs toward a chondrogenic lineage to prompt endochondral ossification to prematurely fuse cranial sutures. By comparing P0-Cre;caBmpr1a and Wnt1-Cre;caBmpr1a mice at the stage of neural crest formation, we found more cell death of cranial NCCs in P0-Cre;caBmpr1a than Wnt1-Cre;caBmpr1a mice at the developing facial primordia. These findings may provide a platform for understanding why mutations of broadly expressed genes result in the premature fusion of limited sutures. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

6.
Dev Biol ; 347(2): 258-70, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20727876

ABSTRACT

The Notch pathway is crucial for a wide variety of developmental processes including the formation of tissue boundaries. That it may function in calvarial suture development and figure in the pathophysiology of craniosynostosis was suggested by the demonstration that heterozygous loss of function of JAGGED1 in humans can cause Alagille syndrome, which has craniosynostosis as a feature. We used conditional gene targeting to examine the role of Jagged1 in the development of the skull vault. We demonstrate that Jagged1 is expressed in a layer of mesoderm-derived sutural cells that lie along the osteogenic-non-osteogenic boundary. We show that inactivation of Jagged1 in the mesodermal compartment of the coronal suture, but not in the neural crest compartment, results in craniosynostosis. Mesodermal inactivation of Jagged1 also results in changes in the identity of sutural cells prior to overt osteogenic differentiation, as well as defects in the boundary between osteogenic and non-osteogenic compartments at the coronal suture. These changes, surprisingly, are associated with increased expression of Notch2 and the Notch effector, Hes1, in the sutural mesenchyme. They are also associated with an increase in nuclear ß-catenin. In Twist1 mutants, Jagged1 expression in the suture is reduced substantially, suggesting an epistatic relationship between Twist1 and Jagged1. Consistent with such a relationship, Twist1-Jagged1 double heterozygotes exhibit a substantial increase in the severity of craniosynostosis over individual heterozygotes. Our results thus suggest that Jagged1 is an effector of Twist1 in coronal suture development.


Subject(s)
Calcium-Binding Proteins/physiology , Cranial Sutures/embryology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Nuclear Proteins/physiology , Twist-Related Protein 1/physiology , Animals , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Craniosynostoses/embryology , Craniosynostoses/genetics , Epistasis, Genetic , Female , Gene Expression Regulation, Developmental , Heterozygote , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mesoderm/embryology , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Models, Biological , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Osteogenesis/genetics , Osteogenesis/physiology , Penetrance , Pregnancy , Serrate-Jagged Proteins , Twist-Related Protein 1/deficiency , Twist-Related Protein 1/genetics
7.
Dev Biol ; 343(1-2): 28-39, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20398647

ABSTRACT

In an effort to understand the morphogenetic forces that shape the bones of the skull, we inactivated Msx1 and Msx2 conditionally in neural crest. We show that Wnt1-Cre inactivation of up to three Msx1/2 alleles results in a progressively larger defect in the neural crest-derived frontal bone. Unexpectedly, in embryos lacking all four Msx1/2 alleles, the large defect is filled in with mispatterned bone consisting of ectopic islands of bone between the reduced frontal bones, just anterior to the parietal bones. The bone is derived from neural crest, not mesoderm, and, from DiI cell marking experiments, originates in a normally non-osteogenic layer of cells through which the rudiment elongates apically. Associated with the heterotopic osteogenesis is an upregulation of Bmp signaling in this cell layer. Prevention of this upregulation by implantation of noggin-soaked beads in head explants also prevented heterotopic bone formation. These results suggest that Msx genes have a dual role in calvarial development: They are required for the differentiation and proliferation of osteogenic cells within rudiments, and they are also required to suppress an osteogenic program in a cell layer within which the rudiments grow. We suggest that the inactivation of this repressive activity may be one cause of Wormian bones, ectopic bones that are a feature of a variety of pathological conditions in which calvarial bone development is compromised.


Subject(s)
Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Neural Crest/cytology , Osteogenesis/genetics , Skull/embryology , Animals , Body Patterning , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Homeodomain Proteins/metabolism , MSX1 Transcription Factor/metabolism , Mice , Mice, Inbred C57BL , Neural Crest/embryology , Neural Crest/metabolism
8.
Nat Commun ; 12(1): 4797, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376651

ABSTRACT

Sutures separate the flat bones of the skull and enable coordinated growth of the brain and overlying cranium. The coronal suture is most commonly fused in monogenic craniosynostosis, yet the unique aspects of its development remain incompletely understood. To uncover the cellular diversity within the murine embryonic coronal suture, we generated single-cell transcriptomes and performed extensive expression validation. We find distinct pre-osteoblast signatures between the bone fronts and periosteum, a ligament-like population above the suture that persists into adulthood, and a chondrogenic-like population in the dura mater underlying the suture. Lineage tracing reveals an embryonic Six2+ osteoprogenitor population that contributes to the postnatal suture mesenchyme, with these progenitors being preferentially affected in a Twist1+/-; Tcf12+/- mouse model of Saethre-Chotzen Syndrome. This single-cell atlas provides a resource for understanding the development of the coronal suture and the mechanisms for its loss in craniosynostosis.


Subject(s)
Cranial Sutures/metabolism , Gene Expression Regulation, Developmental , Osteogenesis/genetics , Single-Cell Analysis/methods , Transcriptome/genetics , Acrocephalosyndactylia/embryology , Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cranial Sutures/cytology , Cranial Sutures/embryology , Dura Mater/cytology , Dura Mater/embryology , Dura Mater/metabolism , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice, Knockout , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/metabolism , RNA-Seq/methods , Skull/cytology , Skull/embryology , Skull/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
9.
Hepatology ; 49(3): 998-1011, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19085956

ABSTRACT

UNLABELLED: The knowledge concerning fetal hepatic stellate cells (HSCs) is scarce, and their cell lineage and functions are largely unknown. The current study isolated fetal liver mesenchymal cells from a mouse expressing beta-galactosidase under the control of Msx2 promoter by fluorescence-activated cell sorting (FACS) and surveyed marker genes by microarray analysis. Based on the location and immunostaining with conventional and newly disclosed markers, we have identified three distinct populations of fetal liver mesenchymal cells expressing both desmin and p75 neurotrophin receptor (p75NTR): HSCs in the liver parenchyma; perivascular mesenchymal cells expressing alpha-smooth muscle actin (alpha-SMA); and submesothelial cells associated with the basal lamina beneath mesothelial cells and expressing activated leukocyte cell adhesion molecule (ALCAM) and platelet-derived growth factor receptor alpha. A transitional cell type from the submesothelial cell phenotype to fetal HSCs was also identified near the liver surface. Mesothelial cells expressed podoplanin and ALCAM. Ki-67 staining showed that proliferative activity of the submesothelial cells is higher than that of mesothelial cells and transitional cells. Using anti-ALCAM antibodies, submesothelial and mesothelial cells were isolated by FACS. The ALCAM(+) cells expressed hepatocyte growth factor and pleiotrophin. In culture, the ALCAM(+) cells rapidly acquired myofibroblastic morphology and alpha-SMA expression. The ALCAM(+) cells formed intracellular lipid droplets when embedded in collagen gel and treated with retinol, suggesting the potential for ALCAM(+) cells to differentiate to HSCs. Finally, we demonstrated that fetal HSCs, submesothelial cells, and perivascular mesenchymal cells are all derived from mesoderm by using MesP1-Cre and ROSA26 reporter mice. CONCLUSION: Fetal HSCs, submesothelial cells, and perivascular mesenchymal cells are mesodermal in origin, and ALCAM(+) submesothelial cells may be a precursor for HSCs in developing liver.


Subject(s)
Endothelium, Vascular/cytology , Epithelial Cells/cytology , Hepatic Stellate Cells/cytology , Liver/cytology , Liver/embryology , Mesenchymal Stem Cells/cytology , Organogenesis/physiology , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Cells, Cultured , Desmin/metabolism , Endothelium, Vascular/metabolism , Epithelial Cells/metabolism , Female , Hepatic Stellate Cells/metabolism , Homeodomain Proteins/metabolism , Lac Operon/genetics , Liver/metabolism , Male , Membrane Glycoproteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Receptors, Nerve Growth Factor/metabolism
10.
Elife ; 82019 12 23.
Article in English | MEDLINE | ID: mdl-31869306

ABSTRACT

The vertebrate skull varies widely in shape, accommodating diverse strategies of feeding and predation. The braincase is composed of several flat bones that meet at flexible joints called sutures. Nearly all vertebrates have a prominent 'coronal' suture that separates the front and back of the skull. This suture can develop entirely within mesoderm-derived tissue, neural crest-derived tissue, or at the boundary of the two. Recent paleontological findings and genetic insights in non-mammalian model organisms serve to revise fundamental knowledge on the development and evolution of this suture. Growing evidence supports a decoupling of the germ layer origins of the mesenchyme that forms the calvarial bones from inductive signaling that establishes discrete bone centers. Changes in these relationships facilitate skull evolution and may create susceptibility to disease. These concepts provide a general framework for approaching issues of homology in cases where germ layer origins have shifted during evolution.


Subject(s)
Biological Evolution , Mesoderm/growth & development , Neural Crest/growth & development , Skull/growth & development , Animals , Cranial Sutures/growth & development , Cranial Sutures/pathology , Humans , Skull/pathology
11.
BMC Dev Biol ; 8: 75, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18667074

ABSTRACT

BACKGROUND: Msx1 and Msx2, which belong to the highly conserved Nk family of homeobox genes, display overlapping expression patterns and redundant functions in multiple tissues and organs during vertebrate development. Msx1 and Msx2 have well-documented roles in mediating epithelial-mesenchymal interactions during organogenesis. Given that both Msx1 and Msx2 are crucial downstream effectors of Bmp signaling, we investigated whether Msx1 and Msx2 are required for the Bmp-induced endothelial-mesenchymal transformation (EMT) during atrioventricular (AV) valve formation. RESULTS: While both Msx1-/- and Msx2-/- single homozygous mutant mice exhibited normal valve formation, we observed hypoplastic AV cushions and malformed AV valves in Msx1-/-; Msx2-/- mutants, indicating redundant functions of Msx1 and Msx2 during AV valve morphogenesis. In Msx1/2 null mutant AV cushions, we found decreased Bmp2/4 and Notch1 signaling as well as reduced expression of Has2, NFATc1 and Notch1, demonstrating impaired endocardial activation and EMT. Moreover, perturbed expression of chamber-specific genes Anf, Tbx2, Hand1 and Hand2 reveals mispatterning of the Msx1/2 double mutant myocardium and suggests functions of Msx1 and Msx2 in regulating myocardial signals required for remodelling AV valves and maintaining an undifferentiated state of the AV myocardium. CONCLUSION: Our findings demonstrate redundant roles of Msx1 and Msx2 in regulating signals required for development of the AV myocardium and formation of the AV valves.


Subject(s)
Endocardial Cushions/embryology , Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Myocardium/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Body Patterning , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Endocardial Cushions/cytology , Endocardium/cytology , Endocardium/embryology , Gene Expression , Gene Expression Regulation, Developmental , Heart Valves/cytology , Heart Valves/embryology , Mesoderm/cytology , Mesoderm/embryology , Mice , Mice, Inbred BALB C , NFATC Transcription Factors/genetics , Organogenesis , Receptor, Notch1/genetics , Transforming Growth Factor beta/genetics
12.
Mech Dev ; 124(9-10): 729-45, 2007.
Article in English | MEDLINE | ID: mdl-17693062

ABSTRACT

The homeobox genes Msx1 and Msx2 function as transcriptional regulators that control cellular proliferation and differentiation during embryonic development. Mutations in the Msx1 and Msx2 genes in mice disrupt tissue-tissue interactions and cause multiple craniofacial malformations. Although Msx1 and Msx2 are both expressed throughout the entire development of the frontal bone, the frontal bone defect in Msx1 or Msx2 null mutants is rather mild, suggesting the possibility of functional compensation between Msx1 and Msx2 during early frontal bone development. To investigate this hypothesis, we generated Msx1(-/-);Msx2(-/-) mice. These double mutant embryos died at E17 to E18 with no formation of the frontal bone. There was no apparent defect in CNC migration into the presumptive frontal bone primordium, but differentiation of the frontal mesenchyme and establishment of the frontal primordium was defective, indicating that Msx1 and Msx2 genes are specifically required for osteogenesis in the cranial neural crest lineage within the frontal bone primordium. Mechanistically, our data suggest that Msx genes are critical for the expression of Runx2 in the frontonasal subpopulation of cranial neural crest cells and for differentiation of the osteogenic lineage. This early function of the Msx genes is likely independent of the Bmp signaling pathway.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/physiology , Frontal Bone/embryology , Homeodomain Proteins/physiology , MSX1 Transcription Factor/physiology , Neural Crest/cytology , Animals , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , DNA-Binding Proteins/genetics , Frontal Bone/cytology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Mice , Mice, Knockout , Osteogenesis/genetics
13.
J Vis Exp ; (140)2018 10 09.
Article in English | MEDLINE | ID: mdl-30371662

ABSTRACT

Neural crest cells (NCCs) are migrating multipotent stem cells that can differentiate into different cell types and give rise to multiple tissues and organs. The O9-1 cell line is derived from the endogenous mouse embryonic NCCs and maintains its multipotency. However, under specific culture conditions, O9-1 cells can differentiate into different cell types and be utilized in a wide range of research applications. Recently, with the combination of mouse studies and O9-1 cell studies, we have shown that the Hippo signaling pathway effectors Yap and Taz play important roles in neural crest-derived craniofacial development. Although the culturing process for O9-1 cells is more complicated than that used for other cell lines, the O9-1 cell line is a powerful model for investigating NCCs in vitro. Here, we present a protocol for culturing the O9-1 cell line to maintain its stemness, as well as protocols for differentiating O9-1 cells into different cell types, such as smooth muscle cells and osteoblasts. In addition, protocols are described for performing gene loss-of-function studies in O9-1 cells by using CRISPR-Cas9 deletion and small interfering RNA-mediated knockdown.


Subject(s)
Cell Culture Techniques , Multipotent Stem Cells/cytology , Neural Crest/cytology , Animals , CRISPR-Cas Systems , Cell Differentiation , Cell Line , Mice , Myocytes, Smooth Muscle , Osteoblasts , RNA, Small Interfering/genetics
14.
Elife ; 72018 10 25.
Article in English | MEDLINE | ID: mdl-30375332

ABSTRACT

Cranial sutures separate the skull bones and house stem cells for bone growth and repair. In Saethre-Chotzen syndrome, mutations in TCF12 or TWIST1 ablate a specific suture, the coronal. This suture forms at a neural-crest/mesoderm interface in mammals and a mesoderm/mesoderm interface in zebrafish. Despite this difference, we show that combinatorial loss of TCF12 and TWIST1 homologs in zebrafish also results in specific loss of the coronal suture. Sequential bone staining reveals an initial, directional acceleration of bone production in the mutant skull, with subsequent localized stalling of bone growth prefiguring coronal suture loss. Mouse genetics further reveal requirements for Twist1 and Tcf12 in both the frontal and parietal bones for suture patency, and to maintain putative progenitors in the coronal region. These findings reveal conservation of coronal suture formation despite evolutionary shifts in embryonic origins, and suggest that the coronal suture might be especially susceptible to imbalances in progenitor maintenance and osteoblast differentiation.


Subject(s)
Acrocephalosyndactylia/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Craniosynostoses/genetics , Twist-Related Protein 1/genetics , Acrocephalosyndactylia/pathology , Animals , Bone Development , Craniosynostoses/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Humans , Mice , Mutation , Neural Crest/growth & development , Neural Crest/pathology , Osteogenesis/genetics , Zebrafish/genetics
15.
Sci Rep ; 7(1): 2497, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28566723

ABSTRACT

Whereas Jagged1-Notch2 signaling is known to pattern the sensorineural components of the inner ear, its role in middle ear development has been less clear. We previously reported a role for Jagged-Notch signaling in shaping skeletal elements derived from the first two pharyngeal arches of zebrafish. Here we show a conserved requirement for Jagged1-Notch2 signaling in patterning the stapes and incus middle ear bones derived from the equivalent pharyngeal arches of mammals. Mice lacking Jagged1 or Notch2 in neural crest-derived cells (NCCs) of the pharyngeal arches display a malformed stapes. Heterozygous Jagged1 knockout mice, a model for Alagille Syndrome (AGS), also display stapes and incus defects. We find that Jagged1-Notch2 signaling functions early to pattern the stapes cartilage template, with stapes malformations correlating with hearing loss across all frequencies. We observe similar stapes defects and hearing loss in one patient with heterozygous JAGGED1 loss, and a diversity of conductive and sensorineural hearing loss in nearly half of AGS patients, many of which carry JAGGED1 mutations. Our findings reveal deep conservation of Jagged1-Notch2 signaling in patterning the pharyngeal arches from fish to mouse to man, despite the very different functions of their skeletal derivatives in jaw support and sound transduction.


Subject(s)
Alagille Syndrome/genetics , Hearing Loss, Sensorineural/genetics , Jagged-1 Protein/genetics , Receptor, Notch2/genetics , Alagille Syndrome/physiopathology , Animals , Ear, Middle/growth & development , Ear, Middle/pathology , Gene Expression Regulation, Developmental/genetics , Hearing Loss, Sensorineural/pathology , Humans , Mice , Mice, Knockout , Neural Crest/growth & development , Neural Crest/pathology , Signal Transduction/genetics
16.
Mech Dev ; 117(1-2): 115-22, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12204252

ABSTRACT

Mouse embryos lacking the retinoic acid (RA) receptors RARalpha1 and RARbeta suffer from a failure to properly septate (divide) the early outflow tract of the heart into distinct aortic and pulmonary channels, a phenotype termed persistent truncus arteriosus. This phenotype is associated with a failure in the development of the cardiac neural crest cell lineage, which normally forms the aorticopulmonary septum. In this study, we examined the fate of the neural crest lineage in RARalpha1/RARbeta mutant embryos by crossing with the Wnt1-cre and conditional R26R alleles, which together constitute a genetic lineage marker for the neural crest. We find that the number, migration, and terminal fate of the cardiac neural crest is normal in mutant embryos; however, the specific function of these cells in forming the aorticopulmonary septum is impaired. We furthermore show that the neural crest cells themselves do not utilize retinoid receptors and do not respond to RA during this process, but rather that the phenotype is cell non-autonomous for the neural crest cell lineage. This suggests that an alternative tissue in the vicinity of the outflow tract of the heart responds directly to RA, and thereby induces or permits the neural crest cell lineage to initiate aorticopulmonary septation.


Subject(s)
Fetal Heart/cytology , Fetal Heart/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Receptors, Retinoic Acid/genetics , Animals , Cell Count , Cell Movement , Fetal Heart/embryology , Gene Expression Regulation, Developmental , Gestational Age , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Mice , Mice, Knockout , Neural Crest/embryology , Phenotype , Receptors, Retinoic Acid/deficiency , Retinoic Acid Receptor alpha , Rhombencephalon/embryology , Rhombencephalon/metabolism , Truncus Arteriosus, Persistent/embryology , Truncus Arteriosus, Persistent/genetics , Truncus Arteriosus, Persistent/metabolism
17.
Curr Top Dev Biol ; 115: 131-56, 2015.
Article in English | MEDLINE | ID: mdl-26589924

ABSTRACT

The skull vault is a complex, exquisitely patterned structure that plays a variety of key roles in vertebrate life, ranging from the acquisition of food to the support of the sense organs for hearing, smell, sight, and taste. During its development, it must meet the dual challenges of protecting the brain and accommodating its growth. The bones and sutures of the skull vault are derived from cranial neural crest and head mesoderm. The frontal and parietal bones develop from osteogenic rudiments in the supraorbital ridge. The coronal suture develops from a group of Shh-responsive cells in the head mesoderm that are collocated, with the osteogenic precursors, in the supraorbital ridge. The osteogenic rudiments and the prospective coronal suture expand apically by cell migration. A number of congenital disorders affect the skull vault. Prominent among these is craniosynostosis, the fusion of the bones at the sutures. Analysis of the pathophysiology underling craniosynostosis has identified a variety of cellular mechanisms, mediated by a range of signaling pathways and effector transcription factors. These cellular mechanisms include loss of boundary integrity, altered sutural cell specification in embryos, and loss of a suture stem cell population in adults. Future work making use of genome-wide transcriptomic approaches will address the deep structure of regulatory interactions and cellular processes that unify these seemingly diverse mechanisms.


Subject(s)
Cranial Sutures/embryology , Craniosynostoses/embryology , Morphogenesis , Skull/embryology , Animals , Cranial Sutures/metabolism , Craniosynostoses/genetics , Craniosynostoses/physiopathology , Gene Expression Regulation, Developmental , Humans , Mesoderm/embryology , Mesoderm/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Signal Transduction/genetics , Skull/metabolism
18.
Invest Ophthalmol Vis Sci ; 44(6): 2404-12, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12766037

ABSTRACT

PURPOSE: Microphthalmia is a relatively common ocular malformation. Molecular mechanisms that lead to this dire condition are largely unknown. Msx genes have been shown to be expressed in the developing eye. In the Msx1;Msx2, double mutant mouse, eye development arrests early in embryogenesis. To investigate possible functions of Msx2 in early ocular development, we created transgenic animals that overexpress Msx2. METHODS: Msx2 transgenic embryos and nontransgenic littermates were examined histopathologically. The effect of Msx2 overexpression on retinal cell proliferation was assayed by bromodeoxyuridine (BrdU) incorporation and immunohistochemical staining. Apoptosis was determined by TUNEL labeling. Expression of retina and retinal pigmented epithelium (RPE)-specific genes was investigated by performing in situ hybridization or immunohistochemical staining. RESULTS: Forced expression of the Msx2 gene resulted in optic nerve aplasia and microphthalmia in all transgenic animals. In developing retinas of Msx2 transgenic animals, proliferation was significantly reduced and increased numbers of retinal cells underwent apoptosis. Marker analysis showed suppression of Bmp4 and induction of Bmp7 gene expression in the optic vesicle. Ectopic concurrent expression of the RPE cell markers Cx43 and Trp-2 in the neural retinal layer suggests cell fate respecification. CONCLUSION: These results indicate that forced expression of Msx2 perturbs BMP signaling in the developing eye and is accompanied by an increase in retinal cell death and a reduction in cell proliferation. Thus, deregulated Msx2 gene expression may be a plausible genetic mechanism by which the autosomal dominant form of congenital microphthalmia may arise.


Subject(s)
Apoptosis/genetics , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Microphthalmos/genetics , Optic Nerve/abnormalities , Retina/pathology , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Bromodeoxyuridine/metabolism , Cell Division , Female , Gene Expression , In Situ Hybridization , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Microphthalmos/metabolism , Microphthalmos/pathology , Optic Nerve/pathology , RNA, Messenger/metabolism , Retina/embryology , Retina/metabolism
19.
Nat Genet ; 45(3): 304-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23354436

ABSTRACT

Craniosynostosis, the premature fusion of the cranial sutures, is a heterogeneous disorder with a prevalence of ∼1 in 2,200 (refs. 1,2). A specific genetic etiology can be identified in ∼21% of cases, including mutations of TWIST1, which encodes a class II basic helix-loop-helix (bHLH) transcription factor, and causes Saethre-Chotzen syndrome, typically associated with coronal synostosis. Using exome sequencing, we identified 38 heterozygous TCF12 mutations in 347 samples from unrelated individuals with craniosynostosis. The mutations predominantly occurred in individuals with coronal synostosis and accounted for 32% and 10% of subjects with bilateral and unilateral pathology, respectively. TCF12 encodes one of three class I E proteins that heterodimerize with class II bHLH proteins such as TWIST1. We show that TCF12 and TWIST1 act synergistically in a transactivation assay and that mice doubly heterozygous for loss-of-function mutations in Tcf12 and Twist1 have severe coronal synostosis. Hence, the dosage of TCF12-TWIST1 heterodimers is critical for normal coronal suture development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Craniosynostoses , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics , Acrocephalosyndactylia/complications , Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/pathology , Animals , Cranial Sutures/growth & development , Cranial Sutures/pathology , Craniosynostoses/complications , Craniosynostoses/genetics , Craniosynostoses/pathology , Dimerization , Exome , Gene Expression Regulation, Developmental , Heterozygote , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Sequence Analysis, DNA , Transcriptional Activation
20.
Stem Cells Dev ; 21(17): 3069-80, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-22889333

ABSTRACT

Cranial neural crest cells give rise to ectomesenchymal derivatives such as cranial bones, cartilage, smooth muscle, dentin, as well as melanocytes, corneal endothelial cells, and neurons and glial cells of the peripheral nervous system. Previous studies have suggested that although multipotent stem-like cells may exist during the course of cranial neural crest development, they are transient, undergoing lineage restriction early in embryonic development. We have developed culture conditions that allow cranial neural crest cells to be grown as multipotent stem-like cells. With these methods, we obtained 2 independent cell lines, O9-1 and i10-1, which were derived from mass cultures of Wnt1-Cre; R26R-GFP-expressing cells. These cell lines can be propagated and passaged indefinitely, and can differentiate into osteoblasts, chondrocytes, smooth muscle cells, and glial cells. Whole-genome expression profiling of O9-1 cells revealed that this line stably expresses stem cell markers (CD44, Sca-1, and Bmi1) and neural crest markers (AP-2α, Twist1, Sox9, Myc, Ets1, Dlx1, Dlx2, Crabp1, Epha2, and Itgb1). O9-1 cells are capable of contributing to cranial mesenchymal (osteoblast and smooth muscle) neural crest fates when injected into E13.5 mouse cranial tissue explants and chicken embryos. These results suggest that O9-1 cells represent multipotent mesenchymal cranial neural crest cells. The O9-1 cell line should serve as a useful tool for investigating the molecular properties of differentiating cranial neural crest cells.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Cell Line , Neural Crest/cytology , Skull/cytology , Animals , Biomarkers/metabolism , Cell Movement , Chick Embryo , Culture Media/metabolism , Flow Cytometry , Gene Expression Profiling , Hyaluronan Receptors/metabolism , Mice , Microinjections , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Osteogenesis , Skull/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL