Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Cell ; 184(1): 243-256.e18, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33417861

ABSTRACT

Craniosynostosis results from premature fusion of the cranial suture(s), which contain mesenchymal stem cells (MSCs) that are crucial for calvarial expansion in coordination with brain growth. Infants with craniosynostosis have skull dysmorphology, increased intracranial pressure, and complications such as neurocognitive impairment that compromise quality of life. Animal models recapitulating these phenotypes are lacking, hampering development of urgently needed innovative therapies. Here, we show that Twist1+/- mice with craniosynostosis have increased intracranial pressure and neurocognitive behavioral abnormalities, recapitulating features of human Saethre-Chotzen syndrome. Using a biodegradable material combined with MSCs, we successfully regenerated a functional cranial suture that corrects skull deformity, normalizes intracranial pressure, and rescues neurocognitive behavior deficits. The regenerated suture creates a niche into which endogenous MSCs migrated, sustaining calvarial bone homeostasis and repair. MSC-based cranial suture regeneration offers a paradigm shift in treatment to reverse skull and neurocognitive abnormalities in this devastating disease.


Subject(s)
Cognition/physiology , Cranial Sutures/physiopathology , Craniosynostoses/physiopathology , Regeneration/physiology , Skull/physiopathology , Animals , Behavior, Animal/drug effects , Cognition/drug effects , Craniosynostoses/genetics , Dura Mater/pathology , Dura Mater/physiopathology , Gelatin/pharmacology , Gene Expression Profiling , Hand Strength , Intracranial Pressure/drug effects , Intracranial Pressure/physiology , Locomotion/drug effects , Mesenchymal Stem Cells/drug effects , Methacrylates/pharmacology , Mice, Inbred C57BL , Motor Activity/drug effects , Organ Size/drug effects , Regeneration/drug effects , Skull/pathology , Twist-Related Protein 1/metabolism , Wnt Signaling Pathway/drug effects
2.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38108472

ABSTRACT

Nerves play important roles in organ development and tissue homeostasis. Stem/progenitor cells differentiate into different cell lineages responsible for building the craniofacial organs. The mechanism by which nerves regulate stem/progenitor cell behavior in organ morphogenesis has not yet been comprehensively explored. Here, we use tooth root development in mouse as a model to investigate how sensory nerves regulate organogenesis. We show that sensory nerve fibers are enriched in the dental papilla at the initiation of tooth root development. Through single cell RNA-sequencing analysis of the trigeminal ganglion and developing molar, we reveal several signaling pathways that connect the sensory nerve with the developing molar, of which FGF signaling appears to be one of the important regulators. Fgfr2 is expressed in the progenitor cells during tooth root development. Loss of FGF signaling leads to shortened roots with compromised proliferation and differentiation of progenitor cells. Furthermore, Hh signaling is impaired in Gli1-CreER;Fgfr2fl/fl mice. Modulation of Hh signaling rescues the tooth root defects in these mice. Collectively, our findings elucidate the nerve-progenitor crosstalk and reveal the molecular mechanism of the FGF-SHH signaling cascade during tooth root morphogenesis.


Subject(s)
Tooth , Animals , Mice , Molar , Morphogenesis/genetics , Odontogenesis/genetics , Tooth Root
3.
Development ; 150(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36825984

ABSTRACT

Craniofacial morphogenesis requires complex interactions involving different tissues, signaling pathways, secreted factors and organelles. The details of these interactions remain elusive. In this study, we have analyzed the molecular mechanisms and homeostatic cellular activities governing soft palate development to improve regenerative strategies for individuals with cleft palate. We have identified canonical Wnt signaling as a key signaling pathway primarily active in cranial neural crest (CNC)-derived mesenchymal cells surrounding soft palatal myogenic cells. Using Osr2-Cre;ß-cateninfl/fl mice, we show that Wnt signaling is indispensable for mesenchymal cell proliferation and subsequently for myogenesis through mediating ciliogenesis. Specifically, we have identified that Wnt signaling directly regulates expression of the ciliary gene Ttll3. Impaired ciliary disassembly leads to differentiation defects in mesenchymal cells and indirectly disrupts myogenesis through decreased expression of Dlk1, a mesenchymal cell-derived pro-myogenesis factor. Moreover, we show that siRNA-mediated reduction of Ttll3 expression partly rescues mesenchymal cell proliferation and myogenesis in the palatal explant cultures from Osr2-Cre;ß-cateninfl/fl embryos. This study highlights the role of Wnt signaling in palatogenesis through the control of ciliary homeostasis, which establishes a new mechanism for Wnt-regulated craniofacial morphogenesis.


Subject(s)
Cleft Palate , Wnt Signaling Pathway , Mice , Animals , Wnt Signaling Pathway/physiology , Palate , Cleft Palate/genetics , Cell Differentiation , Palate, Soft , Homeostasis , Gene Expression Regulation, Developmental
4.
Development ; 148(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33766930

ABSTRACT

Stem cells self-renew or give rise to transit-amplifying cells (TACs) that differentiate into specific functional cell types. The fate determination of stem cells to TACs and their transition to fully differentiated progeny is precisely regulated to maintain tissue homeostasis. Arid1a, a core component of the switch/sucrose nonfermentable complex, performs epigenetic regulation of stage- and tissue-specific genes that is indispensable for stem cell homeostasis and differentiation. However, the functional mechanism of Arid1a in the fate commitment of mesenchymal stem cells (MSCs) and their progeny is not clear. Using the continuously growing adult mouse incisor model, we show that Arid1a maintains tissue homeostasis through limiting proliferation, promoting cell cycle exit and differentiation of TACs by inhibiting the Aurka-Cdk1 axis. Loss of Arid1a overactivates the Aurka-Cdk1 axis, leading to expansion of the mitotic TAC population but compromising their differentiation ability. Furthermore, the defective homeostasis after loss of Arid1a ultimately leads to reduction of the MSC population. These findings reveal the functional significance of Arid1a in regulating the fate of TACs and their interaction with MSCs to maintain tissue homeostasis.


Subject(s)
Aurora Kinase A/metabolism , CDC2 Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Incisor/embryology , Mesenchymal Stem Cells/metabolism , Mitosis , Signal Transduction , Transcription Factors/metabolism , Animals , Aurora Kinase A/genetics , CDC2 Protein Kinase/genetics , DNA-Binding Proteins/genetics , Mice , Mice, Transgenic , Transcription Factors/genetics
5.
Development ; 148(2)2021 01 21.
Article in English | MEDLINE | ID: mdl-33323370

ABSTRACT

The control of size and shape is an important part of regulatory process during organogenesis. Tooth formation is a highly complex process that fine-tunes the size and shape of the tooth, which are crucial for its physiological functions. Each tooth consists of a crown and one or more roots. Despite comprehensive knowledge of the mechanism that regulates early tooth crown development, we have limited understanding of the mechanism regulating root patterning and size during development. Here, we show that Ror2-mediated non-canonical Wnt signaling in the dental mesenchyme plays a crucial role in cell proliferation, and thereby regulates root development size in mouse molars. Furthermore, Cdc42 acts as a potential downstream mediator of Ror2 signaling in root formation. Importantly, activation of Cdc42 can restore cell proliferation and partially rescue the root development size defects in Ror2 mutant mice. Collectively, our findings provide novel insights into the function of Ror2-mediated non-canonical Wnt signaling in regulating tooth morphogenesis, and suggest potential avenues for dental tissue engineering.


Subject(s)
Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Tooth Root/embryology , Tooth Root/metabolism , Wnt Signaling Pathway , cdc42 GTP-Binding Protein/metabolism , Animals , Cell Differentiation , Cell Proliferation , Female , Male , Mesoderm/embryology , Mice , Mice, Mutant Strains , Morphogenesis , Odontoblasts/cytology , Odontoblasts/metabolism , Tooth Root/cytology
6.
PLoS Genet ; 17(2): e1009320, 2021 02.
Article in English | MEDLINE | ID: mdl-33596195

ABSTRACT

Mammalian tooth crown formation has long served as a model for investigating how patterning and morphogenesis are orchestrated during development. However, the mechanism underlying root patterning and morphogenesis remains poorly understood. In this study, we find that Lhx6 labels a subpopulation of root progenitor cells in the apical dental mesenchyme, which is closely associated with furcation development. Loss of Lhx6 leads to furcation and root number defects, indicating that Lhx6 is a key root patterning regulator. Among the multiple cellular events regulated by Lhx6 is the odontoblast fate commitment of progenitor cells, which it controls in a cell-autonomous manner. Specifically, Lhx6 loss leads to elevated expression of the Wnt antagonist Sfrp2 and down-regulation of Wnt signaling in the furcation region, while overactivation of Wnt signaling in Lhx6+ progenitor cells partially restore the furcation defects in Lhx6-/- mice. Collectively, our findings have important implications for understanding organ morphogenesis and future strategies for tooth root regeneration.


Subject(s)
Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/genetics , Mesenchymal Stem Cells/metabolism , Molar/metabolism , Morphogenesis/genetics , Nerve Tissue Proteins/genetics , Tooth Root/metabolism , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Female , LIM-Homeodomain Proteins/metabolism , Male , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Genetic , Molar/cytology , Molar/growth & development , Nerve Tissue Proteins/metabolism , Tooth Root/cytology , Tooth Root/growth & development , Transcription Factors/metabolism
7.
Development ; 147(18)2020 09 21.
Article in English | MEDLINE | ID: mdl-32958507

ABSTRACT

The FaceBase Consortium was established by the National Institute of Dental and Craniofacial Research in 2009 as a 'big data' resource for the craniofacial research community. Over the past decade, researchers have deposited hundreds of annotated and curated datasets on both normal and disordered craniofacial development in FaceBase, all freely available to the research community on the FaceBase Hub website. The Hub has developed numerous visualization and analysis tools designed to promote integration of multidisciplinary data while remaining dedicated to the FAIR principles of data management (findability, accessibility, interoperability and reusability) and providing a faceted search infrastructure for locating desired data efficiently. Summaries of the datasets generated by the FaceBase projects from 2014 to 2019 are provided here. FaceBase 3 now welcomes contributions of data on craniofacial and dental development in humans, model organisms and cell lines. Collectively, the FaceBase Consortium, along with other NIH-supported data resources, provide a continuously growing, dynamic and current resource for the scientific community while improving data reproducibility and fulfilling data sharing requirements.


Subject(s)
Dental Research/methods , Facial Bones/physiology , Skull/physiology , Animals , Databases, Factual , Humans , Reproducibility of Results , Research Personnel
8.
Dev Biol ; 441(1): 191-203, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29981310

ABSTRACT

Cleft palate is one of the most common craniofacial congenital defects in humans. It is associated with multiple genetic and environmental risk factors, including mutations in the genes encoding signaling molecules in the sonic hedgehog (Shh) pathway, which are risk factors for cleft palate in both humans and mice. However, the function of Shh signaling in the palatal epithelium during palatal fusion remains largely unknown. Although components of the Shh pathway are localized in the palatal epithelium, specific inhibition of Shh signaling in palatal epithelium does not affect palatogenesis. We therefore utilized a hedgehog (Hh) signaling gain-of-function mouse model, K14-Cre;R26SmoM2, to uncover the role of Shh signaling in the palatal epithelium during palatal fusion. In this study, we discovered that constitutive activation of Hh signaling in the palatal epithelium results in submucous cleft palate and persistence of the medial edge epithelium (MEE). Further investigation revealed that precise downregulation of Shh signaling is required at a specific time point in the MEE during palatal fusion. Upregulation of Hh signaling in the palatal epithelium maintains the proliferation of MEE cells. This may be due to a dysfunctional p63/Irf6 regulatory loop. The resistance of MEE cells to apoptosis is likely conferred by enhancement of a cell adhesion network through the maintenance of p63 expression. Collectively, our data illustrate that persistent Hh signaling in the palatal epithelium contributes to the etiology and pathogenesis of submucous cleft palate through its interaction with a p63/Irf6-dependent biological regulatory loop and through a p63-induced cell adhesion network.


Subject(s)
Embryo, Mammalian/metabolism , Epithelial Cells/metabolism , Hedgehog Proteins/metabolism , Palate/embryology , Signal Transduction/physiology , Animals , Cell Adhesion/physiology , Embryo, Mammalian/cytology , Epithelial Cells/cytology , Hedgehog Proteins/genetics , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mice , Mice, Transgenic , Palate/cytology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
9.
Hum Mol Genet ; 26(5): 860-872, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28069795

ABSTRACT

Ciliopathies are pleiotropic human diseases resulting from defects of the primary cilium, and these patients often have cleft lip and palate. IFT88 is required for the assembly and function of the primary cilia, which mediate the activity of key developmental signaling pathways. Through whole exome sequencing of a family of three affected siblings with isolated cleft lip and palate, we discovered that they share a novel missense mutation in IFT88 (c.915G > C, p.E305D), suggesting this gene should be considered a candidate for isolated orofacial clefting. In order to evaluate the function of IFT88 in regulating craniofacial development, we generated Wnt1-Cre;Ift88fl/fl mice to eliminate Ift88 specifically in cranial neural crest (CNC) cells. Wnt1-Cre;Ift88fl/flpups died at birth due to severe craniofacial defects including bilateral cleft lip and palate and tongue agenesis, following the loss of the primary cilia in the CNC-derived palatal mesenchyme. Loss of Ift88 also resulted in a decrease in neural crest cell proliferation during early stages of palatogenesis as well as a downregulation of the Shh signaling pathway in the palatal mesenchyme. Importantly, Osr2KI-Cre;Ift88fl/flmice, in which Ift88 is lost specifically in the palatal mesenchyme, exhibit isolated cleft palate. Taken together, our results demonstrate that IFT88 has a highly conserved function within the primary cilia of the CNC-derived mesenchyme in the lip and palate region in mice and is a strong candidate as an orofacial clefting gene in humans.


Subject(s)
Cleft Lip/genetics , Embryonic Development/genetics , Tumor Suppressor Proteins/genetics , Wnt1 Protein/genetics , Animals , Cell Proliferation/genetics , Cleft Lip/pathology , Gene Expression Regulation, Developmental , Humans , Mesoderm/growth & development , Mesoderm/pathology , Mice , Neural Crest/growth & development , Neural Crest/metabolism , Neural Crest/pathology , Palate/growth & development , Palate/pathology , Signal Transduction , Tumor Suppressor Proteins/biosynthesis , Wnt1 Protein/biosynthesis
10.
Development ; 142(19): 3374-82, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26293299

ABSTRACT

Coordination between the Hertwig's epithelial root sheath (HERS) and apical papilla (AP) is crucial for proper tooth root development. The hedgehog (Hh) signaling pathway and Nfic are both involved in tooth root development; however, their relationship has yet to be elucidated. Here, we establish a timecourse of mouse molar root development by histological staining of sections, and we demonstrate that Hh signaling is active before and during root development in the AP and HERS using Gli1 reporter mice. The proper pattern of Hh signaling activity in the AP is crucial for the proliferation of dental mesenchymal cells, because either inhibition with Hh inhibitors or constitutive activation of Hh signaling activity in transgenic mice leads to decreased proliferation in the AP and shorter roots. Moreover, Hh activity is elevated in Nfic(-/-) mice, a root defect model, whereas RNA sequencing and in situ hybridization show that the Hh attenuator Hhip is downregulated. ChIP and RNAscope analyses suggest that Nfic binds to the promoter region of Hhip. Treatment of Nfic(-/-) mice with Hh inhibitor partially restores cell proliferation, AP growth and root development. Taken together, our results demonstrate that an Nfic-Hhip-Hh signaling pathway is crucial for apical papilla growth and proper root formation. This discovery provides insight into the molecular mechanisms regulating tooth root development.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/metabolism , Morphogenesis/physiology , NFI Transcription Factors/metabolism , Signal Transduction/physiology , Tooth Root/growth & development , Animals , Base Sequence , Chromatin Immunoprecipitation , DNA Primers/genetics , Galactosides , In Situ Hybridization , Indoles , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Transgenic , Microscopy, Fluorescence , Molar/growth & development , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Tamoxifen , X-Ray Microtomography , Zinc Finger Protein GLI1
11.
Development ; 141(4): 909-17, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24496627

ABSTRACT

Clefting of the soft palate occurs as a congenital defect in humans and adversely affects the physiological function of the palate. However, the molecular and cellular mechanism of clefting of the soft palate remains unclear because few animal models exhibit an isolated cleft in the soft palate. Using three-dimensional microCT images and histological reconstruction, we found that loss of TGFß signaling in the palatal epithelium led to soft palate muscle defects in Tgfbr2(fl/fl);K14-Cre mice. Specifically, muscle mass was decreased in the soft palates of Tgfbr2 mutant mice, following defects in cell proliferation and differentiation. Gene expression of Dickkopf (Dkk1 and Dkk4), negative regulators of WNT-ß-catenin signaling, is upregulated in the soft palate of Tgfbr2(fl/fl);K14-Cre mice, and WNT-ß-catenin signaling is disrupted in the palatal mesenchyme. Importantly, blocking the function of DKK1 and DKK4 rescued the cell proliferation and differentiation defects in the soft palate of Tgfbr2(fl/fl);K14-Cre mice. Thus, our findings indicate that loss of TGFß signaling in epithelial cells compromises activation of WNT signaling and proper muscle development in the soft palate through tissue-tissue interactions, resulting in a cleft soft palate. This information has important implications for prevention and non-surgical correction of cleft soft palate.


Subject(s)
Cleft Palate/physiopathology , Epithelial-Mesenchymal Transition/physiology , Muscle, Skeletal/embryology , Palate, Soft/embryology , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/physiology , Animals , Cleft Palate/etiology , DNA Primers/genetics , Imaging, Three-Dimensional , Immunoblotting , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Microarray Analysis , Protein Serine-Threonine Kinases/genetics , Real-Time Polymerase Chain Reaction , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , X-Ray Microtomography
12.
Dev Biol ; 400(2): 180-90, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25722190

ABSTRACT

Growth factor signaling regulates tissue-tissue interactions to control organogenesis and tissue homeostasis. Specifically, transforming growth factor beta (TGFß) signaling plays a crucial role in the development of cranial neural crest (CNC) cell-derived bone, and loss of Tgfbr2 in CNC cells results in craniofacial skeletal malformations. Our recent studies indicate that non-canonical TGFß signaling is activated whereas canonical TGFß signaling is compromised in the absence of Tgfbr2 (in Tgfbr2(fl/fl);Wnt1-Cre mice). A haploinsufficiency of Tgfbr1 (aka Alk5) (Tgfbr2(fl/fl);Wnt1-Cre;Alk5(fl/+)) largely rescues craniofacial deformities in Tgfbr2 mutant mice by reducing ectopic non-canonical TGFß signaling. However, the relative involvement of canonical and non-canonical TGFß signaling in regulating specific craniofacial bone formation remains unclear. We compared the size and volume of CNC-derived craniofacial bones (frontal bone, premaxilla, maxilla, palatine bone, and mandible) from E18.5 control, Tgfbr2(fl/fl);Wnt1-Cre, and Tgfbr2(fl/fl);Wnt1-Cre;Alk5(fl/+)mice. By analyzing three dimensional (3D) micro-computed tomography (microCT) images, we found that different craniofacial bones were restored to different degrees in Tgfbr2(fl/fl);Wnt1-Cre;Alk5(fl/+) mice. Our study provides comprehensive information on anatomical landmarks and the size and volume of each craniofacial bone, as well as insights into the extent that canonical and non-canonical TGFß signaling cascades contribute to the formation of each CNC-derived bone. Our data will serve as an important resource for developmental biologists who are interested in craniofacial morphogenesis.


Subject(s)
Bone Development , Facial Bones/embryology , Skull/embryology , Animals , Facial Bones/anatomy & histology , Imaging, Three-Dimensional , Mice , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , X-Ray Microtomography
13.
Hum Mol Genet ; 23(1): 182-93, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23975680

ABSTRACT

Mutations in transforming growth factor beta (TGFß) receptor type II (TGFBR2) cause Loeys-Dietz syndrome, characterized by craniofacial and cardiovascular abnormalities. Mice with a deletion of Tgfbr2 in cranial neural crest cells (Tgfbr2(fl/fl);Wnt1-Cre mice) develop cleft palate as the result of abnormal TGFß signaling activation. However, little is known about metabolic processes downstream of TGFß signaling during palatogenesis. Here, we show that Tgfbr2 mutant palatal mesenchymal cells spontaneously accumulate lipid droplets, resulting from reduced lipolysis activity. Tgfbr2 mutant palatal mesenchymal cells failed to respond to the cell proliferation stimulator sonic hedgehog, derived from the palatal epithelium. Treatment with p38 mitogen-activated protein kinase (MAPK) inhibitor or telmisartan, a modulator of p38 MAPK activation and lipid metabolism, blocked abnormal TGFß-mediated p38 MAPK activation, restoring lipid metabolism and cell proliferation activity both in vitro and in vivo. Our results highlight the influence of alternative TGFß signaling on lipid metabolic activities, as well as how lipid metabolic defects can affect cell proliferation and adversely impact palatogenesis. This discovery has broader implications for the understanding of metabolic defects and potential prevention of congenital birth defects.


Subject(s)
Benzimidazoles/pharmacology , Benzoates/pharmacology , Cleft Palate/drug therapy , Lipid Metabolism/drug effects , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Cell Proliferation/drug effects , Cleft Palate/embryology , Cleft Palate/pathology , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Telmisartan , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Development ; 140(6): 1220-30, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23406900

ABSTRACT

Cleft palate is one of the most common human birth defects and is associated with multiple genetic and environmental risk factors. Although mutations in the genes encoding transforming growth factor beta (TGFß) signaling molecules and interferon regulatory factor 6 (Irf6) have been identified as genetic risk factors for cleft palate, little is known about the relationship between TGFß signaling and IRF6 activity during palate formation. Here, we show that TGFß signaling regulates expression of Irf6 and the fate of the medial edge epithelium (MEE) during palatal fusion in mice. Haploinsufficiency of Irf6 in mice with basal epithelial-specific deletion of the TGFß signaling mediator Smad4 (Smad4(fl/fl);K14-Cre;Irf6(+/R84C)) results in compromised p21 expression and MEE persistence, similar to observations in Tgfbr2(fl/fl);K14-Cre mice, although the secondary palate of Irf6(+/R84C) and Smad4(fl/fl);K14-Cre mice form normally. Furthermore, Smad4(fl/fl);K14-Cre;Irf6(+/R84C) mice show extra digits that are consistent with abnormal toe and nail phenotypes in individuals with Van der Woude and popliteal pterygium syndromes, suggesting that the TGFß/SMAD4/IRF6 signaling cascade might be a well-conserved mechanism in regulating multiple organogenesis. Strikingly, overexpression of Irf6 rescued p21 expression and MEE degeneration in Tgfbr2(fl/fl);K14-Cre mice. Thus, IRF6 and SMAD4 synergistically regulate the fate of the MEE, and TGFß-mediated Irf6 activity is responsible for MEE degeneration during palatal fusion in mice.


Subject(s)
Epistasis, Genetic , Interferon Regulatory Factors/genetics , Palate/embryology , Smad4 Protein/genetics , Transforming Growth Factor beta/pharmacology , Animals , Animals, Newborn , Cell Fusion , Cells, Cultured , Embryo, Mammalian , Epistasis, Genetic/drug effects , Epistasis, Genetic/physiology , Female , Humans , Interferon Regulatory Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organogenesis/drug effects , Organogenesis/genetics , Palate/drug effects , Palate/metabolism , Pregnancy , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism
15.
Proc Natl Acad Sci U S A ; 109(51): 20853-8, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23213213

ABSTRACT

One of the hurdles for practical application of induced pluripotent stem cells (iPSC) is the low efficiency and slow process of reprogramming. Octamer-binding transcription factor 4 (Oct4) has been shown to be an essential regulator of embryonic stem cell (ESC) pluripotency and key to the reprogramming process. To identify small molecules that enhance reprogramming efficiency, we performed a cell-based high-throughput screening of chemical libraries. One of the compounds, termed Oct4-activating compound 1 (OAC1), was found to activate both Oct4 and Nanog promoter-driven luciferase reporter genes. Furthermore, when added to the reprogramming mixture along with the quartet reprogramming factors (Oct4, Sox2, c-Myc, and Klf4), OAC1 enhanced the iPSC reprogramming efficiency and accelerated the reprogramming process. Two structural analogs of OAC1 also activated Oct4 and Nanog promoters and enhanced iPSC formation. The iPSC colonies derived using the Oct4-activating compounds along with the quartet factors exhibited typical ESC morphology, gene-expression pattern, and developmental potential. OAC1 seems to enhance reprogramming efficiency in a unique manner, independent of either inhibition of the p53-p21 pathway or activation of the Wnt-ß-catenin signaling. OAC1 increases transcription of the Oct4-Nanog-Sox2 triad and Tet1, a gene known to be involved in DNA demethylation.


Subject(s)
Benzamides/pharmacology , Cellular Reprogramming/drug effects , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/metabolism , Pyridines/pharmacology , Pyrroles/pharmacology , Animals , Benzamides/chemistry , Cell Differentiation , Chemistry, Pharmaceutical/methods , DNA Methylation , DNA-Binding Proteins/metabolism , Drug Design , Fibroblasts/metabolism , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Factor 4 , Mice , Mixed Function Oxygenases , Nanog Homeobox Protein , Proto-Oncogene Proteins/metabolism , Pyridines/chemistry , Pyrroles/chemistry , SOXB1 Transcription Factors/metabolism
16.
J Biol Chem ; 288(41): 29760-70, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23950180

ABSTRACT

Microglossia is a congenital birth defect in humans and adversely impacts quality of life. In vertebrates, tongue muscle derives from the cranial mesoderm, whereas tendons and connective tissues in the craniofacial region originate from cranial neural crest (CNC) cells. Loss of transforming growth factor ß (TGFß) type II receptor in CNC cells in mice (Tgfbr2(fl/fl);Wnt1-Cre) causes microglossia due to a failure of cell-cell communication between cranial mesoderm and CNC cells during tongue development. However, it is still unclear how TGFß signaling in CNC cells regulates the fate of mesoderm-derived myoblasts during tongue development. Here we show that activation of the cytoplasmic and nuclear tyrosine kinase 1 (ABL1) cascade in Tgfbr2(fl/fl);Wnt1-Cre mice results in a failure of CNC-derived cell differentiation followed by a disruption of TGFß-mediated induction of growth factors and reduction of myogenic cell proliferation and differentiation activities. Among the affected growth factors, the addition of fibroblast growth factor 4 (FGF4) and neutralizing antibody for follistatin (FST; an antagonist of bone morphogenetic protein (BMP)) could most efficiently restore cell proliferation, differentiation, and organization of muscle cells in the tongue of Tgfbr2(fl/fl);Wnt1-Cre mice. Thus, our data indicate that CNC-derived fibroblasts regulate the fate of mesoderm-derived myoblasts through TGFß-mediated regulation of FGF and BMP signaling during tongue development.


Subject(s)
Muscles/metabolism , Neural Crest/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Tongue/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Female , Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 4/metabolism , Fibroblast Growth Factor 4/pharmacology , Follistatin/genetics , Follistatin/metabolism , Follistatin/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Immunoblotting , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Muscles/embryology , Neural Crest/cytology , Neural Crest/embryology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Skull/embryology , Skull/metabolism , Tongue/cytology , Tongue/embryology , Transforming Growth Factor beta/pharmacology
17.
bioRxiv ; 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38915513

ABSTRACT

IRF6 is a key genetic determinant of syndromic and non-syndromic cleft lip and palate. The ability to interrogate post-embryonic requirements of Irf6 has been hindered, as global Irf6 ablation in the mouse causes neonatal lethality. Prior work analyzing Irf6 in mouse models defined its role in the embryonic surface epithelium and periderm where it is required to regulate cell proliferation and differentiation. Several reports have also described Irf6 gene expression in other cell types, such as muscle, and neuroectoderm. However, analysis of a functional role in non-epithelial cell lineages has been incomplete due to the severity and lethality of the Irf6 knockout model and the paucity of work with a conditional Irf6 allele. Here we describe the generation and characterization of a new Irf6 floxed mouse model and analysis of Irf6 ablation in periderm and neural crest lineages. This work found that loss of Irf6 in periderm recapitulates a mild Irf6 null phenotype, suggesting that Irf6-mediated signaling in periderm plays a crucial role in regulating embryonic development. Further, conditional ablation of Irf6 in neural crest cells resulted in an anterior neural tube defect of variable penetrance. The generation of this conditional Irf6 allele allows for new insights into craniofacial development and new exploration into the post-natal role of Irf6.

18.
Cell Stem Cell ; 31(6): 904-920.e6, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38703771

ABSTRACT

Mesenchymal stem cells (MSCs) reside in niches to maintain tissue homeostasis and contribute to repair and regeneration. Although the physiological functions of blood and lymphatic vasculature are well studied, their regulation of MSCs as niche components remains largely unknown. Using adult mouse incisors as a model, we uncover the role of Trp53 in regulating vascular composition through THBS2 to maintain mesenchymal tissue homeostasis. Loss of Trp53 in GLI1+ progeny increases arteries and decreases other vessel types. Platelet-derived growth factors from arteries deposit in the MSC region and interact with PDGFRA and PDGFRB. Significantly, PDGFRA+ and PDGFRB+ cells differentially contribute to defined cell lineages in the adult mouse incisor. Collectively, our results highlight Trp53's importance in regulating the vascular niche for MSCs. They also shed light on how different arterial cells provide unique cues to regulate MSC subpopulations and maintain their heterogeneity. Furthermore, they provide mechanistic insight into MSC-vasculature crosstalk.


Subject(s)
Incisor , Mesenchymal Stem Cells , Signal Transduction , Tumor Suppressor Protein p53 , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Tumor Suppressor Protein p53/metabolism , Incisor/cytology , Incisor/metabolism , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism
19.
Nat Commun ; 15(1): 4614, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816354

ABSTRACT

ARID1B haploinsufficiency in humans causes Coffin-Siris syndrome, associated with developmental delay, facial dysmorphism, and intellectual disability. The role of ARID1B has been widely studied in neuronal development, but whether it also regulates stem cells remains unknown. Here, we employ scRNA-seq and scATAC-seq to dissect the regulatory functions and mechanisms of ARID1B within mesenchymal stem cells (MSCs) using the mouse incisor model. We reveal that loss of Arid1b in the GLI1+ MSC lineage disturbs MSCs' quiescence and leads to their proliferation due to the ectopic activation of non-canonical Activin signaling via p-ERK. Furthermore, loss of Arid1b upregulates Bcl11b, which encodes a BAF complex subunit that modulates non-canonical Activin signaling by directly regulating the expression of activin A subunit, Inhba. Reduction of Bcl11b or non-canonical Activin signaling restores the MSC population in Arid1b mutant mice. Notably, we have identified that ARID1B suppresses Bcl11b expression via specific binding to its third intron, unveiling the direct inter-regulatory interactions among BAF subunits in MSCs. Our results demonstrate the vital role of ARID1B as an epigenetic modifier in maintaining MSC homeostasis and reveal its intricate mechanistic regulatory network in vivo, providing novel insights into the linkage between chromatin remodeling and stem cell fate determination.


Subject(s)
DNA-Binding Proteins , Mesenchymal Stem Cells , Repressor Proteins , Transcription Factors , Animals , Mice , Activins/metabolism , Cell Proliferation , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Repressor Proteins/metabolism , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Zinc Finger Protein GLI1
20.
Bone Res ; 12(1): 37, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38910207

ABSTRACT

Stem/progenitor cells differentiate into different cell lineages during organ development and morphogenesis. Signaling pathway networks and mechanotransduction are important factors to guide the lineage commitment of stem/progenitor cells during craniofacial tissue morphogenesis. Here, we used tooth root development as a model to explore the roles of FGF signaling and mechanotransduction as well as their interaction in regulating the progenitor cell fate decision. We show that Fgfr1 is expressed in the mesenchymal progenitor cells and their progeny during tooth root development. Loss of Fgfr1 in Gli1+ progenitors leads to hyperproliferation and differentiation, which causes narrowed periodontal ligament (PDL) space with abnormal cementum/bone formation leading to ankylosis. We further show that aberrant activation of WNT signaling and mechanosensitive channel Piezo2 occurs after loss of FGF signaling in Gli1-CreER;Fgfr1fl/fl mice. Overexpression of Piezo2 leads to increased osteoblastic differentiation and decreased Piezo2 leads to downregulation of WNT signaling. Mechanistically, an FGF/PIEZO2/WNT signaling cascade plays a crucial role in modulating the fate of progenitors during root morphogenesis. Downregulation of WNT signaling rescues tooth ankylosis in Fgfr1 mutant mice. Collectively, our findings uncover the mechanism by which FGF signaling regulates the fate decisions of stem/progenitor cells, and the interactions among signaling pathways and mechanotransduction during tooth root development, providing insights for future tooth root regeneration.


Subject(s)
Fibroblast Growth Factors , Mechanotransduction, Cellular , Tooth Root , Wnt Signaling Pathway , Animals , Wnt Signaling Pathway/physiology , Tooth Root/growth & development , Tooth Root/metabolism , Tooth Root/cytology , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Mice , Cell Differentiation , Stem Cells/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Ion Channels
SELECTION OF CITATIONS
SEARCH DETAIL