Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Protein Expr Purif ; 179: 105797, 2021 03.
Article in English | MEDLINE | ID: mdl-33242573

ABSTRACT

Myogenesis is an important and complicated biological process, especially during the process of embryonic development. The homeoprotein Msx1 is a crucial transcriptional repressor of myogenesis and maintains myogenic precursor cells in an undifferentiated, proliferative state. However, the molecular mechanism through which Msx1 coordinates myogenesis remains to be elucidated. Here, we determine the interacting partner proteins of Msx1 in myoblast cells by a proteomic screening method. Msx1 is found to interact with 55 proteins, among which our data demonstrate that the cooperation of Runt-related transcription factor 1 (Runx1) with Msx1 is required for myoblast cell differentiation. Our findings provide important insights into the mechanistic roles of Msx1 in myoblast cell differentiation, and lays foundation for the myogenic differentiation process.


Subject(s)
Cell Differentiation/physiology , Core Binding Factor Alpha 2 Subunit , MSX1 Transcription Factor , Myoblasts , Animals , Cell Line , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/physiology , Gene Knockout Techniques , MSX1 Transcription Factor/chemistry , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , MSX1 Transcription Factor/physiology , Mice , Myoblasts/cytology , Myoblasts/metabolism
2.
Int J Mol Sci ; 21(12)2020 Jun 25.
Article in English | MEDLINE | ID: mdl-32630554

ABSTRACT

Prognostic factors are of great interest in patients with endometrial cancer. One potential factor could be the protein MSX1, a transcription repressor, that has an inhibitory effect on the cell cycle. For this study, endometrioid endometrial carcinomas (n = 53), clear cell endometrial carcinomas (n = 6), endometrioid ovarian carcinomas (n = 19), and clear cell ovarian carcinomas (n = 11) were immunochemically stained for the protein MSX1 and evaluated using the immunoreactive score (IRS). A significant stronger expression of MSX1 was found in endometrioid endometrial carcinomas (p < 0.001), in grading 2 (moderate differentiation) (p = 0.001), and in tumor material of patients with no involvement of lymph nodes (p = 0.031). Correlations were found between MSX1 expression and the expression of ß-Catenin, p21, p53, and the steroid receptors ERα, ERß, PRα, and PRß. A significant (p = 0.023) better survival for patients with an MSX1 expression in more than 10% of the tumor cells was observed for endometrioid endometrial carcinomas (21.3 years median survival (MSX1-positive) versus 17.3 years (MSX1-negative)). Although there is evidence that MSX1 expression correlates with improved long-term survival, further studies are necessary to evaluate if MSX1 can be used as a prognostic marker.


Subject(s)
Endometrial Neoplasms/metabolism , MSX1 Transcription Factor/metabolism , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , DNA Methylation/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Humans , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/physiology , Middle Aged , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Uterine Neoplasms/pathology , Uterus/metabolism , Uterus/pathology
3.
J Invest Dermatol ; 138(1): 141-149, 2018 01.
Article in English | MEDLINE | ID: mdl-28927893

ABSTRACT

Melanoma cells share many biological properties with neural crest stem cells. Here we show that the homeodomain transcription factor MSX1, which is significantly correlated with melanoma disease progression, reprograms melanocytes and melanoma cells toward a neural crest precursor-like state. MSX1-reprogrammed normal human melanocytes express the neural crest marker p75 and become multipotent. MSX1 induces a phenotypic switch in melanoma, which is characterized by an oncogenic transition from an E-cadherin-high nonmigratory state toward a ZEB1-high invasive state. ZEB1 up-regulation is responsible for the MSX1-induced migratory phenotype in melanoma cells. Depletion of MSX1 significantly inhibits melanoma metastasis in vivo. These results show that neural crest-like reprogramming achieved by a single factor is a critical process for melanoma progression.


Subject(s)
Cell Transformation, Neoplastic/pathology , Cellular Reprogramming/physiology , MSX1 Transcription Factor/physiology , Melanocytes/pathology , Melanoma/pathology , Skin Neoplasms/pathology , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Cell Movement , Dermis/cytology , Dermis/pathology , Disease Progression , HEK293 Cells , Human Embryonic Stem Cells , Humans , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Liver Neoplasms/secondary , MSX1 Transcription Factor/genetics , Melanoma/mortality , Melanoma/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/metabolism , Neural Crest/physiology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Nerve Growth Factor/metabolism , Skin Neoplasms/mortality , Xenograft Model Antitumor Assays , Zinc Finger E-box-Binding Homeobox 1/metabolism
4.
Eur J Oral Sci ; 126(1): 1-12, 2018 02.
Article in English | MEDLINE | ID: mdl-29148101

ABSTRACT

Homeobox protein MSX-1 (hereafter referred to as MSX-1) is essential for early tooth-germ development. Tooth-germ development is arrested at bud stage in Msx1 knockout mice, which prompted us to study the functions of MSX-1 beyond this stage. Here, we investigated the roles of MSX-1 during late bell stage. Mesenchymal cells of the mandibular first molar were isolated from mice at embryonic day (E)17.5 and cultured in vitro. We determined the expression levels of ß-catenin, bone morphogenetic protein 2 (Bmp2), Bmp4, and lymphoid enhancer-binding factor 1 (Lef1) after knockdown or overexpression of Msx1. Our findings suggest that knockdown of Msx1 promoted expression of Bmp2, Bmp4, and Lef1, resulting in elevated differentiation of odontoblasts, which was rescued by blocking the expression of these genes. In contrast, overexpression of Msx1 decreased the expression of Bmp2, Bmp4, and Lef1, leading to a reduction in odontoblast differentiation. The regulation of Bmp2, Bmp4, and Lef1 by Msx1 was mediated by the Wnt/ß-catenin signaling pathway. Additionally, knockdown of Msx1 impaired cell proliferation and slowed S-phase progression, while overexpression of Msx1 also impaired cell proliferation and prolonged G1-phase progression. We therefore conclude that MSX-1 maintains cell proliferation by regulating transition of cells from G1-phase to S-phase and prevents odontoblast differentiation by inhibiting expression of Bmp2, Bmp4, and Lef1 at the late bell stage via the Wnt/ß-catenin signaling pathway.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/physiology , Lymphoid Enhancer-Binding Factor 1/metabolism , MSX1 Transcription Factor/physiology , Mesenchymal Stem Cells/metabolism , Odontogenesis/physiology , Tooth Germ/cytology , Animals , Female , Fetus , Flow Cytometry , In Vitro Techniques , Mice , Mice, Inbred ICR , Pregnancy , Wnt Signaling Pathway/physiology , beta Catenin/metabolism
5.
Organogenesis ; 13(4): 141-155, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28933666

ABSTRACT

Before the secretion of hard dental tissues, tooth germs undergo several distinctive stages of development (dental lamina, bud, cap and bell). Every stage is characterized by specific proliferation patterns, which is regulated by various morphogens, growth factors and homeodomain proteins. The role of MSX homeodomain proteins in odontogenesis is rather complex. Expression domains of genes encoding for murine Msx1/2 during development are observed in tissues containing highly proliferative progenitor cells. Arrest of tooth development in Msx knockout mice can be attributed to impaired proliferation of progenitor cells. In Msx1 knockout mice, these progenitor cells start to differentiate prematurely as they strongly express cyclin-dependent kinase inhibitor p19INK4d. p19INK4d induces terminal differentiation of cells by blocking the cell cycle in mitogen-responsive G1 phase. Direct suppression of p19INK4d by Msx1 protein is, therefore, important for maintaining proliferation of progenitor cells at levels required for the normal progression of tooth development. In this study, we examined the expression patterns of MSX1, MSX2 and p19INK4d in human incisor tooth germs during the bud, cap and early bell stages of development. The distribution of expression domains of p19INK4d throughout the investigated period indicates that p19INK4d plays active role during human tooth development. Furthermore, comparison of expression domains of p19INK4d with those of MSX1, MSX2 and proliferation markers Ki67, Cyclin A2 and pRb, indicates that MSX-mediated regulation of proliferation in human tooth germs might not be executed by the mechanism similar to one described in developing tooth germs of wild-type mouse.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p19/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Incisor/embryology , MSX1 Transcription Factor/physiology , Cell Cycle , Cell Proliferation , Cyclin A2/metabolism , Humans , Ki-67 Antigen/physiology , Protein Domains , Retinoblastoma Protein/metabolism , Stem Cells/cytology
6.
Arch Oral Biol ; 75: 8-13, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27951410

ABSTRACT

OBJECTIVE: Inherited congenital anomalies in tooth number, particularly hypodontia are relatively common. Although substantial progress has been made that permits a better understanding of the causes of tooth agenesis, overall knowledge of the phenotype:genotype correlations in this anomaly are still lacking. The aim in this study was to identify the causal gene mutation(s) in a family of two sisters with severe hypodontia (oligodontia) including 2nd premolars and 1st and 3rd molars, using whole exome sequencing (WES). METHODS: WES was performed using in-solution hybridization, followed by massively parallel sequencing. RESULTS: A frameshift insertion of 7 basepairs (GCAAGTT) in the homebox of MSX1 gene located in the exon 2 in heterozygous state has been identified in both sisters (NM_002448:exon2:c.572_573ins GCAAGTT: p.F191fs). CONCLUSION: We conclude that this frameshift mutation in the homeodomain (which plays an essential role in DNA binding) of MSX1 gene is responsible for tooth agenesis in this family. This expands the phenotype-genotype correlation associated with MSX1 mutations.


Subject(s)
Anodontia/genetics , Frameshift Mutation/genetics , Genes, Homeobox , MSX1 Transcription Factor/genetics , Mutagenesis, Insertional , Adult , Anodontia/diagnostic imaging , Anodontia/pathology , Base Sequence , Bicuspid/abnormalities , Bicuspid/diagnostic imaging , Female , Heterozygote , Homeodomain Proteins/genetics , Humans , MSX1 Transcription Factor/physiology , Molar/abnormalities , Molar/diagnostic imaging , Radiography, Panoramic , Exome Sequencing
7.
Congenit Anom (Kyoto) ; 56(1): 12-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26754466

ABSTRACT

The tympanic membrane is a thin layer that originates from the ectoderm, endoderm, and mesenchyme. Molecular-genetic investigations have revealed that interaction between epithelial and mesenchymal cells in the pharyngeal arches is essential for development of the tympanic membrane. We have recently reported that developmental mechanisms underlying the tympanic membrane seem to be different between mouse and chicken, suggesting that the tympanic membrane evolved independently in mammals and non-mammalian amniotes. In this review, we summarize previous studies of tympanic membrane formation in the mouse. We also discuss its formation in amniotes from an evolutionary point of view.


Subject(s)
Tympanic Membrane/embryology , Animals , Biological Evolution , Gene Expression Regulation, Developmental , Goosecoid Protein/physiology , Homeodomain Proteins/physiology , Humans , MSX1 Transcription Factor/physiology , Mammals , Tympanic Membrane/abnormalities , Tympanic Membrane/metabolism
8.
J Biol Chem ; 290(24): 15337-49, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25931120

ABSTRACT

Embryonic diapause is a reproductive strategy widespread in the animal kingdom. This phenomenon is defined by a temporary arrest in blastocyst growth and metabolic activity within a quiescent uterus without implantation until the environmental and maternal milieu become favorable for pregnancy to progress. We found that uterine Msx expression persists during diapause across species; their inactivation in the mouse uterus results in termination of diapause with the development of implantation-like responses ("pseudoimplantation") that ultimately succumbed to resorption. To understand the cause of this failure, we compared proteome profiles between floxed and Msx-deleted uteri. In deleted uteri, several functional networks, including transcription/translation, ubiquitin-proteasome, inflammation, and endoplasmic reticulum stress, were dysregulated. Computational modeling predicted intersection of these pathways on an enhanced inflammatory signature. Further studies showed that this signature was reflected in increased phosphorylated IκB levels and nuclear NFκB in deleted uteri. This was associated with enhanced proteasome activity and endoplasmic reticulum stress. Interestingly, treatment with anti-inflammatory glucocorticoid (dexamethasone) reduced the inflammatory signature with improvement of the diapause phenotype. These findings highlight an unexpected role of uterine Msx in limiting aberrant inflammatory responses to maintain embryonic diapause.


Subject(s)
Embryonic Development/genetics , Genes, Homeobox , Homeodomain Proteins/physiology , Inflammation/genetics , MSX1 Transcription Factor/physiology , Muscle, Smooth/pathology , Uterus/pathology , Animals , Female , Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Mice , Mice, Knockout , Pregnancy , Uterus/metabolism
9.
Int J Dev Biol ; 57(11-12): 829-36, 2013.
Article in English | MEDLINE | ID: mdl-24623074

ABSTRACT

The neural crest (NC) comprises a transient and multipotent embryonic cell population, which gives rise to a wide variety of cell types, including craniofacial cartilage, melanocytes, and neurons and glia of the peripheral nervous system. The NC is induced by the integrated action of Wnt, FGF, and BMP signaling, and its cell fates are subsequently specified by a genetic cascade of specific transcription factors. Here we describe a critical role of AWP1 in NC induction during Xenopus early development. Xenopus AWP1 (XAWP1) was found to be expressed in the presumptive preplacodal ectoderm, neural tissue, and posterior dorsal mesoderm, but was absent in the neural fold along the anterior-posterior axis of the neurulae. Notably, XAWP1 was induced by FGF8a in naïve ectodermal tissue. XAWP1-depleted embryos exhibited defects in pigmentation, craniofacial cartilage, and in the dorsal fin. A knockdown of XAWP1 impaired both endogenous and the FGF8a or Wnt8-induced expression of NC markers without affecting mesoderm formation. Furthermore, NC induction inhibited by XAWP1 depletion was rescued by co-expression of activating forms of beta-catenin or TCF3. In addition, overexpression of XAWP1, in concert with BMP inhibition, induced the expression of neural plate border specifiers, Pax3 and Msx1, and these regulatory factors recovered NC induction in the XAWP1-depleted embryos. Beta-catenin stability and Wnt-responsive reporter activity were also impaired in AWP1-depleted cells. Taken together, these results suggest that XAWP1 functions as a mediator of Wnt signaling to regulate NC specification.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Neural Crest/embryology , Xenopus Proteins/physiology , Xenopus laevis/embryology , Animals , Bone Morphogenetic Proteins/physiology , Cartilage/physiology , Cell Lineage , Gene Expression Profiling , Gene Expression Regulation, Developmental , MSX1 Transcription Factor/physiology , Mesoderm/physiology , PAX3 Transcription Factor , Paired Box Transcription Factors/physiology , Signal Transduction , Transcription Factor 3/physiology , Wnt Proteins/physiology , Xenopus laevis/physiology , beta Catenin/physiology
10.
Biochem J ; 444(3): 573-80, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22455953

ABSTRACT

DKK1 (dickkopf 1 homologue) is a secreted inhibitor of the Wnt signalling pathway and a critical modulator of tumour promotion and the tumour microenvironment. However, mechanisms regulating DKK1 expression are understudied. DNAJB6 {DnaJ [HSP40 (heat-shock protein 40 kDa)] homologue, subfamily B, member 6} is an HSP40 family member whose expression is compromised during progression of breast cancer and melanoma. Inhibition of the Wnt/ß-catenin signalling pathway by up-regulation of DKK1 is one of the key mechanisms by which DNAJB6 suppresses tumour metastasis and EMT (epithelial-mesenchymal transition). Analysis of the DKK1 promoter to define the cis-site responsible for its up-regulation by DNAJB6 revealed the presence of two binding sites for a transcriptional repressor, MSX1 (muscle segment homeobox 1). Our investigations showed that MSX1 binds the DKK1 promoter and inhibits DKK1 transcription. Interestingly, silencing DNAJB6 resulted in up-regulation of MSX1 concomitant with increased stabilization of ß-catenin. ChIP (chromatin immunoprecipitation) studies revealed that ß-catenin binds the MSX1 promoter and stabilization of ß-catenin elevates MSX1 transcription, indicating that ß-catenin works as a transcription co-activator for MSX1. Functionally, exogenous expression of MSX1 in DNAJB6-expressing cells promotes the mesenchymal phenotype by suppression of DKK1. Thus we have identified a novel regulatory mechanism of DNAJB6-mediated DKK1 transcriptional up-regulation that can influence EMT. DKK1 is a feedback regulator of ß-catenin levels and thus our studies also define an additional negative control of this ß-catenin/DKK1 feedback loop by MSX1, which may potentially contribute to excessive stabilization of ß-catenin.


Subject(s)
HSP40 Heat-Shock Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , MSX1 Transcription Factor/physiology , Molecular Chaperones/physiology , Nerve Tissue Proteins/physiology , Wnt Signaling Pathway/physiology , beta Catenin/physiology , Animals , Binding Sites/physiology , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Protein Stability , Wnt Proteins/physiology
11.
Development ; 138(24): 5393-402, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22071108

ABSTRACT

The mechanisms regulating germ line sex determination and meiosis initiation are poorly understood. Here, we provide evidence for the involvement of homeobox Msx transcription factors in foetal meiosis initiation in mammalian germ cells. Upon meiosis initiation, Msx1 and Msx2 genes are strongly expressed in the foetal ovary, possibly stimulated by soluble factors found there: bone morphogenetic proteins Bmp2 and Bmp4, and retinoic acid. Analysis of Msx1/Msx2 double mutant embryos revealed a majority of undifferentiated germ cells remaining in the ovary and, importantly, a decrease in the number of meiotic cells. In vivo, the Msx1/Msx2 double-null mutation prevented full activation of Stra8, a gene required for meiosis. In F9 cells, Msx1 can bind to Stra8 regulatory sequences and Msx1 overexpression stimulates Stra8 transcription. Collectively, our data demonstrate for the first time that some homeobox genes are required for meiosis initiation in the female germ line.


Subject(s)
Homeodomain Proteins/physiology , MSX1 Transcription Factor/physiology , Meiosis/physiology , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Mice , Mutation , Organ Culture Techniques , Ovary/physiology , Proteins/metabolism , Tretinoin/metabolism , Tretinoin/physiology
12.
Dev Biol ; 353(2): 344-53, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21420399

ABSTRACT

Mammalian tooth development depends on activation of odontogenic potential in the presumptive dental mesenchyme by the Msx1 and Pax9 transcription factors. We recently reported that the zinc finger transcription factor Osr2 was expressed in a lingual-to-buccal gradient pattern surrounding the developing mouse molar tooth germs and mice lacking Osr2 developed supernumerary teeth lingual to their molars. We report here generation of a gene-targeted mouse strain that allows conditional inactivation of Pax9 and subsequent activation of expression of Osr2 in the developing tooth mesenchyme from the Pax9 locus. Expression of Osr2 from one copy of the Pax9 gene did not disrupt normal tooth development but was sufficient to suppress supernumerary tooth formation in the Osr2(-/-) mutant mice. We found that endogenous Osr2 mRNA expression was significantly downregulated in the developing tooth mesenchyme in Pax9(del/del) mice. Mice lacking both Osr2 and Pax9 exhibited early tooth developmental arrest with significantly reduced Bmp4 and Msx1 mRNA expression in the developing tooth mesenchyme, similar to that in Pax9(del/del) mutants but in contrast to the rescue of tooth morphogenesis in Msx1(-/-)Osr2(-/-) double mutant mice. Furthermore, we found that Osr2 formed stable protein complexes with the Msx1 protein and interacted weakly with the Pax9 protein in co-transfected cells. These data indicate that Osr2 acts downstream of Pax9 and patterns the mesenchymal odontogenic field through protein-protein interactions with Msx1 and Pax9 during early tooth development.


Subject(s)
MSX1 Transcription Factor/physiology , Odontogenesis/physiology , Paired Box Transcription Factors/physiology , Transcription Factors/physiology , Animals , Base Sequence , Bone Morphogenetic Protein 4/genetics , DNA/genetics , Female , Gene Expression Regulation, Developmental , Gene Knockout Techniques , MSX1 Transcription Factor/deficiency , MSX1 Transcription Factor/genetics , Male , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Odontogenesis/genetics , PAX9 Transcription Factor , Paired Box Transcription Factors/deficiency , Paired Box Transcription Factors/genetics , Pregnancy , Protein Interaction Mapping , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tooth Germ/embryology , Tooth Germ/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
13.
Genesis ; 48(11): 645-55, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20824629

ABSTRACT

The Msx and Dlx families of homeobox proteins are important regulators for embryogenesis. Loss of Msx1 in mice results in multiple developmental defects including craniofacial malformations. Although Dlx5 is widely expressed during embryonic development, targeted null mutation of Dlx5 mainly affects the development of craniofacial bones. Msx1 and Dlx5 show overlapping expression patterns during frontal bone development. To investigate the functional significance of Msx1/Dlx5 interaction in regulating frontal bone development, we generated Msx1 and Dlx5 double null mutant mice. In Msx1(-/-) ;Dlx5(-/-) mice, the frontal bones defect was more severe than that of either Msx1(-/-) or Dlx5(-/-) mice. This aggravated frontal bone defect suggests that Msx1 and Dlx5 function synergistically to regulate osteogenesis. This synergistic effect of Msx1 and Dlx5 on the frontal bone represents a tissue specific mode of interaction of the Msx and Dlx genes. Furthermore, Dlx5 requires Msx1 for its expression in the context of frontal bone development. Our study shows that Msx1/Dlx5 interaction is crucial for osteogenic induction during frontal bone development.


Subject(s)
Frontal Bone/embryology , Homeodomain Proteins/physiology , MSX1 Transcription Factor/physiology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation , Cells, Cultured , Embryo, Mammalian , Epistasis, Genetic/physiology , Frontal Bone/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Knockout , Morphogenesis/genetics , Morphogenesis/physiology , Osteogenesis/genetics , Osteogenesis/physiology
14.
Dev Biol ; 340(2): 438-49, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20123092

ABSTRACT

Developmental abnormalities of craniofacial structures and teeth often occur sporadically and the underlying genetic defects are not well understood, in part due to unknown gene-gene interactions. Pax9 and Msx1 are co-expressed during craniofacial development, and mice that are single homozygous mutant for either gene exhibit cleft palate and an early arrest of tooth formation. Whereas in vitro assays have demonstrated that protein-protein interactions between Pax9 and Msx1 can occur, it is unclear if Pax9 and Msx1 interact genetically in vivo during development. To address this question, we compounded the Pax9 and Msx1 mutations and observed that double homozygous mutants exhibit an incompletely penetrant cleft lip phenotype. Moreover, in double heterozygous mutants, the lower incisors were consistently missing and we find that transgenic BMP4 expression partly rescues this phenotype. Reduced expression of Shh and Bmp2 indicates that a smaller "incisor field" forms in Pax9(+/-);Msx1(+/-) mutants, and dental epithelial growth is substantially reduced after the bud to cap stage transition. This defect is preceded by drastically reduced mesenchymal expression of Fgf3 and Fgf10, two genes that encode known stimulators of epithelial growth during odontogenesis. Consistent with this result, cell proliferation is reduced in both the dental epithelium and mesenchyme of double heterozygous mutants. Furthermore, the developing incisors lack mesenchymal Notch1 expression at the bud stage and exhibit abnormal ameloblast differentiation on both labial and lingual surfaces. Thus, Msx1 and Pax9 interact synergistically throughout lower incisor development and affect multiple signaling pathways that influence incisor size and symmetry. The data also suggest that a combined reduction of PAX9 and MSX1 gene dosage in humans may increase the risk for orofacial clefting and oligodontia.


Subject(s)
Gene Expression Regulation, Developmental , Lip/embryology , MSX1 Transcription Factor/genetics , Morphogenesis/genetics , Paired Box Transcription Factors/genetics , Tooth/metabolism , Animals , Embryo, Mammalian , Heterozygote , Immunohistochemistry , In Situ Hybridization , MSX1 Transcription Factor/metabolism , MSX1 Transcription Factor/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred Strains , Mice, Knockout , Odontogenesis/genetics , PAX9 Transcription Factor , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/physiology
15.
Reproduction ; 139(5): 857-70, 2010 May.
Article in English | MEDLINE | ID: mdl-20176746

ABSTRACT

This study was conducted to investigate the effect of suppressing transcription factor gene MSX1 on the development of in vitro produced bovine oocytes and embryos, and identify its potential target genes regulated by this gene. Injection of long double-stranded RNA (LdsRNA) and small interfering RNA (siRNA) at germinal vesicle stage oocyte reduced MSX1 mRNA expression by 73 and 37% respectively at metaphase II stage compared with non-injected controls. Similarly, injection of the same anti-sense oligomers at zygote stage reduced MSX1 mRNA expression by 52 and 33% at 8-cell stage compared with non-injected controls. Protein expression was also reduced in LdsRNA- and siRNA-injected groups compared with non-injected controls at both stages. Blastocysts rates were 33, 28, 20 and 18% in non-injected control, scrambled RNA (scRNA), LdsRNA- and siRNA-injected groups respectively. Cleavage rates were also significantly reduced in Smartpool siRNA (SpsiRNA)-injected group (53.76%) compared with scRNA-injected group (57.76%) and non-injected control group (61%). Large-scale gene expression analysis showed that 135 genes were differentially regulated in SpsiRNA-injected group compared with non-injected controls, of which 54 and 81 were down- and up-regulated respectively due to suppression of MSX1. Additionally, sequence homology mapping and gene enrichment analysis with known human pathway information identified several functional modules that were affected due to suppression of MSX1. In conclusion, suppression of MSX1 affects oocyte maturation, embryo cleavage rate and the expression of several genes, suggesting its potential role in the development of bovine preimplantation embryos.


Subject(s)
Blastocyst/physiology , Embryonic Development/genetics , Gene Expression Regulation, Developmental , MSX1 Transcription Factor/genetics , Suppression, Genetic , Animals , Cattle , Female , Fertilization in Vitro , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , MSX1 Transcription Factor/chemistry , MSX1 Transcription Factor/physiology , Male , Metaphase , Microinjections , Oligonucleotide Array Sequence Analysis , Oocytes/cytology , Oocytes/physiology , RNA, Double-Stranded , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , RNA, Small Interfering , Sequence Homology, Nucleic Acid , Time Factors , Zygote/physiology
16.
Cancer Lett ; 289(2): 195-207, 2010 Mar 28.
Article in English | MEDLINE | ID: mdl-19815336

ABSTRACT

Neuroblastoma is the most common extra-cranial solid childhood cancer; it arises from neural crest-derived cells of the sympathetic nervous system. The anomalous regulation of embryonic developmental pathways like Delta-Notch and Wnt has been implicated in aberrant cell growth and differentiation in many (childhood) tumours. We have previously found regulation of Delta-Notch pathway genes by the MSX1 neural crest development gene in a neuroblastoma cell line, and significant correlations between these genes in neuroblastic tumours. However, a clear role for the Wnt pathway in neuroblastic tumours has not yet been determined. We now analyze the complete spectrum of genes regulated by inducible expression of MSX1 in the SJNB8 neuroblastoma cell line using Affymetrix expression profiling. We show that MSX1 induces the expression of four different Wnt pathway inhibitor genes: Dickkopf 1-3 (DKK1-3) and secreted frizzled-related protein 1 (SFRP1), and provide evidence that high expression of two of these genes correlates with good prognosis. We were able to demonstrate that both the canonical Wnt3 and the alternative Wnt5A ligands are highly expressed in neuroblastic tumours and cell lines, and specifically activate the DVL3 Wnt co-receptor protein in SJNB8 neuroblastoma cells. These results suggest involvement of MSX1 in Wnt signalling and demonstrate activity of the more upstream Wnt pathway in neuroblastic cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation, Neoplastic/physiology , Intercellular Signaling Peptides and Proteins/genetics , MSX1 Transcription Factor/physiology , Neuroblastoma/genetics , Phosphoproteins/genetics , Wnt Proteins/genetics , Blotting, Northern , Blotting, Western , Chemokines , Dishevelled Proteins , Gene Expression Profiling , Humans , Luciferases/metabolism , Membrane Proteins/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , Signal Transduction , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/metabolism , Tumor Cells, Cultured , Wnt-5a Protein , Wnt3 Protein , beta Catenin/genetics , beta Catenin/metabolism
17.
Sci Signal ; 2(85): pe53, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19706870

ABSTRACT

The organization and renewal capacity of teeth vary greatly among vertebrates. Mammals have only one row of teeth that are renewed at most once, whereas many nonmammalian species have multirowed dentitions and show remarkable capacity to replace their teeth throughout life. Although knowledge on the genetic basis of tooth morphogenesis has increased exponentially over the past 20 years, little is known about the molecular mechanisms controlling sequential initiation of multiple tooth rows or restricting tooth development to one row in mammals. Mouse genetics has revealed a pivotal role for the transcription factor Osr2 in this process. Loss of Osr2 caused expansion of the expression domain of Bmp4, a well-known activator of tooth development, leading to the induction of supernumerary teeth in a manner resembling the initiation of a second tooth row in nonmammalian species.


Subject(s)
Dentition , Mice/anatomy & histology , Odontogenesis/physiology , Vertebrates/anatomy & histology , Animals , Bone Morphogenetic Protein 4/physiology , Ectoderm/physiology , Gene Expression Regulation, Developmental , Hedgehog Proteins/physiology , Homeodomain Proteins/physiology , Humans , MSX1 Transcription Factor/deficiency , MSX1 Transcription Factor/physiology , Mammals/anatomy & histology , Mammals/embryology , Mammals/genetics , Mammals/growth & development , Mesoderm/physiology , Mice/embryology , Mice/growth & development , Mice, Knockout , Odontogenesis/genetics , Regeneration/genetics , Regeneration/physiology , Species Specificity , Tooth, Supernumerary/embryology , Tooth, Supernumerary/genetics , Transcription Factors/deficiency , Transcription Factors/physiology , Vertebrates/embryology , Vertebrates/genetics , Vertebrates/growth & development , Homeobox Protein PITX2
18.
Dev Biol ; 331(2): 189-98, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19422820

ABSTRACT

In tetrapods, the anteroposterior (AP) patterning of the limb is under the control of the antagonistic activities of the secreted factor Sonic hedgehog (Shh) and Gli3R, the truncated repressor form of the transcription factor Gli3. In this report, we show that Msx1 and Msx2 are targets and downstream effectors of Gli3R. Consequently, in Shh null mutants, Msx genes are overexpressed and, furthermore, partially responsible for the limb phenotype. This is exemplified by the fact that reducing Msx activity in Shh mutants partially restores a normal limb development. Finally, we show that the main action of the Msx genes, in both normal and Shh(-/-) limb development, is to control cell death in the mesenchyme. We propose that, in the limb, Msx genes act downstream of the Shh/Gli3 pathway by transducing BMP signaling and that, in the absence of Shh signaling, their deregulation contributes to the extensive apoptosis that impairs limb development.


Subject(s)
Apoptosis/physiology , Extremities/physiology , Hedgehog Proteins/physiology , Homeodomain Proteins/physiology , Kruppel-Like Transcription Factors/physiology , MSX1 Transcription Factor/physiology , Mesoderm/physiology , Nerve Tissue Proteins/physiology , Animals , Bone Morphogenetic Proteins/physiology , Extremities/embryology , Hedgehog Proteins/genetics , Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Mesoderm/embryology , Mice , Mice, Knockout , Mutation , Zinc Finger Protein Gli3
19.
Eur J Oral Sci ; 116(2): 113-23, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18353004

ABSTRACT

Enamel formation is orchestrated by the sequential expression of genes encoding enamel matrix proteins; however, the mechanisms sustaining the spatio-temporal order of gene transcription during amelogenesis are poorly understood. The aim of this study was to characterize the cis-regulatory sequences necessary for normal expression of enamelin (Enam). Several enamelin transcription regulatory regions, showing high sequence homology among species, were identified. DNA constructs containing 5.2 or 3.9 kb regions upstream of the enamelin translation initiation site were linked to a LacZ reporter and used to generate transgenic mice. Only the 5.2-Enam-LacZ construct was sufficient to recapitulate the endogenous pattern of enamelin tooth-specific expression. The 3.9-Enam-LacZ transgenic lines showed no expression in dental cells, but ectopic beta-galactosidase activity was detected in osteoblasts. Potential transcription factor-binding sites were identified that may be important in controlling enamelin basal promoter activity and in conferring enamelin tissue-specific expression. Our study provides new insights into regulatory mechanisms governing enamelin expression.


Subject(s)
Amelogenesis/genetics , Dental Enamel Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Organ Specificity/genetics , Regulatory Sequences, Nucleic Acid/physiology , Transcription Factors/physiology , Ameloblasts/metabolism , Animals , Binding Sites , Cell Line, Transformed , Core Binding Factor Alpha 1 Subunit/physiology , Dental Enamel Proteins/biosynthesis , Genes, Reporter , MSX1 Transcription Factor/physiology , Mice , Mice, Transgenic , Promoter Regions, Genetic , beta-Galactosidase/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...