Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 326
Filtrar
Más filtros

Intervalo de año de publicación
1.
Reprod Biomed Online ; 48(6): 103815, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582043

RESUMEN

RESEARCH QUESTION: What is the effect of micro-RNA (miR)-21-5p-loaded bone marrow mesenchymal stem cell-derived exosomes (miR-21-Exo) on autoimmune premature ovarian insufficiency (POI)? DESIGN: The Cell Counting Kit 8 (CCK8) assay, fluorescence-activated cell sorting, western blotting, quantitative reverse transcriptase (qRT)-PCR and enzyme-linked immunosorbent assay (ELISA) verified the effect of miR-21-Exo on interferon-γ (IFN-γ)-induced KGN cells. qRT-PCR, western blotting and dual-luciferase reporter gene assays verified that miR-21-Exo mediated Msh homeobox 1 (MSX1) regulation of the Notch signalling pathway and that miR-21 interacted directly with MSX1. The effects of miR-21-Exo on the ovaries were verified by monitoring of the oestrous cycle, haematoxylin and eosin staining, follicle counts, ELISA, immunohistochemistry, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling (TUNEL), western blotting and qRT-PCR. RESULTS: The results showed that miR-21-Exo promoted IFN-γ-induced KGN cell proliferation and hormone synthesis, and inhibited apoptosis. Using dual-luciferase reporter gene assays, miR-21 and MSX1 were shown to have direct interactions. Moreover, the findings elucidated that miR-21-Exo inhibited cell apoptosis and promoted hormone synthesis by mediating MSX1 to regulate the Notch signalling pathway. miR-21-Exo restored the ovarian structure in a mouse model of autoimmune POI, promoted endocrine function and proliferation, and inhibited apoptosis and inflammation in vivo. CONCLUSIONS: This study demonstrates that miR-21-Exo regulates the MSX1-mediated Notch signalling pathway to inhibit granulosa cell apoptosis and improve hormone synthesis function, providing insight into a potential mechanism of molecular therapy for the treatment of autoimmune POI.


Asunto(s)
Exosomas , Factor de Transcripción MSX1 , Células Madre Mesenquimatosas , MicroARNs , Insuficiencia Ovárica Primaria , Femenino , MicroARNs/metabolismo , MicroARNs/genética , Insuficiencia Ovárica Primaria/metabolismo , Insuficiencia Ovárica Primaria/genética , Animales , Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Factor de Transcripción MSX1/metabolismo , Factor de Transcripción MSX1/genética , Humanos , Ovario/metabolismo , Enfermedades Autoinmunes/metabolismo , Apoptosis , Proliferación Celular
2.
Biochem Biophys Res Commun ; 642: 75-82, 2023 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-36566565

RESUMEN

The right and left mandibular processes derived from the first branchial arch grow toward the midline and fuse to create the rostral tip region of the mandible during mandibular development. Severe and mild cases of failure in this process results in rare median cleft of the lower lip and cleft chin, respectively. The detailed molecular mechanisms of mandibular tip formation are unknown. We hypothesize that the Msx1 gene is involved in mandibular tip development, because Msx1 has a central role in other craniofacial morphogenesis processes, such as teeth and the secondary palate development. Normal Msx1 expression was observed in the rostral end of the developing mandible; however, a reduced expression of Msx1 was observed in the soft tissue of the mandibular tip than in the lower incisor bud region. The rostral tip of the right and left mandibular processes was unfused in both control and Msx1-null (Msx1-/-) mice at embryonic day (E) 12.5; however, a complete fusion of these processes was observed at E13.5 in the control. The fused processes exhibited a conical shape in the control, whereas the same region remained bifurcated in Msx1-/-. This phenotype occurred with 100% penetrance and was not restored at subsequent stages of development. Furthermore, Meckel's cartilage in addition to the outline surface soft tissues was also unfused and bifurcated in Msx1-/- from E14.5 onward. The expression of phosho-Smad1/5, which is a mediator of bone morphogenetic protein (Bmp) signaling, was downregulated in the mandibular tip of Msx1-/- at E12.5 and E13.5, probably due to the downregulated Bmp4 expression in the neighboring lower incisor bud. Cell proliferation was significantly reduced in the midline region of the mandibular tip in Msx1-/- at the same developmental stages in which downregulation of pSmad was observed. Our results indicate that Msx1 is indispensable for proper mandibular tip development.


Asunto(s)
Factor de Transcripción MSX1 , Diente , Ratones , Animales , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Mandíbula , Diente/metabolismo , Morfogénesis/genética , Transducción de Señal
3.
Development ; 147(21)2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32467233

RESUMEN

Nonsyndromic clefts of the lip and palate are common birth defects resulting from gene-gene and gene-environment interactions. Mutations in human MSX1 have been linked to orofacial clefting and we show here that Msx1 deficiency causes a growth defect of the medial nasal process (Mnp) in mouse embryos. Although this defect alone does not disrupt lip formation, Msx1-deficient embryos develop a cleft lip when the mother is transiently exposed to reduced oxygen levels or to phenytoin, a drug known to cause embryonic hypoxia. In the absence of interacting environmental factors, the Mnp growth defect caused by Msx1 deficiency is modified by a Pax9-dependent 'morphogenetic regulation', which modulates Mnp shape, rescues lip formation and involves a localized abrogation of Bmp4-mediated repression of Pax9 Analyses of GWAS data revealed a genome-wide significant association of a Gene Ontology morphogenesis term (including assigned roles for MSX1, MSX2, PAX9, BMP4 and GREM1) specifically for nonsyndromic cleft lip with cleft palate. Our data indicate that MSX1 mutations could increase the risk for cleft lip formation by interacting with an impaired morphogenetic regulation that adjusts Mnp shape, or through interactions that inhibit Mnp growth.


Asunto(s)
Hipoxia/embriología , Hipoxia/metabolismo , Labio/embriología , Factor de Transcripción MSX1/deficiencia , Morfogénesis , Animales , Proteína Morfogenética Ósea 4/metabolismo , Labio Leporino/embriología , Labio Leporino/genética , Labio Leporino/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Genoma , Proteínas de Homeodominio/metabolismo , Humanos , Hipoxia/genética , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Morfogénesis/genética , Mutación/genética , Nariz/embriología , Oxígeno/metabolismo , Factor de Transcripción PAX9/metabolismo , Fenitoína , Respiración , Regulación hacia Arriba/genética
4.
Development ; 147(14)2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32665245

RESUMEN

Amputation of a salamander limb triggers a regeneration process that is perfect. A limited number of genes have been studied in this context and even fewer have been analyzed functionally. In this work, we use the BMP signaling inhibitor LDN193189 on Ambystoma mexicanum to explore the role of BMPs in regeneration. We find that BMP signaling is required for proper expression of various patterning genes and that its inhibition causes major defects in the regenerated limbs. Fgf8 is downregulated when BMP signaling is blocked, but ectopic injection of either human or axolotl protein did not rescue the defects. By administering LDN193189 treatments at different time points during regeneration, we show clearly that limb regeneration progresses in a proximal to distal fashion. This demonstrates that BMPs play a major role in patterning of regenerated limbs and that regeneration is a progressive process like development.


Asunto(s)
Ambystoma mexicanum/metabolismo , Proteínas Anfibias/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Extremidades/fisiología , Regeneración/fisiología , Transducción de Señal , Ambystoma mexicanum/crecimiento & desarrollo , Proteínas Anfibias/genética , Animales , Proteínas Morfogenéticas Óseas/genética , Proliferación Celular/efectos de los fármacos , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Regeneración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo
5.
Oral Dis ; 29(8): 3168-3172, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36478500

RESUMEN

OBJECTIVE: Muscle segment homeobox gene 1 (MSX1) is widely expressed in craniofacial development and tooth formation. The aim of this study was to report a novel MSX1 mutation in a Chinese family with selective tooth agenesis and abnormal median maxillary labial frenum (MMLF). MATERIALS AND METHODS: Mutation analysis was carried out by whole exome sequencing. The pMD18-T vector was used to verify the mutations. PubMed and Human Gene Mutation Database were searched to analyze the relationship between the mutations in MSX1 and related phenotypes. RESULTS: A novel heterozygous mutation (c.75delG) in MSX1 was detected in the proband and her mother. They presented as oligodontia and lower attached hypertrophy median maxillary labial frenum. 60 MSX1 mutations from 39 reports did not declare malformed MMLF except our cases. Meanwhile, we found that the types and sites of MSX1 mutations may affect the selectivity of tooth agenesis and orofacial cleft. CONCLUSION: This study suggests malformed MMLF as a new phenotype of MSX1 mutation and a specific relationship between MSX1 genotype and phenotype.


Asunto(s)
Anodoncia , Labio Leporino , Fisura del Paladar , Humanos , Femenino , Estudios Retrospectivos , Frenillo Labial , Labio Leporino/genética , Linaje , Anodoncia/genética , Mutación , Factor de Transcripción MSX1/genética
6.
Arch Gynecol Obstet ; 308(1): 273-279, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37101223

RESUMEN

PURPOSE: The human endometrium consists of different layers (basalis and functionalis) and undergoes different phases throughout the menstrual cycle. In a former paper, our research group was able to describe MSX1 as a positive prognosticator in endometrial carcinomas. The aim of this study was to examine the MSX1 expression in healthy endometrial tissue throughout the different phases to gain more insight on the mechanics of MSX-regulation in the female reproductive system. MATERIALS AND METHODS: In this retrospective study, we investigated a total of 17 normal endometrial tissues (six during proliferative phase and five during early and six during late secretory phase). We used immunohistochemical staining and an immunoreactive score (IRS) to evaluate MSX1 expression. We also investigated correlations with other proteins, that have already been examined in our research group using the same patient collective. RESULTS: MSX1 is expressed in glandular cells during the proliferative phase and downregulated at early and late secretory phase (p = 0.011). Also, a positive correlation between MSX1 and the progesterone-receptor A (PR-A) (correlation coefficient (cc) = 0.0671; p = 0.024), and the progesterone receptor B (PR-B) (cc = 0.0691; p = 0.018) was found. A trend towards negative correlation was recognized between MSX1 and Inhibin Beta-C-expression in glandular cells (cc = - 0.583; p-value = 0.060). CONCLUSION: MSX1 is known as a member of the muscle segment homeobox gene family. MSX1 is a p53-interacting protein and overexpression of homeobox MSX1 induced apoptosis of cancer cells. Here we show that MSX1 is expressed especially in the proliferative phase of glandular epithelial tissue of the normal endometrium. The found positive correlation between MSX1 and progesterone receptors A and B confirms the results of a previous study on cancer tissue by our research group. Because MSX1 is known to be downregulated by progesterone, the found correlation of MSX1 and both PR-A and -B may represent a direct regulation of the MSX1 gene by a PR-response element. Here further investigation would be of interest.


Asunto(s)
Neoplasias Endometriales , Progesterona , Humanos , Femenino , Progesterona/metabolismo , Estudios Retrospectivos , Endometrio/metabolismo , Ciclo Menstrual/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo
7.
Am J Orthod Dentofacial Orthop ; 163(3): 368-377, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36494218

RESUMEN

INTRODUCTION: Morphologic features of the mandible are influenced by the genes of each individual. Mandible size is important to orthodontists because the mandible is the mechanism by which the lower face influences facial esthetics and dental function. To date, no biological marker has been identified that indicates eventual mandible size. This study aimed to correlate the expression of DLX5, DLX6, EDN1, HAND2, PRRX1, and MSX1 to mandible size. METHODS: Fifty-nine orthodontic patients aged >6 years who had available cephalometric radiographs were studied. Patients were classified on the basis of condylion-to-gnathion measurements. Messenger RNA was isolated from saliva and subjected to real-time quantitative polymerase chain reaction. RESULTS: Threshold cycle values for subjects with small mandibles (>1 standard deviation [SD] from the mean) had the least expression of DLX6 and MSX1. Threshold cycle values for subjects with large mandibles (>1 SD) had less expression of DLX6 and MSX1 than subjects within 1 SD but more than those with small mandibles. CONCLUSIONS: DLX6 and MSX1 are related to mandible development and size. This finding could be used to improve treatment planning for medical and dental professionals seeking to understand the impact of genetics on bone growth.


Asunto(s)
Maloclusión de Angle Clase III , Saliva , Humanos , Estudios Transversales , Mandíbula , Cefalometría , Proteínas de Homeodominio/metabolismo , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo
8.
Nucleic Acids Res ; 48(20): 11452-11467, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33080014

RESUMEN

Msh homeobox (Msx) is a subclass of homeobox transcriptional regulators that control cell lineage development, including the early stage of vertebrate limb development, although the underlying mechanisms are not clear. Here, we demonstrate that Msx1 promotes the proliferation of myoblasts and mesenchymal stem cells (MSCs) by enhancing mitogen-activated protein kinase (MAPK) signaling. Msx1 directly binds to and upregulates the expression of fibroblast growth factor 9 (Fgf9) and Fgf18. Accordingly, knockdown or antibody neutralization of Fgf9/18 inhibits Msx1-activated extracellular signal-regulated kinase 1/2 (Erk1/2) phosphorylation. Mechanistically, we determined that the phosphorylation of Msx1 at Ser136 is critical for enhancing Fgf9 and Fgf18 expression and cell proliferation, and cyclin-dependent kinase 1 (CDK1) is apparently responsible for Ser136 phosphorylation. Furthermore, mesenchymal deletion of Msx1/2 results in decreased Fgf9 and Fgf18 expression and Erk1/2 phosphorylation, which leads to serious defects in limb development in mice. Collectively, our findings established an important function of the Msx1-Fgf-MAPK signaling axis in promoting cell proliferation, thus providing a new mechanistic insight into limb development.


Asunto(s)
Proliferación Celular , Extremidades/embriología , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Sistema de Señalización de MAP Quinasas , Factor de Transcripción MSX1/metabolismo , Animales , Proteína Quinasa CDC2/metabolismo , Línea Celular , Factor 9 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/genética , Factor de Transcripción MSX1/química , Factor de Transcripción MSX1/genética , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Mioblastos/citología , Mioblastos/enzimología , Mioblastos/metabolismo , Fosforilación , Serina/metabolismo
9.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35055037

RESUMEN

The osteoblast differentiation capacity of mesenchymal stem cells must be tightly regulated, as inadequate bone mineralization can lead to osteoporosis, and excess bone formation can cause the heterotopic ossification of soft tissues. The balanced protein level of Msh homeobox 1 (MSX1) is critical during normal osteogenesis. To understand the factors that prevent MSX1 protein degradation, the identification of deubiquitinating enzymes (DUBs) for MSX1 is essential. In this study, we performed loss-of-function-based screening for DUBs regulating MSX1 protein levels using the CRISPR/Cas9 system. We identified ubiquitin-specific protease 11 (USP11) as a protein regulator of MSX1 and further demonstrated that USP11 interacts and prevents MSX1 protein degradation by its deubiquitinating activity. Overexpression of USP11 enhanced the expression of several osteogenic transcriptional factors in human mesenchymal stem cells (hMSCs). Additionally, differentiation studies revealed reduced calcification and alkaline phosphatase activity in USP11-depleted cells, while overexpression of USP11 enhanced the differentiation potential of hMSCs. These results indicate the novel role of USP11 during osteogenic differentiation and suggest USP11 as a potential target for bone regeneration.


Asunto(s)
Sistemas CRISPR-Cas , Diferenciación Celular/genética , Enzimas Desubicuitinizantes/genética , Estudio de Asociación del Genoma Completo/métodos , Osteogénesis/genética , Tioléster Hidrolasas/genética , Enzimas Desubicuitinizantes/metabolismo , Regulación de la Expresión Génica , Humanos , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteolisis , Medicina Regenerativa , Tioléster Hidrolasas/metabolismo , Factores de Transcripción/metabolismo , Ubiquitinación
10.
BMC Oral Health ; 22(1): 16, 2022 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-35065635

RESUMEN

BACKGROUND: This study aimed to investigate the genetic association of specific Single Nucleotide Polymorphisms (SNPs) within the muscle segment homeobox gene 1 (MSX1) with susceptibility to the peg-shaped teeth in 36 Jordanian Arab families and case-control samples in the Jordanian Arab population. METHODS: This cohort involved 108 individuals (36 trios families), which were used for family-based genetic study. Additionally, 56 patients and 57 controls were used for case-control study. Genomic DNA samples from both families and case-control were extracted according to distinguished processes. Then, polymerase chain reaction technique (PCR) was conducted using specific primers for the axons of the MSX1. Moreover, DNA sequencing genotyping method analysis of SNPs was used to detect specified SNPs in the MSX1 linked with peg-shaped teeth. Hardy-Weinberg Equilibrium and Chi-square were used to evaluate the data quality and the presence of any genotypic error. In addition, Transmission Disequilibrium Test (TDT) was used identify family-based association in which trios of parents and proband are used. RESULTS: The results of this study showed fourteen polymorphic sites in this gene, eight of them (rs121913129, rs104893852, rs104893853, rs121913130, rs104893850, rs1095, rs3775261, and rs1042484) were none-polymorphic. Meanwhile, the minor allele frequencies of the rest of the SNPs were polymorphic (rs8670, rs12532, rs3821949, rs4464513, rs1907998, and rs6446693). However, none of these SNPs were associated with peg-shaped teeth. Moreover, the haplotype genetic analysis revealed that there was no genetic association with peg-shaped teeth disorder susceptibility (P > 0.05) in the Jordanian families of Arab descent. CONCLUSIONS: The present findings can be used in estimation of prevalence of peg-shaped teeth in the Jordanian population. However, our findings revealed that there is no evidence that the MSX1 polymorphisms had a crucial role in the peg-shaped teeth phenomenon, emphasizing that other genes might have this role. These findings are beneficial for clinicians to comprehensively understand the molecular aspects of teeth abnormalities.


Asunto(s)
Factor de Transcripción MSX1 , Anomalías Dentarias , Estudios de Casos y Controles , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Jordania , Factor de Transcripción MSX1/genética , Polimorfismo de Nucleótido Simple/genética , Anomalías Dentarias/genética
11.
BMC Dev Biol ; 21(1): 14, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34615475

RESUMEN

BACKGROUND: Successful embryogenesis relies on the coordinated interaction between genes and tissues. The transcription factors Pax9 and Msx1 genetically interact during mouse craniofacial morphogenesis, and mice deficient for either gene display abnormal tooth and palate development. Pax9 is expressed specifically in the pharyngeal endoderm at mid-embryogenesis, and mice deficient for Pax9 on a C57Bl/6 genetic background also have cardiovascular defects affecting the outflow tract and aortic arch arteries giving double-outlet right ventricle, absent common carotid arteries and interruption of the aortic arch. RESULTS: In this study we have investigated both the effect of a different genetic background and Msx1 haploinsufficiency on the presentation of the Pax9-deficient cardiovascular phenotype. Compared to mice on a C57Bl/6 background, congenic CD1-Pax9-/- mice displayed a significantly reduced incidence of outflow tract defects but aortic arch defects were unchanged. Pax9-/- mice with Msx1 haploinsufficiency, however, have a reduced incidence of interrupted aortic arch, but more cases with cervical origins of the right subclavian artery and aortic arch, than seen in Pax9-/- mice. This alteration in arch artery defects was accompanied by a rescue in third pharyngeal arch neural crest cell migration and smooth muscle cell coverage of the third pharyngeal arch arteries. Although this change in phenotype could theoretically be compatible with post-natal survival, using tissue-specific inactivation of Pax9 to maintain correct palate development whilst inducing the cardiovascular defects was unable to prevent postnatal death in the mutant mice. Hyoid bone and thyroid cartilage formation were abnormal in Pax9-/- mice. CONCLUSIONS: Msx1 haploinsufficiency mitigates the arch artery defects in Pax9-/- mice, potentially by maintaining the survival of the 3rd arch artery through unimpaired migration of neural crest cells to the third pharyngeal arches. With the neural crest cell derived hyoid bone and thyroid cartilage also being defective in Pax9-/- mice, we speculate that the pharyngeal endoderm is a key signalling centre that impacts on neural crest cell behaviour highlighting the ability of cells in different tissues to act synergistically or antagonistically during embryo development.


Asunto(s)
Sistema Cardiovascular , Haploinsuficiencia , Factor de Transcripción MSX1 , Animales , Región Branquial , Factor de Transcripción MSX1/genética , Ratones , Ratones Noqueados , Cresta Neural , Factor de Transcripción PAX9 , Fenotipo
12.
Clin Genet ; 99(4): 493-502, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33249565

RESUMEN

Like all developmental processes, odontogenesis is highly complex and dynamically regulated, with hundreds of genes co-expressed in reciprocal networks. Tooth agenesis (missing one or more/all teeth) is a common human craniofacial anomaly and may be caused by genetic variations and/or environmental factors. Variants in PAX9, MSX1, AXIN2, EDA, EDAR, and WNT10A genes are associated with tooth agenesis. Currently, variants in ATF1, DUSP10, CASC8, IRF6, KDF1, GREM2, LTBP3, and components and regulators of WNT signaling WNT10B, LRP6, DKK, and KREMEN1 are at the forefront of interest. Due to the interconnectedness of the signaling pathways of carcinogenesis and odontogenesis, tooth agenesis could be a suitable marker for early detection of cancer predisposition. Variants in genes associated with tooth agenesis could serve as prognostic or therapeutic targets in cancer. This review aims to summarize existing knowledge of development and clinical genetics of teeth. Concurrently, the review proposes possible approaches for future research in this area, with particular attention to roles in monitoring, early diagnosis and therapy of tumors associated with defective tooth development.


Asunto(s)
Anodoncia/genética , Neoplasias/genética , Anodoncia/epidemiología , Biomarcadores de Tumor , Carcinogénesis , Carcinoma/epidemiología , Carcinoma/genética , Neoplasias Colorrectales/epidemiología , Neoplasias Colorrectales/genética , Detección Precoz del Cáncer , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Factor de Transcripción MSX1/genética , Neoplasias/epidemiología , Síndromes Neoplásicos Hereditarios/epidemiología , Síndromes Neoplásicos Hereditarios/genética , Odontogénesis , Neoplasias Ováricas/epidemiología , Neoplasias Ováricas/genética , Factor de Transcripción PAX9/genética , Transducción de Señal/genética , Neoplasias Gástricas/epidemiología , Neoplasias Gástricas/genética , Decoloración de Dientes , Vía de Señalización Wnt/genética
13.
Protein Expr Purif ; 179: 105797, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33242573

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Factor de Transcripción MSX1 , Mioblastos , Animales , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Técnicas de Inactivación de Genes , Factor de Transcripción MSX1/química , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Factor de Transcripción MSX1/fisiología , Ratones , Mioblastos/citología , Mioblastos/metabolismo
14.
Int J Mol Sci ; 22(11)2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071193

RESUMEN

Cutaneous melanoma represents one of the deadliest types of skin cancer. The prognosis strongly depends on the disease stage, thus early detection is crucial. New therapies, including BRAF and MEK inhibitors and immunotherapies, have significantly improved the survival of patients in the last decade. However, intrinsic and acquired resistance is still a challenge. In this review, we discuss two major aspects that contribute to the aggressiveness of melanoma, namely, the embryonic origin of melanocytes and melanoma cells and cellular plasticity. First, we summarize the physiological function of epidermal melanocytes and their development from precursor cells that originate from the neural crest (NC). Next, we discuss the concepts of intratumoral heterogeneity, cellular plasticity, and phenotype switching that enable melanoma to adapt to changes in the tumor microenvironment and promote disease progression and drug resistance. Finally, we further dissect the connection of these two aspects by focusing on the transcriptional regulators MSX1, MITF, SOX10, PAX3, and FOXD3. These factors play a key role in NC initiation, NC cell migration, and melanocyte formation, and we discuss how they contribute to cellular plasticity and drug resistance in melanoma.


Asunto(s)
Plasticidad de la Célula/fisiología , Resistencia a Antineoplásicos/fisiología , Melanoma/metabolismo , Cresta Neural/metabolismo , Neoplasias Cutáneas/metabolismo , Acrilonitrilo/análogos & derivados , Acrilonitrilo/farmacología , Compuestos de Anilina/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Diferenciación Celular , Movimiento Celular , Resistencia a Antineoplásicos/genética , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor de Transcripción MSX1/genética , Melanocitos/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción PAX3/genética , Fenotipo , Pirimidinonas/farmacología , Factores de Transcripción SOXE/genética , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología
15.
Cleft Palate Craniofac J ; 58(6): 697-706, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34047208

RESUMEN

OBJECTIVE: Cleft palate is among the most frequent congenital defects in humans. While gene-environment multifactorial threshold models have been proposed to explain this cleft palate formation, only a few experimental models have verified this theory. This study aimed to clarify whether gene-environment interaction can cause cleft palate through a combination of specific genetic and environmental factors. METHODS: Msx1 heterozygosity in mice (Msx1+/-) was selected as a genetic factor since human MSX1 gene mutations may cause nonsyndromic cleft palate. As an environmental factor, hypoxic stress was induced in pregnant mice by administration of the antiepileptic drug phenytoin, a known arrhythmia inducer, during palatal development from embryonic day (E) 11 to E14. Embryos were dissected at E13 for histological analysis or at E17 for recording of the palatal state. RESULTS: Phenytoin administration downregulated cell proliferation in palatal processes in both wild-type and Msx1+/- embryos. Bone morphogenetic protein 4 (Bmp4) expression was slightly downregulated in the anterior palatal process of Msx1+/- embryos. Although Msx1+/- embryos do not show cleft palate under normal conditions, phenytoin administration induced a significantly higher incidence of cleft palate in Msx1+/- embryos compared to wild-type littermates. CONCLUSION: Our data suggest that cleft palate may occur because of the additive effects of Bmp4 downregulation as a result of Msx1 heterozygosity and decreased cell proliferation upon hypoxic stress. Human carriers of MSX1 mutations may have to take more precautions during pregnancy to avoid exposure to environmental risks.


Asunto(s)
Fisura del Paladar , Factor de Transcripción MSX1 , Estrés Oxidativo , Animales , Fisura del Paladar/inducido químicamente , Fisura del Paladar/genética , Factor de Transcripción MSX1/genética , Ratones , Hueso Paladar , Fenitoína , Transducción de Señal
16.
Eur J Orthod ; 43(4): 478-485, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-33200192

RESUMEN

OBJECTIVE: Verify the presence of association between four variables-transforming growth factor α (TGFA; C/T rs1523305), interferon regulatory factor 6 (IRF6; A/C rs2013162), muscle segment homeobox 1 (MSX1; A/G rs12532), and dental anomalies-with skeletal malocclusion by comparing these four variables with Angle Classes I, II, and III, and normal, hyperdivergent, and hypodivergent growth patterns. METHODS: A total of 505 orthodontic records of patients older than 8 years were evaluated. The sample consisted of 285 (56.4 per cent) females, 220 (43.6 per cent) males, 304 (60.2 per cent) Whites (the rest were mixed Blacks with Whites), with a mean age of 20.28 (±10.35) years (ranging from 8 to 25 years). Eight cephalometric points, which served as the anatomical framework for obtaining angles and cephalometric measurements, were used for skeletal characterization using the Dolphin Software. Samples of saliva were collected and the DNA was extracted, diluted and quantified. Markers in TGFA, IRF6, and MSX1 were used and genotypes were obtained using TaqMan chemistry. Odds ratio (OR) and 95 per cent confidence interval (CI) calculations, chi-square, Fisher's Exact, Mann-Whitney, and correlation coefficient tests (significance level: 95 per cent) were performed. Bonferroni correction was applied and an alpha of 0.0006 was considered statistically significant. RESULTS: There was no statistically significant associations between markers in TGFA or IRF6 with skeletal malocclusions. Tooth agenesis was associated with facial convexity (P < 0.001). MSX1 was associated with Class II skeletal malocclusion (P = 0.0001, OR = 0.6, CI = 0.46-0.78). CONCLUSION: Individuals with tooth agenesis were more likely to have a convex face. MSX1 was associated with Class II skeletal malocclusion.


Asunto(s)
Maloclusión Clase II de Angle , Maloclusión Clase I de Angle , Maloclusión , Cefalometría , Femenino , Humanos , Factores Reguladores del Interferón , Factor de Transcripción MSX1/genética , Masculino , Factor de Crecimiento Transformador alfa
17.
Ann Hum Genet ; 84(1): 97-101, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31469409

RESUMEN

Mutations of MSX1 have been associated with nonsyndromic hypodontia. To seek the causal gene mutation sites in a family with nonsyndromic oligodontia, whole-exome sequencing (WES) was performed to seek the causative locus of the family. The candidate mutation was further identified by Sanger sequencing afterward. Two mutations of MSX1 were found both in the proband and her mother. One novel heterozygous missense mutation (c.C667G, p.R223G) of MSX1 inherited from the asymptomatic mother with mosaic mutation was located in the highly conserved fragment of exon 2. The other was a synonymous mutation (c.C348T, p.G116G) in exon 1, which had been reported. The novel maternal heterozygous missense mutation (c.C667G, p.R223G) was likely to be the major reason for nonsyndromic oligodontia in the family. This is the first mosaic variant that has been recorded of the MSX1 gene. Our study expands the phenotype-genotype correlation associated with MSX1 variants. Our study also suggests that the determination of the mosaicism is essential for precise genetic counseling if a disease appears to arise de novo.


Asunto(s)
Anodoncia/etiología , Heterocigoto , Factor de Transcripción MSX1/genética , Mosaicismo , Mutación , Anodoncia/patología , Niño , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Linaje , Fenotipo
18.
Hum Genet ; 139(10): 1261-1272, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32318854

RESUMEN

Nonsyndromic cleft lip with or without cleft palate (NSCLP) is a common birth defect for which only ~ 20% of the underlying genetic variation has been identified. Variants in noncoding regions have been increasingly suggested to contribute to the missing heritability. In this study, we investigated whether variation in craniofacial enhancers contributes to NSCLP. Candidate enhancers were identified using VISTA Enhancer Browser and previous publications. Prioritization was based on patterning defects in knockout mice, deletion/duplication of craniofacial genes in animal models and results of whole exome/whole genome sequencing studies. This resulted in 20 craniofacial enhancers to be investigated. Custom amplicon-based sequencing probes were designed and used for sequencing 380 NSCLP probands (from multiplex and simplex families of non-Hispanic white (NHW) and Hispanic ethnicities) using Illumina MiSeq. The frequencies of identified variants were compared to ethnically matched European (CEU) and Los Angeles Mexican (MXL) control genomes and used for association analyses. Variants in mm427/MSX1 and hs1582/SPRY1 showed genome-wide significant association with NSCLP (p ≤ 6.4 × 10-11). In silico analysis showed that these enhancer variants may disrupt important transcription factor binding sites. Haplotypes involving these enhancers and also mm435/ABCA4 were significantly associated with NSCLP, especially in NHW (p ≤ 6.3 × 10-7). Importantly, groupwise burden analysis showed several enhancer combinations significantly over-represented in NSCLP individuals, revealing novel NSCLP pathways and supporting a polygenic inheritance model. Our findings support the role of craniofacial enhancer sequence variation in the etiology of NSCLP.


Asunto(s)
Labio Leporino/genética , Fisura del Paladar/genética , Elementos de Facilitación Genéticos , Predisposición Genética a la Enfermedad , Variación Genética , Herencia Multifactorial , Transportadoras de Casetes de Unión a ATP/genética , Animales , Enfermedades Asintomáticas , Labio Leporino/etnología , Labio Leporino/patología , Fisura del Paladar/etnología , Fisura del Paladar/patología , Embrión de Mamíferos , Femenino , Estudios de Asociación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Hispánicos o Latinos , Humanos , Factor de Transcripción MSX1/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Linaje , Fosfoproteínas/genética , Estados Unidos , Población Blanca
19.
Int J Mol Sci ; 21(12)2020 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-32630554

RESUMEN

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.


Asunto(s)
Neoplasias Endometriales/metabolismo , Factor de Transcripción MSX1/metabolismo , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patología , Metilación de ADN/genética , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Femenino , Humanos , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/fisiología , Persona de Mediana Edad , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias Uterinas/patología , Útero/metabolismo , Útero/patología
20.
J Neurosci ; 38(7): 1662-1676, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29321139

RESUMEN

The embryonic formation of midbrain dopaminergic (mDA) neurons in vivo provides critical guidelines for the in vitro differentiation of mDA neurons from stem cells, which are currently being developed for Parkinson's disease cell replacement therapy. Bone morphogenetic protein (BMP)/SMAD inhibition is routinely used during early steps of stem cell differentiation protocols, including for the generation of mDA neurons. However, the function of the BMP/SMAD pathway for in vivo specification of mammalian mDA neurons is virtually unknown. Here, we report that BMP5/7-deficient mice (Bmp5-/-; Bmp7-/-) lack mDA neurons due to reduced neurogenesis in the mDA progenitor domain. As molecular mechanisms accounting for these alterations in Bmp5-/-; Bmp7-/- mutants, we have identified expression changes of the BMP/SMAD target genes MSX1/2 (msh homeobox 1/2) and SHH (sonic hedgehog). Conditionally inactivating SMAD1 in neural stem cells of mice in vivo (Smad1Nes) hampered the differentiation of progenitor cells into mDA neurons by preventing cell cycle exit, especially of TH+SOX6+ (tyrosine hydroxylase, SRY-box 6) and TH+GIRK2+ (potassium voltage-gated channel subfamily-J member-6) substantia nigra neurons. BMP5/7 robustly increased the in vitro differentiation of human induced pluripotent stem cells and induced neural stem cells to mDA neurons by up to threefold. In conclusion, we have identified BMP/SMAD signaling as a novel critical pathway orchestrating essential steps of mammalian mDA neurogenesis in vivo that balances progenitor proliferation and differentiation. Moreover, we demonstrate the potential of BMPs to improve the generation of stem-cell-derived mDA neurons in vitro, highlighting the importance of sequential BMP/SMAD inhibition and activation in this process.SIGNIFICANCE STATEMENT We identify bone morphogenetic protein (BMP)/SMAD signaling as a novel essential pathway regulating the development of mammalian midbrain dopaminergic (mDA) neurons in vivo and provide insights into the molecular mechanisms of this process. BMP5/7 regulate MSX1/2 (msh homeobox 1/2) and SHH (sonic hedgehog) expression to direct mDA neurogenesis. Moreover, the BMP signaling component SMAD1 controls the differentiation of mDA progenitors, particularly to substantia nigra neurons, by directing their cell cycle exit. Importantly, BMP5/7 increase robustly the differentiation of human induced pluripotent and induced neural stem cells to mDA neurons. BMP/SMAD are routinely inhibited in initial stages of stem cell differentiation protocols currently being developed for Parkinson's disease cell replacement therapies. Therefore, our findings on opposing roles of the BMP/SMAD pathway during in vitro mDA neurogenesis might improve these procedures significantly.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Neuronas Dopaminérgicas/fisiología , Mesencéfalo/fisiología , Células-Madre Neurales , Neurogénesis/fisiología , Células Madre Pluripotentes , Transducción de Señal/fisiología , Proteínas Smad/fisiología , Animales , Proteína Morfogenética Ósea 5/genética , Proteína Morfogenética Ósea 5/metabolismo , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/metabolismo , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Mesencéfalo/citología , Ratones , Ratones Noqueados , Proteína Smad1/genética , Proteína Smad1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA