Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Genet ; 15: 1391804, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39221224

RESUMEN

17q12 deletion syndrome is a chromosomal abnormality, where there is a small missing piece (deletion) of genetic material on the long arm (q) of chromosome 17. Sign and symptoms can vary widely among different patients. Recently, a patient was diagnosed with 17q12 deletion syndrome in our hospital, and the clinical characteristics presented as absence of the right kidney, compensatory hypertrophy of the left kidney, multiple small cysts in the left kidney, pancreatic atrophy, hypomagnesemia, bowed uterus, multiple follicular cysts in both lobes of the thyroid gland, and maturity-onset diabetes of the young type 5 (MODY-5). A 1.5-Mb deletion with haploinsufficiency for 20 genes within the 17q12 region was found through copy number variation (CNV) analysis based on metagenomic next-generation sequencing (mNGS) technology. In addition to HNF1B absence, the LIM-class homeobox 1 transcription factor (LHX1) and GGNBP2 absence was also involved in regulation of kidney development and the reproductive system through bioinformatics analysis. The inheriting risk of 17q12 deletion syndrome is about 50%, and it is recommended to provide genetic counseling to all patients who are suspected or diagnosed with the syndrome.

2.
Heliyon ; 10(13): e32457, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39027525

RESUMEN

Background: Premature birth (PTB) is a major cause of neonatal mortality and has enduring consequences. LIM Homeobox 1 (LHX1) is vital in embryonic organogenesis, while Inositol-Requiring Enzyme 1 (IRE-1) regulates endoplasmic reticulum stress (ERS). This study explores whether IRE-1 impacts PTB via LHX1 modulation. Methods: We analyzed LHX1 expression in placental samples from PTB patients and examined its impact on the viability, migration, invasion, and apoptosis of the human placental trophoblast cell line HTR8/Svneo, particularly when treated with the ERS inducer tunicamycin (TM). We also assessed the levels of ERS-related genes and autophagy activation in response to LHX1 deficiency. To gain mechanistic insights, we evaluated the ERS-mediated activation of the IRE-1/XBP1/CHOP signaling pathway in LHX1-silenced HTR8/Svneo cells. Additionally, we examined the transcriptional activation of IRE-1 and the binding of LHX1 to the IRE-1 promoter in HTR8/Svneo cells. We overexpressed IRE-1 in LHX1-silenced HTR8/Svneo cells to assess its effects on cell viability, migration, invasion, apoptosis, and autophagy. Finally, we induced LHX1 knockdown in mice through intraperitoneal injections of tunicamycin (TM) and Sh-LHX1 over a 24-h period to evaluate PTB symptoms. Results: We observed LHX1 overexpression in placental tissue from PTB cases and TM-induced HTR8/Svneo cells. LHX1 depletion enhanced cell viability, migration, and invasion while reducing autophagy and apoptosis. This reduction in LHX1 led to decreased levels of IRE-1, XBP1, CHOP, and other ERS-related genes, indicating LHX1's role in ERS induction and the activation of the IRE-1/XBP1/CHOP pathway. Mechanistically, LHX1 was found to bind to the IRE-1 promoter, inducing its transcriptional activation. Notably, overexpressing IRE-1 counteracted the impact of LHX1 depletion on trophoblast cell behavior, suggesting that LHX1 modulates IRE-1. In line with our in vitro studies, LHX1 knockdown ameliorated PTB symptoms in TM-treated mice. Conclusion: LHX1 contributes to the progression of PTB by regulating the IRE-1-XBP1-CHOP pathway.

3.
Stem Cell Reports ; 19(7): 973-992, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38942030

RESUMEN

Genetic differences between pluripotent stem cell lines cause variable activity of extracellular signaling pathways, limiting reproducibility of directed differentiation protocols. Here we used human embryonic stem cells (hESCs) to interrogate how exogenous factors modulate endogenous signaling events during specification of foregut endoderm lineages. We find that transforming growth factor ß1 (TGF-ß1) activates a putative human OTX2/LHX1 gene regulatory network which promotes anterior fate by antagonizing endogenous Wnt signaling. In contrast to Porcupine inhibition, TGF-ß1 effects cannot be reversed by exogenous Wnt ligands, suggesting that induction of SHISA proteins and intracellular accumulation of Fzd receptors render TGF-ß1-treated cells refractory to Wnt signaling. Subsequently, TGF-ß1-mediated inhibition of BMP and Wnt signaling suppresses liver fate and promotes pancreas fate. Furthermore, combined TGF-ß1 treatment and Wnt inhibition during pancreatic specification reproducibly and robustly enhance INSULIN+ cell yield across hESC lines. This modification of widely used differentiation protocols will enhance pancreatic ß cell yield for cell-based therapeutic applications.


Asunto(s)
Proteínas Morfogenéticas Óseas , Diferenciación Celular , Endodermo , Células Madre Embrionarias Humanas , Vía de Señalización Wnt , Humanos , Endodermo/citología , Endodermo/metabolismo , Diferenciación Celular/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
4.
J Cell Sci ; 136(11)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37305998

RESUMEN

Besides assembling nuclear pore complexes, the conduits of nuclear transport, many nucleoporins also contribute to chromatin organization and gene expression, with critical roles in development and pathologies. We previously reported that Nup133 and Seh1, two components of the Y-complex subassembly of the nuclear pore scaffold, are dispensable for mouse embryonic stem cell viability but required for their survival during neuroectodermal differentiation. Here, a transcriptomic analysis revealed that Nup133 regulates a subset of genes at early stages of neuroectodermal differentiation, including Lhx1 and Nup210l, which encodes a newly validated nucleoporin. These genes are also misregulated in Nup133ΔMid neuronal progenitors, in which nuclear pore basket assembly is impaired. However, a four-fold reduction of Nup133 levels, despite also affecting basket assembly, is not sufficient to alter Nup210l and Lhx1 expression. Finally, these two genes are also misregulated in Seh1-deficient neural progenitors, which only show a mild reduction in nuclear pore density. Together these data reveal a shared function of Y-complex nucleoporins in gene regulation during neuroectodermal differentiation, apparently independent of nuclear pore basket integrity.


Asunto(s)
Proteínas de Complejo Poro Nuclear , Poro Nuclear , Animales , Ratones , Proteínas de Complejo Poro Nuclear/genética , Poro Nuclear/genética , Regulación de la Expresión Génica , Perfilación de la Expresión Génica , Células Madre Embrionarias de Ratones
5.
Am J Med Genet A ; 191(3): 870-877, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36548033

RESUMEN

Chromosomal band 17q12 is a gene-rich region flanked by segmental duplications, making the region prone to deletions and duplications via the non-allelic homologous recombination mechanism. While deletions cause a well-described disorder with a specific phenotype called renal cysts and diabetes mellitus, the phenotype caused by reciprocal duplications is less specific, primarily because of variable expressivity, and incomplete penetrance. We present an unusual family with four children carrying the 17q12 microduplication inherited from their clinically healthy mother, who was a carrier of both the duplication and, interestingly, also of an atypical deletion of the 17q12 region. The duplication was inherited from her diabetic father and the deletion from her diabetic mother who also suffered from a renal disorder. Clinical manifestations in the family were variable, but all children showed some degree of a neurodevelopmental disorder, such as epilepsy, intellectual disability, delayed speech development, or attention deficit disorder. The simultaneous occurrence of a deletion and duplication in the same chromosomal region in one family is very rare, and to our knowledge, individuals carrying both a deletion and a duplication of this region have never been described.


Asunto(s)
Anomalías Múltiples , Discapacidad Intelectual , Femenino , Humanos , Deleción Cromosómica , República Checa , Anomalías Múltiples/genética , Fenotipo , Discapacidad Intelectual/genética , Duplicación Cromosómica/genética
6.
J Dev Biol ; 10(4)2022 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-36412639

RESUMEN

The first event of differentiation and morphogenesis in the optic vesicle (OV) is specification of the neural retina (NR) and retinal pigment epithelium (RPE), separating the inner and outer layers of the optic cup, respectively. Here, we focus on a basic helix-loop-helix gene, BHLHE40, which has been shown to be expressed by the developing RPE in mice and zebrafish. Firstly, we examined the expression pattern of BHLHE40 in the developing chicken eye primordia by in situ hybridization. Secondly, BHLHE40 overexpression was performed with in ovo electroporation and its effects on optic cup morphology and expression of NR and RPE marker genes were examined. Thirdly, we examined the expression pattern of BHLHE40 in LHX1-overexpressed optic cup. BHLHE40 expression emerged in a subset of cells of the OV at Hamburger and Hamilton stage 14 and became confined to the outer layer of the OV and the ciliary marginal zone of the retina by stage 17. BHLHE40 overexpression in the prospective NR resulted in ectopic induction of OTX2 and repression of VSX2. Conversely, BHLHE40 was repressed in the second NR after LHX1 overexpression. These results suggest that emergence of BHLHE40 expression in the OV is involved in initial RPE specification and that BHLHE40 plays a role in separation of the early OV domains by maintaining OTX2 expression and antagonizing an NR developmental program.

7.
Clinics (Sao Paulo) ; 77: 100103, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36116266

RESUMEN

OBJECTIVES: To investigate the expression of LHX1 and its role as a biomarker in the diagnosis and prognosis of Uterine Corpus Endometrial Carcinoma (UCEC). METHODS: The Cancer Genome Atlas (TCGA) database was used to detect the expression level of LHX1 in UCEC cells and tissues, and to find out the effect of LHX1 on prognosis. Co-expressed genes were then identified by Spearman correlation analysis, and the protein-protein interaction network was constructed using Cytoscape software. The R "clusterProfiler" package was used to conduct Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. A series of in vitro experiments were performed to evaluate LHX1 expression and detect UCEC cell proliferation, invasion, and migration. Western blotting was used to determine the effect of LHX1 on expression levels of Epithelial-Mesenchymal Transition (EMT)-related proteins. RESULTS: LHX1 was upregulated in UCEC tissues and correlated with poor overall survival and disease-specific survival outcomes. Functional enrichment analysis suggested that genes co-expressed with LHX1 were enriched in cell adhesion. The expression of LHX1 was positively correlated with the expression levels of genes related to EMT induction and invasion. LHX1 can enhance the proliferation, migration, and invasion activities of UCEC cells in vitro, and alter the expression levels of EMT-related proteins. CONCLUSION: LHX1 expression was highly upregulated in UCEC cells and tissues, which was correlated with the prognosis of patients with UCEC. LHX1 may regulate UCEC progression at least in part by modulating EMT induction.


Asunto(s)
Neoplasias Endometriales , Transición Epitelial-Mesenquimal , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Pronóstico
8.
Stem Cell Res ; 61: 102780, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35395624

RESUMEN

Spermatogonial stem cells (SSCs) originate from gonocytes that differentiate from primordial germ cells (PGCs). In the developing mouse testis, expression of the gene LIM homeobox 1 (Lhx1) marks the most undifferentiated SSCs, which has not yet been reported for spermatogonia-like cells generated in vitro. Previously, it was shown that a chemical intervention in male mouse embryonic stem (ES) cells in serum culture, including Sirtuin 1 (SIRT1) inhibitor Ex-527, DNA methyltransferase (DNMT) inhibitor RG-108 and electrophilic redox cycling compound tert-butylhydroquinone (tBHQ), was associated with molecular markers of PGC to gonocyte differentiation. Here, we report the in vitro differentiation of male mouse ES cells, cultured under dual chemical inhibition of GSK3ß and MEK (2i) with leukemia inhibitory factor (LIF) (2iL) and serum, into cells with spermatogonia-like morphology (CSMs) and population-averaged expression of spermatogonia-specific genes by removal of 2iL and a specific schedule of twice daily partial medium replacement. Combination of this new protocol with the previously reported chemical intervention increased population-averaged gene expression of Lhx1 in the resulting CSMs. Furthermore, we detected single CSMs with strong nuclear LHX1/5 protein signal only in the chemical intervention group. We propose that further investigation of CSMs may provide new insights into male germline development.


Asunto(s)
Células Madre Embrionarias de Ratones , Espermatogonias , Animales , Diferenciación Celular/genética , Expresión Génica , Genes Homeobox , Proteínas con Homeodominio LIM/genética , Masculino , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Clinics ; 77: 100103, 2022. graf
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1404314

RESUMEN

Abstract Objectives: To investigate the expression of LHX1 and its role as a biomarker in the diagnosis and prognosis of Uterine Corpus Endometrial Carcinoma (UCEC). Methods: The Cancer Genome Atlas (TCGA) database was used to detect the expression level of LHX1 in UCEC cells and tissues, and to find out the effect of LHX1 on prognosis. Co-expressed genes were then identified by Spearman correlation analysis, and the protein-protein interaction network was constructed using Cytoscape software. The R "clusterProfiler" package was used to conduct Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. A series of in vitro experiments were performed to evaluate LHX1 expression and detect UCEC cell proliferation, invasion, and migration. Western blotting was used to determine the effect of LHX1 on expression levels of Epithelial-Mesenchymal Transition (EMT)-related proteins. Results: LHX1 was upregulated in UCEC tissues and correlated with poor overall survival and disease-specific survival outcomes. Functional enrichment analysis suggested that genes co-expressed with LHX1 were enriched in cell adhesion. The expression of LHX1 was positively correlated with the expression levels of genes related to EMT induction and invasion. LHX1 can enhance the proliferation, migration, and invasion activities of UCEC cells in vitro, and alter the expression levels of EMT-related proteins. Conclusion: LHX1 expression was highly upregulated in UCEC cells and tissues, which was correlated with the prognosis of patients with UCEC. LHX1 may regulate UCEC progression at least in part by modulating EMT induction.

10.
Curr Top Dev Biol ; 145: 113-166, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34074528

RESUMEN

The field of molecular embryology started around 1990 by identifying new genes and analyzing their functions in early vertebrate embryogenesis. Those genes encode transcription factors, signaling molecules, their regulators, etc. Most of those genes are relatively highly expressed in specific regions or exhibit dramatic phenotypes when ectopically expressed or mutated. This review focuses on one of those genes, Lim1/Lhx1, which encodes a transcription factor. Lim1/Lhx1 is a member of the LIM homeodomain (LIM-HD) protein family, and its intimate partner, Ldb1/NLI, binds to two tandem LIM domains of LIM-HDs. The most ancient LIM-HD protein and its partnership with Ldb1 were innovated in the metazoan ancestor by gene fusion combining LIM domains and a homeodomain and by creating the LIM domain-interacting domain (LID) in ancestral Ldb, respectively. The LIM domain has multiple interacting interphases, and Ldb1 has a dimerization domain (DD), the LID, and other interacting domains that bind to Ssbp2/3/4 and the boundary factor, CTCF. By means of these domains, LIM-HD-Ldb1 functions as a hub protein complex, enabling more intricate and elaborate gene regulation. The common, ancestral role of LIM-HD proteins is neuron cell-type specification. Additionally, Lim1/Lhx1 serves crucial roles in the gastrula organizer and in kidney development. Recent studies using Xenopus embryos have revealed Lim1/Lhx1 functions and regulatory mechanisms during development and regeneration, providing insight into evolutionary developmental biology, functional genomics, gene regulatory networks, and regenerative medicine. In this review, we also discuss recent progress at unraveling participation of Ldb1, Ssbp, and CTCF in enhanceosomes, long-distance enhancer-promoter interactions, and trans-interactions between chromosomes.


Asunto(s)
Proteínas con Homeodominio LIM/metabolismo , Animales , Biología Evolutiva , Genómica , Humanos , Unión Proteica , Medicina Regenerativa
11.
EXCLI J ; 19: 1038-1051, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32788915

RESUMEN

Colorectal cancer (CRC) is the third most common type of cancer and the fourth leading cause of cancer related deaths worldwide. The Histone Deacetylase 8 (HDAC8) gene is a gene with unique features which can be used as a potential target for drug design. The LHX1 transcription factor is an important transcription factor for this gene. The aim of this study was to investigate the effect of sodium butyrate (NaB) as a histone deacetylase inhibitor (HDACi) on the expression of the HDAC8 gene in the colorectal cancer cell line, and the molecular docking of the LHX1 transcription factor with NaB. For this purpose, HCT-116 and HT-29 cell lines were treated with different concentrations of NaB (6.25 mM to 150 mM) at 24, 48 and 72 hours. Subsequently, RNA was extracted from the treated and untreated cells and cDNA was synthesized. Quantitative Real-Time-PCR was done to investigate the mRNA expression of HDAC8. Molecular docking was also performed to investigate the interaction between NaB and LHX1. Based on Real-time-PCR results, the concentration of 150 mM of NaB after 24 hours in HT-29 and HCT-116 cell lines caused a significant reduction in mRNA expression of HDAC8 (P<0.05). After 48 hours of treatment, there was a significant decrease in the mRNA expression of HDAC8 at all concentrations (P<0.05). The docking results showed that LHX1 and NaB interacted best at the lowest energy levels. Our results also showed that NaB bonded strongly to LHX1. In addition, our results demonstrated that NaB bound to the LHX1 transcription factor and inhibited the function of this factor and consequently decreased the transcription from the HDAC8 gene which resulted in cell death. Future studies are needed to assess the likely molecular mechanisms of NaB action on gene expression.

12.
Epigenetics ; 15(11): 1259-1274, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32441560

RESUMEN

Apart from the conventional view of repressive promoter methylation, the DNA methyltransferase 1 (DNMT1) was recently described to modulate gene expression through a variety of interactions with diverse epigenetic key players. We here investigated the DNMT1-dependent transcriptional control of the homeobox transcription factor LHX1, which we previously identified as an important regulator in cortical interneuron development. We found that LHX1 expression in embryonic interneurons originating in the embryonic pre-optic area (POA) is regulated by non-canonic DNMT1 function. Analysis of histone methylation and acetylation revealed that both epigenetic modifications seem to be implicated in the control of Lhx1 gene activity and that DNMT1 contributes to their proper establishment. This study sheds further light on the regulatory network of cortical interneuron development including the complex interplay of epigenetic mechanisms.


Asunto(s)
Código de Histonas , Interneuronas/metabolismo , Proteínas con Homeodominio LIM/genética , Factores de Transcripción/genética , Animales , Línea Celular Tumoral , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Endogámicos C57BL , Área Preóptica/citología , Área Preóptica/embriología , Área Preóptica/metabolismo , Factores de Transcripción/metabolismo
13.
Dev Growth Differ ; 61(5): 327-336, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31111476

RESUMEN

Development of an embryo is driven by a series of molecular instructions that control the differentiation of tissue precursor cells and shape the tissues into major body parts. LIM homeobox 1 (LHX1) is a transcription factor that plays a major role in the development of the embryonic head of the mouse. Loss of LHX1 function disrupts the morphogenetic movement of head tissue precursors and impacts on the function of molecular factors in modulating the activity of the WNT signaling pathway. LHX1 acts with a transcription factor complex to regulate the transcription of target genes in multiple phases of development and in a range of embryonic tissues of the mouse and Xenopus. Determining the interacting factors and transcriptional targets of LHX1 will be key to unraveling the ensemble of factors involved in head development and building a head gene regulatory network.


Asunto(s)
Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Redes Reguladoras de Genes , Cabeza/embriología , Proteínas con Homeodominio LIM/metabolismo , Animales , Redes Reguladoras de Genes/genética , Humanos , Proteínas con Homeodominio LIM/deficiencia , Proteínas con Homeodominio LIM/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Genesis ; 56(9): e23246, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30114334

RESUMEN

Development of the embryonic head is driven by the activity of gene regulatory networks of transcription factors. LHX1 is a homeobox transcription factor that plays an essential role in the formation of the embryonic head. The loss of LHX1 function results in anterior truncation of the embryo caused by the disruption of morphogenetic movement of tissue precursors and the dysregulation of WNT signaling activity. Profiling the gene expression pattern in the Lhx1 mutant embryo revealed that tissues in anterior germ layers acquire posterior tissue characteristics, suggesting LHX1 activity is required for the allocation and patterning of head precursor tissues. Here, we used LHX1 as an entry point to delineate its transcriptional targets and interactors and construct a LHX1-anchored gene regulatory network. Using a gain-of-function approach, we identified genes that immediately respond to Lhx1 activation. Meta-analysis of the datasets of LHX1-responsive genes and genes expressed in the anterior tissues of mouse embryos at head-fold stage, in conjunction with published Xenopus embryonic LHX1 (Xlim1) ChIP-seq data, has pinpointed the putative transcriptional targets of LHX1 and an array of genetic determinants functioning together in the formation of the mouse embryonic head.


Asunto(s)
Redes Reguladoras de Genes , Genes Homeobox , Cabeza/embriología , Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Células Germinativas/fisiología , Transcripción Genética , Xenopus laevis/embriología
15.
Med Genet ; 30(1): 3-11, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29527097

RESUMEN

The Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome [MIM 277000] is characterised by the absence of a uterus and vagina in otherwise phenotypically normal women with karyotype 46,XX. Clinically, the MRKH can be subdivided into two subtypes: an isolated or type I form can be delineated from a type II form, which is characterised by extragenital malformations. The so-called Müllerian hypoplasia, renal agenesis, cervicothoracic somite dysplasia (MURCS) association can be seen as the most severe phenotypic outcome. The MRKH syndrome affects at least 1 in 4000 to 5000 female new-borns. Although most of the cases are sporadic, familial clustering has also been described, indicating a genetic cause of the disease. However, the mode of inheritance is autosomal-dominant inheritance with reduced penetrance. High-resolution array-CGH and MLPA analysis revealed recurrent aberrations in different chromosomal regions such as TAR susceptibility locus in 1q21.1, chromosomal regions 16p11.2, and 17q12 and 22q11.21 microduplication and -deletion regions in patients with MRKH. Sequential analysis of the genes LHX1, TBX6 and RBM8A, which are located in chromosomal regions 17q12, 16p11.2 and 1q21.1, yielded in the detection of MRKH-associated mutations. In a subgroup of patients with signs of hyperandrogenaemia mutations of WNT4 have been found to be causative. Analysis of another member of the WNT family, WNT9B, resulted in the detection of some causative mutations in MRKH patients.

16.
Genetics ; 208(2): 673-686, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29187504

RESUMEN

Studying genes involved in organogenesis is often difficult because many of these genes are also essential for early development. The allotetraploid frog, Xenopus laevis, is commonly used to study developmental processes, but because of the presence of two homeologs for many genes, it has been difficult to use as a genetic model. Few studies have successfully used CRISPR in amphibians, and currently there is no tissue-targeted knockout strategy described in Xenopus The goal of this study is to determine whether CRISPR/Cas9-mediated gene knockout can be targeted to the Xenopus kidney without perturbing essential early gene function. We demonstrate that targeting CRISPR gene editing to the kidney and the eye of F0 embryos is feasible. Our study shows that knockout of both homeologs of lhx1 results in the disruption of kidney development and function but does not lead to early developmental defects. Therefore, targeting of CRISPR to the kidney may not be necessary to bypass the early developmental defects reported upon disruption of Lhx1 protein expression or function by morpholinos, antisense RNA, or dominant negative constructs. We also establish a control for CRISPR in Xenopus by editing a gene (slc45a2) that when knocked out results in albinism without altering kidney development. This study establishes the feasibility of tissue-specific gene knockout in Xenopus, providing a cost-effective and efficient method for assessing the roles of genes implicated in developmental abnormalities that is amenable to high-throughput gene or drug screening techniques.


Asunto(s)
Silenciador del Gen , Xenopus laevis/genética , Animales , Sistemas CRISPR-Cas , Edición Génica , Técnicas de Inactivación de Genes , Marcación de Gen , Riñón/metabolismo , Proteínas con Homeodominio LIM/genética , Especificidad de Órganos/genética , Fenotipo , ARN Guía de Kinetoplastida , Factores de Transcripción/genética , Proteínas de Xenopus/genética
17.
Dev Dyn ; 246(9): 657-669, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28598520

RESUMEN

BACKGROUND: Pitx3 plays a well understood role in directing development of lens, muscle fiber, and dopaminergic neurons; however, in Xenopus laevis, it may also play a role in early gastrulation and somitogenesis. Potential downstream targets of pitx3 possess multiple binding motifs that would not be readily accessible by conventional promoter analysis. RESULTS: We isolated and characterized pitx3 target genes lhx1 and xnr5 using a novel three-fluor flow cytometry tool that was designed to dissect promoters with multiple binding sites for the same transcription factor. This approach was calibrated using a known pitx3 target gene, Tyrosine hydroxylase. CONCLUSIONS: We demonstrate how flow cytometry can be used to detect gene regulatory changes with exquisite precision on a cell-by-cell basis, and establish that in HEK293 cells, pitx3 directly activates lhx1 and represses xnr5. Developmental Dynamics 246:657-669, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Ligandos de Señalización Nodal/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Proteínas con Homeodominio LIM/genética , Ligandos de Señalización Nodal/genética , Factores de Transcripción/genética , Proteínas de Xenopus/genética , Xenopus laevis
18.
Oncotarget ; 8(5): 8785-8790, 2017 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-28061432

RESUMEN

Congenital absence of the uterus and vagina (CAUV) is the most extreme female Müllerian duct abnormality. Several researches proposed that genetic factors contributed to this disorder, whereas the precise genetic mechanism is far from full elucidation. Here, utilizing whole-exome sequencing (WES), we identified one novel missense mutation in LHX1 (NM_005568: c.G1108A, p.A370T) in one of ten unrelated patients diagnosed with CAUV. This mutation was absent from public databases and our internal database. Through the luciferase reporter analysis, we found that the mutation could change the transcriptional activity of LHX1 and its effect on the regulation of the downstream target gene GSC, which might be associated with urogenital system development. In short, we concluded that the LHX1 may be a pathogenic gene of CAUV. Our results demonstrate the power of whole exome sequencing and gene prioritization approach as diagnostic tools in clinical practice that help make genetic diagnosis of CAUV.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/genética , Anomalías Congénitas/genética , Proteínas con Homeodominio LIM/genética , Conductos Paramesonéfricos/anomalías , Mutación Missense , Factores de Transcripción/genética , Trastornos del Desarrollo Sexual 46, XX/diagnóstico , Trastornos del Desarrollo Sexual 46, XX/metabolismo , Anomalías Congénitas/diagnóstico , Anomalías Congénitas/metabolismo , Análisis Mutacional de ADN , Femenino , Regulación de la Expresión Génica , Estudios de Asociación Genética , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Proteína Goosecoide/genética , Proteína Goosecoide/metabolismo , Células HEK293 , Humanos , Proteínas con Homeodominio LIM/metabolismo , Conductos Paramesonéfricos/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección , Secuenciación del Exoma
19.
Curr Biol ; 27(1): 128-136, 2017 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-28017605

RESUMEN

The suprachiasmatic nucleus (SCN) is the central circadian clock in mammals. It is entrained by light but resistant to temperature shifts that entrain peripheral clocks [1-5]. The SCN expresses many functionally important neuropeptides, including vasoactive intestinal peptide (VIP), which drives light entrainment, synchrony, and amplitude of SCN cellular clocks and organizes circadian behavior [5-16]. The transcription factor LHX1 drives SCN Vip expression, and cellular desynchrony in Lhx1-deficient SCN largely results from Vip loss [17, 18]. LHX1 regulates many genes other than Vip, yet activity rhythms in Lhx1-deficient mice are similar to Vip-/- mice under light-dark cycles and only somewhat worse in constant conditions. We suspected that LHX1 targets other than Vip have circadian functions overlooked in previous studies. In this study, we compared circadian sleep and temperature rhythms of Lhx1- and Vip-deficient mice and found loss of acute light control of sleep in Lhx1 but not Vip mutants. We also found loss of circadian resistance to fever in Lhx1 but not Vip mice, which was partially recapitulated by heat application to cultured Lhx1-deficient SCN. Having identified VIP-independent functions of LHX1, we mapped the VIP-independent transcriptional network downstream of LHX1 and a largely separable VIP-dependent transcriptional network. The VIP-independent network does not affect core clock amplitude and synchrony, unlike the VIP-dependent network. These studies identify Lhx1 as the first gene required for temperature resistance of the SCN clockworks and demonstrate that acute light control of sleep is routed through the SCN and its immediate output regions.


Asunto(s)
Relojes Circadianos , Redes Reguladoras de Genes , Proteínas con Homeodominio LIM/fisiología , Sueño , Factores de Transcripción/fisiología , Péptido Intestinal Vasoactivo/fisiología , Vigilia , Animales , Ritmo Circadiano , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Calor , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fotoperiodo , Transducción de Señal , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/metabolismo
20.
Curr Top Dev Biol ; 117: 497-521, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26969997

RESUMEN

The embryonic head is the first major body part to be constructed during embryogenesis. The allocation and the assembly of the progenitor tissues, which start at gastrulation, are accompanied by the spatiotemporal activity of transcription factors and signaling pathways that drives lineage specification, germ layer formation, and cell/tissue movement. The morphogenesis, regionalization, and patterning of the brain and craniofacial structures rely on the function of LIM-domain, homeodomain, and basic helix-loop-helix transcription factors. These factors constitute the central nodes of a gene regulatory network (GRN) which encompasses and intersects with signaling pathways involved with head formation. It is predicted that the functional output of this "head GRN" impacts on cellular function and cell-cell interactions that are essential for lineage differentiation and tissue modeling, which are key processes underpinning the formation of the head.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Embrión de Mamíferos/citología , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Cabeza/embriología , Animales , Embrión de Mamíferos/metabolismo , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA