Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Reprod Biol Endocrinol ; 21(1): 46, 2023 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-37194006

RESUMO

Efficient evaluation of the primordial follicle pool (PFP) of mammalian models is an essential subject in biomedical research relating to ovarian physiology and pathogenesis. Our recent study has identified a gene signature including Sohlh1, Nobox, Lhx8, Tbpl2, Stk31, Padi6, and Vrtn strongly correlated with ovarian reserve by using bioinformatics analysis. Aimed to investigate the validity of these candidate biomarkers for evaluating the PFP, we utilized an OR comparison model to decode the relationship between the numbers of PFP and candidate biomarkers in the present study. Our results suggest that these biomarkers Sohlh1, Nobox, Lhx8, Tbpl2, Stk31, Padi6, and Vrtn possess independent potential to evaluate the number of the PFP. And the combination of Sohlh1 and Lhx8 can be used as the optimal biomarkers for rapid assessment of the PFP in the murine ovary. Our findings provide a new perspective for evaluating the PFP of the ovary in animal studies and the clinic.


Assuntos
Folículo Ovariano , Fatores de Transcrição , Animais , Feminino , Camundongos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Folículo Ovariano/fisiologia , Ovário , Fatores de Transcrição/genética
2.
Genet Med ; 24(11): 2274-2284, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36029299

RESUMO

PURPOSE: The genetic causes of oocyte maturation arrest leading to female infertility are largely unknown, and no population-based genetic analysis has been applied in cohorts of patients with infertility. We aimed to identify novel pathogenic genes causing oocyte maturation arrest by using a gene-based burden test. METHODS: Through comparison of exome sequencing data from 716 females with infertility characterized by oocyte maturation arrest and 3539 controls, we performed a gene-based burden test and identified a novel pathogenic gene LHX8. Splicing event was evaluated using a minigene assay, expression of LHX8 protein was assessed in HeLa cells, and nuclear subcellular localization was determined in both HeLa cells and mouse oocytes. RESULTS: A total of 5 heterozygous loss-of-function LHX8 variants were identified from 6 independent families (c.389+1G>T, c.412C>T [p.Arg138∗], c.282C>A [p.Cys94∗]; c.257dup [p.Tyr86∗]; and c.180del, [p.Ser61Profs∗30]). All the identified variants in LHX8 produced truncated LHX8 protein and resulted in loss of LHX8 nuclear localization in both HeLa cells and mouse oocytes. CONCLUSION: By combining genetic evidence and functional evaluations, we identified a novel pathogenic gene LHX8 and established the causative relationship between LHX8 haploinsufficiency and female infertility characterized by oocyte maturation arrest.


Assuntos
Infertilidade Feminina , Feminino , Humanos , Camundongos , Animais , Infertilidade Feminina/genética , Infertilidade Feminina/patologia , Células HeLa , Oogênese/genética , Oócitos , Sequenciamento do Exoma
3.
J Cell Mol Med ; 25(6): 3051-3062, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33580754

RESUMO

The homeobox gene, LIM-homeobox 8 (Lhx8), has previously been identified as an essential transcription factor for dental mesenchymal development. However, how Lhx8 itself is regulated and regulates odontogenesis remains poorly understood. In this study, we employed an RNAscope assay to detect the co-expression pattern of Lhx8 and Suv39h1 in the dental mesenchyme, which coincided with the dynamic expression profiles of the early epithelium signal of Fibroblast Growth Factor 8 (FGF8) and the later mesenchymal signal Bone Morphogenetic Protein 2 (BMP2). Moreover, FGF8 activated Lhx8, whereas BMP2 repressed Lhx8 expression at the transcriptional level. The high expression of Lhx8 in the early dental mesenchyme maintained the cell fate in an undifferentiated status by interacting with Suv39h1, a histone-lysine N-methyltransferase constitutively expressed in the dental mesenchyme. Further in the ex vivo organ culture model, the knockdown of Suv39h1 significantly blocked the function of Lhx8 and FGF8. Mechanistically, Lhx8/Suv39h1 recognized the odontoblast differentiation-related genes and repressed gene expression via methylating H3K9 on their promoters. Taken together, our data here suggest that Lhx8/Suv39h1 complex is inversely regulated by epithelium-mesenchymal signals, balancing the differentiation and proliferation of dental mesenchyme via H3K9 methylation.


Assuntos
Proteína Morfogenética Óssea 2/genética , Diferenciação Celular/genética , Fator 8 de Crescimento de Fibroblasto/genética , Proteínas com Homeodomínio LIM/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteína Morfogenética Óssea 2/metabolismo , Proliferação de Células , Polpa Dentária/citologia , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Histonas , Humanos , Imuno-Histoquímica , Metilação , Camundongos , Complexos Multiproteicos/metabolismo , Odontogênese/genética , Ligação Proteica
4.
Biochem Biophys Res Commun ; 530(2): 418-424, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32546349

RESUMO

LIM homeobox 8 (LHX8) is expressed during embryonic development of craniofacial tissues, including bone and teeth. In a previous study, the overexpression of LHX8 inhibited osteodifferentiation of human dental pulp stem cells (DPSCs). In this study, a cDNA microarray analysis was performed to reveal the molecular changes which occur in response to LHX8 overexpression in DPSCs and discover possible targets for an osteoinductive agent. There were 345 differentially expressed genes (DEGs) in response to osteoinductive signaling and 53 DEGs in response to LHX8 overexpression and osteoinductive signaling, respectively. Thirty-eight genes were common in both conditions, and among these, genes upregulated in LHX8 DPSCs but downregulated in osteodifferentiated DPSCs were chosen. Five of them had commercial inhibitors available. Among the tested inhibitors, ML323, which target DNA-binding protein inhibitor ID-1, promoted osteodifferentiation of DPSCs. In conclusion, inhibition of ID-1 led to increased osteogenesis of human DPSCs.


Assuntos
Polpa Dentária/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Células-Tronco/efeitos dos fármacos , Proteases Específicas de Ubiquitina/antagonistas & inibidores , Adulto , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Polpa Dentária/citologia , Polpa Dentária/metabolismo , Feminino , Humanos , Proteínas com Homeodomínio LIM/genética , Masculino , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Transcriptoma/efeitos dos fármacos , Proteases Específicas de Ubiquitina/genética , Regulação para Cima/efeitos dos fármacos , Adulto Jovem
5.
Cereb Cortex ; 29(6): 2653-2667, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29878134

RESUMO

Immature neurons generated by the subpallial MGE tangentially migrate to the cortex where they become parvalbumin-expressing (PV+) and somatostatin (SST+) interneurons. Here, we show that the Sp9 transcription factor controls the development of MGE-derived cortical interneurons. SP9 is expressed in the MGE subventricular zone and in MGE-derived migrating interneurons. Sp9 null and conditional mutant mice have approximately 50% reduction of MGE-derived cortical interneurons, an ectopic aggregation of MGE-derived neurons in the embryonic ventral telencephalon, and an increased ratio of SST+/PV+ cortical interneurons. RNA-Seq and SP9 ChIP-Seq reveal that SP9 regulates MGE-derived cortical interneuron development through controlling the expression of key transcription factors Arx, Lhx6, Lhx8, Nkx2-1, and Zeb2 involved in interneuron development, as well as genes implicated in regulating interneuron migration Ackr3, Epha3, and St18. Thus, Sp9 has a central transcriptional role in MGE-derived cortical interneuron development.


Assuntos
Córtex Cerebral/citologia , Interneurônios/citologia , Eminência Mediana/embriologia , Neurogênese/fisiologia , Proteínas de Ligação a RNA/metabolismo , Animais , Movimento Celular/fisiologia , Córtex Cerebral/embriologia , Interneurônios/metabolismo , Eminência Mediana/citologia , Camundongos , Fatores de Transcrição/metabolismo
6.
Toxicol Appl Pharmacol ; 317: 33-40, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28089945

RESUMO

Zearalenone (ZEA) is an estrogenic mycotoxin mainly produced as a secondary metabolite by numerous species of Fusarium. Previous work showed that ZEA had a negative impact on domestic animals with regard to reproduction. The adverse effects and the mechanisms of ZEA on mammalian ovarian folliculogenesis remain largely unknown, particularly its effect on primordial follicle formation. Thus, we investigated the biological effects of ZEA exposure on murine ovarian germ cell cyst breakdown and primordial follicle assembly. Our results demonstrated that newborn mouse ovaries exposed to 10 or 30µM ZEA in vitro had significantly less germ cell numbers compared to the control group. Moreover, the presence of ZEA in vitro increased the numbers of TUNEL and γH2AX positive cells within mouse ovaries and the ratio of mRNA levels of the apoptotic genes Bax/Bcl-2. Furthermore, ZEA exposure reduced the mRNA of oocyte specific genes such as LIM homeobox 8 (Lhx8), newborn ovary homeobox (Nobox), spermatogenesis and oogenesis helix-loop-helix (Sohlh2), and factor in the germline alpha (Figlα) in a dose dependent manner. Exposure to ZEA led to remarkable changes in the Lhx8 3'-UTR DNA methylation dynamics in oocytes and severely impaired folliculogenesis in ovaries after transplantation under the kidney capsules of immunodeficient mice. In conclusion, ZEA exposure impairs mouse primordial follicle formation in vitro.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Estrogênios não Esteroides/toxicidade , Proteínas com Homeodomínio LIM/biossíntese , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Fatores de Transcrição/biossíntese , Zearalenona/toxicidade , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo/fisiologia , Feminino , Regulação da Expressão Gênica , Proteínas com Homeodomínio LIM/antagonistas & inibidores , Camundongos , Camundongos SCID , Folículo Ovariano/crescimento & desenvolvimento , Fatores de Transcrição/antagonistas & inibidores
7.
Int J Mol Sci ; 16(10): 25759-72, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26516845

RESUMO

Bone morphogenetic protein 15 (BMP15) is secreted by the mammalian oocytes and is indispensable for ovarian follicular development, ovulation, and fertility. To determine the regulation mechanism of BMP15 gene, the regulatory sequence of porcine BMP15 was investigated in this study. The cloned BMP15 promoter retains the cell-type specificity, and is activated in cells derived from ovarian tissue. The luciferase assays in combination with a series of deletion of BMP15 promoter sequence show that the -427 to -376 bp region of BMP15 promoter is the primary regulatory element, in which there are a number of transcription factor binding sites, including LIM homeobox 8 (LHX8), newborn ovary homeobox gene (NOBOX), and paired-like homeodomain transcription factor 1 (PITX1). Determination of tissue-specific expression reveals that LHX8, but not PITX1 and NOBOX, is exclusively expressed in pig ovary tissue and is translocated into the cell nuclei. Overexpression of LHX8 in Chinese hamster ovary (CHO) cells could significantly promote BMP15 promoter activation. This study confirms a key regulatory element that is located in the proximal region of BMP15 promoter and is regulated by the LHX8 factor.


Assuntos
Proteína Morfogenética Óssea 15/genética , Regiões Promotoras Genéticas , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Cricetinae , Cricetulus , Proteínas com Homeodomínio LIM/metabolismo , Dados de Sequência Molecular , Fatores de Transcrição Box Pareados/metabolismo , Ligação Proteica , Suínos , Ativação Transcricional
8.
Heliyon ; 9(9): e19878, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37809754

RESUMO

Osteoporosis (OP), a common systemic bone metabolism disease with a high incidence rate, is a serious health risk factor. Osteogenic differentiation balance is regulated by bone marrow mesenchymal stem cells (BMSCs) and plays a key role in OP occurrence and progression. Although, LIM homeobox 8 (Lhx8) has been identified to affect BMSCs osteogenic differentiation, its roles in OP and the associated mechanism remains unclear. Here, we aimed to elucidate the role and mechanism of Lhx8 in the osteogenic differentiation of BMSCs. BMSCs isolated from wild type and OP Sprague-Dawley rats were cultured and confirmed via flow cytometry and microscopy. Based on dual-luciferase reporter assay, BMSCs were transfected with miR-142-5p mimics and miR-NC (negative control). Real-time quantitative reverse transcription polymerase chain reaction and Western blot analyses were performed to determine the role of Lhx8 in BMSCs osteogenic differentiation. Lhx8 expression was significantly reduced in OP, whereas that of miR-142-5p, a possible Lhx8 regulator, was significantly upregulated. Dual-luciferase reporter assay demonstrated that miR-142-5p exerted a direct targeted regulatory effect on Lhx8. Moreover, miR-142-5p mimics significantly inhibited BMSCs osteogenic differentiation as well as Lhx8 expression in vitro, indicating that miR-142-5p may be involved in BMSCs osteogenic differentiation via Lhx8 expression regulation and may serve as a potential diagnostic target for OP. Overall, these findings indicated that miR-142-5p inhibits BMSCs osteogenic differentiation by suppressing Lhx8. These may serve as a foundation for further studies on OP treatment based on miR-142-5p targeting.

9.
Elife ; 122023 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-36602025

RESUMO

Non-coding RNAs exert diverse functions in many cell types. In addition to transcription factors from coding genes, non-coding RNAs may also play essential roles in shaping and directing the fate of germ cells. The presence of many long non-coding RNAs (lncRNAs) which are specifically expressed in the germ cells during human gonadal development were reported and one divergent lncRNA, LNC1845, was functionally characterized. Comprehensive bioinformatic analysis of these lncRNAs indicates that divergent lncRNAs occupied the majority of female and male germ cells. Integrating lncRNA expression into the bioinformatic analysis also enhances the cell-type classification of female germ cells. Functional dissection using in vitro differentiation of human pluripotent stem cells to germ cells revealed the regulatory role of LNC1845 on a transcription factor essential for ovarian follicle development, LHX8, by modulating the levels of histone modifications, H3K4me3 and H3K27Ac. Hence, bioinformatical analysis and experimental verification provide a comprehensive analysis of lncRNAs in developing germ cells and elucidate how an lncRNA function as a cis regulator during human germ cell development.


Assuntos
RNA Longo não Codificante , Feminino , Humanos , Masculino , Diferenciação Celular/genética , Regulação da Expressão Gênica , Células Germinativas/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Gene Expr Patterns ; 43: 119227, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34861428

RESUMO

Craniofacial development is controlled by a large number of genes, which interact with one another to form a complex gene regulatory network (GRN). Key components of GRN are signaling molecules and transcription factors. Therefore, identifying targets of core transcription factors is an important part of the overall efforts toward building a comprehensive and accurate model of GRN. LHX6 and LHX8 are transcription factors expressed in the oral mesenchyme of the first pharyngeal arch (PA1), and they are crucial regulators of palate and tooth development. Previously, we performed genome-wide transcriptional profiling and chromatin immunoprecipitation to identify target genes of LHX6 and LHX8 in PA1, and described a set of genes repressed by LHX. However, there has not been any discussion of the genes positively regulated by LHX6 and LHX8. In this paper, we revisited the above datasets to identify candidate positive targets of LHX in PA1. Focusing on those with known connections to craniofacial development, we performed RNA in situ hybridization to confirm the changes in expression in Lhx6;Lhx8 mutant. We also confirmed the binding of LHX6 to several putative enhancers near the candidate target genes. Together, we have uncovered novel connections between Lhx and other important regulators of craniofacial development, including Eya1, Barx1, Rspo2, Rspo3, and Wnt11.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Maxila/metabolismo , Palato/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Gene ; 817: 146201, 2022 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-35063574

RESUMO

Lhx8, belonging to the LIM-Homebox family, is involved in the tooth, nervous system, and primordial follicles development in mammals. However, little is known about the regulatory roles of lhx8 in teleosts. In this study, two lhx8 duplicates were identified in Paralichthys olivaceus, termed Polhx8a and Polhx8b, respectively. Bioinformatic analysis showed that Polhx8a was more likely to be a teleost-specific paralog. According to expression analysis, Polhx8a transcripts were almost exclusively concentrated in the oocytes, while Polhx8b was weakly expressed in the spleen, gill, and some facial organs, indicating sub-functionalization of this gene pair during evolution. Furthermore, Polhx8a mRNA level elevated from perinucleolar oocyte (PNO) stage to vitellogenic oocyte (VO) stage transition and changed after exogenous hormone stimulation, proving that Polhx8a was involved in the oocyte development and could be regulated by sex hormones. Yeast two-hybrid, bimolecular fluorescence complementation (BiFC) and co-immunoprecipitation (co-IP) experiments captured the positive protein interactions between PoLhx8a and the other two oocyte-specific transcription factors: PoFigla and PoNobox. After knocking down lhx8a in embryos or adult ovaries in vivo, the expression of oocyte-associated genes was significantly down-regulated (P < 0.05). Our findings suggest the evolution and functional differentiation of lhx8 genes, and shed light on the potential role of lhx8a in protein interactions and gene regulation in teleosts.


Assuntos
Proteínas de Peixes/genética , Linguado/genética , Proteínas com Homeodomínio LIM/genética , Animais , Evolução Molecular , Feminino , Proteínas de Peixes/fisiologia , Linguado/fisiologia , Técnicas de Silenciamento de Genes/veterinária , Células HeLa , Humanos , Proteínas com Homeodomínio LIM/fisiologia , Masculino , Oogênese/genética , Sintenia
12.
Stem Cell Res Ther ; 12(1): 339, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34112263

RESUMO

BACKGROUND: The spatiotemporal regulation of essential genes is crucial for controlling the growth and differentiation of cells in a precise manner during regeneration. Recently, optogenetics was considered as a potent technology for sophisticated regulation of target genes, which might be a promising tool for regenerative medicine. In this study, we used an optogenetic control system to precisely regulate the expression of Lhx8 to promote efficient bone regeneration. METHODS: Quantitative real-time PCR and western blotting were used to detect the expression of Lhx8 and osteogenic marker genes. Alkaline phosphatase staining and alizarin red staining were used to detect alkaline phosphatase activity and calcium nodules. A customized optogenetic expression system was constructed to regulate Lhx8, of which the expression was activated in blue light but not in dark. We also used a critical calvarial defect model for the analysis of bone regeneration in vivo. Moreover, micro-computed tomography (micro-CT), three-dimensional reconstruction, quantitative bone measurement, and histological and immunohistochemistry analysis were performed to investigate the formation of new bone in vivo. RESULTS: During the osteogenic differentiation of BMSCs, the expression levels of Lhx8 increased initially but then decreased thereafter. Lhx8 promoted the early proliferation of BMSCs but inhibited subsequent osteogenic differentiation. The optogenetic activation of Lhx8 in BMSCs in the early stages of differentiation by blue light stimulation led to a significant increase in cell proliferation, thus allowing a sufficient number of differentiating BMSCs to enter the later osteogenic differentiation stage. Analysis of the critical calvarial defect model revealed that the pulsed optogenetic activation of Lhx8 in transplanted BMSCs over a 5-day period led to a significant increase in the generation of bone in vivo. CONCLUSIONS: Lhx8 plays a critical role in balancing proliferation and osteogenic differentiation in BMSCs. The optogenetic activation of Lhx8 expression at early stage of BMSCs differentiation led to better osteogenesis, which would be a promising strategy for precise bone regeneration.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Células da Medula Óssea , Regeneração Óssea , Diferenciação Celular , Células Cultivadas , Optogenética , Microtomografia por Raio-X
13.
Life Sci ; 260: 118388, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32890602

RESUMO

Damage to the cholinergic system in central nervous system injuries such as traumatic brain injury (TBI) and neurodegenerative diseases leads to impaired learning and cognition. Neural stem cells (NSCs) have self-renewal capacity and multi-directional differentiation potential and considered the best source of cells for cell replacement therapy. However, how to promote the differentiation of NSCs into neurons is a major challenge in current research. Lhx8 has a specific effect on the development of the cholinergic nervous system, but its exact function is unclear. In this study, we found that Lhx8 could regulate the expression of Growth arrest-specific (GAS)5 which has been implicated in cancer but was less studied in the nervous system. Additionally, results from PCR, fluorescence in situ hybridization, and immunocytochemical analyses showed that GAS5 is mainly expressed in the cytoplasm of hippocampal neural stems cells and promotes their differentiation into neurons; the Morris water maze test demonstrated that GAS5 overexpression restored learning and memory in rats with cholinergic injury. These findings indicate that GAS5, which is regulated by Lhx8, improve brain function following cholinergic nerve injury.


Assuntos
Lesões Encefálicas/fisiopatologia , Neurônios Colinérgicos/patologia , Proteínas com Homeodomínio LIM/metabolismo , Aprendizagem/fisiologia , Memória/fisiologia , Células-Tronco Neurais/patologia , RNA Longo não Codificante/genética , Fatores de Transcrição/metabolismo , Acetilcolina/metabolismo , Animais , Colina O-Acetiltransferase/metabolismo , Neurônios Colinérgicos/metabolismo , Regulação da Expressão Gênica , Proteínas com Homeodomínio LIM/genética , Células-Tronco Neurais/metabolismo , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Fatores de Transcrição/genética
14.
Epigenomics ; 12(18): 1569-1578, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32938193

RESUMO

Aim: To evaluate the triage performance of six host-cell DNA methylation markers derived from two genome-wide discovery screens for detection of cervical precancer (cervical intraepithelial neoplasia 3 [CIN]) and cancer. Materials & methods: Human papillomavirus-positive cervical scrapes of controls (≤CIN1; n = 352) and women diagnosed with CIN3 (n = 175) or cervical cancer (n = 50) were analyzed for methylation of ASCL1, LHX8, ST6GALNAC5, GHSR, SST and ZIC1. Results: Methylation levels increased significantly with disease severity (all markers p < 0.001). Three markers (ASCL1, LHX8, ZIC1) showed receiver operating characteristic curves with area under the curve >0.800 after leave-one-out cross-validation. Bi-marker panel ASCL1/LHX8 had highest area under the curve (0.882), and detected 83.4% of CIN3 and all cervical cancers at specificity of 82.4%. Conclusion: All six methylation markers showed an equivalent, high performance for the triage of human papillomavirus-positive women using cervical scrapes with complementarity between markers.


Assuntos
Metilação de DNA , Displasia do Colo do Útero/genética , Neoplasias do Colo do Útero/genética , Adulto , Alphapapillomavirus/isolamento & purificação , Biomarcadores Tumorais , Feminino , Genoma Humano , Humanos , Neoplasias do Colo do Útero/diagnóstico , Displasia do Colo do Útero/diagnóstico
15.
Transl Neurodegener ; 7: 29, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30410751

RESUMO

BACKGROUND: Cell replacement therapy has been envisioned as a promising treatment for neurodegenerative diseases. Due to the ethical concerns of ESCs-derived neural progenitor cells (NPCs) and tumorigenic potential of iPSCs, reprogramming of somatic cells directly into multipotent NPCs has emerged as a preferred approach for cell transplantation. METHODS: Mouse astrocytes were reprogrammed into NPCs by the overexpression of transcription factors (TFs) Foxg1, Sox2, and Brn2. The generation of subtypes of neurons was directed by the force expression of cell-type specific TFs Lhx8 or Foxa2/Lmx1a. RESULTS: Astrocyte-derived induced NPCs (AiNPCs) share high similarities, including the expression of NPC-specific genes, DNA methylation patterns, the ability to proliferate and differentiate, with the wild type NPCs. The AiNPCs are committed to the forebrain identity and predominantly differentiated into glutamatergic and GABAergic neuronal subtypes. Interestingly, additional overexpression of TFs Lhx8 and Foxa2/Lmx1a in AiNPCs promoted cholinergic and dopaminergic neuronal differentiation, respectively. CONCLUSIONS: Our studies suggest that astrocytes can be converted into AiNPCs and lineage-committed AiNPCs can acquire differentiation potential of other lineages through forced expression of specific TFs. Understanding the impact of the TF sets on the reprogramming and differentiation into specific lineages of neurons will provide valuable strategies for astrocyte-based cell therapy in neurodegenerative diseases.

16.
Methods Mol Biol ; 1622: 101-109, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28674804

RESUMO

Techniques for controlling the expression of a specific gene in embryonic stem cells are effective and important for clarifying the functions of the gene. Regarding differentiation of cells into nervous system components, these techniques would play key roles in elucidating, not only the differentiation mechanisms of neuronal and glial cells but also how neuronal phenotypes are determined. In this chapter, we describe a RNA interference method for suppressing cholinergic differentiation in murine embryonic stem cells by knockdown of expression of the transcription factor L3/Lhx8, a Lim homeobox gene family protein. This method will greatly facilitate functional analyses of the factors involved in neuronal differentiation and regeneration and will contribute to cell transplantation studies.


Assuntos
Acetilcolina/metabolismo , Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Homeodomínio LIM/genética , Interferência de RNA , Fatores de Transcrição/genética , Animais , Morte Celular/genética , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Plasmídeos/genética , Fatores de Transcrição/metabolismo , Transfecção
17.
Curr Top Dev Biol ; 123: 1-47, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28236964

RESUMO

WNK (With-No-Lysine (K)) kinases are serine-threonine kinases characterized by an atypical placement of a catalytic lysine within the kinase domain. Mutations in human WNK1 or WNK4 cause an autosomal dominant syndrome of hypertension and hyperkalemia, reflecting the fact that WNK kinases are critical regulators of renal ion transport processes. Here, the role of WNKs in the regulation of ion transport processes in vertebrate and invertebrate renal function, cellular and organismal osmoregulation, and cell migration and cerebral edema will be reviewed, along with emerging literature demonstrating roles for WNKs in cardiovascular and neural development, Wnt signaling, and cancer. Conserved roles for these kinases across phyla are emphasized.


Assuntos
Doença , Desenvolvimento Embrionário , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Modelos Biológicos , Neoplasias/enzimologia , Proteínas Serina-Treonina Quinases/química , Transdução de Sinais
18.
Biomaterials ; 63: 35-46, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26081866

RESUMO

LIM homeobox 8 (Lhx8) is a highly conserved transcriptional factor with recently illustrated roles in cholinergic and GABAergic differentiation, and is expressed in neural crest derived craniofacial tissues during development. However, Lhx8 functions and signaling pathways are largely elusive. Here we showed that Lhx8 regulates dental mesenchyme differentiation and function via Wnt and TGFß pathways. Lhx8 expression was restricted to dental mesenchyme from E11.5 to a peak at E14.5, and absent in dental epithelium. By reconstituting dental epithelium and mesenchyme in an E16.5 tooth organ, Lhx8 knockdown accelerated dental mesenchyme differentiation; conversely, Lhx8 overexpression attenuated dentin formation. Lhx8 overexpressed adult human dental pulp stem/progenitor cells in ß-tricalcium phosphate cubes attenuated mineralized matrix production in vivo. Gene profiling revealed that postnatal dental pulp stem/progenitor cells upon Lhx8 overexpression modified matrix related gene expression including Dspp, Cola1 and osteocalcin. Lhx8 transcriptionally activated Wnt and TGFß pathways, and its attenuation upregulated multiple dentinogenesis genes. Together, Lhx8 regulates dentin development and regeneration by fine-turning Wnt and TGFß signaling.


Assuntos
Dentinogênese , Proteínas com Homeodomínio LIM/metabolismo , Odontogênese , Regeneração , Dente/fisiologia , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , Adulto , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/transplante , Animais , Fosfatos de Cálcio/química , Diferenciação Celular , Células Cultivadas , Polpa Dentária/citologia , Polpa Dentária/metabolismo , Polpa Dentária/transplante , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Proteínas com Homeodomínio LIM/genética , Camundongos , Transdução de Sinais , Alicerces Teciduais/química , Dente/citologia , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/genética , Proteínas Wnt/genética
19.
J Biosci Bioeng ; 119(3): 260-6, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25239070

RESUMO

Expression of the LIM homeodomain transcription factor Lhx8 is restricted to and up-regulated in the mesenchyme of the upper face prominence before lip fusion. Msx1/2 acts in early development to control cell proliferation and differentiation. Deficiency of these genes is associated with nonsyndromic cleft lip with/without cleft palate. Since retinoid is a potential patterning influence on the developing face, we have examined whether retinoic acid (RA) signaling regulated Lhx8, Msx1 and Msx2 transcription through fibroblast growth factor (FGF) signals in the maxillary prominence. Application of exogenous RA caused severe defects of the maxilla. Citral also induced a specific loss of derivatives from the maxillary prominences by blocking RA synthesis. Real-time RT-PCR and semi-quantitative RT-PCR analysis of the maxillary mesenchyme revealed that the expressions of Lhx8, Msx1 and Msx2 were significantly down-regulated by RA as well as by citral. The downregulated Lhx8 was rescued by combined treatment with FGF-8b, which indicated a downstream of RA signaling. FGF-8b induced up-regulated Lhx8 expression whereas SU5402, a pan-FGF family antagonist, down-regulated and caused defective maxillary morphogenesis and cleft lip. Our data suggest that Lhx8 is regulated by RA signaling through FGF signals and the level window of RA and FGF-8b could control the upper jaw morphogenesis.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Arcada Osseodentária/efeitos dos fármacos , Arcada Osseodentária/embriologia , Proteínas com Homeodomínio LIM/metabolismo , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia , Monoterpenos Acíclicos , Animais , Diferenciação Celular/efeitos dos fármacos , Embrião de Galinha , Regulação para Baixo/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/antagonistas & inibidores , Fator 8 de Crescimento de Fibroblasto/farmacologia , Proteínas de Homeodomínio/metabolismo , Arcada Osseodentária/metabolismo , Fator de Transcrição MSX1/metabolismo , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Monoterpenos/farmacologia , Morfogênese/efeitos dos fármacos , Pirróis/farmacologia , Transdução de Sinais/efeitos dos fármacos
20.
Neurosci Lett ; 559: 184-8, 2014 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-24316404

RESUMO

Lhx8 is a transcription factor for cholinergic differentiation. Our previous experiments found upregulation of Lhx8 promoted cholinergic neuronal differentiation of hippocampal neural stem/progenitor cells or hippocampal newborn neurons in vitro. However, the role of Lhx8 in VAChT expression and ACh release is still less understood. In this report, we transfected Lhx8 cDNA into neuronal cell line SHSY5Y by lentiviral vectors to acquire the cells which stably expressed high level of Lhx8. Using this cell model, we provided experimental evidence that increasing Lhx8 upregulated the expression of ChAT and VAChT, and also increased the ACh release in culture medium. We suggested that Lhx8 overexpression is a useful strategy to increase the release of ACh and maybe of therapeutic value to neurodegenerative diseases.


Assuntos
Acetilcolina/metabolismo , Regulação da Expressão Gênica , Proteínas com Homeodomínio LIM/genética , Neurônios/metabolismo , Fatores de Transcrição/genética , Regulação para Cima/genética , Proteínas Vesiculares de Transporte de Acetilcolina/genética , Linhagem Celular Tumoral , Humanos , Proteínas com Homeodomínio LIM/biossíntese , Distribuição Aleatória , Fatores de Transcrição/biossíntese , Proteínas Vesiculares de Transporte de Acetilcolina/biossíntese
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa