Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Sleep Breath ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39186098

ABSTRACT

BACKGROUND: As the most common sleep disorder, chronic insomnia disorder (CID) has become a global health burden to the public. However, it remains unclear about the pathogenesis of this disease. Epigenetic changes may provide important insights into the gene-environment interaction in CID. Therefore, this study was conducted to investigate the DNA methylation pattern in CID and reveal the epigenetic mechanism of this disease. METHODS: In this study, whole blood DNA was extracted from 8 CID patients (the CID group) and 8 healthy controls (the control group), respectively. Besides, genome-wide DNA methylation was detected by Illumina Human Methylation 850 K Beadchip. Moreover, the sleep quality and insomnia severity were evaluated by the Pittsburgh Sleep Quality Index (PSQI) and Insomnia Severity Index (ISI), respectively. RESULTS: A total of 369 differentially methylated positions (DMPs) and 23 differentially methylated regions (DMRs) were identified between the CID and control groups. LHX6 was identified as the most important differentially methylated gene (DMG). The Gene Ontology (GO) analysis results corroborated that DMPs were significantly enriched in 105 GO terms, including cell signaling, homogenous cell adhesion of plasma membrane adhesion molecules, nervous system development, cell adhesion, and calcium ion binding. In addition, it was demonstrated that DMPs were significantly enriched in the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including the hippo signaling pathway, Ras signaling pathway, and vitamin B6 metabolism. The DMR-related GO analysis results revealed the positive regulation of protein kinase activities. CONCLUSIONS: DNA methylation plays a critical role in the development of CID, and LHX6 is validated to be an important DMG.

2.
Schizophr Res ; 267: 113-121, 2024 May.
Article in English | MEDLINE | ID: mdl-38531158

ABSTRACT

A decreased expression of specific interneuron subtypes, containing either the calcium binding protein parvalbumin (PV) or the neurotransmitter somatostatin (SST), are observed in the cortex and hippocampus of both patients with schizophrenia and rodent models used to study the disorder. Moreover, preclinical studies suggest that this loss of inhibitory function is a key pathological mechanism underlying the symptoms of schizophrenia. Interestingly, decreased expression of Lhx6, a key transcriptional regulator specific to the development and migration of PV and SST interneurons, is seen in human postmortem studies and following multiple developmental disruptions used to model schizophrenia preclinically. These results suggest that disruptions in interneuron development in utero may contribute to the pathology of the disorder. To recapitulate decreased Lhx6 expression during development, we used in utero electroporation to introduce an Lhx6 shRNA plasmid and knockdown Lhx6 expression in the brains of rats on gestational day 17. We then examined schizophrenia-like neurophysiological and behavioral alterations in the offspring once they reached adulthood. In utero Lhx6 knockdown resulted in increased ventral tegmental area (VTA) dopamine neuron population activity and a sex-specific increase in locomotor response to a psychotomimetic, consistent with positive symptomology of schizophrenia. However, Lhx6 knockdown had no effect on social interaction or spatial working memory, suggesting behaviors associated with negative and cognitive symptom domains were unaffected. These results suggest that knockdown of Lhx6 during development results in neurophysiological and behavioral alterations consistent with the positive symptom domain of schizophrenia in adult rats.


Subject(s)
Disease Models, Animal , LIM-Homeodomain Proteins , Schizophrenia , Transcription Factors , Animals , Schizophrenia/metabolism , Schizophrenia/physiopathology , Schizophrenia/genetics , Female , Male , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Rats , Pregnancy , Gene Knockdown Techniques , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiopathology , Interneurons/metabolism , Interneurons/physiology , Rats, Sprague-Dawley , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Small Interfering
3.
Sleep ; 47(3)2024 Mar 11.
Article in English | MEDLINE | ID: mdl-37599437

ABSTRACT

To determine how a waking brain falls asleep researchers have monitored and manipulated activity of neurons and glia in various brain regions. While imaging Gamma-Aminobutyric Acid (GABA) neurons in the zona incerta (ZI) we found a subgroup that anticipates onset of NREM sleep (Blanco-Centurion C, Luo S, Vidal-Ortiz A, Swank C, Shiromani PJ. Activity of a subset of vesicular GABA-transporter neurons in the ventral ZI anticipates sleep onset. Sleep. 2021;44(6):zsaa268. doi:10.1093/sleep/zsaa268.). To differentiate the GABA subtype we now image and optogenetically manipulate the ZI neurons containing the transcription factor, Lhx6. In the first study, Lhx6-cre mice (n = 5; female = 4) were given rAAV-DJ-EF1a-DIO-GCaMP6M into the ZI (isofluorane anesthesia), a GRIN lens implanted, and 21days later sleep and fluorescence in individual Lhx6 neurons were recorded for 4 hours. Calcium fluorescence was detected in 132 neurons. 45.5% of the Lhx6 neurons were REM-max; 30.3% were wake-max; 11.4% were wake + REM max; 9% were NREM-max; and 3.8% had no change. The NREM-max group of neurons fluoresced 30 seconds ahead of sleep onset. The second study tested the effects of unilateral optogenetic stimulation of the ZI Lhx6 neurons (n = 14 mice) (AAV5-Syn-FLEX-rc[ChrimsonR-tdTomato]. Stimulation at 1 and 5 Hz (1 minute on- 4 minutes off) significantly increased percent REM sleep during the 4 hours stimulation period (last half of day cycle). The typical experimental approach is to stimulate neurons in both hemispheres, but here we found that low-frequency stimulation of ZI Lhx6 neurons in one hemisphere is sufficient to shift states of consciousness. Detailed mapping combined with mechanistic testing is necessary to identify local nodes that can shift the brain between wake-sleep states.


Subject(s)
Red Fluorescent Protein , Sleep, REM , Zona Incerta , Mice , Female , Animals , Sleep, REM/physiology , Zona Incerta/physiology , Optogenetics , Sleep/physiology , Neurons , gamma-Aminobutyric Acid
4.
Anat Rec (Hoboken) ; 306(4): 905-917, 2023 04.
Article in English | MEDLINE | ID: mdl-36583474

ABSTRACT

A recent report suggested that LIM homeobox 6 (Lhx6) + GABA-releasing neurons of the ventral zona incerta (VZI) promote sleep, particularly paradoxical sleep (PS). While their potential involvement in sleep still needs to be firmly confirmed, little is known about their specific input/output connections with widespread brain regions, including those involved in sleep. Thus, the present study was designed to examine whether Lhx6-expressing neurons (in parallel to intermingled MCH-expressing ones) may send efferent projections to cholinergic and/or monoaminergic nuclei from basal forebrain (BF) to brainstem (BS). Based on the present observations, the proportions of Lhx6+ neuronal projection to the BF and BS cholinergic nuclei over the total number of Lhx6+ VZI cells were approximately 5.9% and 6.9%, respectively. Likewise, the proportions of Lhx6+ neuronal projection to the dorsal raphe and locus coeruleus over the total number of Lhx6+ VZI cells were about 4.3% and 3.9%, respectively. In addition, Lhx6+ cells projecting to the cholinergic or monoaminergic nuclei were scattered along the entire dorsal-to-ventral extent of the VZI. Based on the present as well as our previous observations, it is suggested that Lhx6+ VZI neurons might play an important role in the regulation of PS, partly via the neural network involving the cholinergic as well as monoaminergic nuclei of the rat.


Subject(s)
Zona Incerta , Rats , Animals , Genes, Homeobox , Brain Stem/physiology , GABAergic Neurons , Cholinergic Agents
5.
J Clin Lab Anal ; 36(7): e24540, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35698314

ABSTRACT

BACKGROUND: Preeclampsia (PE) is a serious complication of pregnancy, with a global incidence of about 2%-8%. It is one of the important causes of morbidity and mortality among the pregnant women and perinatal infants. Circular RNA (circRNA) has been confirmed to play an important regulatory role in PE. This study aimed to evaluate the role of hsa_circ_0008726 in the occurrence and development of PE. METHODS: The expression of hsa_circ_0008726 in PE placental tissue and blood was detected by qRT-PCR. CCK-8, wound closure, and Transwell assay were used to measure cell proliferation, migration, and invasion. Bioinformatics prediction, Western blotting, and dual-luciferase reporter gene detection were used to explore the mechanism of hsa_circ_0008726 in HTR8/SVneo cells. RESULTS: The expression level of circ_0008726 in the placental tissue and blood samples of PE patients was significantly higher than that of normal controls. The overexpression of circ_0008726 can significantly inhibit the proliferation, migration, and invasion ability of HTR-8/SVneo cells. While the silence of circ_0008726 showed an opposite effect. Furthermore, hsa_circ_0008726 can modulate the expression of LHX6 by adsorbing miR-1290. CONCLUSION: Hsa_circ_000872 can regulate LHX6 by adsorbing miR-1290 to inhibit PE progression, thus establishing hsa_circ_000872 as a potential target for predicting and treating PE.


Subject(s)
MicroRNAs , Pre-Eclampsia , Cell Movement/genetics , Cell Proliferation/genetics , Female , Humans , LIM-Homeodomain Proteins , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Placenta/metabolism , Pregnancy , Signal Transduction/genetics , Transcription Factors , Trophoblasts/metabolism
6.
Cancer Med ; 11(19): 3657-3673, 2022 10.
Article in English | MEDLINE | ID: mdl-35384355

ABSTRACT

LIM homeobox 6 (LHX6) has been reported to be downregulated and inhibits cell proliferation in various cancers. Alternative splicing of LHX6 leads to six annotated isoforms, which can be found in the NCBI database. However, the expression patterns and potential roles of these isoforms remain poorly characterized in cervical cancer. Here, we demonstrated that the LHX6 isoforms containing exon 12 (LHX6EX(+12) group) and isoforms lacking exon 12 (LHX6EX(-12) group) were differentially expressed in cervical tissue by qRT-PCR. The mRNA expression level of LHX6EX(+12) group was higher than that of LHX6EX(-12) group in cervical cancer tissue. Knockdown of LHX6EX(+12) group and all LHX6 isoforms (LHX6All group) inhibited cell growth, increased cell apoptosis, and induced cell cycle arrest from G0/G1 phase to S phase in vitro. Consistently, overexpression of the LHX6EX(+12) group promoted cervical cancer cell proliferation in vitro. In contrast, no significant differences in cell proliferation were found between LHX6EX(-12) isoform knockdown group and its control. RNA-sequencing suggested that the LHX6EX(+12) isoform group might exert its cancer-promoting effects in cervical cancer via regulating MAPK signaling pathway. Downregulation of the LHX6EX(+12) group significantly suppressed the phosphorylation of MRK, ERK, JNK, and P38 at the protein level. We also identified some unique biological processes and signaling pathways in which each isoform group might be involved. In summary, our results indicated that LHX6EX(+12) isoform group was the dominant oncogenic type of LHX6 in cervical cancer, which may be a new biomarker and a potential precise therapeutic target for cervical cancer in the future.


Subject(s)
Uterine Cervical Neoplasms , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Exons , Female , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Nerve Tissue Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA , RNA, Messenger , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Uterine Cervical Neoplasms/genetics
7.
Cells ; 11(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-35269475

ABSTRACT

GABAergic interneurons control the neural circuitry and network activity in the brain. The dysfunction of cortical interneurons, especially those derived from the medial ganglionic eminence, contributes to neurological disease states. Pluripotent stem cell-derived interneurons provide a powerful tool for understanding the etiology of neuropsychiatric disorders, as well as having the potential to be used as medicine in cell therapy for neurological conditions such as epilepsy. Although large numbers of interneuron progenitors can be readily induced in vitro, the generation of defined interneuron subtypes remains inefficient. Using CRISPR/Cas9-assisted homologous recombination in hPSCs, we inserted the coding sequence of mEmerald and mCherry fluorescence protein, respectively, downstream that of the LHX6, a gene required for, and a marker of medial ganglionic eminence (MGE)-derived cortical interneurons. Upon differentiation of the LHX6-mEmerald and LHX6-mCherry hPSCs towards the MGE fate, both reporters exhibited restricted expression in LHX6+ MGE derivatives of hPSCs. Moreover, the reporter expression responded to changes of interneuron inductive cues. Thus, the LHX6-reporter lines represent a valuable tool to identify molecules controlling human interneuron development and design better interneuron differentiation protocols as well as for studying risk genes associated with interneuronopathies.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Cell Line , Humans , Induced Pluripotent Stem Cells/metabolism , Interneurons/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Median Eminence/metabolism , Nerve Tissue Proteins/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Gene Expr Patterns ; 43: 119227, 2022 03.
Article in English | MEDLINE | ID: mdl-34861428

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Developmental , Transcription Factors , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Maxilla/metabolism , Palate/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Environ Toxicol ; 37(3): 435-445, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34773443

ABSTRACT

Colorectal cancer (CRC) is one of the prevalent types of human malignancies and ranks as the second leading cause of cancer-associated death worldwide. Dysregulated miRNAs have been promulgated as oncogenes or tumor-suppressive genes participating in the initiation and progression of CRC. A recent study reported that miR-346 was highly expressed in CRC patients. However, the biological role and underlying mechanism of miR-346 in CRC remain elusive. qRT-PCR and western blot assays were employed to detect miR-346 and LIM homeobox domain 6 (LHX6) expression in CRC cells. Cell proliferation was evaluated by CCK-8 and BrdU assays. Apoptosis was evaluated by TUNEL assay. The interaction between miR-346 and LHX6 was assessed by luciferase reporter assay. Results showed that miR-346 expression was increased and LHX6 expression was reduced in CRC cells. miR-346 knockdown and LHX6 overexpression inhibited proliferation and promoted apoptosis of CRC cells. Additionally, we found that miR-346 negatively regulated LHX6 expression in CRC cells by directly targeting LHX6. LHX6 knockdown partially attenuated anti-miR-346-induced proliferation reduction and apoptosis promotion in CRC cells. Furthermore, miR-346 knockdown inhibited the protein kinase B (Akt)/mechanistic target of rapamycin (mTOR) pathway in CRC cells by targeting LHX6. The present study indicated that miR-346 knockdown repressed cell growth in CRC cells by upregulating LHX6, and this was associated with inactivation of the Akt/mTOR pathway.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , LIM-Homeodomain Proteins/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Transcription Factors/genetics , Up-Regulation
10.
Mol Ther Nucleic Acids ; 26: 760-772, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34729246

ABSTRACT

Communication between the maternal uterus and the embryo is vital for a successful pregnancy. Exosomes, subtypes of extracellular vesicles comprising many bioactive factors, regulate the early stages of pregnancy, specifically during embryo implantation. Nevertheless, the mechanism by which exosomal microRNAs (miRNAs) derived from placental trophoblasts regulate embryo implantation remains elusive. We isolated and identified exosomes derived from placental trophoblast cells (HTR8/SVneo). Subsequently, we evaluated the loading miRNA in exosomes by small RNA sequencing. Consequently, we showed that trophoblast cell-derived exosomes could transfer to endometrial epithelial cells. Besides, these exosomes promoted the epithelial-mesenchymal transition (EMT) as well as migration of endometrial cells and were implicated in the regulation of inflammation. Further, the specific miRNAs were screened in exosomes, and as a result, miRNA (miR)-1290 was enriched specifically in exosomes. miR-1290 promoted the expression of inflammatory factors (interleukin [IL]-6 and IL-8) and migration of endometrial epithelial cells. In addition, exosomal miR-1290 promoted angiogenesis in vitro. More importantly, by targeting LHX6, trophoblast HTR8/SVneo cell-derived exosomal miR-1290 promoted the EMT process of endometrial epithelial cell HEC-1-A. Altogether, our findings provide novel insights into the mechanism of trophoblast cell-derived exosomes during embryo implantation.

11.
Onco Targets Ther ; 13: 10983-10994, 2020.
Article in English | MEDLINE | ID: mdl-33149613

ABSTRACT

BACKGROUND: miR-214 has been reported to contribute to erlotinib resistance in non-small-cell lung cancer (NSCLC) through targeting LHX6; however, the molecular mechanisms underlying the involvement of LHX6 in mediating the resistance to EGFR-TKIs in erlotinib-resistant NSCLC HCC827 (HCC827/ER) cells remain unknown. This study aimed to investigate the mechanisms responsible for the contribution of LHX6 to EGFR-TKIs resistance in HCC827/ER cells. MATERIALS AND METHODS: HCC827/ER cells were generated by erlotinib treatment at a dose-escalation scheme. LHX6 knockout or overexpression was modeled in HCC827 and HCC827/ER cells, and then erlotinib IC50 values were measured. The cell migration ability was evaluated using a transwell migration assay, and the TCF/LEF luciferase activity was assessed with a TCF/LEF reporter luciferase assay. LHX6, ß-catenin and Cyclin D1 expression was quantified using qPCR and Western blotting assays. In addition, the LHX6 expression was detected in lung cancer and peri-cancer specimens using immunohistochemical staining, and the associations of LHX expression with the clinicopathological characteristics of lung cancer were evaluated. RESULTS: Lower LHX6 expression was detected in HCC827/ER cells than in HCC827 cells (P < 0.0001), while higher ß-catenin expression was seen in HCC827/ER cells than in HCC827 cells (P < 0.001). LHX6 knockout increased erlotinib resistance and cell migration ability in HCC827 cells, and LHX6 overexpression inhibited erlotinib resistance and cell migration ability in HCC827/ER cells. In addition, LHX6 mediated erlotinib resistance and cell migration ability in HCC827/ER cells via the Wnt/ß-catenin pathway. Immunohistochemical staining showed lower LHX6 expression in lung cancer specimens relative to peri-cancer specimens, and there were no associations of LHX6 expression with pathologic stage, gender, age or tumor size in lung cancer patients (P > 0.05). CONCLUSION: LHX6 down-regulation may induce EGFR-TKIs resistance and increase the migration ability of HCC827/ER cells via activation of the Wnt/ß-catenin pathway.

12.
Brain Res ; 1748: 147125, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32931819

ABSTRACT

There was a recent report suggesting that LIM homeobox 6 (Lhx6)+ GABA-releasing neurons of the ventral zona incerta (ZI) promote sleep. We demonstrated in the previous study that Lhx6+ ZI neurons are activated during paradoxical sleep (PS) hypersomnia which was induced by 48-hour PS deprivation, implying their roles in the control of PS like melanin-concentrating hormone (MCH) cells. Since the core portion of the lateral supramammillary nucleus (SUMl) is the major hypothalamic area activating the dentate gyrus as well as other limbic cortices during PS, we examined in the present study whether Lhx6+ ZI cells provide efferent projections to the SUMl, using the retrograde-tracing method. The majority of Lhx6+ neurons projecting to the SUMl occupied the ventral border (or ventral one-third) of the ventral ZI. Based on the quantitative analysis, the mean number of retrogradely-labeled Lhx6+ neurons was comparable to that of retrogradely-labeled MCH cells in the ZI. However, the total (i.e., single- plus double-labeled) number of Lhx6+ cells was approximately three times larger than that of MCH cells in the ZI. Thus, the proportion (about 7.8%) of retrogradely-labeled Lhx6+ neurons over the total Lhx6+ cells was approximately one-third of the percentage (about 20.9%) of retrogradely-labeled MCH neurons over the total MCH cells. On the other hand, a combination of retrogradely-labeled, Lhx6 and MCH cells occupied approximately 43.7% of the total retrogradely-labeled neurons in the ventral ZI. The present observations suggested that Lhx6+ neurons in the ventral ZI might play an important role in the regulation of PS, partly via the neural network involving the SUMl.


Subject(s)
Hypothalamus, Posterior/metabolism , LIM-Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Zona Incerta/metabolism , Animals , Neural Pathways/metabolism , Rats , Rats, Sprague-Dawley
13.
Biochem Biophys Res Commun ; 526(3): 626-632, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32248980

ABSTRACT

Epigenetic gene silencing by aberrant DNA methylation leads to loss of key cellular pathways in tumorigenesis. DNA methylation-mediated silenced genes in pancreatic cancer were searched for using the methyl-CpG targeted transcriptional activation (MeTA) method, and LHX6 (LIM homeobox 6), a transcription factor involved in embryogenesis and head development, was selected as a strong candidate gene. LHX6 was downregulated in most of the pancreatic cancer cell lines (83%, 10/12), mainly through promoter hypermethylation and histone deacetylation. Furthermore, LHX6 was methylated in primary pancreatic cancer specimens (57%, 16/28) in a tumor-specific manner. Re-expression of LHX6 inhibited colony formation and proliferation in LHX6 low-expressing pancreatic cancer cell lines, PK-1 and PK-9. In contrast, knockdown of LHX6 accelerated cell proliferation in LHX6 high-expressing pancreatic cancer cell lines, PCI-35 and MIA PaCa-2. In order to analyze LHX6 downstream genes, we performed microarray analyses using LHX6 inducible PK-1 and PK-9 and found that LHX6 induction upregulated several genes that had tumor suppressive functions. Among these, we focused on TFPI2 (Tissue factor pathway inhibitor-2) and found that TFPI2 was greatly downregulated in all twelve pancreatic cancer cell lines. Our present results suggest that epigenetic inactivation of LHX6 plays an important role in pancreatic tumorigenesis by promoting cell proliferation through aberrant transcriptional regulation of several cancer-related genes.


Subject(s)
Gene Expression Regulation, Neoplastic , LIM-Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Pancreatic Neoplasms/genetics , Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Gene Silencing , Humans , Pancreatic Neoplasms/pathology
14.
Cancers (Basel) ; 11(12)2019 Dec 02.
Article in English | MEDLINE | ID: mdl-31810245

ABSTRACT

In human epithelial ovarian cancer (EOC), various miRNAs can function as either oncogenes or tumor suppressor genes. We investigated miRNAs known to be involved in EOC progression and analyzed their expression in tissues and serum-derived exosomes from benign serous cystadenoma, borderline serous tumor, low-grade serous ovarian cancer, and high-grade serous ovarian cancer patients (HGSO). The HGSO group was divided based on the platinum-free interval, which is defined as the duration from the completion of platinum-based chemotherapy to recurrence. We also analyzed the mRNA levels of target genes that candidate miRNAs might regulate in patient tissues. miR-214-3p was highly expressed in tissues and exosomes derived from EOC with high malignancy and also found to regulate the expression of LIM homeobox domain 6 (LHX6) mRNA. Serum exosomal levels of miR-214-3p were significantly increased in platinum-resistant HGSO (25.2-fold, p < 0.001) compared to the exosomal expression of benign tumor patients. On transfection of miR-214-3p inhibitor in EOC cells, cell proliferation was inhibited while apoptotic cell death was increased. Collectively, we suggest that miR-214-3p in serum exosomes can be a potential biomarker for the diagnosis and prognosis of ovarian tumor, and its inhibition can be a supportive treatment for EOC.

15.
Exp Cell Res ; 385(1): 111649, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31562861

ABSTRACT

Exosomes are present within the local hypoxic tumor microenvironment, where they are able to transfer microRNAs between cells, thereby, effectively mediating cell-cell communication. Hypoxia plays a pivotal role in the progression of many tumor types such as hepatocellular carcinoma (HCC), but how hypoxia-induced exosomes in HCC affect HCC cells remains uncertain. In the present study, we found that hypoxic conditions induced increased exosomal production by HCC cells, and these exosomes, in turn, enhanced the proliferation, migration, and invasiveness in addition to epithelial-to-mesenchymal transition (EMT) in HCC cells under normoxic conditions. When we analyzed these exosomes, we found that miR-1273f were present at higher levels under hypoxic conditions, and we determined that this miRNA was responsible for directly replicating the effects of hypoxic exosomes within HCC cells, in addition to activating the Wnt/ß-catenin signaling. We finally identified LHX6, which is a known inhibitor of the Wnt/ß-catenin pathway, to be a miR-1273f target. These results, thus, provide evidence that hypoxic conditions can lead HCC cells to express increased exosomes that facilitate miR-1273f expression in normoxic cells, thereby enhancing their malignant phenotype at least in part by targeting LHX6 for downregulation.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Proliferation/genetics , Exosomes/genetics , Hypoxia/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Neoplasm Metastasis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/pathology , Neoplasm Metastasis/pathology , Tumor Microenvironment/genetics , Wnt Signaling Pathway/genetics
16.
Gene Expr Patterns ; 34: 119065, 2019 12.
Article in English | MEDLINE | ID: mdl-31376504

ABSTRACT

PURPOSE: To investigate the expression patterns of LIM Homeobox 6 (Lhx6) in the adult and developing mouse retina. METHODS: The Lhx6-GFP knock-in allele was used to activate constitutive expression of a GFP reporter in Lhx6 expressing cells. Double labeling with GFP and retinal markers in the mouse retina at postnatal day 56 (P56) was performed to identify the cell types expressing Lhx6. To determine the neuronal cell types that express Lhx6, double labeling with GFP and various retinal markers was employed in the differentiating retina at P7 and P15. RESULTS: GFP + Lhx6 lineage cells were determined in Brn3a + retinal ganglion cells (RGCs), ChAT + amacrine cells (ACs), and Islet-class LIM-homeodomain 1 (Isl1+) ACs in the mouse retina at P56. In the ganglion cell layer (GCL), Lhx6 was expressed in Brn3a + RGCs but not Brn3b + RGCs at P15. Moreover, in the inner nuclear layer (INL), Lhx6 was not expressed in Bhlhb5+ ACs at P15. However, Lhx6 was weakly expressed in Glyt1+ ACs and Pax6+ ACs, and strongly expressed in Isl1+ and ChAT + ACs at P15. CONCLUSION: Lhx6 was expressed in RGCs and ACs in both the adult and developing mouse retina.


Subject(s)
LIM-Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Retina/growth & development , Retina/metabolism , Transcription Factors/genetics , Age Factors , Amacrine Cells/metabolism , Animals , Cell Lineage , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Developmental/genetics , LIM-Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Retinal Ganglion Cells/metabolism , Transcription Factors/metabolism , Transcriptome/genetics
17.
Environ Pollut ; 252(Pt A): 216-226, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31151060

ABSTRACT

Microcystins (MCs) have been shown to be carcinogenic by animal and cellular experiments and found to be associated with the development of human hepatocellular carcinoma (HCC) through epidemiological studies. However, the molecular mechanism of microcystin-LR (MC-LR) induced HCC is still unclear. This study is determined to clarify the role and mechanism of LHX6 in MC-LR-induced hepatocarcinogenesis. Using the previously established MC-LR-induced malignant transformation model in L02 cells, we screened out LHX6, homeobox gene that was significantly changed. We found that LHX6 was significantly down-regulated in MC-LR treated L02 cells and the liver tissue of rats treated for 35 weeks with 10 µg/kg body weight of MC-LR. Expression of LHX6 in human tumor tissue was significantly down-regulated in high MC-LR-exposure group. LHX6 was hypermethylated in MC-LR treated L02 cells and up-regulated after treatment with 10 µM of 5-aza-2'-deoxycytidine. Furthermore, overexpression of LHX6 inhibited proliferation, invasion and migration of malignantly transformed L02 cells in vitro and in vivo, while knockdown of LHX6 resulted in an opposite phenotype. In addition, we found that up-regulation of P53 and Bax resulted in apoptosis, and that down-regulation of CTNNB1 and MMP7 led to migration of MC-LR treated L02 cells. Blockade of P53 and CTNNB1 by its inhibitor significantly diminished the effect of LHX6. These genes were working together during the process of MC-LR-induced hepatocarcinogenesis. Our study demonstrated for the first time that LHX6 gene expression is regulated by DNA methylation and can inhibit the proliferation, invasion and migration through Wnt/ß-catenin and P53 signaling pathways during the MC-LR-induced hepatocarcinogenesis. This result may suggest that LHX6 gene can be used as a potential target gene and a biomarker for liver cancer treatment.


Subject(s)
Carcinoma, Hepatocellular/chemically induced , Cell Transformation, Neoplastic/chemically induced , LIM-Homeodomain Proteins/metabolism , Liver Neoplasms/chemically induced , Microcystins/toxicity , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , DNA Methylation/drug effects , Decitabine/pharmacology , Epigenesis, Genetic , Humans , LIM-Homeodomain Proteins/genetics , Matrix Metalloproteinase 7/metabolism , Nerve Tissue Proteins/genetics , Rats , Signal Transduction , Transcription Factors/genetics , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Up-Regulation , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
18.
Cell Rep ; 27(13): 3752-3759.e4, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31242409

ABSTRACT

Neurexins are key synaptic organizers that are expressed in thousands of alternatively spliced isoforms. Because transsynaptic neurexin interactions with different postsynaptic molecules are largely isoform dependent, a cell type-level census of different neurexin isoforms could predict molecular interactions relating to synapse identity and function. Using single-cell transcriptomics to study the origin of neurexin diversity in multiple murine mature and embryonic cell types, we have discovered shared neurexin expression patterns in developmentally related cells. By comparing neurexin profiles in immature embryonic neurons, we show that neurexin profiles are specified during early development and remain unchanged throughout neuronal maturation. Thus, our findings reveal ontogenetic stability and provide a cell type-level census of neurexin isoform expression in the cortex.


Subject(s)
Alternative Splicing , Cerebral Cortex/metabolism , Neural Cell Adhesion Molecules , RNA-Seq , Single-Cell Analysis , Animals , Cerebral Cortex/cytology , Female , Male , Mice , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Optogenetics , Protein Stability
19.
Cereb Cortex ; 29(6): 2653-2667, 2019 06 01.
Article in English | MEDLINE | ID: mdl-29878134

ABSTRACT

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.


Subject(s)
Cerebral Cortex/cytology , Interneurons/cytology , Median Eminence/embryology , Neurogenesis/physiology , RNA-Binding Proteins/metabolism , Animals , Cell Movement/physiology , Cerebral Cortex/embryology , Interneurons/metabolism , Median Eminence/cytology , Mice , Transcription Factors/metabolism
20.
J Exp Neurosci ; 12: 1179069518784277, 2018.
Article in English | MEDLINE | ID: mdl-30013387

ABSTRACT

The mammalian cortex consists of two main neuronal types: the principal excitatory pyramidal neurons (PNs) and the inhibitory interneurons (INs). The interplay between these two neuronal populations - which drive excitation and inhibition (E/I balance), respectively - is crucial for controlling the overall activity in the brain. A number of neurological and psychiatric disorders have been associated with changes in E/I balance. It is not surprising, therefore, that neural networks employ several different mechanisms to maintain their firing rates at a stable level, collectively referred as homeostatic forms of plasticity. Here, we share our views on how the size of IN populations may provide an early homeostatic checkpoint for controlling brain activity. In a recent paper published in Cell Reports, we demonstrate that the extent of IN apoptosis during a critical early postnatal period is plastic, cell type specific, and can be reduced in a cell-autonomous manner by acute increases in neuronal activity. We propose that a critical interplay between the physiological state of the network and its cellular units fine-tunes the size of IN populations with the aim of stabilizing network activity.

SELECTION OF CITATIONS
SEARCH DETAIL