Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Heliyon ; 10(16): e35722, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39220970

ABSTRACT

Sevoflurane, an inhalation anesthetic, has been shown to suppress cancer development. In this study, we investigated the specific mechanisms involving sevoflurane, zinc-finger CCCH-type containing 13 (ZC3H13), and lncRNA DLX6-AS1 in gastric cancer (GC) progression, focusing on the N6-methyladenosine (m6A) modification of long non-coding RNAs (lncRNAs). We used quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses to measure the levels of ZC3H13 and lncRNA DLX6-AS1 in GC tissues and cells. Furthermore, we conducted Cell Counting Kit-8, colony formation, Transwell, and tumor xenograft assays to evaluate changes in GC cell malignancy following cell transfection and sevoflurane treatment. Additionally, actinomycin D, methylated RNA immunoprecipitation, and qRT-PCR assays were performed to examine the regulatory effects of ZC3H13 on the DLX6-AS1 m6A modification. We detected elevated levels of ZC3H13 in GC samples, while ZC3H13 silencing inhibited GC cell proliferation, migration, and invasion. Silencing ZC3H13 also enhanced the inhibitory effects of sevoflurane on GC cell malignancy. Moreover, we found that the increased expression of DLX6-AS1 in GC cells could be suppressed by ZC3H13 through the mediation of the m6A modification of DLX6-AS1, thereby reducing DLX6-AS1 stability. In conclusion, ZC3H13 knockdown enhances the inhibitory effect of sevoflurane on GC cell malignancy by inducing DLX6-AS1 m6A modification. Our findings may help identify potential therapeutic targets for the treatment of GC.

2.
Int Endod J ; 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38973098

ABSTRACT

AIM: The regenerative capacity of dental pulp relies on the odonto/osteogenic differentiation of dental pulp cells (DPCs), but dynamic microenvironmental changes hinder the process. Bone morphogenetic protein 9 (BMP9) promotes differentiation of DPCs towards an odonto/osteogenic lineage, forming dentinal-like tissue. However, the molecular mechanism underlying its action remains unclear. This study investigates the role of DLX6 antisense RNA 1 (DLX6-AS1) in odonto/osteogenic differentiation induced by BMP9. METHODOLOGY: Custom RT2 profiler PCR array, quantitative Real-Time PCR (qRT-PCR) and western blots were used to investigate the expression pattern of DLX6-AS1 and its potential signal axis. Osteogenic ability was evaluated using alkaline phosphatase and alizarin red S staining. Interactions between lncRNA and miRNA, as well as miRNA and mRNA, were predicted through bioinformatic assays, which were subsequently validated via RNA immunoprecipitation and dual luciferase reporter assays. Student's t-test or one-way ANOVA with post hoc Tukey HSD tests were employed for data analysis, with a p-value of less than .05 considered statistically significant. RESULTS: DLX6-AS1 was upregulated upon BMP9 overexpression in DPCs, thereby promoting odonto/osteogenic differentiation. Additionally, miR-128-3p participated in BMP9-induced odonto/osteogenic differentiation by interacting with the downstream signal MAPK14. Modifying the expression of miR-128-3p and transfecting pcMAPK14/siMAPK14 had a rescue impact on odonto/osteogenic differentiation downstream of DLX6-AS1. Lastly, miR-128-3p directly interacted with both MAPK14 and DLX6-AS1. CONCLUSIONS: DLX6-AS1 could regulate the odonto/osteogenic differentiation of DPCs under the control of BMP9 through the miR-128-3p/MAPK14 axis.

3.
Neurol Res ; 46(8): 706-716, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38735062

ABSTRACT

OBJECTIVE: Cerebral ischemia is a neurological disorder that leads to permanent disability. This research focuses on exploring the ameliorative effects of lipid nanoparticle (LNP)-encapsulated lncRNA DLX6-AS1 knockdown in cerebral ischemic injury via the Nrf2/HO-1/NLRP3 axis. METHODS: LNP-encapsulated lncRNA DLX6-AS1 was prepared. Cerebral ischemic injury mouse models were established utilizing middle cerebral artery occlusion (MCAO). The mice were treated by intravenous injection of LNP-encapsulated lncRNA DLX6-AS1. The neurological deficits, Inflammatory factor levels, pathological characteristics were observed. In vitro N2a cell oxygen and glucose deprivation (OGD) models were established, and the cells were treated with LNP-encapsulated lncRNA DLX6-AS1 or Nrf2 inhibitor (ML385). Cell viability and apoptosis were tested. DLX6-AS1, Nrf2, HO-1, and NLRP3 expression levels were assessed. RESULTS: LncRNA DLX6-AS1 levels were elevated in the brain tissues of mice with cerebral ischemic injury and OGD-induced N2a cells. LNP-encapsulated DLX6-AS1 siRNA (si-DLX6-AS1) improved neurological deficit scores, reduced the levels of inflammatory factors, improved brain tissue pathological damage, and raised the number of survival neurons in CA1. LNP-encapsulated si-DLX6-AS1 ameliorated the OGD-induced N2a cell viability decrease and apoptosis rate increase, and ML385 (Nrf2 inhibitor) reversed the ameliorative effects of LNP-encapsulated si-DLX6-AS1. In cerebral ischemic injury mice and OGD-induced N2a cells, Nrf2 and HO-1 levels were reduced and NLRP3 levels were increased. LNP-encapsulated si-DLX6-AS1 raised Nrf2 and HO-1 levels and reduced NLRP3 levels. Nrf2 inhibitor ML385 treatment reversed the ameliorative effects of LNP-encapsulated si-DLX6-AS1 on OGD-induced N2a cell viability and apoptosis. CONCLUSION: Lipid nanoparticle-encapsulated si-DLX6-AS1 ameliorates cerebral ischemic injury via the Nrf2/HO-1/NLRP3 axis.


Subject(s)
Brain Ischemia , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Nanoparticles , RNA, Long Noncoding , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Nanoparticles/administration & dosage , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Mice , Brain Ischemia/metabolism , Male , Mice, Inbred C57BL , Gene Knockdown Techniques/methods , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/genetics , Infarction, Middle Cerebral Artery , Membrane Proteins/metabolism , Membrane Proteins/genetics , Apoptosis/drug effects , Lipids , Liposomes , Heme Oxygenase-1
4.
Comb Chem High Throughput Screen ; 27(1): 118-126, 2024.
Article in English | MEDLINE | ID: mdl-37143278

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is recognized as one of the frequently diagnosed malignancies, and numerous microRNAs (miRs) are identified to be active in CRC. OBJECTIVE: This work aimed to clarify the effect of miR-141-3p on the radiosensitivity of CRC cells. METHODS: Firstly, CRC cell lines were cultured and applied to construct radiation-resistant CRC cells via X-ray treatment. The expression levels of miR-141-3p and long non-coding RNA DLX6 antisense RNA 1 (lncRNA DLX6-AS1) in CRC cells were measured using real-time quantitative polymerase chain reaction. After transfection with miR-141-3p mimics and 24 h treatment with 6- MV X-ray (0, 2, 4, 6 Gy), the survival fraction (SF) and the colony formation ability of CRC cells were determined using the cell counting kit-8 and colony formation methods. The interactions between miR-141-3p and DLX6-AS1 were analyzed using the dual-luciferase assay. The impact of miR-141-3p on DLX6-AS1 stability was detected after adding actinomycin-D. The role of DLX6- AS1 in the radiosensitivity of CRC cells was explored by transfecting oe-DLX6-AS1 into radiation- resistant CRC cells overexpressing miR-141-3p. RESULTS: The relative expression levels of miR-141-3p were downregulated in CRC cells and further declined in radiation-resistant cells. Upregulation of miR-141-3p relative expression reduced SF and the colony formation ability while amplifying the radiosensitivity of radiation-resistant CRC cells. miR-141-3p directly bound to DLX6-AS1 to reduce DLX6-AS1 stability, and therefore downregulated DLX6-AS1 expression. DLX6-AS1 overexpression counteracted the role of miR- 141-3p overexpression in amplifying the radiosensitivity of radiation-resistant CRC cells. CONCLUSION: miR-141-3p binding to DLX6-AS1 significantly decreased DLX6-AS1 stability and expression, promoting the radiosensitivity of CRC cells.


Subject(s)
MicroRNAs , Cell Proliferation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Up-Regulation , Cell Line , Radiation Tolerance/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics
5.
Animals (Basel) ; 13(17)2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37685013

ABSTRACT

Cleft lip and palate (CLP) is a well-known congenital defect in dogs, characterized by abnormal communication between the oral and nasal cavities. Its incidence rate is high and affects all dog breeds. The etiology of CLP is thought to be multifactorial, caused by both genetic and environmental factors. In this study, four puppies out of seven from a single litter of Staffordshire Bull Terrier dogs with craniofacial abnormalities were anatomically and genetically examined. Classical anatomical preparation, dyed-latex-injection of the arterial vessels, and cone-beam computed tomography were used. The puppies showed variations in their observable abnormalities: three of them had a complete cleft of the palate on both sides, while one puppy had a cleft on the right side only. Cytogenetic analysis showed a normal diploid chromosome number (2n = 78,XX or 78,XY) in the studied animals. Known genomic variants of CLP were examined in the ADAMTS20, DLX6, and MYH3 genes, but no mutations were identified. Further studies are needed to identify the breed-specific genetic variants associated with canine CLP.

6.
Noncoding RNA Res ; 8(4): 605-614, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37767112

ABSTRACT

Long non-coding RNA (lncRNA) distal-less homeobox 6 antisense RNA 1 (DLX6-AS1) is elevated in a variety of cancers, including non-small cell lung cancer (NSCLC) and cervical cancer. Although it was found that the microRNA-16-5p (miR-16), which is known to regulate autophagy and apoptosis, had been downregulated in similar cancers. Recent research has shown that in tumors with similar characteristics, DLX6-AS1 acts as a sponge for miR-16 expression. However, the cell death-related molecular mechanism of the DLX6-AS1/miR-16 axis has yet to be investigated. Therefore, we propose a dynamic Boolean model to investigate gene regulation in cell death processes via the DLX6-AS1/miR-16 axis. We found the finest concordance when we compared our model to many experimental investigations including gain-of-function genes in NSCLC and cervical cancer. A unique positive circuit involving BMI1/ATM/miR-16 is also something we predict. Our results suggest that this circuit is essential for regulating autophagy and apoptosis under stress signals. Thus, our Boolean network enables an evident cell-death process coupled with NSCLC and cervical cancer. Therefore, our results suggest that DLX6-AS1 targeting may boost miR-16 activity and thereby restrict tumor growth in these cancers by triggering autophagy and apoptosis.

7.
Genes (Basel) ; 14(8)2023 07 26.
Article in English | MEDLINE | ID: mdl-37628577

ABSTRACT

Split Hand-Foot Malformation (SHFM) is a congenital limb defect characterized by a median cleft of the hands and/or feet due to the absence/hypoplasia of the central rays. It may occur as part of a syndromic condition or as an isolated malformation. The most common of the six genetic loci identified for this condition is correlated to SHFM1 and maps in the 7q21q22 region. SHFM1 is characterized by autosomal dominant transmission, incomplete penetrance and variable expressivity. Associated features often include hearing loss, intellectual disability/developmental delay and craniofacial abnormalities. Disruption of the DLX5/DLX6 genes, mapping within the SHFM1 locus, is now known to be responsible for the phenotype. Through SNP array, we analyzed a patient affected by SHFM1 associated with deafness and an abnormality of the inner ear (incomplete partition type I); we identified a deletion in 7q21, not involving the DLX5/6 genes, but including exons 15 and 17 of DYNC1I1, known to act as exonic enhancers (eExons) of the DLX5/6 genes. We further demonstrated the role of DYNC1I1 eExons in regulating DLX5/6 expression by means of showing a reduced expression of the DLX5/6 genes through RT-PCR in a patient-derived lymphoblastoid cell line. Furthermore, our data and a review of published cases do not support the hypothesis that DLX5/6 are imprinted in humans. This work is an example of how the disruption of regulatory elements can be responsible for congenital malformations.


Subject(s)
Deafness , Limb Deformities, Congenital , Humans , Genes, Homeobox , Lower Extremity , Limb Deformities, Congenital/genetics , Deafness/genetics , Transcription Factors/genetics , Homeodomain Proteins/genetics
8.
J Orthop Case Rep ; 13(4): 49-52, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37193374

ABSTRACT

Introduction: Authors report a rare case report about split hand and foot malformation (SHFM) also sometimes referred to as ectrodactyly. Case Report: The patient with hand and foot malformations presented to casualty. A 60-year-old male was brought with alleged history of road traffic accident with tenderness and deformity in left thigh. On further physical examination, a malformation was present in bilateral feet and right hand. Plain radiographs were taken after emergency primary management which revealed a fracture of shaft of femur of the left side and absence of 2nd and 3rd phalanges in bilateral feet and lobster claw like malformation in the right hand. The patient was further investigated and operated with femur interlocking nail and later discharged under stable condition. Screening for other congenital defects was done. Conclusion: Patients with SHFM should undergo screening for other congenital anomalies. Electrocardiogram, 2D ECHO, chest radiograph, and ultrasonography abdomen should be done. Genetic analysis ideally should be done to identify mutations involved. Surgical intervention is only required when patient demands improved function of limb.

9.
Biotechnol Genet Eng Rev ; : 1-12, 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37078565

ABSTRACT

Hepatic carcinoma (HCC) is one of the most common malignant tumors worldwide, and the prognosis of HCC patients is often poor. Long-chain non-coding RNA (lncRNA) distal-less homeobox 6 antisense 1 (DLX6-AS1) has been shown to be involved in the pathogenesis of various cancers. This study aims to investigate the expression of DLX6-AS1 in HCC patients and its prognostic value. The serum DLX6-AS1 was quantified using a reverse transcription-polymerase chain reaction (RT-PCR) assay in both HCC patients and healthy individuals, and the correlation of DLX6-AS1 with clinicopathological features of HCC patients, as well as the diagnostic and prognostic value of DLX6-AS1 for HCC patients, were analyzed. The results showed that the expression of serum DLX6-AS1 in HCC patients was significantly higher than that of healthy individuals (P < 0.05), and DLX6-AS1 was related to tumor differentiation, pathological staging, and lymph node metastasis (all P < 0.05). Patients with high DLX6-AS1 expression showed significantly higher mortality than those with low DLX6-AS1 expression, and the DLX6-AS1 expression in dead patients was significantly higher than that in living patients. Furthermore, the AUC of DLX6-AS1 for poor prognosis of HCC patients was larger than 0.8. The univariate analysis revealed that the poor prognosis of HCC patients was related to pathological staging, lymph node metastasis, differentiation, and DLX6-AS1 expression (all P < 0.05), and the Cox multivariate analysis revealed that pathological staging, lymph node metastasis, differentiation, and DLX6-AS1 expression were independent risk factors for poor prognosis of HCC patients (all P < 0.05). These findings suggest that DLX6-AS1 may be a promising target for diagnosis, treatment, and prognosis prediction of HCC patients.

10.
Oncol Rep ; 49(2)2023 Feb.
Article in English | MEDLINE | ID: mdl-36601767

ABSTRACT

Long non­coding RNAs (lncRNAs) are common in the human body. Misregulated lncRNA expression can cause a variety of diseases in the human body. The present study aimed to investigate the effect of lncRNA differentiation antagonizing non­protein­coding RNA (DANCR) on glioma proliferation and autophagy through the microRNA (miR)­33b/distal­less homeobox 6 (DLX6)/autophagy­related 7 (ATG7) axis. Reverse transcription­quantitative PCR was used to detect DANCR and miR­33b expression. Cell Counting Kit­8 assay and flow cytometry were used to detect cell proliferation and apoptosis, respectively. Transmission electron microscopy was used to determine the autophagy level by observing intracellular autophagosomes. A western blot assay was used to detect protein expression levels and determine the level of autophagy in different cells. The binding sites of miR­33b and DANCR or DLX6 were detected using a dual­luciferase reporter assay. A chromatin immunoprecipitation assay confirmed DLX6 as a transcript of ATG7. In vivo tumorigenesis of glioma cells was validated in nude mice. DANCR and DLX6 were highly expressed in glioma cells, while miR­33b showed low expression in glioma cells. DANCR reduced the targeted binding of miR­33b to DLX6 by sponging miR­33b. The result verified that DANCR could promote ATG7 protein expression through miR­33b/DLX6, promote intracellular autophagy and proliferation and reduce apoptosis. The present study identified the role of the DANCR/miR­33b/DLX6/ATG7 axis in regulating autophagy, proliferation, and apoptosis in glioma cells, providing new ideas for glioma treatment.


Subject(s)
Glioma , MicroRNAs , RNA, Long Noncoding , Animals , Mice , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mice, Nude , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Autophagy/genetics , Apoptosis/genetics , Glioma/genetics , Cell Line, Tumor , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism
11.
Front Endocrinol (Lausanne) ; 13: 916173, 2022.
Article in English | MEDLINE | ID: mdl-35909540

ABSTRACT

Dlx5 and Dlx6 encode distal-less homeodomain transcription factors that are present in the genome as a linked pair at a single locus. Dlx5 and Dlx6 have redundant roles in craniofacial, skeletal, and uterine development. Previously, we performed a transcriptome comparison for anti-Müllerian hormone (AMH)-induced genes expressed in the Müllerian duct mesenchyme of male and female mouse embryos. In that study, we found that Dlx5 transcripts were nearly seven-fold higher in males compared to females and Dlx6 transcripts were found only in males, suggesting they may be AMH-induced genes. Therefore, we investigated the role of Dlx5 and Dlx6 during AMH-induced Müllerian duct regression. We found that Dlx5 was detected in the male Müllerian duct mesenchyme from E14.5 to E16.5. In contrast, in female embryos Dlx5 was detected in the Müllerian duct epithelium. Dlx6 expression in Müllerian duct mesenchyme was restricted to males. Dlx6 expression was not detected in female Müllerian duct mesenchyme or epithelium. Genetic experiments showed that AMH signaling is necessary for Dlx5 and Dlx6 expression. Müllerian duct regression was variable in Dlx5 homozygous mutant males at E16.5, ranging from regression like controls to a block in Müllerian duct regression. In E16.5 Dlx6 homozygous mutants, Müllerian duct tissue persisted primarily in the region adjacent to the testes. In Dlx5-6 double homozygous mutant males Müllerian duct regression was also found to be incomplete but more severe than either single mutant. These studies suggest that Dlx5 and Dlx6 act redundantly to mediate AMH-induced Müllerian duct regression during male differentiation.


Subject(s)
Genes, Homeobox , Mullerian Ducts , Animals , DNA-Binding Proteins/genetics , Female , Homeodomain Proteins/genetics , Male , Mice , Mullerian Ducts/metabolism , Sex Differentiation , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Front Genet ; 13: 871988, 2022.
Article in English | MEDLINE | ID: mdl-35719380

ABSTRACT

Accumulating evidence has revealed the vital regulatory roles of lncRNA DLX6-AS1 in various tumors at pre-transcriptional, transcriptional, and post-transcriptional levels, which makes it a potential prognosis factor and therapeutic target. In addition, the presence of lncRNA DLX6-AS1 in the exosomes of peripheral blood of patients with tumors may also contribute to it being a possible cancer-related biomarker. However, most literature studies are devoted to studying the effect of lncRNA DLX6-AS1 as a sponging molecule of miRNAs, the research of which is likely to get stuck into a dilemma. Literature studies published already have demonstrated an exciting cell malignant phenotype inhibition with the knockdown of lncRNA DLX6-AS1 in various tumor cell lines. With the comprehensive development of delivery systems, high-throughput sequencing, and aptamers, the problems of finding novel research methods and exploring the therapeutic options which are based on lncRNA DLX6-AS1 in vivo could come into a period to deal with. This review aims to summarize the research statuses of lncRNA DLX6-AS1, discuss other study methodologies and therapeutic strategies on it, which might be of help to the deep learning of lncRNA DLX6-AS1 and its application from basic to clinical research.

13.
Behav Brain Res ; 431: 113923, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35550840

ABSTRACT

Parkinson's disease (PD) is a prevailing neurodegenerative disorder. This study discussed the mechanism of lncRNA distal-less homeobox 6 antisense 1 (DLX6-AS1) on inflammatory responses in PD. With healthy male C57BL/6 mice (8-10 weeks) and BV2 microglia as study subjects, we established PD models in vivo/in vitro by injection of 1-methyl-4-phenyl-2, 3, 6-tetrahydropyridine (MPTP) for 4 weeks and treatment of lipopolysaccharide (LPS) for 24 h, respectively. DLX6-AS1 expression in PD mice and BV2 microglia was examined using reverse transcription quantitative-polymerase chain reaction and then down-regulated via stereotaxic catheter injection or cell transfection to evaluate its effect on neurological function. Meanwhile, the cell number of TH+ /Caspase3 + /IBA1 + in substantia nigra, cell viability, and apoptosis rate of BV2 microglia, inflammatory levels, and NLR family pyrin domain containing 3 (NLRP3) inflammasome were determined using immunohistochemistry, MTT assay, flow cytometry, ELIZA assay, and Western blot. The binding relationship between miR-223-3p and DLX6-AS1/Neuropilin-1 (NRP1) was verified by dual-luciferase assay and RNA immunoprecipitation assay. After down-regulation of DLX6-AS1, we down-regulated/overexpressed miR-223-3p/NRP1 levels in BV2 microglia. DLX6-AS1 was overexpressed in PD mice. Silencing DLX6-AS1 improved neurological function and alleviated microglial inflammation in PD mice. Specifically, the latency of mice falling from the rotating rod was longer, and the latency of climbing rod test was shorter; TH+ cells increased, while Caspase3 + /IBA1 + cells decreased; the levels of inflammatory were lowered. Silencing DLX6-AS1 inhibited LPS-induced inflammation of BV2 microglia. DLX6-AS1 acted as the ceRNA of miR-223-3p to promote NRP1. Down-regulation of miR-223-3p or overexpression of NRP1 partially annulled the effect of silencing DLX6-AS1 on BV2 microglial inflammation. Overall, DLX6-AS1 promotes the microglial inflammatory response in PD through the ceRNA mechanism of miR-223-3p/NRP1.


Subject(s)
MicroRNAs , Parkinson Disease , RNA, Long Noncoding , Animals , Homeodomain Proteins/genetics , Humans , Inflammation/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Microglia/metabolism , Neuropilin-1/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , RNA, Long Noncoding/genetics
14.
Front Cell Dev Biol ; 10: 746443, 2022.
Article in English | MEDLINE | ID: mdl-35281110

ABSTRACT

Long non-coding RNAs (lncRNAs) are a heterogeneous group of ncRNAs with characteristic size of more than 200 nucleotides. An increasing number of lncRNAs have been found to be dysregulated in many human diseases particularly cancer. However, their role in carcinogenesis is not precisely understood. DLX6-AS1 is an lncRNAs which has been unveiled to be up-regulated in various number of cancers. In different cell studies, DLX6-AS1 has shown oncogenic role via promoting oncogenic phenotype of cancer cell lines. Increase in tumor cell proliferation, migration, invasion, and EMT while suppressing apoptosis in cancer cells are the effects of DLX6-AS1 in development and progression of cancer. In the majority of cell experiment, mediator miRNAs have been identified which are sponged and negatively regulated by DLX6-AS1, and they in turn regulate expression of a number of transcription factors, eventually affecting signaling pathways involved in carcinogenesis. These pathways form axes through which DLX6-AS1 promotes carcinogenicity of cancer cells. Xenograft animal studies, also have confirmed enhancing effect of DLX6-AS1 on tumor growth and metastasis. Clinical evaluations in cancerous patients have also shown increased expression of DLX6-AS1 in tumor tissues compared to healthy tissues. High DLX6-AS1 expression has shown positive association with advanced clinicopathological features in cancerous patients. Survival analyses have demonstrated correlation between high DLX6-AS1 expression and shorter survival. In cox regression analysis, DLX6-AS1 has been found as an independent prognostic factor for patients with various types of cancer.

15.
Kidney Blood Press Res ; 47(3): 177-184, 2022.
Article in English | MEDLINE | ID: mdl-35038705

ABSTRACT

INTRODUCTION: Diabetic nephropathy (DN) is the leading cause of kidney failure worldwide. To explore the pathogenesis and effective biological target of DN is beneficial to seeking novel treatment strategies. OBJECTIVE: This study aimed to investigate the role of the lncRNA Dlx6os1/SOX6/EZH2 axis in DN progression. METHODS: PAS staining was performed to evaluate extracellular matrix accumulation; ELISA was carried out to assess the levels of urine microalbumin and blood glucose concentration; RT-qPCR was carried out to detect the levels of lncRNA Dlx6os1, TNF-α, IL-1ß, IL-6, SOX6, and EZH2. Western blot was performed to assess the levels of Col-IV, FN, TGF-ß1, and SOX6 proteins. RIP assay was carried out to verify the interaction between lncRNA Dlx6os1 and EZH2. ChIP-qPCR was conducted to verify the interaction between EZH2 and SOX6 promoter. RESULTS: Our results illustrated that lncRNA Dlx6os1 was highly expressed in DN mice and HG-induced SV40 MES13 cells. LncRNA Dlx6os1 knockdown inhibited HG-induced SV40 MES13 cell proliferation, fibrosis, and inflammatory cytokine release. LncRNA Dlx6os1 inhibited SOX6 expression by recruiting EZH2 in HG-SV40 MES13 cells, and SOX6 mediated the effects of lncRNA Dlx6os1 on proliferation, fibrosis, and inflammatory factor release of HG-induced SV40 MES13 cells. CONCLUSION: LncRNA Dlx6os1 accelerates the progression of DN by epigenetically repressing SOX6 via recruiting EZH2.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , RNA, Long Noncoding , Animals , Cell Proliferation , Diabetic Nephropathies/pathology , Enhancer of Zeste Homolog 2 Protein , Fibrosis , Mice , RNA, Long Noncoding/genetics , SOXD Transcription Factors
16.
Expert Rev Mol Med ; 23: e17, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34823630

ABSTRACT

Long non-coding RNAs (lncRNAs) have important roles in regulating the expression of genes and act as biomarkers in the initial development of different cancers. Increasing research studies have verified that dysregulation of lncRNAs occurs in various pathological processes including tumorigenesis and cancer progression. Among the different lncRNAs, DLX6-AS1 has been reported to act as an oncogene in the development and prognoses of different cancers, by affecting many different signalling pathways. This review summarises and analyses the recent research studies describing the biological functions of DLX6-AS1, its overall effect on signalling pathways and the molecular mechanisms underlying its action on the expression of genes in multiple human cancers. Our critical analysis suggests that different signalling pathways associated to this lncRNA may be used as a biomarker for diagnosis, or targets of treatment in cancers.


Subject(s)
MicroRNAs , Neoplasms , RNA, Long Noncoding , Cell Proliferation , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Neoplasms/genetics , Oncogenes/genetics , RNA, Long Noncoding/genetics
17.
Hum Cell ; 34(6): 1642-1652, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34508305

ABSTRACT

Long non-coding RNAs (lncRNAs) are a kind of translational-repressor RNAs composed of more than 200 nucleotides and formerly considered as "transcriptional noise". Recently studies have shown that lncRNAs could bind to multiple biomolecules such as DNA, transcription factors, RNA, chromatin complexes and proteins, and regulate target gene expression at multi-levels, thus playing an essential role in human tumors. DLX6-AS1, a recently discovered oncogenic lncRNA, is highly expressed in various human tumors, including lung cancer, liver cancer and pancreatic cancer. This paper mainly reviewed the regulatory mechanism of DLX6-AS1 as a competitive endogenous RNA (ceRNA) in tumor cell proliferation, cell apoptosis, angiogenesis, epithelial-mesenchymal transformation, chemotherapy resistance and metabolic changes. Furthermore, the translational value of DLX6-AS1 in cancer was also elucidated, which suggested its potential as a diagnostic or prognostic biomarker in cancer. In summary, this present article not only makes an in-depth analysis of the expression changes and carcinogenic mechanism of DLX6-AS1 in various human cancers, but also provides a new breakthrough for the diagnosis and treatment of cancers.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Apoptosis/genetics , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression , Homeodomain Proteins/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/therapy , Neovascularization, Pathologic/pathology , Prognosis , Protein Biosynthesis
18.
J Cell Mol Med ; 25(19): 9319-9330, 2021 10.
Article in English | MEDLINE | ID: mdl-34514705

ABSTRACT

Long non-coding RNA DLX6 antisense RNA 1 (DLX6-AS1) lists a critical position in thyroid carcinoma (TC) development. However, the overall comprehension about DLX6-AS1, microRNA (miR)-193b-3p and homeobox A1 (HOXA1) in TC is not thoroughly enough. Concerning to this, this work is pivoted on DLX6-AS1/miR-193b-3p/HOXA1 axis in TC cell growth and autophagy. TC tissues and adjacent normal thyroid tissues were collected, in which expression of DLX6-AS1, miR-193b-3p and HOXA1 was tested, together with their interactions. TC cells were transfected with DLX6-AS1/miR-193b-3p-related oligonucleotides or plasmids to test cell growth and autophagy. Tumorigenesis in nude mice was observed. DLX6-AS1 and HOXA1 were up-regulated, and miR-193b-3p was down-regulated in TC. Depleted DLX6-AS1 or restored miR-193b-3p disturbed cell growth and promoted autophagy. DLX6-AS1 targeted miR-193b-3p and positively regulated HOXA1. miR-193b-3p inhibition mitigated the impaired tumorigenesis induced by down-regulated DLX6-AS1. Tumorigenesis in nude mice was consistent with that in cells. It is clear that DLX6-AS1 depletion hinders TC cell growth and promotes autophagy via up-regulating miR-193b-3p and down-regulating HOXA1.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Homeodomain Proteins/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Transcription Factors/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Disease Models, Animal , Female , Heterografts , Humans , Mice , RNA Interference
19.
Cureus ; 13(2): e13572, 2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33815981

ABSTRACT

Introduction There are various genes that affect craniofacial development and among the important genes that affect jaw development is distal-less homeobox (DLX) 6 genes. The present study was carried out to determine the role of DLX6 gene variations in mandibular deficiency. Methods Thirty subjects having retrognathic mandible were evaluated by clinical examination and assessed using lateral cephalometric radiographs based on cephalometrics for orthognathic surgery (COGS) analysis of hard tissue with N-Pog parameters being less than -13 mm. For the same subjects, saliva samples were taken and sent to biotechnology labs for genetic evaluation. DNA was isolated from salivary samples using a DNA extraction kit and was subjected to polymerase chain reaction (PCR) amplification and sequencing. Single nucleotide polymorphisms (SNP) analysis was done to assess the role of DLX6 gene in these study subjects.  Results  All 30 subjects showed N-POG parameters of COGS analysis for hard tissue to be less than -13mm, confirming retrognathic mandible. SNP analysis of subjects showed no SNPs in any EXON of the DLX6 gene for all 30 study samples. Conclusion  No variations in DLX6 gene were found in the present study. Further studies are required to investigate other genes that could be involved in the cause of retrognathic mandible with a larger sample size and to include subjects in the sample having features other than mandibular retrognathia like hearing loss, abnormal pinnae, ectrodactyly, cleft palate, developmental delay and abnormal teeth to determine the contribution of DLX6 gene variations in mandibular deficiency.

20.
Front Genet ; 12: 616329, 2021.
Article in English | MEDLINE | ID: mdl-33747042

ABSTRACT

Hemifacial microsomia (HM) is a craniofacial congenital defect involving the first and second branchial arch, mainly characterized by ocular, ear, maxilla-zygoma complex, mandible, and facial nerve malformation. HM follows autosomal dominant inheritance. Whole-exome sequencing of a family revealed a missense mutation in a highly conserved domain of ITPR1. ITPR1 is a calcium ion channel. By studying ITPR1's expression pattern, we found that ITPR1 participated in craniofacial development, especially the organs that corresponded to the phenotype of HM. In zebrafish, itpr1b, which is homologous to human ITPR1, is closely related to craniofacial bone formation. The knocking down of itpr1b in zebrafish could lead to a remarkable decrease in craniofacial skeleton formation. qRT-PCR suggested that knockdown of itpr1b could increase the expression of plcb4 while decreasing the mRNA level of Dlx5/6. Our findings highlighted ITPR1's role in craniofacial formation for the first time and suggested that ITPR1 mutation contributes to human HM.

SELECTION OF CITATIONS
SEARCH DETAIL