Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Transl Oncol ; 50: 102139, 2024 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-39395273

RESUMO

LncRNAs affect tumorigenesis, and although the genesis, regulation and physiological mechanism of lncRNAs in gastric cancer (GC) have been reported, the research of lncRNAs still have a lot of value. Through comprehensive bioinformatics analysis, we screened the candidate lncRNA WAC-AS1(WAC-AS1). We analyzed WAC-AS1 expression in GC related tissues and cells using qRT-PCR. WAC-AS1's impact on GC growth and metastasis was investigated. LncRNA WC-AS-miR-204-5p-HOXC8 interaction was established through dual-luciferase reporter, FISH, RIP and RNA pull-down assay. We observed substantial upregulation in WAC-AS1 expression in cells and tissues of GC. WAC-AS1 through miR-204-5p/HOXC8 axis promoted GC proliferation, invasion, and migration. WAC-AS1 plays a cancer-promoting role for promoting the progression of GC.

2.
Discov Med ; 36(182): 559-570, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38531796

RESUMO

BACKGROUND: The epidermal growth factor receptor 2 (HER2) is overexpressed in 30% of breast cancers, and this overexpression is strongly correlated with a poor prognosis. Herceptin is a common treatment for HER2-positive breast cancer; however, cancer cells tend to adapt gradually to the drug, rendering it ineffective. The study revealed an association between the methylation status of the Homeobox C8 (HOXC8) gene and tumor development. Therefore, it is of paramount importance to delve into the interaction between HOXC8 and HER2-positive breast cancer, along with its molecular mechanisms. This exploration holds significant implications for a deeper understanding of the pathophysiological processes underlying HER2-positive breast cancer. METHOD: Tumor tissue and pathological data from patients with HER2-positive breast cancer were systematically collected. Additionally, the human HER2-positive breast cancer cell line, SKBR3, was cultured in vitro to assess both the expression level of HOXC8 and the degree of DNA methylation. The study aimed to explore the relationship between the relative expression of HOXC8 and the clinical characteristics of breast cancer patients. The expression level of HOXC8 and the promoter methylation of HOXC8 were verified by methylation treatment of SKBR3 breast cancer cells. The regulation of HOXC8 was meticulously carried out, leading to the division of the cells into distinct groups. The study further analyzed the expression levels and biological capabilities within each group. Finally, the in vitro and in vivo sensitivity of the cells to Herceptin, a common treatment for HER2-positive breast cancer, was measured to assess the efficacy of the drug. RESULT: In HER2-positive breast cancer cases characterized by poor methylation, there was an up-regulation of HOXC8. Its expression was found to be correlated with key clinical factors such as tumor size, lymph node status, clinical tumor, node, metastasis (cTNM) staging, and Herceptin resistance (p < 0.05). Upon methylation of breast cancer cells, there was a significant decrease in HOXC8 expression (p < 0.05). The study revealed that overexpression of HOXC8 resulted in increased proliferation, cloning, and metastasis of HER2-positive breast cancer cells, along with a reduced apoptosis rate (p < 0.05). Conversely, interference with HOXC8 expression reversed this scenario (p < 0.05). A Herceptin-resistant substrain, POOL2, was established using SKBR3 cells. Animal studies demonstrated that overexpressing HOXC8 accelerated tumor development and enhanced POOL2 cells' resistance to Herceptin (p < 0.05). However, following interference with HOXC8, POOL2 cells exhibited increased responsiveness to Herceptin, leading to a gradual reduction in tumor size (p < 0.05). CONCLUSIONS: In HER2-positive breast cancer, the expression of HOXC8 is elevated in a manner dependent on DNA methylation, and this elevated expression is closely linked to the pathology of the patient. Interfering with HOXC8 expression demonstrates the potential to partially inhibit the development and spread of breast cancer, as well as to alleviate resistance to Herceptin.


Assuntos
Neoplasias da Mama , Animais , Humanos , Feminino , Trastuzumab/genética , Trastuzumab/metabolismo , Trastuzumab/farmacologia , Neoplasias da Mama/patologia , Metilação de DNA , Receptor ErbB-2/metabolismo , Linhagem Celular Tumoral , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/uso terapêutico
3.
J Obstet Gynaecol ; 44(1): 2311658, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38348790

RESUMO

BACKGROUND: Human papillomavirus (HPV) is a risk factor for the occurrence of cervical cancer (CC). Here, we aimed to explore the role of HPV16 in CC and identify the underlying mechanism. METHODS: The expression of miR-23a, HPV16 E6/E7 and homeobox C8 (HOXC8) was measured by quantitative real-time PCR or western blot. Cell viability and migration were evaluated using cell counting kit-8, Transwell and wound healing assays. The targeting relationship between miR-23a and HOXC8 was revealed by dual-luciferase reporter assay. RESULTS: miR-23a was downregulated in HPV16-positive (HPV16+) CC tissues and HPV16+ and HPV18+ cells. Additionally, E6/E7 expression was increased in CC cells. Then, we found that E7, rather than E6, positively regulated miR-23a expression. miR-23a suppressed cell viability and migration, whereas E7 overexpression abrogated this suppression. miR-23a targeted HOXC8, which reversed miR-23a-mediated cell viability and migration. CONCLUSIONS: HPV16 E7-mediated miR-23a suppressed CC cell viability and migration by targeting HOXC8, suggesting a novel mechanism of HPV-induced CC.


Cervical cancer (CC) is a common gynaecological malignancy, and persistent human papillomavirus (HPV) infection, especially HPV16, is a main cause of CC. In this study, we explored the role of HPV16 in CC and the molecular mechanism. We used in vitro study to measure CC cell biological behaviours mediated by HPV16 E7, miR-23a and homeobox C8 (HOXC8). We found that HPV16 E7 promotes CC cell viability and migration. miR-23a expression is decreased in CC cells and inhibits cell viability and migration. HOXC8 is a target of miR-23a that reversed the effects on cellular processes caused by miR-23a. These results showed that miR-23a and HOXC8 may be the therapeutic targets of HPV16 E7-infected CC. What is more, our findings provide new insights into the progression of CC.


Assuntos
MicroRNAs , Infecções por Papillomavirus , Neoplasias do Colo do Útero , Feminino , Humanos , Papillomavirus Humano 16/genética , Linhagem Celular Tumoral , Neoplasias do Colo do Útero/genética , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/genética , Sobrevivência Celular/genética , MicroRNAs/genética , Proteínas de Homeodomínio/genética
4.
Heliyon ; 9(11): e21346, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37885723

RESUMO

Background: Accumulating evidence have highlighted the essential roles of HOX genes in embryonic development and carcinogenesis. As a member of the HOX gene family, the abnormal expression of HOXC8 gene is associated with the progression and metastasis of various tumors. However, potential roles of HOXC8 in colorectal cancer (CRC) prognosis and tumor microenvironment (TME) remodeling remain unclear. Methods: We conducted an integrated analysis of clinical and molecular characteristics, relevant oncogenic and immune regulation roles and drug sensitivity features of HOXC8 in CRC. Results: HOXC8 expression was markedly high expressed in CRC samples compared to normal samples, and the upregulated expression of HOXC8 was associated with poor prognosis. High HOXC8 expression was significantly associated with invasion-related pathways especially epithelial-mesenchymal transition (EMT). In vitro experiments showed significantly up-regulated HOXC8 expression in some CRC cell lines and its promoting effect on EMT and cell proliferation. TME categorization through transcriptomic analysis of CRC patients with high HOXC8 expression identified two different TME subtypes known as immune-enriched with fibrotic subtype and immune-depleted subtype. Patients with immune-enriched, fibrotic subtype exhibited significantly longer progression-free survival (PFS), upregulated PD-L1 and CTLA4 expression and higher TMB than those with the immune-depleted subtype. Conclusions: HOXC8 overexpression was associated with poor prognosis and specific TME subtypes in CRC. This study provided valuable resource for further exploring the potential mechanisms and therapeutic targets of HOX genes in CRC.

5.
J Cancer ; 14(13): 2552-2561, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37670969

RESUMO

Genes of the homeobox (HOX) family encode transcription factors, which play a role in cancer progression. However, their role in gastric cancer has not been adequately evaluated. Herein, we evaluated the genetic changes and mRNA of target genes of the HOX family in gastric cancer patients using publicly available online datasets. We found that HOXC8 was amplified in gastric cancer tissues, and mRNA expression levels were significantly associated with tumor status (P=0.044) and poor overall survival (P<0.01). HOXC8 knockdown significantly reduced the viability of gastric cancer cell lines. HOXC8 modulated the expression of secreted phosphoprotein 1 (SPP1, osteopontin) and phosphorylation of AKT/ERK in gastric cancer cells. Survival analysis demonstrated a decrease in overall survival rates among the high HOXC8/high SPP1 expression group compared with the low HOXC8/low SPP1 expression group. In conclusion, HOXC8 may be an independent prognostic factor and serve as a useful predictive biomarker for gastric cancer.

6.
Animals (Basel) ; 13(16)2023 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-37627406

RESUMO

Transcription factor Homeobox C8 (HOXC8) is identified as a white adipose gene as revealed by expression profile analysis in fat tissues. However, the specific role of HOXC8 in fat accumulation remains to be identified. This study was designed to reveal the effects of HOXC8 on preadipocyte proliferation and differentiation. We first make clear that the expression of HOXC8 is associated with fat contents in muscles, highlighting a role of HOXC8 in fat accumulation. Next, it is demonstrated that HOXC8 promotes the proliferation and differentiation of preadipocytes through gain- and loss-of-function assays in primary cultured porcine preadipocytes. Then, mechanisms underlying the regulation of HOXC8 on preadipocyte proliferation and differentiation are identified with RNA sequencing, and a number of differentially expressed genes (DEGs) in response to HOXC8 knockdown are identified. The top GO (Gene Ontology) terms enriched by DEGs involved in proliferation and differentiation, respectively, are identical. IL-17 signaling pathway is the common one significantly enriched by DEGs involved in preadipocyte proliferation and differentiation, respectively, indicating its importance in mediating fat accumulation regulated by HOXC8. Additionally, we find that the inhibition of proliferation is one of the main processes during preadipocyte differentiation. The results will contribue to further revealing the mechanisms underlying fat accumulation regulated by HOXC8.

7.
Biochem Biophys Res Commun ; 662: 39-46, 2023 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-37099809

RESUMO

Liver fibrosis occurs in any chronic liver disease, where extraordinary increase of extracellular matrix components is caused by the hepatic stellate cell (HSC) activation. HOXC8 has been disclosed to participate inregulating cell proliferation and fibrosis in tumors. However, the role of HOXC8 in liver fibrosis and the underlying molecular mechanisms has not yet been investigated. In this study, we founded that HOXC8 mRNA and protein was elevated in a carbon tetrachloride (CCl4)-induced liver fibrosis mouse model and transforming growth factor-ß (TGF-ß)-treated human (LX-2) HSC cells. Importantly, we observed that downregulating HOXC8 alleviates liver fibrosis and suppressed the fibrogenic gene induction induced by CCl4 in vivo. In addition, inhibition of HOXC8 suppressed the HSC activation and the expression of fibrosis-associated genes (α-SMA and COL1a1) induced by TGF-ß1 in LX-2 cells in vitro, while HOXC8 overexpression had the opposite effects. Mechanistically, we demonstrated HOXC8 activates TGFß1 transcription and enhanced the phosphorylated Smad2/Smad3 levels, suggesting a positive feedback loop between HOXC8 and TGF-ß1 that facilitates TGF-ß signaling and subsequent HSCs activation. Collectively, our data strongly indicated that a HOXC8/TGF-ß1 positive feedback loop plays as a critical role in controlling the HSC activation and in the liver fibrosis process, suggesting that inhibition of HOXC8 may serve as a promoting therapeutic strategy for diseases characterized by liver fibrosis.


Assuntos
Células Estreladas do Fígado , Fator de Crescimento Transformador beta1 , Camundongos , Animais , Humanos , Fator de Crescimento Transformador beta1/metabolismo , Células Estreladas do Fígado/metabolismo , Retroalimentação , Cirrose Hepática/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fibrose , Tetracloreto de Carbono/toxicidade , Fígado/metabolismo , Proteína Smad3/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteína Smad2/metabolismo
8.
Hum Cell ; 36(1): 195-208, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36308681

RESUMO

Gestational diabetes mellitus (GDM) is an increasingly frequent disease occurred during pregnancy. HOXC8 has been disclosed to take part in the regulation of cancers. Additionally, the HOXC8 expression was dramatically decreased in the placenta of pre-eclampsia patients, but its expression and function have not been investigated in GDM. In this work, it was demonstrated that the mRNA and protein expression of HOXC8 was lower in GDM placenta tissues and GDM cell model. In addition, HOXC8 facilitated trophoblast cell proliferation and weakened trophoblast cell mitochondrial apoptosis. HOXC8 enhanced trophoblast cell migration and angiogenesis. Moreover, HOXC8 activated the TGFß1-mediated Notch1 signaling pathway. Results showed that the mRNA and protein expressions of TGFß1 and Notch1 were both lower in the GDM group than that in the NP group. Besides, there were positive correlations among HOXC8, TGFß1 and Notch1. Inhibition of TGFß1 (SB202190 treatment) reversed the effects of HOXC8 on trophoblast cells through modulating cell proliferation, mitochondrial apoptosis, migration and angiogenesis. At last, through in vivo experiments, it was identified that HOXC8 relieved GDM symptoms in vivo. In conclusion, HOXC8 alleviated HG-stimulated damage of trophoblast cells during GDM through activating TGFß1-mediated Notch1 pathway. This discovery may provide a novel and useful bio-target for GDM treatment.


Assuntos
Diabetes Gestacional , Gravidez , Feminino , Humanos , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Trofoblastos/metabolismo , Placenta , Implantação do Embrião , Glucose/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo
9.
Genes (Basel) ; 13(11)2022 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-36421849

RESUMO

An increase in the number of vertebrae can significantly affect the meat production performance of livestock, thus increasing carcass weight, which is of great importance for livestock production. The homeobox gene C8 (HOXC8) has been identified as an essential candidate gene for regulating vertebral development. However, it has not been researched on the Dezhou donkey. This study aimed to verify the Dezhou donkey HOXC8 gene's polymorphisms and assess their effects on multiple vertebral numbers and carcass weight. In this study, the entire HOXC8 gene of the Dezhou donkey was sequenced, SNPs at the whole gene level were identified, and typing was accomplished utilizing a targeted sequencing genotype detection technique (GBTS). Then, a general linear model was used to perform an association study of HOXC8 gene polymorphism loci, multiple vertebral numbers, and carcass weight for screening candidate markers that can be used for molecular breeding of Dezhou donkeys. These findings revealed that HOXC8 included 12 SNPs, all unique mutant loci. The HOXC8 g.15179224C>T was significantly negatively associated with carcass weight (CW) and lumbar vertebrae length (LL) (p < 0.05). The g.15179674G>A locus was shown to be significantly positively associated with the number of lumbar vertebrae (LN) (p < 0.05). The phylogenetic tree constructed for the Dezhou donkey HOXC8 gene and seven other species revealed that the HOXC8 gene was highly conserved during animal evolution but differed markedly among distantly related animals. The results suggest that HOXC8 is a vital gene affecting multiple vertebral numbers and carcass weight in Dezhou donkeys, and the two loci g.15179224C>T and g.15179674G>A may be potential genetic markers for screening and breeding of new strains of high-quality and high-yielding Dezhou donkeys.


Assuntos
Equidae , Genes Homeobox , Animais , Equidae/genética , Filogenia , Polimorfismo de Nucleotídeo Único/genética , Vértebras Lombares
10.
Elife ; 112022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35315772

RESUMO

Spinal motor neurons (MNs) constitute cellular substrates for several movement disorders. Although their early development has received much attention, how spinal MNs become and remain terminally differentiated is poorly understood. Here, we determined the transcriptome of mouse MNs located at the brachial domain of the spinal cord at embryonic and postnatal stages. We identified novel transcription factors (TFs) and terminal differentiation genes (e.g. ion channels, neurotransmitter receptors, adhesion molecules) with continuous expression in MNs. Interestingly, genes encoding homeodomain TFs (e.g. HOX, LIM), previously implicated in early MN development, continue to be expressed postnatally, suggesting later functions. To test this idea, we inactivated Hoxc8 at successive stages of mouse MN development and observed motor deficits. Our in vivo findings suggest that Hoxc8 is not only required to establish, but also maintain expression of several MN terminal differentiation markers. Data from in vitro generated MNs indicate Hoxc8 acts directly and is sufficient to induce expression of terminal differentiation genes. Our findings dovetail recent observations in Caenorhabditis elegans MNs, pointing toward an evolutionarily conserved role for Hox in neuronal terminal differentiation.


Assuntos
Proteínas de Caenorhabditis elegans , Neurônios Motores , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Neurônios Motores/fisiologia , Medula Espinal/metabolismo , Fatores de Transcrição/metabolismo
11.
Anim Sci J ; 92(1): e13661, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34856652

RESUMO

MicroRNAs (miRNAs) regulate adipogenic differentiation in stromal vascular fractions (SVFs) through post-transcriptional regulation of transcription factors and other functional genes. miR-301 and the homeobox C8 (HOXC8) gene are involved in lipid homeostasis; however, their roles in the adipogenic differentiation of ovine SVFs are unknown. Here, we explored the effects of miR-301 and HOXC8 on adipogenic differentiation in ovine SVFs and the regulatory role of miR-301a in HOXC8 expression. Additionally, we evaluated the effect of miR-301a and HOXC8 on the mRNA abundance of adipogenic markers and the ability of ovine SVFs to accumulate lipids. We found that miR-301a regulates adipogenic differentiation in ovine SVFs by directly targeting the 3'-untranslated region of HOXC8, resulting in significant downregulation of the HOXC8 mRNA and protein. Moreover, miR-301a overexpression suppressed adipogenic differentiation in ovine SVFs and significantly inhibited the expression of adipogenesis-related genes-including adiponectin, C/EBPα, PPARγ, and FABP4. Conversely, HOXC8 overexpression in ovine SVFs increased the accumulation of lipid droplets and remarkably promoted the expression of adipogenic markers. Taken together, our results indicate that miR-301a attenuates the adipogenic differentiation of ovine SVFs by targeting HOXC8. These findings improve our understanding of the mechanism of lipid accumulation and metabolism in sheep.


Assuntos
Adipogenia , Regiões 3' não Traduzidas , Adipogenia/genética , Animais , Diferenciação Celular/genética , Genes Homeobox , MicroRNAs/genética , RNA Mensageiro , Ovinos/genética , Fração Vascular Estromal
12.
J Cancer ; 12(8): 2430-2439, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33758619

RESUMO

Accumulating evidence suggest that circRNA RNAs (circRNAs) play important roles in tumor formation and development. circNT5E has been shown to be an oncogenic gene in several types of cancer, and the high expression of circNT5E lead to tumorigenesis and cancer progression. However, the precise role of circNT5E in bladder cancer (Bca) has not been characterized. In this study, we observed that circNT5E expression was augmented in Bca tissues compared with that in adjacent normal tissues, and its expression level was positively associated with larger tumor size and lower survival rate. Further experiments showed that suppression of circNT5E restrained the growth and metastasis of Bca cells in vitro. circNT5E was mainly distributed in the cytoplasm and it captured miR-502-5p to increase HOXC8 mRNA and protein expression. Moreover, decreased miR-502-5p obviously reversed the circNT5E silencing-mediated inhibition of Bca cell growth and migration. Thus, this study suggested that circNT5E may act as a pro-oncogene in the development and progression of Bca and it may become a useful tumor biomarker and promising therapeutic target for Bca treatment.

13.
Front Oncol ; 11: 793678, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35111675

RESUMO

Gastric cancer is a deadly human malignancy and the molecular mechanisms underlying gastric cancer pathophysiology are very complicated. Thus, further investigations are warranted to decipher the underlying molecular mechanisms. With the development of high-throughput screening and bioinformatics, gene expression profiles with large scale have been performed in gastric cancer. In the present study, we mined The Cancer Genome Atlas (TCGA) database and analyzed the gene expression profiles between gastric cancer tissues and normal gastric tissues. A series of differentially expressed lncRNAs, miRNAs and mRNAs between gastric cancer tissues and normal gastric tissues were identified. Based on the differentially expressed genes, we constructed miRNA-mRNA network, lncRNA-mRNA network and transcriptional factors-mRNA-miRNA-lncRNA network. Furthermore, the Kaplan survival analysis showed that high expression levels of EVX1, GBX2, GCM1, HOXC8, HOXC9, HOXC10, HOXC11, HOXC12 and HOXC13 were all significantly correlated with shorter overall survival of the patients with gastric cancer. On the other hand, low expression level of HOXA13 was associated with shorter overall survival of patients with gastric cancer. Among these hub genes, we performed the in vitro functional studies of HOXC8 in the gastric cancer cells. Knockdown of HOXC8 and overexpression of miR-4256 both significantly repressed the gastric cancer cell proliferation and migration, and miR-4256 repressed the expression of HOXC8 via targeting its 3' untranslated region in gastric cancer cells. Collectively, our results revealed that a complex interaction networks of differentially expressed genes in gastric cancer, and further functional studies indicated that miR-4256/HOXC8 may be an important axis in regulating gastric cancer progression.

14.
Cancer Manag Res ; 12: 9159-9171, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33061608

RESUMO

BACKGROUND: Circular RNA (circRNA) has an essential regulatory role in the chemotherapy resistance of cancers. Nevertheless, the role of circRNA nuclear receptor-interacting protein 1 (circNRIP1) in the paclitaxel (PTX) resistance of ovarian cancer (OC) remains unclear. MATERIAL AND METHODS: The circNRIP1, miR-211-5p and homeobox C8 (HOXC8) expression levels were assessed using qRT-PCR. The PTX resistance of cells was measured by 3-(4, 5-dimethylthiazolyl-2-yl)-2-5 diphenyl tetrazolium bromide (MTT) assay. Furthermore, cell proliferation, apoptosis, migration and invasion were detected by colony formation assay, flow cytometry and transwell assay, respectively. Moreover, the protein levels of proliferation, apoptosis, metastasis-related markers and HOXC8 were determined by Western blot (WB) analysis. Tumor xenograft models were constructed to explore the influence of circNRIP1 on OC tumor growth. The interaction between miR-211-5p and circNRIP1 or HOXC8 was confirmed by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. RESULTS: CircNRIP1 was highly expressed in PTX-resistant OC tissues and cells. Silencing of circNRIP1 repressed the PTX resistance of OC cells in vitro and OC tumor in vivo. Furthermore, circNRIP1 sponged miR-211-5p, and miR-211-5p inhibitor could reverse the inhibitory effect of circNRIP1 knockdown on the PTX resistance of OC cells. In addition, miR-211-5p targeted HOXC8, and HOXC8 overexpression could reverse the suppression effect of miR-211-5p on the PTX resistance of OC cells. Additionally, the expression of HOXC8 was regulated by circNRIP1 and miR-211-5p. CONCLUSION: CircNRIP1 silencing could inhibit the PTX resistance of OC via regulating the miR-211-5p/HOXC8 axis, showing that circNRIP1 might be a potential target for OC resistance treatment.

15.
J Assist Reprod Genet ; 37(10): 2535-2544, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32772270

RESUMO

PURPOSE: To evaluate whether miR-148a-3p overexpression is associated with disrupted decidualization of recurrent implantation failure (RIF). METHODS: Endometrial miRNA and mRNA expression profiles during the implantation window derived from women with and without RIF were identified using microarray and RT-qPCR. Immortalized human endometrial stromal cells (HESCs) were cultured for proliferation and in vitro decidualization assays after enhancing miR-148a-3p expression or inhibiting putative target gene homeobox C8 (HOXC8) expression. RT-qPCR, western blot, and luciferase reporter assays were used to confirm the relationship between miR-148a-3p and HOXC8 gene. RESULTS: MiR-148a-3p was significantly upregulated in RIF endometrial tissues. Forced expression of miR-148a-3p notably attenuated HESC in vitro decidualization. Mechanistic studies revealed that miR-148a-3p directly bounds to the HOXC8 3' untranslated region (3'UTR) and suppressed HOXC8 expressions in both mRNA and protein levels. Further investigations demonstrated that inhibition of HOXC8 in HESCs induced similar effects on decidual process as those induced by miR-148a-3p overexpression. CONCLUSION: Taken together, our findings suggested that elevated miR-148a-3p might account for flawed decidualization in RIF by negatively regulating HOXC8, raising the possibility that miR-148a-3p might be a novel therapeutic target in RIF.


Assuntos
Implantação do Embrião/genética , Endométrio/metabolismo , Proteínas de Homeodomínio/genética , MicroRNAs/genética , Proliferação de Células/genética , Células Cultivadas , Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Células Estromais/metabolismo , Células Estromais/patologia
16.
Stem Cell Res Ther ; 11(1): 271, 2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32631410

RESUMO

BACKGROUND: Mesenchymal stem cell (MSC)-based cartilage tissue regeneration is a treatment with great potential. How to enhance the MSC chondrogenic differentiation is a key issue involved in cartilage formation. In the present study, we seek to expound the phenotypes and mechanisms of DLX5 in chondrogenic differentiation function in MSCs. METHODS: Stem cells from apical papilla (SCAPs) were used. The Alcian Blue staining, pellet culture system, and cell transplantation in rabbit knee cartilage defect were used to evaluate the chondrogenic differentiation function of MSCs. Western blot, real-time RT-PCR, and ChIP assays were used to evaluate the molecular mechanisms. RESULTS: DLX5 and HOXC8 expressions were upregulated during chondrogenic differentiation. In vitro results showed that DLX5 and HOXC8 enhanced the expression of chondrogenic markers including collagen II (COL2), collagen V (COL5), and sex-determining region Y box protein 9 (SOX9) and promoted the chondrogenic differentiation and the formation of cartilage clumps in the pellet culture system. Mechanically, DLX5 and HOXC8 formed protein complexes and negatively regulated the LncRNA, LINC01013, via directly binding its promoter. In vivo transplantation experiment showed that DLX5 and HOXC8 could restore the cartilage defect in the rabbit knee model. In addition, knock-down of LINC01013 enhanced the chondrogenic differentiation of SCAPs. CONCLUSIONS: In conclusion, DLX5 and HOXC8 enhance the chondrogenic differentiation abilities of SCAPs by negatively regulating LINC01013 in SCAPs, and provided the potential target for promoting cartilage tissue regeneration.


Assuntos
Condrogênese , Células-Tronco Mesenquimais , Animais , Cartilagem , Diferenciação Celular , Células Cultivadas , Condrogênese/genética , Coelhos , Células-Tronco
17.
J Cell Physiol ; 235(11): 8432-8445, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32246725

RESUMO

Enhancing the functions of mesenchymal stem cells (MSCs) is considered a potential approach for promoting tissue regeneration. In the present study, we investigate the role of HOXC8 in regulating differentiation and migration by using stem cells of the apical papilla (SCAPs). Our results showed that overexpression of HOXC8 suppressed the osteo-/dentinogenic differentiation, as detected by measuring alkaline phosphatase activity, in vitro mineralization, and the expressions of dentin sialophosphoprotein, dentin matrix acidic phosphoprotein 1, bone sialoprotein, runt-related transcription factor 2, and osterix in SCAPs, and inhibited in vivo osteo-/dentinogenesis of SCAPs. In addition, knockdown of HOXC8 promoted the osteo-/dentinogenic differentiation potentials of SCAPs. Mechanically, HOXC8 enhanced KDM1A transcription by directly binding to its promoter. HOXC8 and KDM1A also inhibited the migration and chemotaxis abilities of SCAPs. To sum up, HOXC8 negatively regulated the osteo-/dentinogenic differentiation and migration abilities of SCAPs by directly enhancing KDM1A transcription and indicated that HOXC8 and KDM1A could serve as potential targets for enhancing dental MSC mediated tissue regeneration.


Assuntos
Diferenciação Celular/fisiologia , Histona Desmetilases/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco/metabolismo , Diferenciação Celular/genética , Proliferação de Células/fisiologia , Células Cultivadas , Papila Dentária/metabolismo , Genes Homeobox/fisiologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/fisiologia
18.
Onco Targets Ther ; 13: 757-767, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32158227

RESUMO

OBJECTIVE: Long noncoding RNA small nucleolar RNA host gene 1 (SNHG1) has been reported to be aberrantly expressed and plays an important role in human cancers, including esophageal squamous cell cancer. However, the regulatory mechanism underlying SNHG1 in the progression of esophageal squamous cell cancer is poorly defined. MATERIALS AND METHODS: Fifty-three esophageal squamous cell cancer patients were recruited and overall survival was analyzed. EC9706 and KYSE150 cells were cultured for study in vitro. The expression levels of SNHG1, microRNA (miR)-204 and homeobox c8 (HOXC8) were detected by quantitative real-time polymerase chain reaction and Western blot. Cell cycle distribution, apoptosis, migration and invasion were determined by flow cytometry and transwell assays, respectively. The target interaction among SNHG1, miR-204 and HOXC8 was validated by luciferase reporter assay and RNA immunoprecipitation. Xenograft model was established to investigate the role of SNHG1 in vivo. RESULTS: High expression of SNHG1 was exhibited in esophageal squamous cell cancer and indicated poor outcomes of patients. SNHG1 silence led to cell cycle arrest at G0-G1 phase, inhibition of migration and invasion and increase of apoptosis. miR-204 was validated to sponge by SNHG1 and target HOXC8 in esophageal squamous cell cancer cells. miR-204 knockdown or HOXC8 restoration reversed the inhibitive role of SNHG1 silence in the progression of esophageal squamous cell cancer cells. Furthermore, inhibiting SNHG1 decreased xenograft tumor growth by regulating miR-204 and HOXC8. CONCLUSION: SNHG1 knockdown suppresses migration and invasion but induces apoptosis of esophageal squamous cell cancer cells by increasing miR-204 and decreasing HOXC8.

19.
Onco Targets Ther ; 13: 381-387, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32021283

RESUMO

BACKGROUND: Mixed lineage leukemia (MLL) fusion protein alone exhibits poor histone lysine methyltransferase (HKMT) activity in catalyzing histone H3 Lys4 trimethylation (H3K4me3) in MLL-rearranged acute leukemia. METHODS: To explore the HKMT effect of another regulatory protein within the complex of proteins associated with Set 1 (COMPASS), we analyzed the H3K4me3 modification of the HOXC8 promoter under the action of ASH2L regulation. Small interfering RNA of ASH2L, chromatin immunoprecipitation, real-time-PCR (RT-PCR), and Western blotting were used to detect the expression of specific regions of the HOXC8 promoter, RBBP5, WDR5, MLL, and BRTF in two MLL-rearranged acute leukemia cell lines (RS4:11 and THP-1 cells). RESULTS: The gene and protein expression levels of HOXC8 were significantly downregulated upon treatment with ASH2L-siRNA (as analyzed by targeting specific regions of the HOXC8 promoter located 0 and 3 kb (-3.0 kb) upstream of the transcriptional start site in RSH:11 cells; and -3.0 and -2.0 kb upstream of the transcriptional start site, and +1.4 kb downstream of the transcriptional start site in THP-1 cells). The expression levels of the BRTF, RBBP5, WDR5, and MLL genes were significantly downregulated from the different transcriptional start sites of the HOXC8 promoter in the RSH:11 cell line (P < 0.05). Furthermore, the BPTF and RBBP5 genes were downregulated from the HOXC8 promoter in the THP-1 cell line (P < 0.05). CONCLUSION: Based on these results, we suggest a new concept of histone modification of the ASH2L protein in MLL-rearranged acute leukemia, which cannot carry out methyltransferase activity independently. The protein-protein interactions of ASH2L with other COMPASS members, such as MLL, WDR5, RBBP5, and chromatin remodeling factor BRTF, appear to be essential for its role in the activation of HOXC8 gene transcription.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA