Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Int J Mol Sci ; 25(10)2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38791575

RESUMEN

Chromosomal instability is a hallmark of colorectal carcinogenesis and produces an accumulation of different forms of aneuploidies or broad copy number aberrations. Colorectal cancer is characterized by gain-type broad copy number aberrations, specifically in Chr20, Chr8q, Chr13 and Chr7, but their roles and mechanisms in cancer progression are not fully understood. It has been suggested that broad copy number gains might contribute to tumor development through the so-called caricature transcriptomic effect. We intend to investigate the impact of broad copy number gains on long non-coding RNAs' expression in colorectal cancer, given their well-known role in oncogenesis. The influence of such chromosomal aberrations on lncRNAs' transcriptome profile was investigated by SNP and transcriptome arrays in our series of colorectal cancer samples and cell lines. The correlation between aneuploidies and transcriptomic profiles led us to obtain a class of Over-UpT lncRNAs, which are transcripts upregulated in CRC and further overexpressed in colon tumors bearing specific chromosomal aberrations. The identified lncRNAs can contribute to a wide interaction network to establish the cancer driving effect of gain-type aneuploidies.


Asunto(s)
Aneuploidia , Neoplasias Colorrectales , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Variaciones en el Número de Copia de ADN , Transcriptoma , Femenino , Línea Celular Tumoral , Perfilación de la Expresión Génica , Masculino , Inestabilidad Cromosómica , Persona de Mediana Edad , Aberraciones Cromosómicas , Polimorfismo de Nucleótido Simple
2.
Aging (Albany NY) ; 16(1): 493-517, 2024 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-38194709

RESUMEN

BACKGROUND: Sarcomas are a type of highly heterogeneous malignant tumors originating from mesenchymal tissues. Necroptosis is intricately connected to the oncogenesis and progression of tumors. The main goal of this research is to assess the prognostic value of necroptosis-related lncRNAs (NRlncRNAs) in sarcomas and to develop a risk model based on NRlncRNAs to evaluate prognostic and immune status of the sarcomas. METHODS: We screened NRlncRNAs using the gene co-expression network, developed a prognostic risk model of sarcomas, and then verified the model. Following that, various bioinformatics analysis algorithms were employed to analyze the distinct characteristics of patients of the risk model. Furthermore, the function and regulatory mechanism of NRlncRNA SNHG6 in sarcomas were investigated through osteosarcoma cell experiments, such as qRT-PCR, Western blot, CCK-8, clone formation, and transwell assay. RESULTS: We successfully developed a NRlncRNAs-related prognostic risk model and screened 5 prognosis-related NRlncRNAs, with SNGH6 being the most significant for prognosis of patients. According to results, the significant differences exist in prognosis, clinical characteristics, and tumor immune status among patients of the risk model. The experiments of osteosarcoma cells demonstrated that NRlncRNA SNHG6 knockdown significantly attenuated the cells' proliferation, migration, and invasion. qRT-PCR and WB results showed that SNHG6 regulated AXL and AKT signaling. CONCLUSIONS: We have developed an innovative investigation on NRlncRNAs, which can serve as a reference for diagnosis, therapy, and prognosis of sarcomas. Additionally, we demonstrated that NRlncRNA SNHG6 regulated AXL and AKT signaling in osteosarcoma cells and the proliferation, migration, and invasion of tumor cells.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Línea Celular Tumoral , Proteínas Proto-Oncogénicas c-akt/genética , Necroptosis/genética , Pronóstico , Osteosarcoma/genética , Neoplasias Óseas/genética , Regulación Neoplásica de la Expresión Génica
3.
Environ Toxicol ; 39(5): 2634-2641, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38205902

RESUMEN

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is a widespread inflammatory disease with a high mortality rate. Long noncoding RNAs play important roles in pulmonary diseases and are potential targets for inflammation intervention. METHODS: The expression of small nucleolar RNA host gene 6 (SNHG6) in mouse lung epithelial cell line MLE12 with or without cigarette smoke extract (CSE) treatment was first detected using quantitative reverse-transcription PCR. ELISA was used to evaluate the release of inflammatory cytokines (TNF-α, IL-1ß, and IL-6). The binding site of miR-182-5p with SNHG6 was predicted by using miRanda, which was verified by double luciferase reporter assay. RESULTS: Here, we revealed that SNHG6 was upregulated in CS-exposed MLE12 alveolar epithelial cells and lungs from COPD-model mice. SNHG6 silencing weakened CS-induced inflammation in MLE12 cells and mouse lungs. Mechanistic investigations revealed that SNHG6 could upregulate IκBα kinase through sponging the microRNA miR-182-5p, followed by activated NF-κB signaling. The suppressive effects of SNHG6 silencing on CS-induced inflammation were blocked by an miR-182-5p inhibitor. CONCLUSION: Overall, our findings suggested that SNHG6 regulates CS-induced inflammation in COPD by activating NF-κB signaling, thereby offering a novel potential target for COPD treatment.


Asunto(s)
Fumar Cigarrillos , MicroARNs , Neumonía , Enfermedad Pulmonar Obstructiva Crónica , ARN Largo no Codificante , Ratones , Animales , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Fumar Cigarrillos/efectos adversos , Neumonía/inducido químicamente , Neumonía/genética , MicroARNs/genética , MicroARNs/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Inflamación/genética , Inflamación/metabolismo
4.
Biochim Biophys Acta Rev Cancer ; 1879(1): 189056, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38104909

RESUMEN

Colorectal cancer (CRC) ranks among the leading cancers in terms of incidence and mortality in the Western world. Currently, there are no sufficient diagnostic markers that would enable an early diagnosis and efficient therapy. Unfortunately, a significant number of new CRC cases is detected in late stages, with distant metastases, therefore, new therapeutic approaches, which would alleviate the prognosis for advanced stages of CRC, are highly in demand. SNHG6 belongs to the group of long non-coding RNAs, which are a larger entity of RNAs consisting of >200 nucleotides. SNHG6 is expressed mainly in the cell cytoplasm, where it acts as a regulator of numerous processes: modulation of crucial protein hubs; sponging miRNAs and upregulating the expression of their target mRNAs; and interacting with various cellular pathways including TGF-ß/Smad and Wnt/ß-catenin. SNHG6 is an oncogene, substantially overexpressed in CRC tissues and cancerous cell lines as compared to healthy samples. Its overexpression is associated with higher grade, lymphovascular invasion and tumor size. Taking into consideration the role of SNHG6 in the colorectal tumorigenesis, invasion and metastasis, we summarized its role in CRC and conclude that it could serve as a potential biomarker in CRC diagnosis and prognosis assessment.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , ARN Largo no Codificante , Humanos , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
5.
Pathol Res Pract ; 252: 154914, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37992506

RESUMEN

Long non-coding RNAs (lncRNAs) can be utilized as prognostic indicators of gastric cancer since they can affect several cancer-related processes. Coumarin is a natural product with some useful anti-cancer properties. Here, we measured the expression of selected lncRNAs (RuPAR, SNHG6, CASC11, and their targets, miR-340-5p, p21, E-cadherin, and CDK1) in AGS gastric cancer cells treated with coumarin. MTT test has been utilized for assessing the AGS cells' cell viability after exposure to coumarin. The expression of the lncRNAs (RuPAR, SNHG6, and CASC11) and miR-340-5p was evaluated via qRT-PCR. Western blot analysis has been utilized to determine changes in p21, E-cadherin, and CDK1 expression. Coumarin decreased AGS viability in a dose-dependent manner. The coumarin treated cells had lower levels of the mRNAs known to be targets of lncRNAs SNHG6 and CASC11 compared to control. Additionally, the coumarin group had increased levels of lncRNA RuPAR expression when compared with the control group. Some lncRNA targets, including p21, E-cadherin, and CDK1, showed lower expression in the coumarin group compared to the control by Western blotting. Coumarin could be a promising pharmacological candidate to be included in gastric cancer treatment regimens because it modulates lncRNAs and their targets.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , ARN Largo no Codificante/metabolismo , Proliferación Celular/genética , MicroARNs/genética , Cadherinas/genética , Cumarinas/farmacología
6.
Eur J Med Res ; 28(1): 363, 2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37735423

RESUMEN

RNA sequencing has revealed that a substantial portion of the human genome undergoes transcription, yet a minimal fraction of these transcripts translates into proteins. LncRNAs, RNA molecules less than 200 nt in length, once deemed as transcriptional noise, have now emerged as crucial regulators of numerous cellular processes. This review focuses on the lncRNA SNHG6, aiming to elucidate its biogenesis, the pivotal roles it plays, and its mechanisms in facilitating the hallmarks of cancer. A comprehensive literature review and analysis were undertaken to delve into the biogenesis of SNHG6, its roles in cellular processes, and the mechanisms through which it contributes to the hallmarks of cancer. SNHG6 is a notable lncRNA, observed to be overexpressed in various cancer types; its perturbation has been linked to tumor progression, emphasizing its significance in oncogenesis. This lncRNA contributes to a range of cellular aberrations, influencing transcriptional, post-transcriptional, and epigenetic processes of mRNA, ultimately driving cancerous transformations. LncRNA SNHG6 serves as a potential biomarker and therapeutic target due to its association with tumorigenesis. Understanding its mechanism and role in cancer can pave the way for novel diagnostic and therapeutic strategies.


Asunto(s)
Neoplasias , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Neoplasias/genética , Carcinogénesis/genética , ARN Mensajero
7.
Pharmaceuticals (Basel) ; 16(2)2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-37259332

RESUMEN

Gemcitabine (GEM) is the gold-standard therapeutic regimen for patients with pancreatic cancer (PC); however, patients may receive limited benefits due to the drug resistance of GEM. LncRNA SNHG6 is reported to play key roles in drug resistance, but its role and molecular mechanism in PC remain incompletely understood. We found that LncRNA SNHG6 is drastically downregulated in GEM-resistant PC and is positively correlated with the survival of PC patients. With the help of bioinformatic analysis and molecular approaches, we show that LncRNA SNHG6 can sponge miR-944, therefore causing the upregulation of the target gene KPNA5. In vitro experiments showed that LncRNA SNHG6 and KPNA5 suppress PC cell proliferation and colony formation. The Upregulation of LncRNA SNHG6 and KPNA5 increases the response of GEM-resistant PANC-1 cells to GEM. We also show that the expression of KPNA5 is higher in patients without GEM resistance than in those who developed GEM resistance. In summary, our findings indicate that the LncRNA SNHG6/miR944/KPNA5 axis plays a pivotal role in overcoming GEM resistance, and targeting this axis may contribute to an increasing of the benefits of PC patients from GEM treatment.

8.
Heliyon ; 9(5): e16291, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37234669

RESUMEN

The long non-coding RNA small nucleolar RNA host gene 6 (SNHG6) acts as an oncogene in several cancers, and is highly expressed in ovarian cancer. MiR-543, a tumor suppressor, was expressed lowly in ovarian cancer. However, whether SNHG6 performed its oncogenic role via miR-543 in ovarian cancer, as well as the underlying mechanism is still not clear. In this study, we showed that the levels of SNHG6 and Yes-associated protein 1 (YAP1) were significantly elevated, while the level of miR-543 was significantly decreased, in ovarian cancer tissues compared with adjacent normal samples. We demonstrated that overexpression of SNHG6 significantly promoted the proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) of ovarian cancer cells SKOV3 and A2780. Knockdown of SNHG6 showed the opposite effects. MiR-543 level was negatively correlated with the SNHG6 level in ovarian cancer tissues. SHNG6 overexpression significantly inhibited the expression of miR-543, and SHNG6 knockdown significantly elevated the expression of miR-543 in ovarian cancer cells. The effects of SNHG6 on ovarian cancer cells were abrogated by miR-543 mimic, and strengthened by anti-miR-543. YAP1 was identified as a target of miR-543. Forced expression of miR-543 significantly inhibited the expression of YAP1. Moreover, YAP1 overexpression could reverse the effects of SNHG6 downregulation on the malignant phenotypes of ovarian cancer cells. In summary, our study showed that SNHG6 promoted the malignant phenotypes of ovarian cancer cells via miR-543/YAP1 pathway.

9.
J Obstet Gynaecol Res ; 49(1): 232-242, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36396030

RESUMEN

BACKGROUND: Endometrial dysfunction is closely correlated with the development of multiple severe gynecological disorders including intrauterine adhesion. Accumulating evidence supports that some long non-coding RNAs (lncRNAs) have peptide-coding potential. In this text, the peptide-coding ability of lncRNA SNHG6 was examined. Also, the effects of an SNHG6-encoded peptide on the viability and migration of human endometrial stromal cells (hESCs) and human endometrial epithelial cells (hEECs) and related molecular mechanisms were explored. METHODS: The peptide-encoding potential of SNHG6 was predicted by FuncPEP and getorf databases and validated by western blot assay. Cell viability was tested by cell counting kit-8 assay. Cell migratory ability was examined by wound healing and transwell migration assays. Protein levels of genes were measured by western blot assay. RESULTS: Prediction analysis suggested that SNHG6 had the potential peptide-coding ability and multiple open-reading frames (ORFs). Western blot validated that SNHG6 ORF#1 and ORF#2 could translate into short peptides. SNHG6 ORF#2 overexpression facilitated cell migration and epithelial-mesenchymal transition (EMT) in hESCs and hEECs, while these effects were abrogated by transforming growth factor-beta (TGF-ß)/SMAD signaling inhibitor GW788388. Moreover, GW788388 inhibited the increase of p-SMAD2 and p-SMAD3 levels induced by SNHG6 ORF#2 in hESCs. SNHG6 ORF#2-encoded peptide did not influence endometrial stromal and epithelial cell viability. CONCLUSIONS: LncRNA SNHG6 ORF#1 and ORF#2 could translate into small peptides and SNHG6 ORF#2 overexpression promoted cell migration and EMT by activating the TGF-ß/SMAD pathway in hESCs and hEECs, suggesting the potential roles of SNHG6-encoded peptides in the development of endometrial stromal and epithelial cells and related gynecological diseases.


Asunto(s)
ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transición Epitelial-Mesenquimal/genética , ARN Nucleolar Pequeño/farmacología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento Transformador beta/farmacología , Transducción de Señal , Movimiento Celular/genética , Factores de Crecimiento Transformadores/genética , Factores de Crecimiento Transformadores/metabolismo , Factores de Crecimiento Transformadores/farmacología
10.
Front Oncol ; 12: 1015428, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36212408

RESUMEN

Acquired resistance is a major clinical challenge for tamoxifen-based therapy. In this study, we focused on lncRNA SNHG6 which plays a role in chemoresistance of cancer cells, but has never been investigated in the context of tamoxifen resistance. We found elevated levels of SNHG6 in tamoxifen-resistant estrogen receptor (ER)-positive MCF-7 cells (MCF7TR), relative to naïve MCF-7 cells, as well as in tamoxifen-resistant T47D cells (T47DTR), relative to naïve T47D cells, which correlated with induced vimentin, ZEB1/2 and decreased e-cadherin, thus implicating a role of EMT in SNHG6-mediated tamoxifen resistance. Downregulation of SNHG6, using specific siRNA, sensitized MCF7TR as well as T47DTR cells to tamoxifen along with markedly reduced proliferation, invasion and anchorage-independent clonogenicity. Further, SNHG6 was found to sponge and inhibit miR-101 as the endogenous expression levels of SNHG6 and miR-101 inversely correlated in paired parental and tamoxifen-resistant cells and, moreover, silencing of SNHG6 in tamoxifen-resistant cells resulted in de-repression of miR-101, along with reversal of EMT. SNHG6 expression also directly correlated with increased stem cells markers Sox2, Oct4 and EZH2. miR-101 levels, manipulated by transfections with pre/anti-miR-101 oligos, directly affected tamoxifen sensitivity of ER-positive cells with pre-miR-101 sensitizing MCF7TR and T47DTR cells to tamoxifen whereas anti-miR-101 inducing resistance of parental MCF-7 and T47D cells to tamoxifen. Further, miR-101 was found to attenuate SNHG6-mediated effects on tamoxifen resistance, EMT as well as stem cell markers, thereby making a case for SNHG6-miR-101 axis in tamoxifen resistance of ER-positive breast cancer cells. Thus, lncRNA SNHG6 is a novel modulator of tamoxifen resistance through its sponging of miR-101 and the resulting effects on EMT.

11.
J Mol Histol ; 53(6): 871-882, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36173586

RESUMEN

Recurrent spontaneous abortion (RSA) is a gestational disease with complex pathogenesis, and trophoblast cells are closely involved in the pathogenesis of RSA. This study aimed to explore the regulatory effects and mechanisms of SNHG6 on trophoblast cells. The expression of SNHG6, miR-101-3p, and OTUD3 were detected in villous tissues from patients with unexplained RSA and normal pregnant women with induced abortion by qRT-PCR. The target relationships between miR-101-3p and SNHG6/OTUD3 were confirmed by dual-luciferase reporter assay. The viability, migration, and apoptosis of trophoblast cells were measured by MTT, wound healing, and flow cytometry assays, respectively. Western blot was performed to detect the protein expression of OTUD3, Ki-67, Bax, and Bcl-2. The results showed that SNHG6 and OTUD3 were up-regulated, and miR-101-3p was down-regulated in RSA patients. MiR-101-3p was a target of SNHG6, and OTUD3 was a target of miR-101-3p. There were negative correlations between the expression of miR-101-3p and OTUD3/SNHG6 in RSA patients. In addition, both SNHG6 silencing and miR-101-3p overexpression could increase cell viability and migration, decrease cell apoptosis, up-regulate Ki-67 and Bcl-2, and down-regulate Bax in HTR-8/SVneo cells. The effects of SNHG6 silencing on HTR-8/SVneo cells were reversed by miR-101-3p silencing or OTUD3 overexpression. To sum up, silencing of SNHG6 enhanced the viability and migration, and inhibited the apoptosis of trophoblast cells through regulating miR-101-3p/OTUD3. SNHG6/miR-101-3p/OTUD3 may be potential targets for the prevention of unexplained RSA.


Asunto(s)
Aborto Habitual , MicroARNs , ARN Largo no Codificante , Humanos , Femenino , Embarazo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Trofoblastos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Antígeno Ki-67/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proliferación Celular/genética , Aborto Habitual/metabolismo , Movimiento Celular/genética , Proteasas Ubiquitina-Específicas/metabolismo
12.
Cytokine ; 157: 155914, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35809451

RESUMEN

BACKGROUND: Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in premature infants, and its pathogenesis has not been clarified. Long non-coding RNAs (lncRNA) have important functions in cell bioactivity. However, their role in developmental lung disease remains unclear. OBJECTIVE: The aim of this study was to demonstrate the role of lncRNA SNHG6 (SNHG6) in BPD and its underlying mechanisms. METHODS: The blood of patients with BPD were collected, and BPD model of BEAS-2B cells was established by hyperoxia method. SNHG6, miR-335 and KLF5 mRNA expression were detected by RT-qPCR. Western blot was conducted to measure the levels of apoptosis-related proteins' expression and NF-κB pathway related proteins. BEAS-2B cell viability and apoptosis were assessed by CCK-8 and flow cytometry, respectively. Assay Kit was applied to detect ROS, MDA and SOD levels, respectively. ELISA was performed to assess the levels of inflammatory factors. The binding site of miR-335 with SNHG6 or KLF5 were predicted by using DIANA or TargetScan, and which was verified by double luciferase reporter assay. RESULTS: Firstly, SNHG6 was highly expressed and miR-335 was lowly expressed in BPD model, SNHG6 knockdown and miR-335 mimics both alleviated hyperoxia-induced lung cell injury, and SNHG6 targeted miR-335. Subsequently, KLF5 was targeted by miR-335, and KLF5 promoted lung cell injury via activating NF-κB pathway. Furthermore, SNHG6 mediated lung cell injury via regulating the miR-335/KLF5/NF-κB pathway. CONCLUSION: Our research confirmed that SNHG6 mediated hyperoxia-induced lung cell injury via regulating the miR-335/KLF5/NF-κB pathway. These findings suggest that SNHG6 serves as promising targets for the treatment of newborns with BPD.


Asunto(s)
Hiperoxia , Enfermedades Pulmonares , MicroARNs , ARN Largo no Codificante/genética , Humanos , Hiperoxia/genética , Recién Nacido , Factores de Transcripción de Tipo Kruppel/genética , Pulmón/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , FN-kappa B , ARN Largo no Codificante/metabolismo
13.
Environ Toxicol ; 37(10): 2375-2387, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35785413

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is an interstitial pulmonary disease with slow onset and high mortality. Epithelial-mesenchymal transition (EMT) is a significant condition for tissue fibrosis, and lncRNA-Snhg6 (small nucleolar RNA host gene 6) is related to EMT in some cancer cells, but its role in pulmonary fibrosis remains obscure. Here, we found that TGF-ß1 and Snhg6 were up-regulated in lung tissues of BLM-induced lung fibrosis mouse, and Snhg6 expression was significantly increased in primary lung fibroblasts after BLM treatment. Snhg6 knockdown notably alleviated the pulmonary dysfunction, and the increase of fibrosis area and collagen deposition induced by BLM. MiR-26a-5p was downregulated in BLM-induced fibrotic lung tissues, and it was negatively regulated by Snhg6. Silencing Snhg6 markedly alleviated the TGF-ß1-induced increase in fibrotic marker expression, cell proliferation, migration and differentiation, as well as the nuclear transport of p-Smad2/3 by modulating miR-26a-5p expression in mouse lung fibroblasts. Moreover, overexpressing Snhg6-induced collagen accumulation and fibroblast activation in fibroblasts, which was reversed by treatment with miR-26a-5p mimic or oxymatrine (an inhibitor of TGF-ß1-Smads pathway). Interestingly, silencing Snhg6 in vivo mitigated BLM-driven pulmonary fibrosis by regulating the miR-26a-5p/TGF-ß1-Smads axis. Our data revealed that Snhg6 contributed to the process of BLM-driven lung fibrosis in mouse by modulating the miR-26a-5p/TGF-ß1-Smads axis, suggesting that Snhg6 might be a therapeutic target for lung fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , MicroARNs , ARN Largo no Codificante , Animales , Bleomicina/toxicidad , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
14.
Am J Transl Res ; 14(5): 2861-2873, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35702105

RESUMEN

OBJECTIVE: To investigate the role of lncRNA SNHG6 (SNHG6) in gastric carcinoma (GC) and its relationship with the PI3K/AKT/mTOR signaling pathway in order to provide more comprehensive and reliable reference for the diagnosis and treatment of GC. METHODS: GC patients admitted to our hospital from May 2017 to August 2018 as well as healthy individuals who underwent physical examinations during the same time period were enrolled in this study. The serum SNHG6 level was quantified. Patients were followed up for 3 years to analyze the significance of SNHG6 in the diagnosis and treatment of GC. Finally, in vitro assays were performed to determine the influences of SNHG6 and PI3K/AKT/mTOR signaling pathway on biological behaviors and autophagy ability of GC cells. RESULTS: SNHG6 showed high expression in patients with GC and its expression decreased after therapy. SNHG6 also demonstrated a favorable predictive value for the development of GC and the death of patients. The survival curve suggested that increased SNHG6 indicated a higher risk of death. Additionally, mRNA of PI3K/AKT/mTOR pathway related molecules was highly expressed in GC patients. In in vitro assays, GC cells showed stronger viability and invasion activity and weaker apoptosis and autophagy ability after targeted up-regulation of SNHG6. According to the rescue assay, the effect of up-regulating SNHG6 on GC cells could be completely reversed by suppressing the PI3K/AKT/mTOR pathway. CONCLUSION: With high expression in patients with GC, SNHG6 can promote the development of GC by activating the PI3K/AKT/mTOR signaling pathway and suppressing the autophagy of cells. Therefore, it is a potential breakthrough in the diagnosis and treatment of GC in the future.

15.
Mol Biol Rep ; 49(7): 5911-5917, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35426550

RESUMEN

BACKGROUND: Parkinson's disease (PD) is a neurological condition that is associated with abnormal expression of several transcripts. Vitamin D receptor (VDR) is a possible participant in the pathogenesis of PD. METHODS AND RESULTS: In the present research project, we evaluated expressions of VDR and three functionally associated long non-coding RNAs with this signaling, namely SNHG6, SNHG16 and LINC00346 in PD patients versus normal controls. Level of SNHG6 transcripts was lower in total patients in comparison with total controls (Expression ratio (95% CI) 0.44 (0.17-1.08)) and in male patients compared with male controls (Expression ratio (95% CI) 0.29 (0.13-0.65)). On the other hand, expression of VDR was higher in total patients compared with total controls (Expression ratio (95% CI) 10.86 (4.37-26.72)) and in male patients compared with male controls (Expression ratio (95% CI) 22.16 (6.23-78.8)). There was no significant difference in expression of SNHG16 and LINC00346 between PD patients and controls. Amounts of SNHG6 and VDR transcripts could differentiate total PD patients from total controls with AUC values of 0.66 and 0.86, respectively. CONCLUSIONS: Cumulatively, the results of the present investigation imply dysregulation of VDR signaling in PD and necessitate conduction of further functional studies.


Asunto(s)
Enfermedad de Parkinson , ARN Largo no Codificante , Receptores de Calcitriol/metabolismo , Humanos , Masculino , Enfermedad de Parkinson/genética , ARN Largo no Codificante/genética , Receptores de Calcitriol/genética , Vitamina D
16.
BMC Psychiatry ; 22(1): 178, 2022 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-35279108

RESUMEN

BACKGROUND: Bipolar disorder (BD) is a multifactorial condition. Several signaling pathways affect development of this disorder. With the purpose of exploring the role of vitamin D receptor (VDR) signaling in this disorder, we measured expression of selected mRNA coding genes and long non-coding RNAs (lncRNAs) in this pathway in patients versus normal subjects. METHODS: We measured expression of VDR-associated lncRNAs and mRNAs (SNHG6, MALAT1, Linc00511, Linc00346, VDR and CYP27B1) in the peripheral blood of BD patients vs. healthy individuals. RESULTS: Expression of SNHG6 was significantly higher in cases vs. controls (Posterior beta = 1.29, P value < 0.0001. Subgroup analysis by sex revealed significant results in both subgroups (P value < 0.0001 and P value = 0.023 for males and females, respectively). Expression of CYP27B1 was up-regulated in cases vs. controls (Posterior beta = 0.415, P < 0.0001). Such pattern was also detected among males (P < 0.0001), but not females (P = 0.419). Similarly, MALAT1 and Linc00346 were up-regulated in total cases vs. controls (Posterior beta = 0.694, P < 0.0001 and Posterior beta = 0.4, P = 0.012, respectively) and in male cases compared with male controls (Posterior beta = 0.712, P < 0.0001 and Posterior beta = 0.41, P value = 0.038, respectively). Expression of VDR was up-regulated in total cases compared with controls (Posterior beta = 0.683, P value = 0.001). Finally, expression of Linc00511 was not different between groups. MALAT1, SNHG6, CYP27B1, VDR and Linc00346 had AUC values of 0.95, 0.94, 0.91, 0.85 and 0.83 in differentiation of male patients from controls, respectively. CONCLUSION: The current study suggests VDR-associated genes as possible markers for BD.


Asunto(s)
Trastorno Bipolar , ARN Largo no Codificante , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Trastorno Bipolar/genética , Femenino , Humanos , Masculino , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Transducción de Señal , Vitamina D
17.
J Mol Neurosci ; 72(2): 239-245, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34499334

RESUMEN

Vitamin D receptor (VDR) signaling has been found to contribute to the pathology of numerous neuropsychiatric diseases including schizophrenia. Notably, VDR signaling has a functional relationship with many long non-coding RNAs (lncRNAs) such as SNHG6, LINC00346 and LINC00511. We calculated expression of these lncRNAs in the venous blood of patients with schizophrenia versus healthy individuals. Expression of SNHG6 was significantly higher in cases versus controls (posterior beta = 0.552, adjusted P value < 0.0001). This pattern of expression was detected in both men (posterior beta = 0.556, adjusted P value < 0.0001) and women (posterior beta = 0.31, adjusted P value = 0.005). Expression of LINC00346 was also higher in cases versus controls (posterior beta = 0.497, adjusted P value < 0.0001) and in distinct sex-based comparisons (posterior beta = 0.451, adjusted P value = 0.009 among men and posterior beta = 0.214, P value = 0.004 among women). Expression of LINC00511 was higher in cases versus controls (posterior beta = 0.318, adjusted P value = 0.01). While sex-based comparisons revealed significant difference in expression of LINC00511 among female subgroups (posterior beta = 0.424, adjusted P value = 0.016), such comparison showed no difference among male cases and male controls (adjusted P value = 0.295). The expression levels of SNHG6 distinguished patients with schizophrenia from controls, with AUC = 0.932. LINC00346 and LINC00511 distinguished between the two groups with AUC values of 0.795 and 0.706, respectively. Therefore, these lncRNAs might be used as markers for schizophrenia.


Asunto(s)
ARN Largo no Codificante , Esquizofrenia , Femenino , Humanos , Masculino , ARN Largo no Codificante/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Esquizofrenia/genética , Transducción de Señal , Activación Transcripcional , Regulación hacia Arriba
18.
Cancer Biother Radiopharm ; 37(4): 246-255, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-32935999

RESUMEN

Background: Pituitary adenoma (PA) is a common primary brain tumor with invasive properties. Despite that long noncoding RNA (lncRNA) small nucleolar RNA host gene 6 (SNHG6) exerts oncogenic function in cancer cells and that miR-944 inhibits epithelial-mesenchymal transition (EMT) of cancer cells are well documented, few studies have explored the function and mechanism of SNHG6 and miR-944 in invasive pituitary adenoma (IPA). Materials and Methods: Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the expressions of SNHG6 and miR-944 in PA samples. Human PA cell line HP75 was used as a cell model. The biological effects of SNHG6 and miR-944 on HP75 cells were investigated with cell counting kit-8 (CCK-8) assay, Transwell assay, and scratch healing assay in vitro, respectively. Markers of EMT, including E-cadherin and vimentin, were detected by Western blot. Interactions between SNHG6 and miR-944, miR-944 and RAB11A were determined by bioinformatics analysis, qRT-PCR, and dual luciferase reporter assay. Results: SNHG6 was significantly upregulated in IPA samples, whereas miR-944 was downregulated. SNHG6 markedly promoted viability, migration, invasion, and EMT of PA cells, whereas miR-944 transfection had the opposite effects. SNHG6 could downregulate miR-944, and there was a negative correlation between SNHG6 expression and miR-944 expression in IPA samples. Besides, it was confirmed that miR-944 could pair with the 3'-untranslated region of RAB11A and repress its expression. Conclusions: This study authenticates that the SNHG6/miR-994/RAB11A axis plays a crucial role in regulating proliferation, migration, invasion, and EMT of IPA cells. SNHG6 and miR-994 can serve as novel valuable therapeutic targets for IPA.


Asunto(s)
Adenoma , Transición Epitelial-Mesenquimal , MicroARNs , Neoplasias Hipofisarias , ARN Largo no Codificante , Adenoma/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Invasividad Neoplásica/genética , Neoplasias Hipofisarias/genética , ARN Largo no Codificante/genética
19.
Anim Cells Syst (Seoul) ; 26(6): 369-379, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36605586

RESUMEN

Metabolic reprogramming is an important feature in tumor progression. Long noncoding RNA's (lncRNA) small nucleolar RNA host gene 6 (SNHG6) acts as a proto-oncogene in hepatocellular carcinoma (HCC) but its role in glycolysis is mostly unknown. The role of SNHG6 and Block of proliferation 1 (BOP1) on glycolysis is assessed by glucose uptake, lactate production, oxygen consumptive rate (OCR) and extracellular acidification rate (ECAR) and glycolytic enzyme levels. The regulatory effect of SNHG6 on BOP1 protein was confirmed by Western blotting, MS2 pull-down, RNA pull-down, and RIP assay. SNHG6 and BOP1 levels were increased in HCC tissues and cells. SNHG6 and BOP1 were prognostic factors in HCC patients and significantly correlated to TP53 mutant and tumor grade. SNHG6 promoted proliferation, inhibited apoptosis, enhanced glucose uptake and lactate production, decreased OCR, and increased ECAR in HCC cell lines. SNHG6 could bind the BOP1 protein and enhance its stability. BOP1 overexpression rescued the change of proliferation, apoptosis, and glycolysis in HCCLM3 and SMMC-7721 cells. Our data indicate that SNHG6 accelerates proliferation and glycolysis and inhibits the apoptosis of HCC cell lines by binding the BOP1 protein and enhancing its stability. Both SNHG6 and BOP1 are promising prognostic and therapeutic markers in HCC.

20.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-956919

RESUMEN

Objective:To investigate the effect of lncRNA SNHG6 on the proliferation and radiotherapy sensitivity of cervical cancer SiHa cells and its potential mechanism.Methods:The expression levels of lncRNA SNHG6 and miR-485-3p in cervical cancer tissues, paracancer tissues, SiHa cells and SiHa cells exposed to X-ray were detected. The relationship between lncRNA SNHG6 and miR-485-3p was analyzed. After overexpression or knockdown of SNHG6 and miR-485-3p, cell proliferation ability, number of invasion and apoptosis rate were determined by MTT, Transwell chamber assay and flow cytometry, respectively. The effect of miR-485-3p on the Wnt/β-catenin pathway and the effect of XAV939 on SiHa cell proliferation and radiation sensitivity were analyzed.Results:lncRNA SNHG6 was highly expressed in cervical cancer tissues and SiHa cells, whereas was lowly expressed in X-ray irradiated SiHa cells. miR-485-3p was lowly expressed in cervical cancer tissues and SiHa cells, whereas was highly expressed in X-ray irradiated SiHa cells. lncRNA SNHG6 targeted miR-485-3p. Down-regulation of lncRNA SNHG6 expression inhibited cell proliferation and invasion, and enhanced its sensitivity to X-ray radiotherapy, while miR-485-3p inhibitor transfected cells exerted the opposite effect. The up-regulation of lncRNA SNHG6 promoted the proliferation and invasion of SiHa cells through miR-485-3p, and reduced the sensitivity of radiotherapy. Down-regulation of miR-485-3p activated the Wnt/β-catenin pathway, promoted cell proliferation and invasion of SiHa, and reduced its radiation sensitivity to X-ray.Conclusion:Overexpression of lncRNA SNHG6 targeting miR-485-3p activates the Wnt/β-catenin pathway to regulate the proliferation and radiotherapy sensitivity of SiHa cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA