Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Exp Cell Res ; 434(1): 113848, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37918704

RESUMEN

BACKGROUND: Epilepsy is a neurological disorder characterized by recurrent seizures and is often unresponsive to current treatment options. Ferroptosis, a recently defined iron-dependent regulated cell death, has been suggested as a potential therapeutic target for epilepsy due to its association with oxidative stress. Additionally, circRNA SLC8A1 (circSLC8A1) has been implicated in various neurological disorders and oxidative stress-related diseases but its involvement in epilepsy progression, particularly in relation to ferroptosis and oxidative stress, remains unclear. METHODS: qRT-PCR, Western blot, IHC and ELISA assays were employed to validate the relative expression of targeted genes and proteins. The levels of ROS, iron, LOP and GSH were detected by commercial kits. RNA pull-down and RIP assays were employed to detect the interactions among circSLC8A1, FUS and ATF3. A rat epilepsy model was established for further in vivo confirmation. RESULTS AND CONCLUSION: In this study, we investigated the potential involvement of circSLC8A1 in epilepsy progression and its connection to ferroptosis and oxidative stress. Our findings demonstrate that circSLC8A1 triggers neuronal ferroptosis by stabilizing ATF3 mRNA expression through recruitment with FUS. The induced neuronal ferroptosis contributes to epilepsy progression. These results enhance our understanding of epilepsy pathogenesis and may provide insights for the development of novel therapeutic strategies.


Asunto(s)
Epilepsia , Ferroptosis , Animales , Ratas , Epilepsia/genética , Ferroptosis/genética , Hipocampo , Hierro , Estabilidad del ARN , ARN Circular/genética , Proteína FUS de Unión a ARN/metabolismo
2.
J Proteome Res ; 23(11): 4802-4820, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39441737

RESUMEN

A deeper understanding of colorectal cancer (CRC) biology would help to identify specific early diagnostic markers. Here, we conducted quantitative proteomics on FFPE healthy, adenoma, and adenocarcinoma tissue samples from six stage I sporadic CRC patients to identify dysregulated proteins during early CRC development. Two independent quantitative 10-plex TMT experiments were separately performed. After protein extraction, trypsin digestion, and labeling, proteins were identified and quantified by using a Q Exactive mass spectrometer. A total of 2681 proteins were identified and quantified after data analysis and bioinformatics with MaxQuant and the R program. Among them, 284 and 280 proteins showed significant upregulation and downregulation (expression ratio ≥1.5 or ≤0.67, p-value ≤0.05), respectively, in adenoma and/or adenocarcinoma compared to healthy tissue. Ten dysregulated proteins were selected to study their role in CRC by WB, IHC, TMA, and ELISA using tissue and plasma samples from CRC patients, individuals with premalignant colorectal lesions (adenomas), and healthy individuals. In vitro loss-of-function cell-based assays and in vivo experiments using three CRC cell lines with different metastatic properties assessed the important roles of SLC8A1 and TXNDC17 in CRC and liver metastasis. Additionally, SLC8A1 and TXNDC17 protein levels in plasma possessed the diagnostic ability of early CRC stages.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Colorrectales , Adhesión en Parafina , Proteómica , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Proteómica/métodos , Biomarcadores de Tumor/metabolismo , Masculino , Femenino , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenoma/patología , Adenoma/metabolismo , Línea Celular Tumoral , Persona de Mediana Edad , Regulación Neoplásica de la Expresión Génica , Anciano , Animales
3.
J Gene Med ; 22(11): e3263, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32830397

RESUMEN

BACKGROUND: Osteoporosis is a disease characterized by bone loss, imbalance of bone metabolism and destruction of trabecular microarchitecture. Circular RNAs (circRNAs) have been revealed as important biological regulators in human diseases. The expression characteristics and mechanism of circRNAs in osteoporosis are unclear. METHODS: The binding sites of miR-516b-5p on circ-SLC8A1 and AKAP2 mRNA were predicted using circAtlas (http://circatlas.biols.ac.cn) and miRDB (http://mirdb.org). Target sites of miR-516b-5p on circ-SLC8A1 and AKAP2 mRNA were confirmed by a dual luciferase assay. The relationship between miR-516b-5p and AKAP2 was determined by a quantitative reverse transcriptase-polymerase chain reaction. Alizarin red S staining and alkaline phosphatase staining were used to observe the level of osteogenic differentiation after transfection. RESULTS: The first six circRNAs captured from the 30 circRNAs with highest expression in the bone marrow were examined in a mouse model of osteoporosis and the expression of circ-SLC8A1 was found to be significantly reduced in osteoporosis. Circ-SLC8A1 negatively regulated the expression of miR-516b-5p. Overexpression of circ-SLC8A1 blocked the inhibition of AKAP2 by miR-516b-5p. CONCLUSIONS: Circ-SLC8A1 blocks the inhibitory effect of miR-516b-5p on the downstream target gene AKAP2 and promotes osteoporosis. The findings of the present might help to provide a new strategy for the treatment of osteoporosis.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Biomarcadores/metabolismo , Regulación de la Expresión Génica , Proteínas de la Membrana/metabolismo , MicroARNs/genética , Osteoporosis/patología , ARN Circular/genética , Intercambiador de Sodio-Calcio/genética , Proteínas de Anclaje a la Quinasa A/genética , Animales , Células Cultivadas , Femenino , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Osteoporosis/genética , Osteoporosis/metabolismo , Pronóstico
4.
J Cell Physiol ; 234(6): 9019-9032, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30378115

RESUMEN

Extensive investigations into long noncoding RNAs (lncRNAs) in various diseases and cancers, including acute myocardial infarction (AMI) have been conducted. The current study aimed to investigate the role of lncRNA solute carrier family 8 member A1 antisense RNA 1 (SLC8A1-AS1) in myocardial damage by targeting solute carrier family 8 member A1 (SLC8A1) via cyclic guanosine 3',5'-monophosphate-protein kinase G (cGMP-PKG) signaling pathway in AMI mouse models. Differentially expressed lncRNA in AMI were initially screened and target relationship between lncRNA SLC8A1-AS1 and SLC8A1 was then verified. Infarct size, levels of inflammatory factors, biochemical indicators, and the positive expression of the SLC8A1 protein in AMI were subsequently determined. The expression of SLC8A1-AS1, SLC8A1, PKG1, PKG2, atrial natriuretic peptide, and brain natriuretic peptide was detected to assess the effect of SLC8A1-AS1 on SLC8A1 and cGMP-PKG. The respective contents of superoxide dismutase, lactate dehydrogenase (LDH), and malondialdehyde (MDA) were detected accordingly. Microarray data GSE66360 provided evidence indicating that SLC8A1-AS1 was poorly expressed in AMI. SLC8A1 was verified to be a target gene of lncRNA SLC8A1-AS1. SLC8A1-AS1 upregulation decreased levels of left ventricular end-systolic diameter, -dp/ dt max , interleukin 1ß (IL-1ß), IL-6, transforming growth factor α, nitric oxide, inducible nitric-oxide synthase, endothelial nitric-oxide synthase, infarct size, LDH activity and MDA content, and increased IL-10, left ventricular end-diastolic pressure and + dp/ dt max . Furthermore, the overexpression of SLC8A1-AS1 was noted to elicit an inhibitory effect on the cGMP-PKG signaling pathway via SLC8A1. In conclusion, lncRNA SLC8A1-AS1, by downregulating SLC8A1 and activating the cGMP-PKG signaling pathway, was observed to alleviate myocardial damage, inhibit the release of proinflammatory factors and reduce infarct size, ultimately protecting against myocardial damage.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/enzimología , ARN sin Sentido/metabolismo , ARN Largo no Codificante/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Animales , Línea Celular , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Hemodinámica , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/enzimología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , ARN sin Sentido/genética , ARN Largo no Codificante/genética , Sistemas de Mensajero Secundario , Intercambiador de Sodio-Calcio/genética , Función Ventricular Izquierda
5.
Mol Carcinog ; 56(10): 2258-2266, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28544176

RESUMEN

The SLC8A1 (solute carrier family 8, member 1) gene, encoding Na+ /Ca2+ exchanger, is essential in regulating calcium reabsorption and homeostasis. Calcium homeostasis plays a key role in cell proliferation and apoptosis. We hypothesized that polymorphisms in five calcium-regulating genes (SLC8A1, ATP2B1, CALB1, CALB2, and CABP1) interact with calcium intake in relation to the risk of colorectal neoplasia. A two-phase (discovery and replication) study was conducted within the Tennessee Colorectal Polyp Study, including a total of 1275 cases and 2811 controls. In Phase I, we identified six out of 135 SNPs that significantly interacted with calcium intake in relation to adenoma risk. In Phase II, the calcium intake by rs4952490 (SLC8A1) interaction was replicated (Pinteraction = 0.048). We found an inverse association between calcium intake (1000-2000 mg/day) and colorectal adenomas, particularly for multiple/advanced adenomas, among the G-allele carriers but not among homozygous carriers of the common variant (A) in rs4952490. In the joint analysis of SLC8A1, KCNJ1, and SLC12A1 SNPs, carriers of variant alleles in at least two genes and with calcium intake above the DRI (1000 mg/day) were approximately 30-57% less likely to have adenomas than those whose calcium intake was below the DRI. The association was stronger for multiple/advanced adenomas. No association was found among those who did not carry any variant alleles in these genes when calcium intake was below 2500 mg/day. These findings, if confirmed, may provide a new avenue for the personalized prevention of colorectal adenoma and cancer.


Asunto(s)
Calbindina 1/genética , Calbindina 2/genética , Calcio de la Dieta/administración & dosificación , Neoplasias Colorrectales/genética , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Intercambiador de Sodio-Calcio/genética , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Variantes Farmacogenómicas , Polimorfismo de Nucleótido Simple , Factores de Riesgo
6.
Eur Arch Otorhinolaryngol ; 274(2): 757-763, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27837419

RESUMEN

Aim of this work was to assess the role of polymorphisms belonging to genes involved in the regulation of ionic homeostasis in Caucasian patients with Ménière Disease (MD). We recruited 155 patients with definite Ménière Disease and 186 controls (Control Group 1) without a lifetime history of vertigo, overlapping with patients for age and rate of hypertension. We validated the positive results on 413 Caucasian subjects selected from a European general population (Control Group 2). The clinical history for migraine and hypertension was collected; genomic DNA was characterized for a panel of 33 SNPs encoding proteins involved in ionic transport. We found a higher rate of migraineurs in MD subjects compared to Group 1 (46.8 vs 15.5%, p = 0.00005). Four SNPs displayed differences in MD patients compared to Group 1 controls: rs3746951 and rs2838301 in SIK1 gene, rs434082 and rs487119 in SLC8A1; the p values of Chi-squared test for genotype frequencies are 0.009, 0.023, 0.009 and 0.048, respectively. SLC8A1 gene encodes for Na+-Ca++ exchanger, while SIK1 gene encodes for Salt Inducible Kinase 1, an enzyme associated with Na+-K+ ATPase function. The validation with Control Group 2 displayed that only rs3746951 and rs487119 are strongly associated to MD (p = 0.001 and p = 0.0004, respectively). These data support the hypothesis that a genetically induced dysfunction of ionic transport may act as a predisposing factors to develop MD.


Asunto(s)
Homeostasis/genética , Iones/metabolismo , Enfermedad de Meniere/genética , Población Blanca/genética , Adulto , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Masculino , Enfermedad de Meniere/complicaciones , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Vértigo/complicaciones
7.
Heliyon ; 10(17): e36899, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39263055

RESUMEN

Background: The field of gynaecological immunology has increasingly focused on recurrent spontaneous abortion (RSA). The complex mechanisms underlying the interaction between RSA and cancer are not well understood. Methods: Weighted gene coexpression network analysis (WGCNA), single-cell RNA sequencing (scRNA-seq), and machine learning algorithms were used for the analysis of RSA decidua samples to identify the hub genes. The expression and distribution of the hub genes were subsequently investigated via the pancancer database TCGA. A prognostic prediction was made to assess the impact of the hub genes on the cancer response, mutation burden, immune microenvironment, immune checkpoint, and chemotherapy. In vitro assays were performed to determine whether SLC8A1 influences HTR-8/SVneo cell proliferation, apoptosis and the concentration of calcium ions. Results: SLC8A1 was identified as a hub gene within RSA and was highly expressed in uterine corpus endometrial carcinoma (UCEC). The efficacy of SLC8A1 as a predictive marker was substantiated by calibration curves and the concordance index. The mutation rate of SLC8A1 was found to be 6 % on the basis of the waterfall plot. Immune analysis revealed notable differences in the fractions of T cells and macrophages between the high- and low-expression groups. Patients classified in the low-risk group exhibited enhanced responsiveness to osimertinib, dasatinib, and ibrutinib. The results of in vitro experiments revealed that SLC8A1 promotes proliferation and inhibits the apoptosis and concentration of calcium ions in HTR-8/SVneo cells. Conclusion: These findings suggest that SLC8A1 may serve as a promising prognostic biomarker and potential target for immunotherapy in the context of RSA and UCEC.

8.
Nefrologia (Engl Ed) ; 44(4): 576-581, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39216982

RESUMEN

Autosomal dominant tubulointerstitial kidney disease (ADTKD) comprises a heterogeneous group of rare hereditary kidney diseases characterized by family history of progressive chronic kidney disease (CKD) with bland urine sediment, absence of significant proteinuria and normal or small-sized kidneys. Current diagnostic criteria require identification of a pathogenic variant in one of five genes - UMOD, MUC1, REN, HNF1ß, SEC61A1. The most prevalent form of ADTKD is uromodulin-associated kidney disease (ADTKD-UMOD). Genetic study of a Portuguese family diagnosed with familial juvenile hyperuricemic nephropathy (FJHN), one of the nosological entities in the spectrum of ADTKD, revealed a previously unreported large deletion in UMOD encompassing the entire terminal exon, which strictly cosegregated with CKD and hyperuricemia/gout, establishing the primary diagnosis of ADTKD-UMOD; as well as an ultra-rare nonsense SLC8A1 variant cosegregating with the UMOD deletion in patients that consistently exhibited an earlier onset of clinical manifestations. Since the terminal exon of UMOD does not encode for any of the critical structural domains or amino acid residues of mature uromodulin, the molecular mechanisms underlying the pathogenicity of its deletion are unclear and require further research. The association of the SLC8A1 locus with FJHN was first indicated by the results of a genome-wide linkage analysis in several multiplex families, but those data have not been subsequently confirmed. Our findings in this family revive that hypothesis.


Asunto(s)
Hiperuricemia , Linaje , Uromodulina , Humanos , Uromodulina/genética , Hiperuricemia/genética , Masculino , Femenino , Eliminación de Secuencia , Adulto , Gota/genética , Eliminación de Gen , Enfermedades Renales
9.
Sci Rep ; 14(1): 5233, 2024 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-38433218

RESUMEN

Macrophages act as the first immune defense line of the host against Mycobacterium tuberculosis (Mtb). A previous study showed that circRNA_SLC8A1 was significantly upregulated in Mtb-infected macrophages, but its regulatory mechanism in anti-tuberculosis infection is unclear. Therefore, this study aimed to investigate the role of circRNA_SLC8A1 in the anti-tuberculosis activity of macrophages. We showed that circRNA_SLC8A1 was upregulated in tuberculosis patients. Moreover, the binding sites of miR-20b-5p on circRNA_SLC8A1 and Sequestosome 1 (SQSTM1/p62) mRNA were predicted by StarBase and verified by the double luciferase reporter gene assay. Next, we found that miR-20b-5p expression was decreased, while SQSTM1 protein expression was increased in a time- and dose-dependent manner in the human macrophage U937 in response to Mtb infection. Furthermore, circRNA_SLC8A1 overexpression vector (circRNA_SLC8A1) or shRNA (sh-circRNA_SLC8A1) and/or miR-20b-5p mimic or inhibitor and/or SQSTM1 overexpression vector (SQSTM1) or small interfering RNA (si-SQSTM1) or its corresponding control were transfected into Mtb-infected macrophages. Results showed that overexpression of circRNA_SLC8A1 or miR-20b-5p inhibitor promoted the secretion of pro-inflammatory factors IL-1ß, IL-6, and TNF-α, increased Nitric Oxide (NO) content and inducible nitric oxide synthase (iNOS) expression, inhibited Reactive oxygen species (ROS) production. Cleaved-caspase-3 protein expression, and cell apoptosis, and promoted Mtb survival. Silencing SQSTM1 inhibited secretion of pro-inflammatory factors and activation of the NF-κB pathway. Overexpression of miR-20b-5p blocked the promoting of circ-SLC8A1 on SQSTM1 protein expression. In summary, circRNA_SLC8A1 sponged miR-20b-5p to upregulate SQSTM1/p62 expression and promoted Mtb survival in macrophages through the NF-κB signaling pathway.


Asunto(s)
MicroARNs , Mycobacterium tuberculosis , Humanos , FN-kappa B , Proteína Sequestosoma-1/genética , ARN Circular/genética , Proteínas Relacionadas con la Autofagia , MicroARNs/genética
10.
J Mol Cell Cardiol ; 63: 189-98, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23896226

RESUMEN

Myosin-binding protein C (Mybpc3)-targeted knock-in mice (KI) recapitulate typical aspects of human hypertrophic cardiomyopathy. We evaluated whether these functional alterations can be reproduced in engineered heart tissue (EHT) and yield novel mechanistic information on the function of cMyBP-C. EHTs were generated from cardiac cells of neonatal KI, heterozygous (HET) or wild-type controls (WT) and developed without apparent morphological differences. KI had 70% and HET 20% lower total cMyBP-C levels than WT, accompanied by elevated fetal gene expression. Under standard culture conditions and spontaneous beating, KI EHTs showed more frequent burst beating than WT and occasional tetanic contractions (14/96). Under electrical stimulation (6Hz, 37°C) KI EHTs exhibited shorter contraction and relaxation times and a twofold higher sensitivity to external [Ca(2+)]. Accordingly, the sensitivity to verapamil was 4-fold lower and the response to isoprenaline or the Ca(2+) sensitizer EMD 57033 2- to 4-fold smaller. The loss of EMD effect was verified in 6-week-old KI mice in vivo. HET EHTs were apparently normal under basal conditions, but showed similarly altered contractile responses to [Ca(2+)], verapamil, isoprenaline and EMD. In contrast, drug-induced changes in intracellular Ca(2+) transients (Fura-2) were essentially normal. In conclusion, the present findings in auxotonically contracting EHTs support the idea that cMyBP-C's normal role is to suppress force generation at low intracellular Ca(2+) and stabilize the power-stroke step of the cross bridge cycle. Pharmacological testing in EHT unmasked a disease phenotype in HET. The altered drug response may be clinically relevant.


Asunto(s)
Proteínas Portadoras/genética , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/genética , Miocardio/metabolismo , Ingeniería de Tejidos , Agonistas Adrenérgicos beta/farmacología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Modelos Animales de Enfermedad , Marcación de Gen , Espacio Intracelular/metabolismo , Isoproterenol/farmacología , Ratones , Ratones Transgénicos , Transcriptoma , Verapamilo/farmacología
11.
EMBO Mol Med ; 15(12): e17719, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-37966164

RESUMEN

Metastatic uveal melanomas are highly resistant to all existing treatments. To address this critical issue, we performed a kinome-wide CRISPR-Cas9 knockout screen, which revealed the LKB1-SIK2 module in restraining uveal melanoma tumorigenesis. Functionally, LKB1 loss enhances proliferation and survival through SIK2 inhibition and upregulation of the sodium/calcium (Na+ /Ca2+ ) exchanger SLC8A1. This signaling cascade promotes increased levels of intracellular calcium and mitochondrial reactive oxygen species, two hallmarks of cancer. We further demonstrate that combination of an SLC8A1 inhibitor and a mitochondria-targeted antioxidant promotes enhanced cell death efficacy in LKB1- and SIK2-negative uveal melanoma cells compared to control cells. Our study also identified an LKB1-loss gene signature for the survival prognostic of patients with uveal melanoma that may be also predictive of response to the therapy combination. Our data thus identify not only metabolic vulnerabilities but also new prognostic markers, thereby providing a therapeutic strategy for particular subtypes of metastatic uveal melanoma.


Asunto(s)
Melanoma , Neoplasias de la Úvea , Humanos , Calcio , Proliferación Celular , Melanoma/tratamiento farmacológico , Especies Reactivas de Oxígeno , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/patología
12.
J Cancer Res Clin Oncol ; 149(11): 8335-8344, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37074453

RESUMEN

BACKGROUND: In recent years, morbidity and mortality from colorectal cancer have increased. Colorectal adenoma is the main precancerous lesion. Understanding the pathogenesis of colorectal adenoma will help to improve the early diagnosis rate of colorectal cancer. METHODS: In this case-control study, we focused on three single nucleotide polymorphisms (SNPs) in genes SLC8A1 (rs4952490), KCNJ1 (rs2855798), and SLC12A1 (rs1531916). We analyzed 207 colorectal adenoma patients (112 high-risk cases and 95 low-risk cases) and 212 control subjects by Sanger sequencing. A food frequency questionnaire (FFQ) was used to survey demographic characteristics and dietary nutrition. RESULTS: In the overall analysis, the results suggested that the AA+AG and AG genotype carriers of rs4952490 had a 73.1% and 78% lower risk of colorectal adenoma compared to GG genotype carriers, respectively. However rs2855798 and rs1531916 were not associated with the incidence of colorectal adenoma. Additionally, stratified analysis showed that rs4952490 AA+AG and AG genotypes had a protective effect against low-risk colorectal adenoma in patients aged ≤ 60 years old who were non-smokers. We also observed that when calcium intake was higher than 616 mg/d and patients carried at least one gene with variant alleles there was a protective effect against low-risk colorectal adenoma. CONCLUSIONS: Interactions between dietary calcium intake and calcium reabsorption genes may affect the occurrence and development of colorectal adenoma.


Asunto(s)
Adenoma , Neoplasias Colorrectales , Canales de Potasio de Rectificación Interna , Humanos , Persona de Mediana Edad , Calcio , Calcio de la Dieta , Estudios de Casos y Controles , Polimorfismo de Nucleótido Simple , Genotipo , Neoplasias Colorrectales/patología , Adenoma/genética , Factores de Riesgo , Canales de Potasio de Rectificación Interna/genética , Miembro 1 de la Familia de Transportadores de Soluto 12/genética
13.
Front Oncol ; 12: 898954, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35847849

RESUMEN

Background: Expanding the druggable novel anaplastic lymphoma kinase (ALK) fusions list is crucial to the precise treatment of patients with cancer with positive ALK fusions. The intergenic-ALK fusions accounted for a substantial proportion of ALK fusions. However, they were typically considered of limited clinical significance due to the obscure functional partners. In this case report, a patient carrying intergenic-ALK fusion presents an excellent outcome after taking the new second-generation tyrosine kinase inhibitor (TKI) candidate, WX-0593. Case Presentation: A 47-year-old Chinese female patient diagnosed with IVB lung adenocarcinoma was admitted to the hospital with large dimension lesions in the left lobe of the lung. After 1 week of first line chemotherapy, no response was found. A novel ALK rearrangement generated by a fusion of the intergenic region between SLC8A1 and PKDCC to the intron 19 of ALK was presented after next-generation sequencing and was further confirmed by Sanger's sequencing. High expression of ALK was revealed by immunohistochemistry. The patient was directed to engage in phase III clinical trial (NCT04632758) and received an orally active second-generation ALK inhibitor WX-0593. Over the course of 17 months, the partial response was obtained without significant side effects. Conclusion: In summary, a patient with non-small cell lung cancer harboring a novel intergenic-ALK fusion, whose intergenic breakpoint was located between SLC8A1 and PKDCC, benefited from a potent ALK TKI candidate WX-0593. This finding extended the scope of targetable ALK fusions. More importantly, it highlighted the advantages of next-generation sequencing in identifying rare but functional ALK fusions, which eventually benefit patients.

14.
Front Mol Biosci ; 9: 875805, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35755822

RESUMEN

Recently, circular RNAs (circRNAs) have been extensively studied in animals and plants. circRNAs are generated by backsplicing from the same linear transcripts that are canonically spliced to produce, for example, mature mRNAs. circRNAs exhibit tissue-specific expression and are potentially involved in many diseases, among them cardiovascular diseases. The comprehensive analysis of circRNA expression patterns across larger patient cohorts requires a streamlined and cost-effective workflow designed to meet small input requirements. In this article, we present Lexo-circSeq, a targeted RNA sequencing approach that can profile up to 110 circRNAs and their corresponding linear transcripts in one experiment. We established Lexo-circSeq employing total human heart RNA and show that our protocol can detect depletion of a specific circRNA in hiPSC-derived cardiomyocytes. Finally, Lexo-circSeq was applied to biopsies from patients diagnosed with dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM), respectively. Interestingly, our results indicate that circular-to-linear-ratios for circSLC8A1 and circRBM33 are deregulated in cardiomyopathy.

15.
Open Life Sci ; 17(1): 846-850, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36045716

RESUMEN

ALK fusion genes are diverse. Approximately 30 different ALK fusion protein partners have been described previously, and some of these fusion proteins have been reported to be effective against ALK-tyrosine kinase inhibitor (TKI). ALK rearrangements often occur at a common breakpoint in exon 20 of the genome. SLC8A1-ALK, a novel fusion protein partner, comes from exon 2 of the SLC8A1 gene rearranged with exon 20 of the ALK gene. Here, we reported a patient with advanced lung adenocarcinoma harboring a SLC8A1-ALK fusion who benefited from first-line treatment with alectinib. After 2 months of taking alectinib, the targeted lung lesions and intrahepatic metastases regressed significantly. To date, the patient has achieved nearly 1 year of progression-free survival while taking the drug. Given the diversity of ALK fusion genes and the different efficacy of ALK-TKIs, we believe that this case report has an important clinical reference.

16.
Bioengineered ; 13(5): 12572-12582, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35599603

RESUMEN

Papillary thyroid cancer (PTC) is one of the most prevalent endocrine malignancies and is associated with severe morbidity and high mortality. This study aimed to explore the role of long non-coding RNA (lncRNA) SLC8A1 antisense RNA 1 (SLC8A1-AS1) in the pathogenesis of PTC. In this study, we explored the function of SLC8A1-AS1 in PTC progression. We observed that the expression of SLC8A1-AS1 was downregulated in clinical PTC samples and PTC cell lines compared to that in normal controls. Cell counting kit (CCK)-8 assays demonstrated that the overexpression of SLC8A1-AS1 significantly reduced the proliferation of PTC cells. Consistently, apoptosis of PTC cells was enhanced by SLC8A1-AS1 overexpression. SLC8A1-AS1 overexpression attenuated the invasion and migration of PTC cells. Mechanistically, SLC8A1-AS1 maintained NUMB like endocytic adaptor protein (Numbl) mRNA stability by interacting with FUS RNA Binding Protein (FUS) in PTC cells. Depletion of Numbl reversed the inhibitory effect of SLC8A1-AS1 overexpression on PTC. Thus, we concluded that SLC8A1-AS1 suppresses PTC progression via the FUS/Numbl axis. Our findings provide novel insights into the mechanism underlying SLC8A1-AS1 attenuation of the malignant development of PTC, improving our understanding of the association between lncRNAs and PTC. SLC8A1-AS1 and FUS may be potential targets for PTC treatment.


Asunto(s)
ARN sin Sentido , ARN Largo no Codificante , Proteína FUS de Unión a ARN , Neoplasias de la Tiroides , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , ARN sin Sentido/genética , ARN Largo no Codificante/genética , Proteína FUS de Unión a ARN/genética , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología
17.
Cell Oncol (Dordr) ; 44(1): 219-233, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33001357

RESUMEN

PURPOSE: Ca2+ homeostasis plays a pivotal role in regulating proliferation and apoptosis during cancer development. This study intended to examine the potential tumor-suppressing role of ZNF503 antisense RNA 1 (ZNF503-AS1) in bladder cancer, which may be implicated in the regulation of Ca2+ homeostasis. METHODS: Differentially expressed long non-coding RNAs (lncRNAs) related to bladder cancer were identified using microarray analysis, followed by the verification of transcription factors to which they bind. The relationship between ZNF503-AS1, GATA6 and SLC8A1 was assessed using dual luciferase reporter, RIP and ChIP assays. The expression levels of ZNF503-AS1, GATA6 and SLC8A1 were modulated to examine their effects on the tumorigenic potential, intracellular Ca2+ concentration and Ca2+-ATPase activity in bladder cancer cells. The in vivo tumorigenic ability was validated in nude mice. RESULTS: Microarray-based expression profile analysis of the GEO GSE61615 dataset revealed that the expression of ZNF503-AS1 was decreased in bladder cancer. Subsequently, we found that ZNF503-AS1 can bind to the transcription factor GATA6 to up-regulate the expression of SLC8A1. ZNF503-AS1 and SLC8A1 were found to be down-regulated in both primary bladder cancer tissues and cells. Exogenous overexpression of ZNF503-AS1 or SLC8A1 attenuated bladder cancer cell proliferation, invasion and migration, but promoted their apoptosis, accompanied by decreased Ca2+-ATPase activities and increased intracellular Ca2+ concentrations. Additional in vivo experiments validated the inhibitory effect of ZNF503-AS1 overexpression on the tumorigenic capacity of bladder cancer cells in nude mice. CONCLUSION: ZNF503-AS1 can recruit transcription factor GATA6 to up-regulate SLC8A1 expression, thereby increasing the intracellular Ca2+ concentration and repressing the proliferation, invasion and migration, and enhancing the apoptosis of bladder cancer cells.


Asunto(s)
Calcio/metabolismo , Factor de Transcripción GATA6/metabolismo , Genes Supresores de Tumor , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba/genética , Neoplasias de la Vejiga Urinaria/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Desnudos , Modelos Biológicos , Invasividad Neoplásica , ARN Largo no Codificante/genética , Intercambiador de Sodio-Calcio/metabolismo , Neoplasias de la Vejiga Urinaria/patología
18.
Brain Res Bull ; 176: 112-120, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34474120

RESUMEN

As the most common aggressive malignant tumor in the central nervous system, glioma is still an insurmountable disease in the neural system. The mechanism of carcinogenesis in glioma remains largely unclear. In the present study, we identified a dysregulated long non-coding RNA (lncRNA) solute carrier family 8 member A1 antisense RNA 1 (SLC8A1-AS1) associated with glioma based on The Cancer Genome Atlas (TCGA) data. A validation experiment was conducted to confirm a high expression level of lncRNA SLC8A1-AS1 in glioma tissues. Down-regulation of lncRNA SLC8A1-AS1 suppressed the proliferation, colony formation, migration, and invasion of glioma cells in vitro and in vivo. Moreover, lncRNA SLC8A1-AS1 silencing decreased the activity of the Wnt/ß-catenin pathway and suppressed the epithelial to mesenchymal transition (EMT) in glioma cells. These findings collectively provide novel insights into the function and mechanism of lncRNA SLC8A1-AS1 in the pathogenesis of glioma and highlight its potential as a therapeutic target for glioma intervention.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Movimiento Celular/genética , Glioma/metabolismo , ARN Largo no Codificante/metabolismo , Vía de Señalización Wnt/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioma/genética , Glioma/patología , Humanos , ARN Largo no Codificante/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda