Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Anticancer Drugs ; 35(4): 333-343, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38241194

RESUMEN

The treatment strategy for nonsmall cell lung cancer (NSCLC) has always been a hot topic of concern, and its treatment strategies are also emerging. This experiment wants to know the effects of apolipoprotein C1 (APOC1) in immunotherapy of NSCLC. APOC1 mRNA and protein expression were upregulated in lung cancer tissue of patients with NSCLC. programmed cell death protein 1 (PD-1) mRNA expression was negatively correlated with PD-1 mRNA expression in patients. The survival rate of APOC1 high expression was lower than that of low expression in patients with NSCLC. APOC1 gene reduced the transformation of M2 into M1 macrophages (TMMM). APOC1 gene promoted cell growth, and the gene reduced ferroptosis of NSCLC. APOC1-induced nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (NRF2/HO-1) signaling pathway. Sh-APOC1 gene reduced cell growth in mice of NSCLC through the inhibition of NRF2/HO-1 signaling pathway. The inhibition of NRF2 reduced the TMMM by APOC1. The activation of NRF2 reduced the TMMM by si-APOC1. In conclusion, APOC1 reduced anti-PD-1 immunotherapy of NSCLC via the TMMM by ferroptosis by NRF2/HO-1, suggesting that targeting this mechanism of APOC1 may be a feasible strategy for anti-PD-1 immunotherapy for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Ferroptosis , Neoplasias Pulmonares , Humanos , Ratones , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Receptor de Muerte Celular Programada 1 , Apolipoproteína C-I/metabolismo , Apolipoproteína C-I/farmacología , Macrófagos , Hemo-Oxigenasa 1/genética , ARN Mensajero/metabolismo , Inmunoterapia
2.
Neurol Res ; 45(3): 268-275, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36302088

RESUMEN

OBJECTIVE: One of the apolipoprotein's members, apolipoprotein C1 (ApoC1), is critical in the metabolism of both very-low-density lipoprotein (VLDL) and high-density lipoprotein (HDL) cholesterols. Multiple studies have recently revealed that ApoC1 may be a viable therapeutic target in solid malignancies. However, the motor protein ApoC1's specific role and mechanism in glioblastoma remain unknown. METHODS: In this study, the Cancer Genome Atlas (TCGA) database was used to look at the level of ApoC1 in glioma tissues and normal tissues, as well as how it related to the prognosis of glioma. Glioma cell lines (U87 and U251) were subjected to a wide range of experiments to determine the involvement of ApoC1 in cell proliferation, migration, and invasion. RESULTS: Cell proliferation, migration, and invasion decreased in glioma cell lines when ApoC1 was silenced. Furthermore, ApoC1 increased glioma cell metastasis through the epithelial-mesenchymal transition (EMT), while ApoC1 deletion reduced this impact. Additionally, APOC1 influenced the evolution of glioma by affecting the STAT3 pathway. In addition, APOC1 knockdown reduced the activation of the phosphorylated-total signal transducer and activator of transcription (STAT3) in the glioma cells. ApoC1-induced glioma cell metastatic ability was prevented by niclosamide (a STAT3 inhibitor). CONCLUSIONS: These results uncover that ApoC1 may serve as a biomarker or therapeutic target for future fundamental study or clinical treatment of glioma.


Asunto(s)
Glioblastoma , Glioma , Humanos , Transición Epitelial-Mesenquimal/genética , Apolipoproteína C-I/genética , Apolipoproteína C-I/metabolismo , Glioma/patología , Glioblastoma/genética , Línea Celular Tumoral , Factor de Transcripción STAT3/metabolismo , Movimiento Celular , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica
3.
Exp Cell Res ; 422(2): 113452, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36563923

RESUMEN

Zinc finger protein 460 (ZNF460) is closely related to the progression of a variety of human cancers. However, the biological role of ZNF460 in gastric cancer remains fully unrevealed. This study aimed to investigate the role and potential mechanism of ZNF460 in gastric cancer. In this study, we discovered a significant up-regulation of ZNF460 in gastric cancer and that ZNF460 expression correlated with tumor grade, lymph node metastasis, and H. pylon infection in gastric cancer through UALCAN database. Functionally, Diminished ZNF460 expression inhibited gastric cancer cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) in vitro and suppressed tumor growth in vivo. Mechanistically, ZNF460 combined with apolipoprotein C1 (APOC1) promoter to facilitate APOC1 transcription, and accelerated EMT, thereby promoting the progression of gastric cancer. In conclusion, our study confirmed that ZNF460 promotes gastric cancer progression, which might serve as a novel target for gastric cancer treatment.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patología , Transición Epitelial-Mesenquimal/genética , Apolipoproteína C-I/metabolismo , Proliferación Celular/genética , Invasividad Neoplásica/genética , Movimiento Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo
4.
Cardiovasc Diabetol ; 21(1): 272, 2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36471375

RESUMEN

Apolipoprotein C1 (apoC1) is a small size apolipoprotein whose exact role is not totally clarified but which seems to modulate significantly the metabolism of lipoproteins. ApoC1 is involved in the metabolism of triglyceride-rich lipoproteins by inhibiting the binding of very low density lipoproteins (VLDL) to VLDL-receptor (VLDL-R), to low density lipoprotein receptor (LDL-R) and to LDL receptor related protein (LRP), by reducing the activity of lipoprotein lipase (LPL) and by stimulating VLDL production, all these effects leading to increase plasma triglycerides. ApoC1 takes also part in the metabolism of high density lipoproteins (HDL) by inhibiting Cholesterol Ester Transfer Protein (CETP). The functionality of apoC1 on CETP activity is impaired in diabetes that might account, at least in part, for the increased plasma CETP activity observed in patients with diabetes. Its different effects on lipoprotein metabolism with a possible role in the modulation of inflammation makes the net impact of apoC1 on cardiometabolic risk difficult to figure out and apoC1 might be considered as pro-atherogenic or anti-atherogenic depending on the overall metabolic context. Making the link between total plasma apoC1 levels and the risk of cardio-metabolic diseases is difficult due to the high exchangeability of this small protein whose biological effects might depend essentially on its association with VLDL or HDL. The role of apoC1 in humans is not entirely elucidated and further studies are needed to determine its precise role in lipid metabolism and its possible pleiotropic effects on inflammation and vascular wall biology. In this review, we will present data on apoC1 structure and distribution among lipoproteins, on the effects of apoC1 on VLDL metabolism and HDL metabolism and we will discuss the possible links between apoC1, atherosclerosis and diabetes.


Asunto(s)
Apolipoproteína C-I , Aterosclerosis , Diabetes Mellitus , Lipoproteínas HDL , Lipoproteínas VLDL , Humanos , Apolipoproteína C-I/metabolismo , Aterosclerosis/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Diabetes Mellitus/metabolismo , Inflamación/metabolismo , Lipoproteínas HDL/metabolismo , Lipoproteínas VLDL/metabolismo , Triglicéridos
5.
Pharmacol Res ; 183: 106376, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35914680

RESUMEN

Apolipoprotein C1 (APOC1) has been found to play an essential part in proliferation and metastasis of numerous cancers, but related mechanism has not been elucidated, especially its function and role in tumor immunity. Through systematic pan-cancer analysis, we identified that APOC1 was closely associated with the infiltration of various immune cells in multiple cancers. Besides, APOC1 was significantly co-expressed with the immune checkpoints, major histocompatibility complex (MHC) molecules, chemokines and other immune-related genes. Furthermore, single-cell sequencing analysis suggested that the vast majority of APOC1 was expressed in macrophages or tumor-associated macrophages (TAMs). Additionally, the expression of APOC1 was significantly related to the prognosis of different cancers. Since APOC1 was most significantly abnormally expressed in renal cell cancer (RCC), subsequent experiments were carried out in RCC to explore the role of APOC1 in tumor immunity. The expression of APOC1 was significantly elevated in the tumor and serum of RCC patients. Besides, APOC1 was mainly expressed in the macrophage and it was closely related to the immune cell infiltration of RCC. Co-culture with RCC cells could induce the generation of TAMs with M2 phenotype which be blocked by silencing APOC1. The expression of APOC1 was elevated in the M2 or TAMs and APOC1 promoted M2 polarization of macrophages through interacting with CD163 and CD206. Furthermore, macrophages overexpressing APOC1 promoted the metastasis of RCC cells via secreting CCL5. Together, these data indicate that APOC1 is an immunological biomarker which regulates macrophage polarization and promotes tumor metastasis.


Asunto(s)
Apolipoproteína C-I , Carcinoma de Células Renales , Neoplasias Renales , Activación de Macrófagos , Apolipoproteína C-I/genética , Apolipoproteína C-I/metabolismo , Biomarcadores/metabolismo , Carcinoma de Células Renales/metabolismo , Humanos , Neoplasias Renales/metabolismo , Macrófagos/metabolismo , Metástasis de la Neoplasia , Microambiente Tumoral
6.
Acta Pharmacol Sin ; 43(11): 2977-2992, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35581292

RESUMEN

Glioblastoma (GBM), a malignant brain tumor, is a world-wide health problem because of its poor prognosis and high rates of recurrence and mortality. Apolipoprotein C1 (APOC1) is the smallest of apolipoproteins, implicated in many diseases. Recent studies have shown that APOC1 promotes tumorigenesis and development of several types of cancer. In this study we investigated the role of APOC1 in GBM tumorigenesis. Using in silico assays we showed that APOC1 was highly expressed in GBM tissues and its expression was closely related to GBM progression. We showed that APOC1 protein expression was markedly increased in four GBM cell lines (U251, U138, A172 and U87) compared to the normal brain glia cell lines (HEB, HA1800). In U251 cells, overexpression of APOC1 promoted cell proliferation, migration, invasion and colony information, which was reversed by APOC1 knockdown. APOC1 knockdown also markedly inhibited the growth of GBM xenografts in the ventricle of nude mice. We further demonstrated that APOC1 reduced ferroptosis by inhibiting KEAP1, promoting nuclear translocation of NRF2 and increasing expression of HO-1 and NQO1 in GBM cells. APOC1 also induced ferroptosis resistance by increasing cystathionine beta-synthase (CBS) expression, which promoted trans-sulfuration and increased GSH synthesis, ultimately leading to an increase in glutathione peroxidase-4 (GPX4). Thus, APOC1 plays a key role in GBM tumorigenesis, conferring resistance to ferroptosis, and may be a promising therapeutic target for GBM.


Asunto(s)
Apolipoproteína C-I , Ferroptosis , Glioblastoma , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2 , Animales , Humanos , Ratones , Apolipoproteína C-I/metabolismo , Carcinogénesis/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica , Cistationina betasintasa/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones Desnudos , Factor 2 Relacionado con NF-E2/metabolismo
7.
Aging (Albany NY) ; 13(11): 14968-14988, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34081622

RESUMEN

Liver metastasis is a leading cause of death in patients with colorectal cancer (CRC). Increasing evidence demonstrates that competing endogenous RNA (ceRNA) networks play important roles in malignant cancers. The purpose of this study was to identify molecular markers and build a ceRNA network as a significant predictor of colorectal liver metastases (CRLM). By integrated bioinformatics analysis, we found that apolipoprotein C1 (APOC1) was upregulated in CRLM and associated with prognosis in patients with CRC and thereby established an APOC1-dependent ceRNA network. By survival analysis, expression analysis, and correlation analysis of each element in the ceRNA network, we identified that ZEB1-AS1, miR-335-5p and APOC1 regulated each other. We further experimentally confirmed that ZEB1-AS1 promoted a CRC progression via regulating the expression of miR-335-5p that controlled the expression of APOC1. Our findings indicate that the ZEB1-AS1-miR-335-5p-APOC1 ceRNA regulatory network is significantly valuable for better prognosis of patients with CRC and as a new therapeutic target for the treatment of CRLM.


Asunto(s)
Neoplasias Colorrectales/patología , Redes Reguladoras de Genes , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , MicroARNs/genética , ARN Largo no Codificante/genética , Apolipoproteína C-I/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Humanos , MicroARNs/metabolismo , Invasividad Neoplásica , Pronóstico , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia
8.
Med Sci Monit ; 27: e929347, 2021 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-33591959

RESUMEN

BACKGROUND The aims of this study included 3 aspects: 1) assessing the expression of Apolipoprotein C1 (APOC1) in clear cell renal cell carcinoma (ccRCC) and normal groups; 2) evaluating the prognostic significance of APOC1 expression in the overall survival (OS) of ccRCC patients; and 3) exploring APOC1-related signaling pathways. MATERIAL AND METHODS The APOC1 expression value and clinical data of ccRCC patients were obtained from the cBioPortal database. We then evaluated the association of APOC1 expression with clinical characteristics of ccRCC patients. We also assessed the correlation between APOC1 expression and clinical outcome using Kaplan-Meier method. Our work then verified the independent prognostic factors of ccRCC by Cox regression analysis. Finally, the potential role of genes co-expressed with APOC1 was revealed via functional enrichment analysis. RESULTS Bioinformatic data revealed that APOC1 was expressed at higher levels in ccRCC tissue than in the normal group (all P<0.05). The high expression of APOC1 was associated with unfavorable prognosis of female patients (P<0.01), but not of male patients. APOC1 high expression also shortened the survival time of ccRCC patients age ≥60 years old (P<0.05). Cox regression analysis further indicated that APOC1 expression was an independent prognostic factor for OS of ccRCC patients. Additionally, we found that APOC1 expression was significantly associated with sex, grade, clinical stage, and T stage. Finally, enrichment analysis suggested that APOC1-associated pathways were involved in tumor growth and metastasis. CONCLUSIONS The current study indicated that APOC1 was highly expressed in ccRCC and was significantly associated with key clinical features. APOC1 appears to be an independent prognostic factor in patients with ccRCC. Importantly, APOC1 might be a potential therapeutic target for ccRCC via regulating pathways involved in cell growth and metastasis.


Asunto(s)
Apolipoproteína C-I/genética , Carcinoma de Células Renales/genética , Apolipoproteína C-I/metabolismo , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/metabolismo , Biología Computacional/métodos , Bases de Datos Genéticas , Femenino , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Estimación de Kaplan-Meier , Neoplasias Renales/patología , Masculino , Estadificación de Neoplasias , Pronóstico , Transducción de Señal/genética , Transcriptoma/genética
9.
Am J Hum Genet ; 107(4): 714-726, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32961112

RESUMEN

Transcriptome-wide association studies (TWASs) have been widely used to integrate gene expression and genetic data for studying complex traits. Due to the computational burden, existing TWAS methods do not assess distant trans-expression quantitative trait loci (eQTL) that are known to explain important expression variation for most genes. We propose a Bayesian genome-wide TWAS (BGW-TWAS) method that leverages both cis- and trans-eQTL information for a TWAS. Our BGW-TWAS method is based on Bayesian variable selection regression, which not only accounts for cis- and trans-eQTL of the target gene but also enables efficient computation by using summary statistics from standard eQTL analyses. Our simulation studies illustrated that BGW-TWASs achieved higher power compared to existing TWAS methods that do not assess trans-eQTL information. We further applied BWG-TWAS to individual-level GWAS data (N = ∼3.3K), which identified significant associations between the genetically regulated gene expression (GReX) of ZC3H12B and Alzheimer dementia (AD) (p value = 5.42 × 10-13), neurofibrillary tangle density (p value = 1.89 × 10-6), and global measure of AD pathology (p value = 9.59 × 10-7). These associations for ZC3H12B were completely driven by trans-eQTL. Additionally, the GReX of KCTD12 was found to be significantly associated with ß-amyloid (p value = 3.44 × 10-8) which was driven by both cis- and trans-eQTL. Four of the top driven trans-eQTL of ZC3H12B are located within APOC1, a known major risk gene of AD and blood lipids. Additionally, by applying BGW-TWAS with summary-level GWAS data of AD (N = ∼54K), we identified 13 significant genes including known GWAS risk genes HLA-DRB1 and APOC1, as well as ZC3H12B.


Asunto(s)
Enfermedad de Alzheimer/genética , Apolipoproteína C-I/genética , Genoma Humano , Modelos Estadísticos , Proteínas/genética , Sitios de Carácter Cuantitativo , Ribonucleasas/genética , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Apolipoproteína C-I/metabolismo , Teorema de Bayes , Estudios de Casos y Controles , Simulación por Computador , Femenino , Expresión Génica , Marcadores Genéticos , Estudio de Asociación del Genoma Completo , Cadenas HLA-DRB1/genética , Cadenas HLA-DRB1/metabolismo , Humanos , Masculino , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Proteínas/metabolismo , Ribonucleasas/metabolismo , Transcriptoma
10.
Oncogene ; 39(39): 6203-6217, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32826950

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most common renal cancer and frequently diagnosed at an advanced stage. It is prone to develop unpredictable metastases even with proper treatment. Antiangiogenic therapy is the most effective medical treatment for metastatic ccRCC. Thus, exploration of novel approaches to inhibit angiogenesis and metastasis may potentially lead to a better therapeutic option for ccRCC. Among all the types of cancer, renal cancer samples exhibited the maximum upregulation of ApoC1 as referred to in the Oncomine database. The expression of ApoC1 was increased accompanied by ccRCC progression. A high level of ApoC1 was closely related to poor survival time in ccRCC patients. Furthermore, ApoC1 was over-expressed in the highly invasive ccRCC cells as compared to that in the low-invasive ccRCC cells. Besides, ApoC1 promoted metastasis of ccRCC cells via EMT pathway, whereas depletion of ApoC1 alleviated these effects. ApoC1 as a novel pro-metastatic factor facilitates the activation of STAT3 and enhances the metastasis of ccRCC cells. Meanwhile, ApoC1 in the exosomes were transferred from the ccRCC cells to the vascular endothelial cells and promoted metastasis of the ccRCC cells via activating STAT3. Finally, the metastatic potential of the ccRCC cells driven by ApoC1 was suppressed by DPP-4 inhibition. Our study not only identifies a novel ApoC1-STAT3 pathway in ccRCC metastasis but also provides direction for the exploration of novel strategies to predict and treat metastatic ccRCC in the future.


Asunto(s)
Apolipoproteína C-I/metabolismo , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Factor de Transcripción STAT3/metabolismo , Compuestos de Anilina/farmacología , Apolipoproteína C-I/antagonistas & inhibidores , Apolipoproteína C-I/biosíntesis , Apolipoproteína C-I/genética , Compuestos de Bencilideno/farmacología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Metástasis de la Neoplasia , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/genética , Análisis de Supervivencia , Transcripción Genética , Células Tumorales Cultivadas
11.
Int J Mol Sci ; 20(23)2019 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-31779116

RESUMEN

Apolipoprotein C1 (apoC1), the smallest of all apolipoproteins, participates in lipid transport and metabolism. In humans, APOC1 gene is in linkage disequilibrium with APOE gene on chromosome 19, a proximity that spurred its investigation. Apolipoprotein C1 associates with triglyceride-rich lipoproteins and HDL and exchanges between lipoprotein classes. These interactions occur via amphipathic helix motifs, as demonstrated by biophysical studies on the wild-type polypeptide and representative mutants. Apolipoprotein C1 acts on lipoprotein receptors by inhibiting binding mediated by apolipoprotein E, and modulating the activities of several enzymes. Thus, apoC1 downregulates lipoprotein lipase, hepatic lipase, phospholipase A2, cholesterylester transfer protein, and activates lecithin-cholesterol acyl transferase. By controlling the plasma levels of lipids, apoC1 relates directly to cardiovascular physiology, but its activity extends beyond, to inflammation and immunity, sepsis, diabetes, cancer, viral infectivity, and-not last-to cognition. Such correlations were established based on studies using transgenic mice, associated in the recent years with GWAS, transcriptomic and proteomic analyses. The presence of a duplicate gene, pseudogene APOC1P, stimulated evolutionary studies and more recently, the regulatory properties of the corresponding non-coding RNA are steadily emerging. Nonetheless, this prototypical apolipoprotein is still underexplored and deserves further research for understanding its physiology and exploiting its therapeutic potential.


Asunto(s)
Apolipoproteína C-I/química , Apolipoproteína C-I/metabolismo , Lipoproteínas HDL/metabolismo , Lipoproteínas VLDL/metabolismo , Secuencias de Aminoácidos , Apolipoproteína C-I/genética , Apolipoproteínas E/metabolismo , Mapeo Cromosómico , Regulación de la Expresión Génica , Humanos , Metabolismo de los Lípidos , Unión Proteica , Seudogenes , Receptores de Lipoproteína/metabolismo
12.
Med Mol Morphol ; 52(4): 217-225, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31006040

RESUMEN

Mass spectrometric analysis of glioblastoma cyst fluids has disclosed a protein peak with m/z 6424-6433. Among the proteins, potentially generating this peak are ApoC1 and LuzP6. To further elucidate protein expression of glioblastoma cells, we analyzed MALDI-TOF results of cyst fluid, performed immunohistochemistry and mRNA analysis. MALDI-TOF protein extraction from 24 glioblastoma cyst fluids was performed with a weak cation exchange. 50 glioblastoma samples were stained with two custom-made antibodies against LuzP6 and commercial antibodies against ApoC1, C12orf75 and OCC-1 and analyzed. For mRNA detection, 16 tissue samples were stored in RNAlater, extracted using the miRNeasy kit and reversely transcribed. For 12 patients, synopsis of results from all three examinations was possible. MALDI-TOF confirmed the peak at 6433 Da in 75% of samples. Immunohistochemically, LuzP6 was detected in 92% (LuzP61-29) and 96% (LuzP630-58) of samples and ApoC1 in 66%. Mean mRNA levels were highest for ApoC1, followed by LuzP6. No correlation between mRNA expression, immunohistochemical staining and intensity of the MALDI-TOF peaks was found. An unequivocal identification of one protein as the source for the 6433 peak is not possible, but our results point to ApoC1 and LuzP6 as the underlying proteins.


Asunto(s)
Apolipoproteína C-I/genética , Apolipoproteína C-I/metabolismo , Glioblastoma/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Femenino , Humanos , Inmunohistoquímica/métodos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos
13.
Virology ; 524: 1-9, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30130702

RESUMEN

Previous studies have shown that apolipoprotein C1 (apoC1)-specific antibodies precipitated hepatitis C virus (HCV) and neutralized HCV infectivity, suggesting that apoC1 is a HCV component. However, the importance of apoC1 in the HCV life cycle has not been experimentally examined. In the present study, we sought to determine the role of apoC1 in the HCV infection and morphogenesis by knocking out the apoC1 gene using the CRISPR/Cas9 system. Strikingly, apoC1 gene knockout markedly enhanced apoE expression. As a result, apoC1 gene knockout per se didn't significantly affect HCV infection or morphogenesis, probably ascribing to its redundant functions with apoE. However, knockout of apoC1 gene potentiated the impairment of HCV infection and/or morphogenesis by apoE-specific small interfering RNAs. Additionally, a recombinant apoC1 protein efficiently blocked HCV infection. Collectively, these findings suggest that apoC1 and apoE have redundant functions in the HCV infection and morphogenesis.


Asunto(s)
Apolipoproteína C-I/metabolismo , Apolipoproteínas E/metabolismo , Hepacivirus/fisiología , Hepatitis C/virología , Apolipoproteína C-I/genética , Apolipoproteínas E/genética , Línea Celular , Técnicas de Inactivación de Genes , Hepacivirus/genética , Hepacivirus/crecimiento & desarrollo , Hepacivirus/patogenicidad , Humanos , Morfogénesis , ARN Interferente Pequeño , Proteínas Recombinantes
14.
Leukemia ; 32(7): 1587-1597, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29467488

RESUMEN

Deregulation of key regulators of histone modification is important in the initiation and progression of human leukemia. Acidic leucine-rich nuclear phosphoprotein-32A (ANP32A) participates in histone acetylation and its role in acute myeloid leukemia remains unclear. Here we observed significant upregulation of ANP32A in primary AML cells, which was essential for AML cell proliferation, survival, and colony formation. Integrative analysis of the genome-wide histone H3 acetylation and gene expression demonstrated that ANP32A deficiency reduced histone H3 acetylation, in accordance with changes in gene expression. Notably, significant histone H3 acetylation enrichment was associated with mRNA changes in lipid-related genes, including APOC1, PCSK9, P2RX1, and LPPR3. Indeed, over-expression of APOC1 partially compensated the proliferation-defect phenotype in ANP32A deficient AML cells while APOC1 knockdown alone mimicked the effect of ANP32A deficiency. Collectively, our data indicate that ANP32A is a novel regulator of histone H3 acetylation and promotes leukemogenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Acetilación , Animales , Apolipoproteína C-I/metabolismo , Apoptosis , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Regulación Leucémica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Metabolismo de los Lípidos/genética , Ratones , Proteínas Nucleares , ARN Mensajero/genética , Proteínas de Unión al ARN , Ensayo de Tumor de Célula Madre
15.
Int J Dev Biol ; 61(6-7): 415-425, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28695961

RESUMEN

In vertebrates, the neural crest and placodes originate in the neural border, which is located between the neural plate and epidermal ectoderm. The neural crest and placodes give rise to a vast array of cell types. Formation of neural crest is a multi-step process, in which Wnt signals are used reiteratively, but it is currently not clear if a Wnt signal is required for neural border formation. Here, we have identified apolipoprotein C-I (apoc1) in a screen for genes regulated by Wnt/Ctnnb1 signaling in late blastula stage Xenopus tropicalis embryos. We show that Xenopus laevis apoc1 encodes a small, secreted protein, and is induced by Wnt/Ctnnb1 signaling. Depletion of Apoc1 protein results in a neural border formation defect and loss of border fates, including neural crest cells. However, unlike another Wnt/Ctnnb1 target, gbx2.2, apoc1 is not required for patterning of the neural border. We further show that gbx2.2 and apoc1 are independently regulated by Wnt signaling. Our results thus suggest that Wnt regulates border formation and patterning by distinct genetic mechanisms.


Asunto(s)
Apolipoproteína C-I/metabolismo , Embrión no Mamífero/citología , Cresta Neural/citología , Neurogénesis/fisiología , Proteínas Wnt/metabolismo , Xenopus laevis/crecimiento & desarrollo , beta Catenina/metabolismo , Animales , Apolipoproteína C-I/genética , Tipificación del Cuerpo , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula , Embrión no Mamífero/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/metabolismo , Transducción de Señal , Proteínas Wnt/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo , beta Catenina/genética
16.
Oncol Rep ; 38(3): 1797-1805, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28713944

RESUMEN

Truncated apolipoprotein C-I is a post-translationally modified protein characterized by the loss of threonine and proline residues from the N-terminus of the mature peptide. The truncated peptide is involved in many physiological and pathological processes in vivo and is related to malignant diseases. The aim of the present study was to assess the effects of the truncated peptide on tumorigenesis in neuroblastoma. The truncated peptide was chemically synthesized, and a signal peptide was used as the negative control. The results of the CCK-8 assay showed that the truncated peptide selectively inhibited cell proliferation compared with the signal peptide, and inhibited migration and invasion as determined by wound healing and Transwell assays. Flow cytometry analysis demonstrated that the truncated peptide induced apoptosis and cell cycle arrest in the S phase. Bax, Bim, and tBid upregulation, and Bcl­2 and Bcl­xl downregulation were associated with permeabilization of the mitochondrial membrane, as detected by the JC-1 assay and the release of cytochrome c and apoptosis. Activation of caspase­8 was associated with activation of cell death receptors such as the tumor necrosis factor receptor. PARP cleavage indicated apoptosis, and DNA damage was observed in the TUNEL assay. The results showed that the truncated apoC-I induced apoptosis in neuroblastoma by the extrinsic and intrinsic pathways. The anticancer effects were confirmed in vivo in a xenograft mouse model. In conclusion, the endogenous protein apoC-I may be a new promising therapeutic target to suppress tumor growth.


Asunto(s)
Apolipoproteína C-I/metabolismo , Apoptosis/fisiología , Caspasas/metabolismo , Neuroblastoma/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Puntos de Control del Ciclo Celular/fisiología , Muerte Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Citocromos c/metabolismo , Regulación hacia Abajo/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/metabolismo , Mitocondrias/patología , Neuroblastoma/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Fase S/fisiología , Transducción de Señal/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Regulación hacia Arriba/fisiología
17.
J Pathol ; 241(5): 589-599, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27976371

RESUMEN

Diabetic nephropathy is the leading cause of end-stage renal disease. Diabetic patients have increased plasma concentrations of apolipoprotein C-I (apoCI), and meta-analyses found that a polymorphism in APOC1 is associated with an increased risk of developing nephropathy. To investigate whether overexpressing apoCI contributes to the development of kidney damage, we studied renal tissue and peritoneal macrophages from APOC1 transgenic (APOC1-tg) mice and wild-type littermates. In addition, we examined renal material from autopsied diabetic patients with and without diabetic nephropathy and from autopsied control subjects. We found that APOC1-tg mice, but not wild-type mice, develop albuminuria, renal dysfunction, and glomerulosclerosis with increased numbers of glomerular M1 macrophages. Moreover, compared to wild-type macrophages, stimulated macrophages isolated from APOC1-tg mice have increased cytokine expression, including TNF-alpha and TGF-beta, both of which are known to increase the production of extracellular matrix proteins in mesangial cells. These results suggest that APOC1 expression induces glomerulosclerosis, potentially by increasing the cytokine response in macrophages. Furthermore, we detected apoCI in the kidneys of diabetic patients, but not in control kidneys. Moreover, patients with diabetic nephropathy have significantly more apoCI present in glomeruli compared to diabetic patients without nephropathy, suggesting that apoCI could be involved in the development of diabetic nephropathy. ApoCI co-localized with macrophages. Therefore, apoCI is a promising new therapeutic target for patients at risk of developing nephropathy. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Apolipoproteína C-I/metabolismo , Nefropatías Diabéticas/etiología , Regulación de la Expresión Génica , Fallo Renal Crónico/etiología , Anciano , Albuminuria/etiología , Albuminuria/patología , Animales , Apolipoproteína C-I/genética , Encéfalo/metabolismo , Encéfalo/patología , Citocinas/metabolismo , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Femenino , Humanos , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/patología , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Pulmón/metabolismo , Pulmón/patología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/metabolismo , Miocardio/patología , Páncreas/metabolismo , Páncreas/patología , Bazo/metabolismo , Bazo/patología
18.
PLoS One ; 11(7): e0158611, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27416065

RESUMEN

BACKGROUND: Coats' disease is an uncommon form of retinal telangiectasis, and the identification of novel proteins that contribute to the development of Coats' disease is useful for improving treatment efficacy. Proteomic techniques have been used to study many eye diseases; however, few studies have used proteomics to study the development of Coats' disease. METHODS: Isobaric tagging for relative and absolute protein quantification (iTRAQ) was employed to screen differentially expressed proteins (DEPs) in the aqueous humor (AH) between stage 3A patients (n = 8), stage 3B patients (n = 14), stage 4 patients (n = 2) and control patients (n = 20). Differentially co-expressed proteins (DCPs) were present in all three stages of Coats' disease and were considered disease-specific proteins. These proteins were further analyzed using Gene Ontology (GO) functional annotations. RESULTS: A total of 819 proteins were identified in the AH, 222 of which were significantly differentially expressed (fold change > 2 and P < 0.05) in the samples from at least one stage of Coats' disease. Of the DEPs, 46 were found among all three stages of Coats' disease and the controls; therefore, they were considered Coats' disease-specific proteins (DCPs). A GO classification analysis indicated that the DCPs were closely related to structural molecule activity, cell adhesion molecule binding and receptor binding. Western blotting confirmed the expression levels of haptoglobin and apolipoprotein C-I were significantly up-regulated in Coats' disease. CONCLUSIONS: The 46 Coats' disease-specific proteins may provide additional insights into the mechanism of Coats' disease and represent potential biomarkers for identifying individuals with Coats' disease.


Asunto(s)
Apolipoproteína C-I/metabolismo , Humor Acuoso/metabolismo , Haptoglobinas/metabolismo , Telangiectasia Retiniana/metabolismo , Adolescente , Anciano , Niño , Preescolar , Progresión de la Enfermedad , Humanos , Persona de Mediana Edad , Proteómica , Vasos Retinianos/metabolismo
19.
Med Sci Monit ; 22: 1152-60, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27052600

RESUMEN

BACKGROUND: Breast cancer (BC) is one of the most common cancers and is among the main causes of death in females around the world. Although several serum biomarkers have been identified for breast cancer, due to lack of adequate sensitivity and specificity they do not adequately distinguish BC from confounding conditions. New approaches are urgently needed to improve BC detection and treatment. MATERIAL/METHODS: Eighty serum samples from 20 healthy individuals and 60 patients with BC (22 triple-negative breast cancer, TNBC; 38 non-triple-negative breast cancer, NTNBC) were included. Protein profiling of serum samples was analyzed using surface-enhanced laser desorption/ionization time-of-flight mass spectroscopy (SELDI-TOF-MS). Candidate biomarkers were purified by SDS-PAGE electrophoresis and identified by MALDI-TOF/TOF. RESULTS: The candidate biomarker positioned at 6447.9 m/z was significantly decreased in BC patients. Moreover, the expression intensity of the candidate biomarker was weaker in the TNBC and pre-surgery group compared with the NTNBC and post-surgery group. We ultimately identified the biomarker as apolipoprotein C-I (ApoC-I). Furthermore, we found that ApoC-I peptides inhibited proliferation of human breast cancer cells in vitro and suppressed tumor growth in vivo. CONCLUSIONS: These results suggest that ApoC-I peptides may be a potential diagnostic biomarker and therapeutic approach for BC.


Asunto(s)
Apolipoproteína C-I/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Péptidos/metabolismo , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Apolipoproteína C-I/química , Apoptosis/efectos de los fármacos , Proteínas Sanguíneas/metabolismo , Neoplasias de la Mama/cirugía , Estudios de Casos y Controles , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Ratones Desnudos , Persona de Mediana Edad , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Adulto Joven
20.
Methods ; 81: 86-92, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25752847

RESUMEN

The impetus for discovery and evaluation of protein biomarkers has been accelerated by recent development of advanced technologies for rapid and broad proteome analyses. Mass spectrometry (MS)-based protein assays hold great potential for in vitro biomarker studies. Described here is the development of a multiplex mass spectrometric immunoassay (MSIA) for quantification of apolipoprotein C-I (apoC-I), apolipoprotein C-II (apoC-II), apolipoprotein C-III (apoC-III) and their proteoforms. The multiplex MSIA assay was fast (∼ 40 min) and high-throughput (96 samples at a time). The assay was applied to a small cohort of human plasma samples, revealing the existence of multiple proteoforms for each apolipoprotein C. The quantitative aspect of the assay enabled determination of the concentration for each proteoform individually. Low-abundance proteoforms, such as fucosylated apoC-III, were detected in less than 20% of the samples. The distribution of apoC-III proteoforms varied among samples with similar total apoC-III concentrations. The multiplex analysis of the three apolipoproteins C and their proteoforms using quantitative MSIA represents a significant step forward toward better understanding of their physiological roles in health and disease.


Asunto(s)
Apolipoproteína C-III/sangre , Apolipoproteína C-II/sangre , Apolipoproteína C-I/sangre , Inmunoensayo/métodos , Espectrometría de Masas/métodos , Apolipoproteína C-I/metabolismo , Apolipoproteína C-II/metabolismo , Apolipoproteína C-III/metabolismo , Humanos , Isoformas de Proteínas/sangre , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...