Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
J Clin Lab Anal ; 35(12): e24003, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34676904

RESUMEN

BACKGROUND: Circular RNA (circRNA) affects the occurrence and development of human cancers, but the specific mechanism of hepatocellular carcinoma (HCC) has not yet been fully understood. METHODS: CircRNAs were determined by human circRNA array analysis and quantitative reverse transcription polymerase chain reaction (qRT-PCR). Cell viability, migration, invasion, and other indicators were used for cell function analysis. Knockdown and overexpression techniques were used to explore the mechanism of circCORO1C in the occurrence and development of HCC by RNA sequencing, qRT-PCR, western blot, and other methods. RESULTS: Among the thousands of circRNAs, 1238 circRNAs were significantly changed. As for the top 10 upregulated circRNAs, the expression of circRNAs, hsa_circ_0036412, hsa_circ_0036411, hsa_circ_0028071, hsa_circ_0036409, hsa_circ_0000437, hsa_circ_0021427, hsa_circ_0097182, hsa_circ_0028067, hsa_circ_0006852, and hsa_circ_0003620 were significantly increased. In regard to the top 10 downregulated circRNAs, the expression of hsa_circ_0123629, hsa_circ_0096121, hsa_circ_0038932, hsa-circRNA3310-44, hsa_circ_0045746, hsa_circ_0016836, hsa-circRNA10899-9, hsa_circ_0050116, hsa_circ_0035543, and hsa_circ_0092118 decreased significantly. About these circRNAs, the downregulation of hsa_circ_0006852 (circCORO1C) can inhibit the tumorigenesis of HCC cells in vivo and in vitro, and the overexpression of circCORO1C can enhance the proliferation and metastasis ability of HCC cells. Mechanistically, circCORO1C activated the NF-κB signaling pathway, increased P65 phosphorylation and upregulation of c-Myc and COX-2, leading to increased PD-L1 expression. CONCLUSION: CircCORO1C upregulates c-Myc and COX-2 through NF-κB signaling pathway, leading to the upregulation of PD-L1, which jointly promotes the development of HCC, suggesting that circCORO1C is a promising biomarker and therapeutic target for HCC.


Asunto(s)
Antígeno B7-H1/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , FN-kappa B/genética , ARN Circular/genética , Animales , Antígeno B7-H1/metabolismo , Carcinoma Hepatocelular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Masculino , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Análisis de Secuencia de ARN , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Cell Physiol ; 235(3): 2273-2285, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31595500

RESUMEN

An in-depth understanding of circular RNAs (circRNAs) indicates that they are abundant in the eukaryotic transcriptome. Many circRNAs act as microRNA sponges; thus, they represent a new type of regulatory factor. However, the role of circRNA in colorectal cancer (CRC) remains largely unknown. Low circ_0021977 expression in patients with CRC is associated with higher tug-lymph node metastasis (TNM) stage and poorer prognosis compared with patients with high circ_0021977 expression. Moreover, miR-10b-5p was shown to be a target of circ_0021977, and p21 and p53 are suggested to be putative target genes of miR-10b-5p. The results showed that the circ_0021977/miR-10b-5p/p21&p53 regulatory axis suppresses proliferation, migration, and invasion by CRC cells. This evidence reveals new relationships and brings new highlights to the treatment of CRC.

3.
Mol Cancer ; 19(1): 140, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32907590

RESUMEN

An amendment to this paper has been published and can be accessed via the original article.

4.
Mol Cancer ; 18(1): 25, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30777076

RESUMEN

BACKGROUND: Circular RNAs (circRNAs) are a class of non-coding RNAs with a loop structure, but its functions remain largely unknown. Growing evidence has revealed that circRNAs play a striking role as functional RNAs in the progression of malignant disease. However, the precise role of circRNAs in gastric cancer (GC) remains unclear. METHODS: CircRNAs were determined by human circRNA array analysis and quantitative reverse transcription polymerase reaction. Luciferase reporter, RNA pull down, and fluorescence in situ hybridization assays were employed to test the interaction between circPSMC3 and miR-296-5p. Ectopic over-expression and siRNA-mediated knockdown of circPSMC3, proliferation, migration and invasion in vitro, and in vivo experiment of metastasis were used to evaluate the function of circPSMC3. RESULTS: CircPSMC3 rather than liner PSMC3 mRNA was down-regulated in GC tissues, corresponding plasmas from GC patients as well as GC cell lines compared to normal controls. Lower circPSMC3 expression in GC patients was correlated with higher TNM stage and shorter overall survival. Over-expression of circPSMC3 and miR-296-5p inhibitor could inhibit the tumorigenesis of gastric cancer cells in vivo and vitro whereas co-transfection of circPSMC3 and miRNA-296-5p could counteract this effect. Importantly, we demonstrated that circPSMC3 could act as a sponge of miR-296-5p to regulate the expression of Phosphatase and Tensin Homolog (PTEN), and further suppress the tumorigenesis of gastric cancer cells. CONCLUSION: Our study reveals that circPSMC3 can serve as a novel potential circulating biomarker for detection of GC. CircPSMC3 participates in progression of gastric cancer by sponging miRNA-296-5p with PTEN, providing a new insight into the treatment of gastric cancer.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Adenocarcinoma/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Complejo de la Endopetidasa Proteasomal/genética , ARN/genética , Neoplasias Gástricas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Anciano , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Xenoinjertos , Humanos , Metástasis Linfática , Masculino , Ratones , MicroARNs/metabolismo , Persona de Mediana Edad , Estadificación de Neoplasias , Fosfohidrolasa PTEN/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN/antagonistas & inhibidores , ARN/metabolismo , ARN Circular , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Análisis de Supervivencia
6.
Mol Cancer ; 17(1): 137, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30236115

RESUMEN

CircRNA expression profiles for gastric cancer (GC) were screened using plasma samples from 10 GC patients with different TNM stages and 5 healthy individuals as controls. Results showed lower expression of circ-KIAA1244 in GC tissues, plasmas, and cells compare to normal controls. Further clinical data analysis demonstrated that a decreased expression of circ-KIAA1244 in plasmas was negatively correlated with TNM stage and lymphatic metastasis, and a shorter overall survival time of GC patients. Moreover, we found that circ-KIAA1244 could be detected in GC plasma exosomes and showed no obvious significance compared to the expression level in the corresponding plasmas. This study revealed a GC-tissues-derived circ-KIAA1244 could serve a novel circulating biomarker for detection of GC.


Asunto(s)
Biomarcadores de Tumor , Ácidos Nucleicos Libres de Células , ARN no Traducido/genética , ARN/genética , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética , Estudios de Casos y Controles , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Estadificación de Neoplasias , Pronóstico , ARN Circular , Neoplasias Gástricas/sangre , Neoplasias Gástricas/mortalidad
7.
Cell Physiol Biochem ; 35(2): 571-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25612651

RESUMEN

BACKGROUND: A proliferation-inducing ligand (APRIL) is a tumor-necrosis factor (TNF) family member and is a novel cytokine crucial in sustaining lymphocytic leukemia B cell survival and proliferation. However, its role in gastric cancer (GC) remains unclear. In this study, we investigated the expression pattern and prognostic role of APRIL in GC. METHODS: Expression of APRIL was assessed by immunohistochemistry and real-time PCR. Prognostic role of APRIL expression was evaluated. We also discovered the effect of APRIL on chemo-resistance in GC cells and the underlying mechanisms. RESULTS: APRIL mRNA levels were significantly increased in GC tissues compared with adjacent tissues and high expression levels of APRIL in tumor cells significantly correlated with poor overall survival in patients receiving cisplatin adjuvant treatment. Overexpression of APRIL in AGS cells significantly attenuated the therapeutic efficacy of cisplatin in vitro and in vivo. In contrast, silence of APRIL in SGC7901 cells enhanced cisplatin-induced tumor suppression. Our data further revealed that the canonical NF-κB pathway was involved in APRIL-mediated chemo-resistance. In addition, expression of APRIL was regulated by miR-145 in GC cells. CONCLUSION: APRIL is a novel clinical chemo-resistance biomarker for gastric cancer and might be a promising therapeutic target for GC patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/genética , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/tratamiento farmacológico , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Adulto , Anciano , Línea Celular Tumoral , Femenino , Silenciador del Gen , Humanos , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Resultado del Tratamiento , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
8.
BMC Cancer ; 14: 699, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25248985

RESUMEN

BACKGROUND: Aptamers have emerged as excellent molecular probes for cancer diagnosis and therapy. The aim of the current study was to determine the feasibility of using DNA aptamer cy-apt 20 developed by live cell-SELEX for detecting and targeting gastric cancer. METHODS: The specificity, sensitivity and biostability of cy-apt 20 in detecting gastric cancer were assessed by binding assay, cell fluorescence imaging, and in vivo tumor imaging in animal model in comparison with non-gastric cancers. RESULTS: Flow cytometric analysis showed that cy-apt 20 had higher than 78% of maximal binding rate to gastric cancer cells, much higher than that of non-gastric cancer cells. Cell fluorescence imaging and in vivo tumor imaging showed that the targeting recognition could be visualized by using minimal dose of fluorochrome labeled cy-apt 20. Meanwhile, strong fluorescence signals were detected and lasted for a period of time longer than 50 min in vitro and 240 min in vivo. The fluorescence intensities of gastric cancer were about seven folds in vitro and five folds of that of non-gastric cancers in vivo. CONCLUSION: Our study demonstrated that cy-apt 20 was an excellent molecular probe with high specificity and sensitivity and a certain degree of biostability for molecular recognition and targeting therapy of gastric cancer.


Asunto(s)
Aptámeros de Nucleótidos/uso terapéutico , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/terapia , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Imagen Molecular , Terapia Molecular Dirigida , Técnica SELEX de Producción de Aptámeros , Sensibilidad y Especificidad , Ensayos Antitumor por Modelo de Xenoinjerto
9.
World J Surg Oncol ; 12: 212, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25022995

RESUMEN

BACKGROUND: Recent research displays that breast cancer (BC) is a heterogeneous disease and distinct molecular subtypes yield different prognostic outcomes. METHODS: We conducted a meta-analysis to clarify the role of molecular subtypes in recurrence risk after breast-conserving therapy (BCT). Eligible studies of single- (ER, PR, Her-2, and p53) and triple-molecular (Luminal A, Luminal B, Her-2, triple-negative) subtypes were identified through multiple search strategies. Pooled hazard ratios with 95% confidence intervals were calculated to assess this research topic. RESULTS: Fifteen studies involving 21,645 participants were included in the meta-analysis. Her-2 positive patients had a significantly higher recurrence risk in both overall merge (HR = 1.97, 95% CI: 1.41-2.75) and subtotal merge of local recurrence (LR) (HR = 1.93, 95% CI: 1.34-2.78). Significantly higher risk of recurrence was also observed in p53 positive patients by overall merge (HR = 1.78, 95% CI: 1.49 -2.12) and subtotal merge of LR (HR = 1.73, 95% CI: 1.44-2.07). When setting Luminal A as a baseline, Luminal B, Her-2, and triple-negative all showed significantly increased risk for both LR and distant recurrence (DR). Comparing triple-negative and non-triple-negative subtypes showed the biggest risk for overall recurrence (HR = 3.19, 95% CI: 1.91-5.31) and LR (HR = 3.31, 95% CI: 1.69-6.45). CONCLUSIONS: Our meta-analysis showed significant differences in recurrence risk among various molecular subtypes after BCT. Although Her-2 and p53 positive subtypes can be considered independent prognostic biomarkers for indicating high LR risk, triple-molecular biomarkers showed higher clinical value. Triple-negative subtype showed the highest recurrence risk among all subtypes, and adjuvant chemotherapy should be considered for it.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/patología , Mastectomía Segmentaria/efectos adversos , Recurrencia Local de Neoplasia/diagnóstico , Complicaciones Posoperatorias/diagnóstico , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/cirugía , Femenino , Humanos , Recurrencia Local de Neoplasia/etiología , Recurrencia Local de Neoplasia/metabolismo , Estadificación de Neoplasias , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/metabolismo , Pronóstico , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo
10.
Surgeon ; 12(2): 94-105, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24321839

RESUMEN

BACKGROUND: The aim of this article is to explore the clinical effects between open extraperitoneal approaches and totally extraperitoneal laparoscopic hernioplasty (TEP) in the repair of inguinal hernias. METHODS: The electronic databases Pubmed, Medline, Embase, Web of science and the Cochrane Library were used to search for articles from January 1992 to March 2013. The present meta-analysis pooled the effects of outcomes of a total of 1157 patients with 1377 hernias enrolled into 10 randomized controlled trials and 2 comparative studies. The data was analyzed using the statistic software Stata12.0 and IBM SPSS Statistics 19. RESULTS: Significant advantages of totally extraperitoneal laparoscopic hernioplasty (TEP) compared to the open extraperitoneal approach include a lower incidence of total postoperative complications (Odds Ratio, 0.544; 95% confidence interval, 0.369-0.803), a reduction in urinary problems (0.206[0.064,0.665]), an earlier return to normal activities or work (SMD = -1.798[-3.322,-0.275]), and a shorter length of hospital stay (-1.995 [-2.358,-1.632]). No difference was found in operative time, the incidence of hernia recurrence, chronic pain, intraoperative complications, seromas or hematomas, wound infection and testicular problems between the two techniques. One significant advantage for the open extraperitoneal inguinal hernia repair was a lower incidence of peritoneal tears (46.504 [15.399,140.437]). CONCLUSIONS: Totally extraperitoneal laparoscopic hernioplasty (TEP) and open extraperitoneal mesh repair are equivalent in most of the analyzed outcomes. TEP is associated with shorter hospital stay, quicker return to normal activities or work, lower incidence of total postoperative complications and urinary problems, while the open extraperitoneal method has less incidence of peritoneal tears.


Asunto(s)
Hernia Inguinal/cirugía , Herniorrafia/métodos , Laparoscopía/métodos , Laparotomía/métodos , Mallas Quirúrgicas , Humanos , Peritoneo , Resultado del Tratamiento
11.
Cell Death Discov ; 10(1): 241, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762481

RESUMEN

Programmed cell death-ligand 1 (PD-L1) has a significant role in tumor progression and metastasis, facilitating tumor cell evasion from immune surveillance. PD-L1 can be detected in the tumor cell nucleus and exert an oncogenic effect by nuclear translocation. Colorectal cancer (CRC) progression and liver metastasis (CCLM) are among the most lethal diseases worldwide, but the mechanism of PD-L1 nuclear translocation in CRC and CCLM remains to be fully understood. In this study, using CRISPR-Cas9-based genome-wide screening combined with RNA-seq, we found that the oncogenic factor NUP43 impacted the process of PD-L1 nuclear translocation by regulating the expression level of the PD-L1 chaperone protein IPO5. Subsequent investigation revealed that this process could stimulate the expression of tumor-promoting factor TM4SF1 and further activate the JAK/STAT3 signaling pathway, which ultimately enhanced the transcription of PD-L1, thus establishing a PD-L1-nPD-L1-PD-L1 feedback loop that ultimately promoted CRC progression and CCLM. In conclusion, our study reveals a novel role for nPD-L1 in CRC, identifies the PD-L1-nPD-L1-PD-L1 feedback loop in CRC, and provides a therapeutic strategy for CRC patients.

12.
Cell Death Dis ; 15(2): 107, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38302412

RESUMEN

Programmed cell death 1 ligand 1 (PDL1)/programmed cell death 1 (PD1) blockade immunotherapy provides a prospective strategy for the treatment of colorectal cancer (CRC), but various constraints on the effectiveness of the treatment are still remaining. As reported in previous studies, follistatin-like 3 (FSTL3) could mediate inflammatory response in macrophages by induction lipid accumulation. Herein, we revealed that FSTL3 were overexpressed in malignant cells in the CRC microenvironment, notably, the expression level of FSTL3 was related to tumor immune evasion and the clinical efficacy of anti-PD1 therapy. Further studies determined that hypoxic tumor microenvironment induced the FSTL3 expression via HIF1α in CRC cells, FSTL3 could bind to the transcription factor c-Myc (354-406 amino acids) to suppress the latter's ubiquitination and increase its stability, thereby to up-regulated the expression of PDL1 and indoleamine 2,3-dioxygenase 1 (IDO1). The results in the immunocompetent tumor models verified that FSLT3 knockout in tumor cells increased the proportion of CD8+ T cells in the tumor microenvironment, reduced the proportion of regulatory T cells (CD25+ Foxp3+) and exhausted T cells (PD1+ CD8+), and synergistically improved the anti-PD1 therapy efficacy. To sum up, FSTL3 enhanced c-Myc-mediated transcriptional regulation to promote immune evasion and attenuates response to anti-PD1 therapy in CRC, suggesting the potential of FSTL3 as a biomarker of immunotherapeutic efficacy as well as a novel immunotherapeutic target in CRC.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias Colorrectales , Humanos , Escape del Tumor , Inmunoterapia/métodos , Linfocitos T Reguladores , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Microambiente Tumoral
13.
Tumour Biol ; 34(4): 2309-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23568706

RESUMEN

CIP2A has been regarded as a novel potential therapeutic target for multiple cancers. The aim of this study was to detect CIP2A expression in pancreatic ductal adenocarcinoma (PDA) and to analyze its association with prognosis of PDA patients. The expression of CIP2A and three epithelial-mesenchymal transition (EMT)-related proteins (E-cadherin, N-cadherin, and vimentin) was examined in 96 PDA tissue samples by immunohistochemistry. Fifty-four cases (56.3 %) were defined as positive for CIP2A expression. Immunohistochemistry showed that CIP2A expression was correlated with poor tumor differentiation, TNM stage, and lymph node metastasis. Kaplan-Meier survival analysis showed that patients with CIP2A-positive expression showed lower overall survival rate than those with CIP2A-negative expression. Multivariate analysis showed that CIP2A expression was an independent prognostic factor for PDA patients. Furthermore, positive expression of CIP2A was strongly associated with loss of the epithelial marker E-cadherin and acquisition of the expression of the mesenchymal markers N-cadherin and vimentin. These findings suggest that CIP2A might promote EMT and progression in PDA, and thus may be a potential therapeutic target for patients with PDA.


Asunto(s)
Autoantígenos/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/mortalidad , Transición Epitelial-Mesenquimal , Proteínas de la Membrana/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Autoantígenos/biosíntesis , Biomarcadores de Tumor/metabolismo , Cadherinas/biosíntesis , Carcinoma Ductal Pancreático/patología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Metástasis Linfática , Masculino , Proteínas de la Membrana/biosíntesis , Persona de Mediana Edad , Estadificación de Neoplasias , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/patología , Pronóstico , Sobrevida , Tasa de Supervivencia , Vimentina/biosíntesis
14.
J Oncol ; 2023: 2611105, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36908705

RESUMEN

Background: Approximately 10% of cancer patients worldwide have colorectal cancer (CRC), a prevalent gastrointestinal malignancy with substantial mortality and morbidity. The purpose of this work was to investigate the APOC1 gene's expression patterns in the CRC tumor microenvironment and, using the findings from bioinformatics, to assess the biological function of APOC1 in the development of CRC. Methods: The TCGA portal was employed in this investigation to find APOC1 expression in CRC. Its correlation with other genes and clinicopathological data was examined using the UALCAN database. To validate APOC1's cellular location, the Human Protein was employed. In order to forecast the relationship between APOC1 expression and prognosis in CRC patients, the Kaplan-Meier plotter database was used. TISIDB was also employed to evaluate the connection between immune responses and APOC1 expression in CRC. The interactions of APOC1 with other proteins were predicted using STRING. In order to understand the factors that contribute to liver metastasis from CRC, single-cell RNA sequencing (scRNA-seq) was done on one patient who had the disease. This procedure included sampling preoperative blood and the main colorectal cancer tissues, surrounding colorectal cancer normal tissues, liver metastatic cancer tissues, and normal liver tissues. Finally, an in vitro knockdown method was used to assess how APOC1 expression in tumor-associated macrophages (TAMs) affected CRC cancer cell growth and migration. Results: When compared to paracancerous tissues, APOC1 expression was considerably higher in CRC tissues. The clinicopathological stage and the prognosis of CRC patients had a positive correlation with APOC1 upregulation and a negative correlation, respectively. APOC1 proteins are mostly found in cell cytosols where they may interact with APOE, RAB42, and TREM2. APOC1 was also discovered to have a substantial relationship with immunoinhibitors (CD274, IDO1, and IL10) and immunostimulators (PVR, CD86, and ICOS). According to the results of scRNA-seq, we found that TAMs of CRC tissues had considerably more APOC1 than other cell groups. The proliferation and migration of CRC cells were impeded in vitro by APOC1 knockdown in TAMs. Conclusion: Based on scRNA-seq research, the current study shows that APOC1 was overexpressed in TAMs from CRC tissues. By inhibiting APOC1 in TAMs, CRC progression was reduced in vitro, offering a new tactic and giving CRC patients fresh hope.

15.
Biomed Pharmacother ; 158: 114127, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36516696

RESUMEN

Apolipoprotein E (ApoE) is a multifunctional protein involved in lipid transport and lipoprotein metabolism, mediating lipid distribution/redistribution in tissues and cells. It can also regulate inflammation and immune function, maintain cytoskeleton stability, and improve neural tissue Function. Due to genetic polymorphisms of ApoE (ε2, ε3, and ε4), its three common structural isoforms (ApoE2, ApoE3, ApoE4) are also associated with the risk of many diseases, especially degenerative diseases, such as vascular degenerative diseases including atherosclerosis (AS), coronary heart disease (CHD), and neurodegenerative disease like Alzheimer's disease (AD). The frequency of the ε4 allele and APOE variants were significantly higher than that of the ε2 and ε3 alleles in the patients with CHD or AD. In recent years, ApoE has frequently appeared in tumor research and become a tumor biomarker gradually. It has been found that ApoE is highly expressed in most solid tumor tissues, such as glioblastoma, gastric cancer, pancreatic ductal cell carcinoma, etc. Studies illustrated that ApoE could regulate the polarization changes of macrophages, participate in the construction of tumor immune microenvironment, regulate tumor inflammation and immune response and play a role in tumor progression, invasion, and metastasis. Of course, many functions of ApoE and its relationship with diseases are still under research. By reviewing the structure and function of ApoE from degeneration diseases to tumor neoplasms, we hope to better understand such a biomarker and further explore the value of ApoE in later studies.


Asunto(s)
Enfermedad de Alzheimer , Neoplasias , Enfermedades Neurodegenerativas , Humanos , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteína E4/genética , Neoplasias/genética , Inflamación/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Genotipo , Microambiente Tumoral/genética
16.
Bioengineered ; 13(4): 8277-8290, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35302432

RESUMEN

Gastric cancer (GC) is one of the most common malignancies in the world, and effective therapeutic targets need to be identified for this type of cancer. In this study, circular RNA (circRNA) microarray analysis was utilized to screen differentially expressed circRNA in GC. Using quantitative reverse transcription polymerase chain reaction (qRT-PCR), hsa_circ_0000081 (circRNA-0000081) expression was found to be up-regulated in tissues and cells and was negative correlated with patients' survival time. RNase R and Actinomycin D assays indicated that circRNA-0000081 was significantly more resistant to R enzyme and had a longer half-life than linear RNA. Moreover, the knockdown or overexpression of circRNA-000081 could influence the proliferation, migration, and invasion potential of GC. Finally, dual luciferase reporter, RNA immunoprecipitation, qRT-PCR, and western blotting assays were used to verify the targeting relationship between circRNA-000081 and miRNA-423-5p or miRNA-423-5p and 3-phosphoinositide-dependent kinase 1 (PDPK1). In conclusion, circRNA-0000081 promotes the function of GC through sponging hsa-miR-423-5p to influence PDPK1 expression, which has a promising therapeutic potential for treating patients with GC.


Asunto(s)
MicroARNs , Neoplasias Gástricas , 1-Fosfatidilinositol 4-Quinasa , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Fosfatidilinositoles , ARN Circular/genética , Neoplasias Gástricas/patología
17.
Biomed Pharmacother ; 151: 113157, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35605299

RESUMEN

Zinc (Zn) has an existence within large quantities in the human brain, while accumulating within synaptic vesicle. There is growing evidence that Zn metabolic equilibrium breaking participates into different diseases (e.g., vascular dementia, carcinoma, Alzheimer's disease). Carnosine refers to an endogenic dipeptide abundant in skeletal muscle and brains and exerts a variety of positive influences (e.g., carcinoma resistance, crosslinking resistance, metal chelation and oxidation limitation). A complex of Zn and carnosine, called Zinc-L-carnosine (ZnC), has been extensively employed within Zn supplement therapeutic method and the treating approach for ulcers. ZnC has been shown to play a variety of roles in the body, including inhibiting intracellular reactive oxygen species(ROS) and free radical levels, inhibiting inflammation, supplementing zinc enzymes and promoting wound healing and mucosal cell repair. The present study conducting a reviewing process for the advances of ZnC in tumor adjuvant therapy.


Asunto(s)
Carcinoma , Carnosina , Compuestos Organometálicos , Carcinoma/tratamiento farmacológico , Carnosina/análogos & derivados , Carnosina/farmacología , Carnosina/uso terapéutico , Humanos , Compuestos Organometálicos/farmacología , Zinc/metabolismo , Compuestos de Zinc
18.
Front Surg ; 9: 895982, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35495754

RESUMEN

Background: Protein tyrosine kinase (PTK) signaling pathway has been confirmed to be involved in the proliferation, differentiation and migration of tumor cells. Anlotinib, as a multi-target tyrosine kinase inhibitor, which can inhibit the expression of vascular endothelial growth factor receptor (VEGFR), has been confirmed to have significant therapeutic effects on non-small cell lung cancer, medullary thyroid carcinoma, and soft tissue sarcoma, but the therapeutic effect on gastric cancer (GC) is still unclear. Methods: Anlotinib was screened out of 880 drugs through Cell Counting Kit 8 (CCK-8) technology. TCGA was used to detect the expression of VEGFR in GC, and Kaplan-Meier Plotter was used to analyze the correlation between the expression of VEGFR and the survival rate of GC patients. The impacts exerted by anlotinib to GC cell proliferating, migrating and invading processes were assessed through wound healing assay, transwell assay, and proliferation assay in vitro. In vivo experiments of GC were performed in C57/B6 mouse model to evaluate the function of anlotinib and PD-1 antibody. Results: It was found from more than compunds that anlotinib has a significant inhibitory effect on GC cells. In vitro experiments show that anlotinib can significantly inhibit the proliferation, invasion and proliferation of GC cells. The expression level of VEGFR is related to the prognosis and survival of GC. GC patients with low expression of VEGFR have better survival. Anlotinib can inhibit the expression of PD-L1, and achieve better therapeutic effects after combined with PD-1 antibody. Conclusion: The present study reveals that anlotinib down regulates PD-L1. The combination of anlotinib and PD-1 monoclonal antibody is beneficial to GC therapy.

19.
Cell Death Dis ; 13(12): 1028, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36477408

RESUMEN

Tumor-derived exosomes participate in omental metastatic colonization of ovarian cancer by inducing an adaptive response in the tumor microenvironment. However, cell-cell communication via exosomes between primary tumor cells and the microenvironment of distant omentum and the mechanism of pre-metastatic niche formation are poorly understood. Here, we demonstrated that ETS1-overexpressing ovarian cancer cells secreted larger exosomes with higher laminin levels. In addition, ovarian cancer exosomes could be taken up by omental macrophages through integrin and laminin interaction. Compared with control exosomes, exosomes derived from ETS1-overexpressing ovarian cancer cells (LV-ETS1 Exos) stimulated the polarization of more macrophages toward the M2 phenotype (CD163 marker), as well as the production of more CXCL5 and CCL2 in macrophages, via integrin αvß5/AKT/Sp1 signaling. In vivo experiments showed that LV-ETS1 Exos promoted omental metastasis of ovarian cancer by mediating the tumor-promoting effect of macrophages, which could be neutralized by integrin ανß5 inhibitor cilengitide. These results indicated that ETS1 could drive ovarian cancer cells to release exosomes with higher laminin levels, thereby accelerating the exosome-mediated pro-metastatic effects of omental macrophages via the integrin αvß5/AKT/Sp1 signaling pathway, and the integrin ανß5 inhibitor cilengitide could inhibit omental metastasis of ovarian cancer driven by tumor-derived exosomes.


Asunto(s)
Laminina , Neoplasias Ováricas , Femenino , Humanos , Neoplasias Ováricas/genética , Integrinas , Microambiente Tumoral , Proteína Proto-Oncogénica c-ets-1/genética
20.
Aging (Albany NY) ; 14(21): 8688-8699, 2022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-36375474

RESUMEN

BACKGROUND: A complex of Zn and carnosine, called Zinc-L-carnosine (ZnC), enjoys a wide application as part of a Zn supplement therapeutic method as well as in treating peptic ulcers. However, researches fail to confirm the biological functions possessed by ZnC as well as tumor immune microenvironment in colorectal cancer (CRC). METHODS: Cell counting kit 8(CCK8), 5-ethynyl-2'-deoxyuridine (EdU), transwell and wound healing assays were conducted to study the influence of ZnC in the proliferating, invading and migrating processes of CRC cell lines (HCT116, LOVO) in vitro. The antitumor activity ZnC as well as its effects on tumor immune microenvironment were then assessed using CRC subcutaneous tumors in the C57BL/6 mouse model. RESULTS: According to CCK8, EdU, transwell and wound healing assays, ZnC inhibited CRC cell lines in terms of proliferation, invasion and migration. ZnC could inhibit miR-570 for up-regulating PD-L1 expression. In vivo experiments showed that gavage (100 mg/kg, once every day) of ZnC inhibited the tumor growth of CRC, and the combination of ZnC and anti-PD1 therapy significantly improved the efficacy exhibited by anti-PD1 in treating CRC. In addition, mass cytometry results showed that immunosuppressive cells including regulatory T cells (tregs), bone marrow-derived suppressor cells (MDSC), and M2 macrophages decreased whereas CD8+ T cells elevated after adding ZnC. CONCLUSIONS: The present study reveals that ZnC slows the progression of CRC by inhibiting CRC cells in terms of proliferation, invasion and migration, meanwhile up-regulating PD-L1 expression via inhibiting miR-570. The ZnC-anti-PD1 co-treatment assists in synergically increasing anti-tumor efficacy in CRC therapy.


Asunto(s)
Carnosina , Neoplasias Colorrectales , MicroARNs , Ratones , Animales , Carnosina/farmacología , Carnosina/uso terapéutico , Ratones Endogámicos C57BL , Antígeno B7-H1 , Inmunoterapia , Factores Inmunológicos , Neoplasias Colorrectales/tratamiento farmacológico , Proliferación Celular , Movimiento Celular , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA