Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cancer Lett ; 592: 216926, 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38714291

RESUMO

Gastric cancer (GC) is one of the most common cancer worldwide. Neural invasion (NI) is considered as the symbiotic interaction between nerves and cancers, which strongly affects the prognosis of GC patients. Small extracellular vesicles (sEVs) play a key role in intercellular communication. However, whether sEVs mediate GC-NI remains unexplored. In this study, sEVs release inhibitor reduces the NI potential of GC cells. Muscarinic receptor M3 on GC-derived sEVs regulates their absorption by neuronal cells. The enrichment of sEV-circVAPA in NI-positive patients' serum is validated by serum high throughput sEV-circRNA sequencing and clinical samples. sEV-circVAPA promotes GC-NI in vitro and in vivo. Mechanistically, sEV-circVAPA decreases SLIT2 transcription by miR-548p/TGIF2 and inhibits SLIT2 translation via binding to eIF4G1, thereby downregulates SLIT2 expression in neuronal cells and finally induces GC-NI. Together, this work identifies the preferential absorption mechanism of GC-derived sEVs by neuronal cells and demonstrates a previously undefined role of GC-derived sEV-circRNA in GC-NI, which provides new insight into sEV-circRNA based diagnostic and therapeutic strategies for NI-positive GC patients.

2.
Gastric Cancer ; 27(2): 275-291, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38252226

RESUMO

BACKGROUND: Peritoneal metastasis (PM), one of the most typical forms of metastasis in advanced gastric cancer (GC), indicates a poor prognosis. Exploring the potential molecular mechanism of PM is urgently necessary, as it has not been well studied. E3 ubiquitin ligase has been widely established to exert a biological function in various cancers, but its mechanism of action in GC with PM remains unknown. METHODS: The effect of MIB1 on PM of GC was confirmed in vitro and in vivo. Co-immunoprecipitation (Co-IP) and mass spectrometry demonstrated the association between MIB1 and DDX3X. Western blot, flow cytometry and immunofluorescence determined that DDX3X was ubiquitylated by MIB1 and promoted stemness. We further confirmed that METTL3 promoted the up-regulation of MIB1 by RNA immunoprecipitation (RIP), luciferase reporter assay and other experiments. RESULTS: We observed that the E3 ubiquitin ligase Mind bomb 1 (MIB1) was highly expressed in PMs, and patients with PM with high MIB1 expression showed a worse prognosis than those with low MIB1 expression. Mechanistically, our study demonstrated that the E3 ubiquitin ligase MIB1 promoted epithelial-mesenchymal transition (EMT) progression and stemness in GC cells by degrading DDX3X. In addition, METTL3 mediated m6A modification to stabilize MIB1, which required the m6A reader IGF2BP2. CONCLUSIONS: Our study elucidated the specific molecular mechanism by which MIB1 promotes PM of GC, and suggested that targeting the METTL3-MIB1-DDX3X axis may be a promising therapeutic strategy for GC with PM.


Assuntos
Adenosina/análogos & derivados , Neoplasias Peritoneais , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , Ubiquitina-Proteína Ligases/genética , Linhagem Celular Tumoral , Metiltransferases/genética , Metiltransferases/metabolismo , RNA Helicases DEAD-box/metabolismo , Proteínas de Ligação a RNA
3.
Small ; 20(13): e2308688, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37946695

RESUMO

Lymph node metastasis (LNM) is a significant barrier to the prognosis of patients with gastric cancer (GC). Helicobacter pylori (H. pylori)-positive GC patients experience a higher rate of LNM than H. pylori-negative GC patients. However, the underlying mechanism remains unclear. Based on the findings of this study, H. pylori-positive GC patients have greater lymphangiogenesis and lymph node immunosuppression than H. pylori-negative GC patients. In addition, miR-1246 is overexpressed in the plasma small extracellular vesicles (sEVs) of H. pylori-positive GC patients, indicating a poor prognosis. Functionally, sEVs derived from GC cells infected with H. pylori deliver miR-1246 to lymphatic endothelial cells (LECs) and promote lymphangiogenesis and lymphatic remodeling. Mechanistically, miR-1246 suppresses GSK3ß expression and promotes ß-Catenin and downstream MMP7 expression in LECs. miR-1246 also stabilizes programmed death ligand-1 (PD-L1) by suppressing GSK3ß and induces the apoptosis of CD8+ T cells. Overall, miR-1246 in plasma sEVs may be a novel biomarker and therapeutic target in GC-LNM.


Assuntos
Vesículas Extracelulares , Helicobacter pylori , MicroRNAs , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/metabolismo , Linfangiogênese , Células Endoteliais/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Glicogênio Sintase Quinase 3 beta , MicroRNAs/genética , Vesículas Extracelulares/metabolismo
4.
J Exp Clin Cancer Res ; 42(1): 258, 2023 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-37789385

RESUMO

BACKGROUND: Liver metastasis (LM) is one of the most common distant metastases of gastric cancer (GC). However, the mechanisms underlying the LM of GC (GC-LM) remain poorly understood. This study aimed to identify the tumour-secreted protein associated with GC-LM and to investigate the mechanisms by which this secreted protein remodels the liver microenvironment to promote GC-LM. METHODS: Data-independent acquisition mass spectrometry (DIA-MS), mRNA expression microarray, quantitative real-time PCR, enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) were performed to identify and validate the GC-secreted proteins associated with GC-LM. A modified intrasplenic injection mouse model of LM was used to evaluate the progression and tumour burden of LM in vivo. Flow cytometry, immunofluorescence (IF), western blots (WB) and IHC were performed to validate the pre-metastatic niche (PMN) formation in the pre-modelling mouse models. mRNA sequencing of PMA-treated THP-1 cells with or without lipopolysaccharide binding protein (LBP) treatment was used to identify the functional target genes of LBP in macrophages. Co-immunoprecipitation (Co-IP), WB, ELISA, IF and Transwell assays were performed to explore the underlying mechanism of LBP in inducing intrahepatic PMN formation. RESULTS: LBP was identified as a critical secreted protein associated with GC-LM and correlated with a worse prognosis in patients with GC. LBP activated the TLR4/NF-κB pathway to promote TGF-ß1 secretion in intrahepatic macrophages, which, in turn, activated hepatic satellite cells (HSCs) to direct intrahepatic fibrotic PMN formation. Additionally, TGF-ß1 enhanced the migration and invasion of incoming metastatic GC cells in the liver. Consequently, selective targeting of the TGF-ß/Smad signaling pathway with galunisertib demonstrated its efficacy in effectively preventing GC-LM in vivo. CONCLUSIONS: The results of this study provide compelling evidence that serological LBP can serve as a valuable diagnostic biomarker for the early detection of GC-LM. Mechanistically, GC-derived LBP mediates the crosstalk between primary GC cells and the intrahepatic microenvironment by promoting TGF-ß1 secretion in intrahepatic macrophages, which induces intrahepatic fibrotic PMN formation to promote GC-LM. Importantly, selectively targeting the TGF-ß/Smad signaling pathway with galunisertib represents a promising preventive and therapeutic strategy for GC-LM.


Assuntos
Neoplasias Hepáticas , Neoplasias Gástricas , Animais , Humanos , Camundongos , Neoplasias Hepáticas/genética , RNA Mensageiro , Transdução de Sinais , Neoplasias Gástricas/patologia , Fator de Crescimento Transformador beta1/metabolismo , Microambiente Tumoral
5.
Cell Death Dis ; 14(8): 520, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37582794

RESUMO

Abnormal 5-methylcytosine (m5C) methylation has been proved to be closely related to gastric carcinogenesis, progression, and prognosis. Dysregulated long noncoding RNAs (lncRNAs) participate in a variety of biological processes in cancer. However, to date, m5C-methylated lncRNAs are rarely researched in gastric cancer (GC). Here, we found that RNA cytosine-C(5)-methyltransferase (NSUN2) was upregulated in GC and high NSUN2 expression was associated with poor prognosis. NR_033928 was identified as an NSUN2-methylated and upregulated lncRNA in GC. Functionally, NR_033928 upregulated the expression of glutaminase (GLS) by interacting with IGF2BP3/HUR complex to promote GLS mRNA stability. Increased glutamine metabolite, α-KG, upregulated NR_033928 expression by enhancing its promoter 5-hydroxymethylcytosine (hm5C) demethylation. In conclusion, our results revealed that NSUN2-methylated NR_033928 promoted GC progression and might be a potential prognostic and therapeutic target for GC.


Assuntos
RNA Longo não Codificante , Neoplasias Gástricas , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Glutamina , Glutaminase/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proliferação de Células/genética
6.
J Gastrointest Oncol ; 14(2): 617-625, 2023 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-37201061

RESUMO

Background: In laparoscopic total gastrectomy with overlap esophagojejunostomy (EJS), esophageal 'false track' is easily formed during EJS. In this study, a linear cutter/stapler guiding device (LCSGD) was used in EJS, so that the linear cutting stapler can complete the technical action with high speed and high efficiency in a narrow space, while avoiding the formation of 'false passage', optimizing the quality of common opening and shortening the anastomosis time. The LCSGD is safe and feasible in laparoscopic total gastrectomy overlap EJS, and the clinical effect is satisfactory. Methods: A retrospective, descriptive design was adopted. The clinical data of 10 gastric cancer patients admitted to the Third Department of Surgery of the Fourth Hospital of Hebei Medical University from July 2021 to November 2021 were collected. The cohort comprised 8 males and 2 females aged 50-75 years. Results: (I) The intra-operative conditions: 10 patients received LCSGD-guided overlap EJS after radical laparoscopic total gastrectomy. Both D2 lymphadenectomy and R0 resection were achieved in these patients. No combined multiple organ resection was performed. There was neither conversion to an open thoracic or abdominal procedure nor conversion to other EJS approaches. The average time from the entry of the LCSGD into the abdominal cavity to the completion of the firing of the stapler was 1.8±0.4 minutes, the average time for manual suturing of the EJS common opening was 14.4±2.1 minutes (mean: 18±2 stitches), and the average operative time was 255±52 minutes. (II) The postoperative outcomes: the time to the first ambulation was 1.9±1.4 days, the average time to the first postoperative exhaust/defecation was 3.5±1.3 days, the average time to a semi-liquid diet was 3.6±0.7 days, and the average postoperative hospital stay was 10.4±4.1 days. All patients were smoothly discharged, without any secondary surgery, bleeding, anastomotic fistula, or duodenal stump fistula. (III) Follow-up: The telephone follow-up lasted 9-12 months. No eating disorders or anastomotic stenosis was reported. One patient experienced Visick grade II heartburn, and the condition of the remaining 9 patients was Visick grade I. Conclusions: Application of the LCSGD in overlap EJS after laparoscopic total gastrectomy is safe and feasible, with satisfactory clinical effectiveness.

7.
Cancer Commun (Lond) ; 43(6): 685-705, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37256823

RESUMO

BACKGROUND: N6-methyladenosine (m6 A) modification is the most common modification that occurs in eukaryotes. Although substantial effort has been made in the prevention and treatment of gastric cancer (GC) in recent years, the prognosis of GC patients remains unsatisfactory. The regulatory mechanism between m6 A modification and GC development needs to be elucidated. In this study, we examined m6 A modification and the downstream mechanism in GC. METHODS: Dot blotting assays, The Cancer Genome Atlas analysis, and quantitative real-time PCR (qRT-PCR) were used to measure the m6 A levels in GC tissues. Methylated RNA-immunoprecipitation sequencing and RNA sequencing were performed to identify the targets of m6 A modification. Western blotting, Transwell, wound healing, and angiogenesis assays were conducted to examine the role of centromere protein F (CENPF) in GC in vitro. Xenograft, immunohistochemistry, and in vivo metastasis experiments were conducted to examine the role of CENPF in GC in vivo. Methylated RNA-immunoprecipitation-qPCR, RNA immunoprecipitation-qPCR and RNA pulldown assays were used to verify the m6 A modification sites of CENPF. Gain/loss-of-function and rescue experiments were conducted to determine the relationship between CENPF and the mitogen-activated protein kinase (MAPK) signaling pathway in GC cells. Coimmunoprecipitation, mass spectrometry, qRT-PCR, and immunofluorescence assays were performed to explore the proteins that interact with CENPF and elucidate the regulatory mechanisms between them. RESULTS: CENPF was upregulated in GC and facilitated the metastasis of GC both in vitro and in vivo. Mechanistically, increased m6 A modification of CENPF was mediated by methyltransferase 3, and this modified molecule could be recognized by heterogeneous nuclear ribonucleoprotein A2/B1 (HNRNPA2B1), thereby promoting its mRNA stability. In addition, the metastatic phenotype of CENPF was dependent on the MAPK signaling pathway. Furthermore, CENPF could bind to FAK and promote its localization in the cytoplasm. Moreover, we discovered that high expression of CENPF was related to lymphatic invasion and overall survival in GC patients. CONCLUSIONS: Our findings revealed that increased m6 A modification of CENPF facilitates the metastasis and angiogenesis of GC through the CENPF/FAK/MAPK and epithelial-mesenchymal transition axis. CENPF expression was correlated with the clinical features of GC patients; therefore, CENPF may serve as a prognostic marker of GC.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , RNA Mensageiro/metabolismo , Linhagem Celular Tumoral , Transporte Ativo do Núcleo Celular
8.
Cancer Lett ; 565: 216235, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37209945

RESUMO

Cisplatin (CDDP)-based chemotherapy is the preferred treatment strategy for advanced stage gastric cancer (GC) patients. Despite the efficacy of chemotherapy, the development of chemoresistance negatively affects the prognosis of GC and the underlying mechanism remains poorly understood. Accumulated evidence suggests that mesenchymal stem cells (MSCs) play important roles in drug resistance. The chemoresistance and stemness of GC cells were observed by colony formation, CCK-8, sphere formation and flow cytometry assays. Cell lines and animal models were utilized to investigate related functions. Western blot, quantitative real-time PCR (qRT-PCR) and co-immunoprecipitation were used to explore related pathways. The results showed that MSCs improved the stemness and chemoresistance of GC cells and accounted for the poor prognosis of GC. Natriuretic peptide receptor A (NPRA) was upregulated in GC cells cocultured with MSCs and knockdown of NPRA reversed the MSC-induced stemness and chemoresistance. At the same time, MSCs could be recruited to GC by NPRA, which formed a loop. In addition, NPRA facilitated stemness and chemoresistance through fatty acid oxidation (FAO). Mechanistically, NPRA protected Mfn2 against protein degradation and promoted its mitochondrial localization, which consequently improved FAO. Furthermore, inhibition of FAO with etomoxir (ETX) attenuated MSC-induced CDDP resistance in vivo. In conclusion, MSC-induced NPRA promoted stemness and chemoresistance by upregulating Mfn2 and improving FAO. These findings help us understand the role of NPRA in the prognosis and chemotherapy of GC. NPRA may be a promising target to overcome chemoresistance.


Assuntos
Neoplasias Gástricas , Animais , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Ácidos Graxos , Linhagem Celular Tumoral
9.
Front Oncol ; 13: 1169454, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37182139

RESUMO

Background: For gastric cancer (GC) patients with pylorus outlet obstruction (POO), whether laparoscopic surgery has advantages over open surgery remains unclear. This study aims to investigate the differences between patients with and without POO in open and laparoscopic groups and to determine the differences between laparoscopic distal gastrectomy (LDG) and open distal gastrectomy (ODG) in GC patients with POO. Methods: A total of 241 GC patients with POO who underwent distal gastrectomy at the Department of Gastric Surgery of the First Affiliated Hospital of Nanjing Medical University between 2016 and 2021 were included in this study. A total of 1,121 non-POO patients who underwent laparoscopic surgery and 948 non-POO patients who underwent open surgery from 2016 to 2021 were also enrolled in the study. We compared complication rates and hospital stays between open and laparoscopic groups. Results: There was no significant difference for LDG between GC patients with and without POO regarding the overall complication rates (P = 0.063), the Grade III-V complication rate (P = 0.673), and the anastomotic complication rate (P = 0.497) from 2016 to 2021. The patients with POO had longer preoperative hospital stay (P = 0.001) and postoperative hospital stay (P=0.007) compared to patients without POO. No significant difference was observed for open patients between POO and non-POO patients regarding the overall complication rate (P = 0.357), grade III-V complication rate (P = 1.000), and anastomosis-related complication rate (P = 0.766). Compared with open surgery in GC patients with POO (n = 111), the total complication rate of the LDG group was 16.2%, which was significantly lower than that of the open group (26.1%, P = 0.041). No significant differences in the Grade III-V complication rate (P = 0.574) and anastomotic complication rate (P = 0.587) were observed between laparoscopic and open groups. Patients receiving laparoscopic surgery had shorter postoperative hospital stay than open surgery (P = 0.001). More resected lymph nodes (LNs) were also observed in the laparoscopic group (P = 0.0145). Conclusion: The comorbidity of GC with POO does not increase the complication rate after laparoscopic or open distal gastrectomy. In GC patients with POO, laparoscopic surgery shows advantages over open surgery with a lower overall complication rate, shorter postoperative hospital stay, and more harvested lymph nodes. Laparoscopic surgery is a safe, feasible, and effective treatment for GC with POO.

10.
Cell Death Dis ; 14(4): 260, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-37037818

RESUMO

Neural invasion (NI) is a vital pathological characteristic of gastric cancer (GC), which correlates with tumor recurrence and a worse prognosis. Long noncoding RNAs (lncRNAs) play critical roles in various biological processes. However, the involvement of lncRNAs in NI of GC (GC-NI) remains unclear. DIAPH2-AS1 was upregulated in NI-positive GC tissues, which was confirmed by qRT-PCR. The higher expression of DIAPH2-AS1 predicted NI and worse survival for GC patients. Both in vitro and in vivo experiments, including wound-healing assay, Transwell assay, DRG-GC cells co-culture model, the mouse sciatic nerve model, and the lung metastasis model, indicated that DIAPH2-AS1 promoted the migration, invasion, and NI potential of GC cells. Mechanistically, pulldown assay and RNA immunoprecipitation assay revealed that DIAPH2-AS1 interacted with NSUN2. Subsequent experiments indicated that DIAPH2-AS1 stabilized NSUN2 from ubiquitin-proteasomal degradation via masking the K577 and K579 of NSUN2. The protection of DIAPH2-AS1 on NSUN2 improved the stability of NTN1 mRNA via m5C modification, which finally induced GC-NI. Our work uncovered DIAPH2-AS1 as a novel oncogenic lncRNA in GC-NI and validated the DIAPH2-AS1-NSUN2-NTN1 axis as a potential therapeutic target for NI-positive GC.


Assuntos
Metiltransferases , MicroRNAs , Netrina-1 , RNA Longo não Codificante , Neoplasias Gástricas , Animais , Camundongos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , Recidiva Local de Neoplasia/genética , RNA Longo não Codificante/genética , Neoplasias Gástricas/patologia , Netrina-1/genética , Metiltransferases/genética
11.
Cancer Lett ; 558: 216092, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36806557

RESUMO

Despite many advances in treatment over the past few years, the poor 5-year survival rate and high recurrence rate of gastric cancer (GC) remain unsatisfactory. As the most abundant epigenetic modification in the eukaryotic mRNA, N6-methyladenosine (m6A) methylation participates in tumor progression and tissue development. During tumor progression, DNA damage repair mechanisms can be reprogrammed to give new growth advantages on tumor clones whose genomic integrity is disturbed. Here we detected the elevated SUV39H2 expression in GC tissues and cell lines. Functionally, SUV39H2 promoted GC proliferation and inhibited apoptosis in vitro and in vivo. Mechanistically, METTL3-mediated m6A modification promotes mRNA stability of SUV39H2 in an IGF2BP2 dependent manner, resulting in upregulated mRNA expression of SUV39H2. As a histone methyltransferase, SUV39H2 was verified to increase the phosphorylation level of ATM through transcriptional repression of DUSP6, thereby promoting HRR and ultimately inhibiting GC chemosensitivity to cisplatin. Collectively, these results indicate the specific mechanism of m6A-modified SUV39H2 as a histone methyltransferase promoting HRR to inhibit the chemosensitivity of GC. SUV39H2 is expected to become a key target in the precision targeted therapy of GC.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , Repressão Epigenética , Linhagem Celular Tumoral , Recombinação Homóloga , Histona Metiltransferases/genética , RNA Mensageiro , Metiltransferases/metabolismo , Proteínas de Ligação a RNA/genética , Fosfatase 6 de Especificidade Dupla/genética , Fosfatase 6 de Especificidade Dupla/metabolismo , Histona-Lisina N-Metiltransferase/genética
12.
Cell Death Dis ; 13(3): 266, 2022 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-35338119

RESUMO

Circular RNAs (circRNAs) play vital regulatory roles in the progression of multiple cancers. In our study, transcriptome analysis and self-organizing maps (SOM) were applied to screen backbone circRNAs in gastric cancer (GC). Upon validation of the expression patterns of screened circRNAs, gain- and loss-of-function assays were performed in vitro and in vivo. Underlying mechanisms were investigated using RNA pull-down, luciferase reporter assay and RNA immunoprecipitation. The expression of circTHBS1 was significantly increased in GC and associated with poor prognosis. CircTHBS1 facilitated the malignant behavior and epithelial-to-mesenchymal transition of GC cells. Mechanistically, circTHBS1 sponged miR-204-5p to promote the expression of Inhibin Subunit Beta A (INHBA). Moreover, circTHBS1 could enhance the HuR-mediated mRNA stability of INHBA, which subsequently activated the TGF-ß pathway. Our research identified circTHBS1 as an oncogenic circRNA that enhances GC malignancy by elevating INHBA expression, providing new insight and a feasible target for the diagnosis and treatment of GC.


Assuntos
MicroRNAs , Neoplasias Gástricas , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Subunidades beta de Inibinas , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Circular/genética , RNA Mensageiro/genética , Neoplasias Gástricas/patologia , Trombospondina 1
13.
Clin Transl Med ; 12(1): e707, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35061934

RESUMO

Gastric cancer (GC) ranks third in mortality among all cancers worldwide. Circular RNAs (circRNAs) play an important role in the occurrence and development of gastric cancer. Forkhead box P2 (FOXP2), as a transcription factor, is closely associated with the development of many types of tumours. However, the regulatory network between FOXP2 and circRNAs remains to be explored. In our study, circST3GAL6 was significantly downregulated in GC and was associated with poor prognosis in GC patients. Overexpression of circST3GAL6 inhibited the malignant behaviours of GC cells, which was mediated by inducing apoptosis and autophagy. In addition, we demonstrated that circST3GAL6 regulated FOXP2 through the mir-300 sponge. We further found that FOXP2 inhibited MET Proto-Oncogene (MET), which was the initiating factor that regulated the classic AKT/mTOR pathway of autophagy. In conclusion, our results suggested that circST3GAL6 played a tumour suppressive role in gastric cancer through miR-300/FOXP2 axis and regulated apoptosis and autophagy through FOXP2-mediated transcriptional inhibition of the MET axis, which may become a potential target for GC therapy.


Assuntos
Autofagia/efeitos dos fármacos , Sialiltransferases/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/efeitos dos fármacos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/prevenção & controle , Proteínas Proto-Oncogênicas c-met/efeitos dos fármacos , Sialiltransferases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias Gástricas/prevenção & controle , Serina-Treonina Quinases TOR/efeitos dos fármacos , beta-Galactosídeo alfa-2,3-Sialiltransferase
14.
J Gastrointest Oncol ; 13(6): 2749-2757, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36636057

RESUMO

Background: The development process of gastrointestinal anastomosis is from complex to simple, from two layers to one layer, from extramucosal anastomosis to seromuscular anastomosis. With the rapid development of anastomosis instruments, the anastomosis process becomes more and more convenient. However, relevant studies have shown that related complications such as anastomotic leakage still occur. This study sought to investigate the feasibility and safety of seromuscular layer sutures in the reinforcement of esophagojejunostomy after open radical total gastrectomy. Methods: This study retrospectively analyzed patients who underwent Roux-en-Y esophagojejunostomy after open radical total gastrectomy at The Third Department of Surgery, The Fourth Hospital of Hebei Medical University from April 2019 to May 2020. The inclusion criteria of patients were between 18 and 80 years old; pathology confirmed gastric adenocarcinoma; preoperative imaging showed no distant metastasis and did not receive neoadjuvant therapy; no complex infectious diseases; no blood transfusion was performed before operation. A total of 192 patients were included according to the inclusion criteria. They were divided into the following 2 groups based on whether seromuscular layer suturing of the anastomosis was performed: (I) group A (the simple anastomosis group, n=76); (II) and group B (the seromuscular layer suture group, n=116). The baseline data, surgical data and postoperative complications were compared between the two groups. Results: All patients underwent esophagojejunostomy Roux-en-Y anastomosis after open radical total gastrectomy, and no perioperative deaths occurred. There was no significant difference in baseline data between the two groups. Group B had an earlier time to liquid diet than group A (4.23±0.76 vs. 4.57±0.58 days, P<0.001). The incidence of postoperative anastomotic leakage in group B (1.72%) was lower than that in group A (9.21%), and the difference was statistically significant (P=0.03). The incidence of pleural effusion was lower in group B (15.52%) than group A (32.89%), and the difference was statistically significant (P=0.005). Conclusions: Compared to the simple anastomosis, seromuscular layer sutures after esophagojejunostomy may decrease the rates of postoperative anastomotic leakage and pleural effusion. This suture method is feasible and may provide a new option to increase surgical safety.

15.
Cell Death Dis ; 12(10): 910, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34611143

RESUMO

Gastric cancer remains the third leading cause of cancer-related mortality worldwide. Emerging evidence has shown that circular RNAs (circRNAs) play a critical regulatory role in the occurrence and development of various cancers through sponging miRNAs or acting as RNA-binding protein (RBP) sponges. We found that circUBE2Q2 was significantly upregulated in GC tissues and cell lines. Knockdown of circUBE2Q2 inhibited proliferation, migration, invasion, and glycolysis, and increased autophagy in vitro. In addition, knockdown of circUBE2Q2 inhibited GC tumorigenicity and metastasis potential in vivo. A series of experiments were performed to confirm that circUBE2Q2 regulates GC progression via the circUBE2Q2-miR-370-3p-STAT3 axis and promotes tumor metastasis through exosomal communication. Further in vivo experiments confirmed that, combination treatment of circUBE2Q2 knocking down and STAT3 inhibitor has synergistic effects on the gastric cancer growth inhibition, which provides a possibility to enhance the sensitivity of targeted drugs to gastric cancer through targeting circUBE2Q2. Our findings revealed that circUBE2Q2 may serve as a new proliferation-promoting factor and prognostic marker in gastric cancer.


Assuntos
Autofagia/genética , Progressão da Doença , Glicólise/genética , RNA Circular/metabolismo , Fator de Transcrição STAT3/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Animais , Autofagossomos/metabolismo , Autofagossomos/ultraestrutura , Sequência de Bases , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Exossomos/metabolismo , Exossomos/ultraestrutura , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Masculino , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica , RNA Circular/genética , Carga Tumoral
16.
J Biotechnol ; 322: 29-32, 2020 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-32653638

RESUMO

Over the past decade, 5-aminolevulinic acid (5-ALA) has been highlighted as a promising functional feed additive and immunomodulator for improving the general health, immune response, and resistance to disease of livestock and poultry. However, it is very costly to produce 5-ALA using conventional chemical synthesis methods. Classical microbial fermentation fulfills the criteria of environmental friendliness, but the unsatisfactory titers still hinder actual industrial production. This study aimed to develop a solid-state fermentation (SSF) process that can be used to efficiently enrich feed with 5-ALA at a low cost. First, the endogenous 5-ALA synthase was overexpressed in Saccharomyces cerevisiae via integrating a copy of HEM1 gene into the chromosome and introducing a multi-copy plasmid pRS416-HEM1 which constitutively overexpresses HEM1 gene. The resulting strain ScA3 was able to produce 63.82 mg/L 5-ALA in shake-flask fermentation. After process optimization, a titer of 225.63 mg/kg dry materials, exceeding the usual effective dosage reported in animal trials, was achieved within 48 h through SSF of 20 kg feed in a 90-L steel drum. To our knowledge, this is the first report on combining microbial 5-ALA production with SSF in feed processing, which will hopefully promote the application and popularization of 5-ALA in the feed industry.


Assuntos
Ácido Aminolevulínico/metabolismo , Ração Animal , Fermentação/genética , Saccharomyces cerevisiae , 5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Microbiologia Industrial/métodos , Plasmídeos/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
17.
ACS Appl Mater Interfaces ; 12(13): 15430-15438, 2020 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-32134241

RESUMO

We report an optoelectronic device consisting of a solution-processed indium gallium zinc oxide (IGZO) thin-film transistor and vacuum-deposited small organic molecules. Depending on the configurations of the organic materials, either bulk heterojunction or planar heterojunction (PHJ), the device assumes the functionality of either a photosensor or a photoinduced memory, respectively. Under λ = 625 nm light illumination, the photosensor shows response and recovery time of ∼50 ms, responsivity of ∼5 mA/W, sensitivity above 104, and a linear response. The mechanism of the photoinduced memory is studied experimentally and verified using a device simulation. We find that the memory is due to long charge retention time at the organic PHJ interface which is stable for over 9 days. It is correlated with the low leakage current found in ordered organic junctions having low subgap tail states. The presented integration of the PHJ with the transistor constitutes a new design of write-once-read-many-times memory device that is likely to be attractive for low-cost applications.

18.
Front Vet Sci ; 6: 228, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31355216

RESUMO

Babesia microti, an intraerythrocytic protozoa, can cause an emerging tick-borne disease-Human babesiosis. The parasite can successfully invade host red blood cells owing to the assistance of molecules expressed by babesia. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH), the housekeeping intracellular glycolytic enzyme, can also be expressed in the external of cells, where contributes to binding to several molecules such as plasminogen and actin. In the present study, we identified B. microti GAPDH (BmGAPDH) and generated the recombinant BmGAPDH (rBmGAPDH) via an E. coli expression system. Furthermore, we confirmed its catalytic dehydration activity in vitro. Moreover, we also demonstrated that rBmGAPDH could bind to human plasminogen and mouse α-actin. In addition, we demonstrated that rBmGAPDH could recognize anti-B. microti mouse serum. In conclusion, BmGAPDH is a multifunctional glycolytic enzyme, which can bind to host plasminogen and α-actin.

19.
Parasitol Res ; 118(4): 1103-1112, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30770979

RESUMO

Human babesiosis, a worldwide emerging tick-borne disease, is caused by the intraerythrocytic apicomplexan parasite, babesia. In recent years, the number of infected patients globally has continued to rise, and thus human babesiosis poses a significant public health threat. Therefore, stronger initiatives should be undertaken to prevent further spread and development of this disease. In the present review, we summarize the epidemiology of reported human babesiosis cases in China from 1993 until now. The data show that Babesia microti is the dominant species causing human babesiosis in China and has led to more than 100 human infections thus far, where Babesia crassa-like is the second-most common. Moreover, Guangxi province is the second-most infected area after the Heilongjiang province. We also review the babesia life cycle, manifestation, diagnosis, and treatment. Additionally, we discuss babesiosis prevention strategies to raise public awareness, and also provide suggestions for improved babesiosis control.


Assuntos
Babesia microti/isolamento & purificação , Babesiose , Doenças Transmitidas por Carrapatos/epidemiologia , Animais , Babesiose/diagnóstico , Babesiose/tratamento farmacológico , Babesiose/epidemiologia , Babesiose/prevenção & controle , China/epidemiologia , Geografia , Humanos , Estágios do Ciclo de Vida , Camundongos , Doenças Transmitidas por Carrapatos/parasitologia , Carrapatos/parasitologia
20.
Future Oncol ; 14(15): 1511-1520, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29380639

RESUMO

Quinacrine (QC), a synthetic antimalarial drug, was consistently used worldwide to combat malaria during the last century. Interestingly, later studies revealed that it also displays various additional properties, specifically antitumor activity. QC's antitumor activity occurs via a variety of pathways, including DNA intercalation, angiogenesis inhibition, signal transduction regulation, cell cycle arrest and autophagy induction. In combination with traditional therapies such as chemotherapy and radiotherapy, QC has also displayed synergistic effects against tumors, which may open promising therapeutic avenues. However, the breadth and complexity of its antitumor mechanisms have not yet been fully elucidated. In this review, we have systematically categorized QC's reported antitumor mechanisms from recent studies, to enable a deeper understanding of its antitumor activity.


Assuntos
Antimaláricos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/terapia , Neovascularização Patológica/tratamento farmacológico , Quinacrina/uso terapêutico , Animais , Antimaláricos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Terapia Combinada/métodos , Sinergismo Farmacológico , Humanos , Neoplasias/patologia , Neovascularização Patológica/patologia , Uso Off-Label , Quinacrina/farmacologia , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA