Pancreatic neoplasms

胰腺肿瘤
  • 文章类型: Journal Article
    UNASSIGNED: pancreatic ductal adenocarcinoma (PDAC) is a malignant tumor with a very poor prognosis and a complex tumor microenvironment, which plays a key role in tumor progression and treatment resistance. Glycosylation plays an important role in processes such as cell signaling, immune response and protein stability.
    UNASSIGNED: single-cell RNA sequencing data and spatial transcriptome data were obtained from GSE197177 and GSE224411, respectively, and RNA-seq data and survival information were obtained from UCSC Xena and TCGA. Multiple transcriptomic data were comprehensively analyzed to explore the role of glycosylation processes in tumor progression, and functional experiments were performed to assess the effects of MGAT1 overexpression on PDAC cell proliferation and migration.
    UNASSIGNED: In PDAC tumor samples, the glycosylation level of macrophages was significantly higher than that of normal samples. MGAT1 was identified as a key glycosylation-related gene, and its high expression was associated with better patient prognosis. Overexpression of MGAT1 significantly inhibited the proliferation and migration of PDAC cells and affected intercellular interactions in the tumor microenvironment.
    UNASSIGNED: MGAT1 plays an important role in PDAC by regulating glycosylation levels in macrophages, influencing tumor progression and improving prognosis.MGAT1 is a potential therapeutic target for PDAC and further studies are needed to develop targeted therapeutic strategies against MGAT1 to improve clinical outcomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胰腺癌中的肿瘤内微生物组失衡促进耐受性免疫反应并引发免疫疗法抗性。在这里,我们展示了鼠李糖乳杆菌GG益生菌,配备镓-多酚网络(LGG@Ga-poly),通过调节微生物群-免疫相互作用来增强胰腺癌的免疫治疗。口服后,LGG@Ga-poly靶向胰腺肿瘤,并通过镓促进细菌铁呼吸的破坏选择性根除促进肿瘤的变形杆菌和微生物群衍生的脂多糖。肿瘤内微生物群的这种消除阻碍了肿瘤Toll样受体的激活,从而降低肿瘤细胞的免疫抑制PD-L1和白细胞介素-1β表达,正在减少的免疫耐受髓系群体,并改善肿瘤细胞毒性T淋巴细胞的浸润。此外,LGG@Ga-poly在预防和治疗方面都会阻碍胰腺肿瘤的生长,并放大了雌性小鼠临床前癌症模型中免疫检查点阻断的抗肿瘤功效。总的来说,我们提供的证据表明,针对肿瘤内微生物群的精心设计的生物材料可以有效地增强具有挑战性的胰腺癌的免疫治疗。
    The intratumor microbiome imbalance in pancreatic cancer promotes a tolerogenic immune response and triggers immunotherapy resistance. Here we show that Lactobacillus rhamnosus GG probiotics, outfitted with a gallium-polyphenol network (LGG@Ga-poly), bolster immunotherapy in pancreatic cancer by modulating microbiota-immune interactions. Upon oral administration, LGG@Ga-poly targets pancreatic tumors specifically, and selectively eradicates tumor-promoting Proteobacteria and microbiota-derived lipopolysaccharides through a gallium-facilitated disruption of bacterial iron respiration. This elimination of intratumor microbiota impedes the activation of tumoral Toll-like receptors, thus reducing immunosuppressive PD-L1 and interleukin-1β expression by tumor cells, diminishing immunotolerant myeloid populations, and improving the infiltration of cytotoxic T lymphocytes in tumors. Moreover, LGG@Ga-poly hampers pancreatic tumor growth in both preventive and therapeutic contexts, and amplifies the antitumor efficacy of immune checkpoint blockade in preclinical cancer models in female mice. Overall, we offer evidence that thoughtfully designed biomaterials targeting intratumor microbiota can efficaciously augment immunotherapy for the challenging pancreatic cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:胰腺腺癌(PAAD)通常表现出“冷”或免疫抑制性肿瘤环境,这与对免疫检查点阻断治疗的抗性有关;然而,潜在的机制还没有完全理解。这里,我们旨在提高我们对肿瘤微环境中发生的分子机制的理解,并确定生物标志物,治疗目标,以及改善PAAD治疗的潜在药物。
    方法:根据具有不同疾病结局的免疫热或冷PAAD亚型对患者进行分类。进行Cox回归和加权相关网络分析以构建新的基因标签,在热瘤中被称为下调,预后,和免疫相关基因(DPIRGs),用于通过机器学习(ML)开发PAAD的预后模型。综合分析了DPIRGs在PAAD中的作用,和能够区分PAAD免疫亚型和预测预后的生物标志物基因通过ML鉴定。使用公共单细胞转录组和蛋白质组资源验证生物标志物的表达。通过分子对接研究鉴定了冷肿瘤变热的候选药物和相应的靶蛋白。
    结果:使用DPIRG签名作为输入数据,从137ML组合中选择生存随机森林和偏最小二乘回归Cox的组合,构建优化的PAAD预后模型.通过分析遗传/表观遗传改变,研究了DPIRGs的作用和分子机制。免疫浸润,途径富集,和miRNA调控。生物标志物和潜在的治疗靶点,包括PLEC,TRPV1和ITGB4等,被确认,并验证了生物标志物的细胞类型特异性表达。候选药物,包括沙利度胺,SB-431542和博来霉素A2基于它们有利地调节DPIRG表达的能力进行鉴定。
    结论:通过组合多种ML算法,我们开发了一种在PAAD队列中表现优异的新型预后模型.ML还被证明对于识别生物标志物和改善PAAD患者分层和免疫疗法的潜在靶标是强大的。
    BACKGROUND: Pancreatic adenocarcinomas (PAADs) often exhibit a \"cold\" or immunosuppressive tumor milieu, which is associated with resistance to immune checkpoint blockade therapy; however, the underlying mechanisms are incompletely understood. Here, we aimed to improve our understanding of the molecular mechanisms occurring in the tumor microenvironment and to identify biomarkers, therapeutic targets, and potential drugs to improve PAAD treatment.
    METHODS: Patients were categorized according to immunologically hot or cold PAAD subtypes with distinct disease outcomes. Cox regression and weighted correlation network analysis were performed to construct a novel gene signature, referred to as \'Downregulated in hot tumors, Prognostic, and Immune-Related Genes\' (DPIRGs), which was used to develop prognostic models for PAAD via machine learning (ML). The role of DPIRGs in PAAD was comprehensively analyzed, and biomarker genes able to distinguish PAAD immune subtypes and predict prognosis were identified by ML. The expression of biomarkers was verified using public single-cell transcriptomic and proteomic resources. Drug candidates for turning cold tumors hot and corresponding target proteins were identified via molecular docking studies.
    RESULTS: Using the DPIRG signature as input data, a combination of survival random forest and partial least squares regression Cox was selected from 137 ML combinations to construct an optimized PAAD prognostic model. The effects and molecular mechanisms of DPIRGs were investigated by analysis of genetic/epigenetic alterations, immune infiltration, pathway enrichment, and miRNA regulation. Biomarkers and potential therapeutic targets, including PLEC, TRPV1, and ITGB4, among others, were identified, and the cell type-specific expression of the biomarkers was validated. Drug candidates, including thalidomide, SB-431542, and bleomycin A2, were identified based on their ability to modulate DPIRG expression favorably.
    CONCLUSIONS: By combining multiple ML algorithms, we developed a novel prognostic model with excellent performance in PAAD cohorts. ML also proved to be powerful for identifying biomarkers and potential targets for improved PAAD patient stratification and immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胰头的发育源于胚胎发育过程中腹侧和背侧胰腺原基的融合。理论上,胰头癌的起源也存在于腹侧胰腺和背侧胰腺。在49例胰头癌患者中,通过影像学和病理分类,胰头癌分为起源于腹侧(PHCv)或背胰腺(PHCd)的胰头癌。收集并比较PHCv组和PHCd组的临床资料。结果显示,PHCd组患者的长期生存率比PHCv组患者差(10个月比14.5个月)。同样,无进展生存期(PFS)结果还表明,PHCd组患者的时间短于PHCV组患者(5个月vs9.5个月).进一步分层分析潜在相关因素显示微血管侵犯与不良预后有关,而来源于背胰腺的胰头癌患者更容易发生微血管侵犯。
    The development of the pancreatic head originates from the fusion of the ventral and dorsal pancreatic primordia during embryonic development. Theoretically, the origin of pancreatic head cancer also exists from the ventral pancreas and the dorsal pancreas. Among 49 patients with pancreatic head cancer, pancreatic head cancer was divided into pancreatic head cancer originating from the ventral (PHCv) or dorsal pancreas (PHCd) through imaging and pathological classification. The clinical data was collected and compared between the PHCv group and the PHCd group. The results showed that the patients from the PHCd group had worse long-term survival than those from the PHCv group (10 months vs 14.5 months). Similarly, the progression-free survival (PFS) results also indicate that patients from the PHCd group had a shorter time than those from the PHCv group (5 months vs 9.5 months). Further stratified analysis of potentially related factors showed that microvascular invasion is related to poor prognosis, and patients with pancreatic head cancer derived from the dorsal pancreas are more likely to develop microvascular invasion.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    这项研究的目的是评估识别主要胰管(MPD)累及的导管内乳头状黏液性肿瘤(IPMN)的恶性的临界值,MPD直径为5至10mm。142例患者的临床放射学特征,分析了涉及MPD的IPMNs(n=53)和分支导管(BD)-IPMNs(n=89)。采用Logistic回归分析确定恶性IPMNs和浸润性癌的危险因素。ROC曲线用于确定术前MPD值的不同截止值,以预测浸润性癌和恶性IPMNs的存在。并对预测性能进行了评估。对于涉及MPD的IPMN(5mm7.5mm和碳水化合物抗原19-9(Ca19-9)>37U/ml被发现是单变量恶性IPMNs的预测因子,MPD>7.5mm是MPD相关IPMNs多变量分析的预测因子。MPD(7.5mm)联合Ca19-9鉴别恶性IPMNs的ROC曲线AUC为0.73。MPD(7.5mm)与Ca19-9组合在鉴定涉及MPD的IPMNs中的恶性IPMNs方面表现良好。
    The aim of this study was to evaluate the cutoff value for identifying malignance in main pancreatic duct (MPD)-involved intraductal papillary mucinous neoplasm (IPMN) with an MPD diameter ranging from 5 to 10 mm. Clinical-radiological characteristics of 142 patients, including MPD-involved IPMNs (n = 53) and branch-duct (BD)-IPMNs (n = 89) were analyzed. Logistic regression analysis was used to determine the risk factors of malignant IPMNs and invasive carcinoma. ROC curves were used to identify different cutoffs in terms of preoperative MPD values to predict the presence of invasive carcinoma as well as malignant IPMNs, and the prediction performance was evaluated. For MPD-involved IPMNs (5 mm < MPD < 10 mm), MPD diameter of 7.5 mm for discriminating malignant IPMNs (area under curve [AUC] = 0.67) and 7.7 mm for discriminating invasive IPMNs (AUC = 0.56) were found to be the optimal cutoff values at receiver operating characteristic curve (ROC) analysis. MPD > 7.5 mm and carbohydrate antigen19-9 (Ca19-9) > 37 U/ml were found to be predictors of malignant IPMNs at univariate, and MPD > 7.5 mm was a predictor in multivariate analysis in MPD-involved IPMNs. The AUC of the ROC curve of MPD (7.5 mm) combined with Ca19-9 in identifying malignant IPMNs was 0.73 in MPD-involved IPMNs. MPD (7.5 mm) combined with Ca19-9 performed well in identifying malignant IPMNs in MPD-involved IPMNs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:胰腺癌行根治性胰腺切除术的患者常发生肝转移。除了化疗,已经探索了针对肝脏病变的各种局部治疗方法。然而,关于射频消融(RFA)作为肝转移的局部治疗方法的研究有限。因此,我们进行了这项回顾性研究以提供临床证据.
    方法:这是一个单中心,回顾性,队列研究。根治性胰十二指肠切除术后,32例患者发生异时性肝转移,病灶少于3个,其中最大的直径小于3厘米。这些患者接受了化疗和RFA的联合治疗。化疗8周后,患者因肝脏病变接受RFA治疗.进行了额外的化疗,监测患者的肿瘤状态和生存率。这项研究的主要终点是总生存期(OS)。使用Cox风险模型分析影响OS的因素。
    结果:在32例患者中,平均OS为28.4个月.单因素和多因素Cox回归分析显示肝转移时间(以月为单位)(HR=0.04,95%CI:0.01至0.19;P<0.001),肝转移的数量(HR=7.08,95%CI:1.85至27.08,P=0.004),和PD(进行性疾病)对第二轮化疗的反应(HR=29.50,95%CI:1.46至597.27;P=0.027)是生存率较差的独立预测因素。
    结论:对于胰十二指肠切除术后肝转移患者,RFA联合化疗是安全的。早期复发(≤12个月),三个肝转移性病变,对第二轮化疗的反应差与低生存率相关。
    OBJECTIVE: Hepatic metastasis frequently occurs in patients who have undergone radical pancreatic resection for pancreatic cancer. Besides chemotherapy, various local treatment approaches targeting hepatic lesions have been explored. However, research on radiofrequency ablation (RFA) as a localized therapy for hepatic metastasis is limited. Therefore, we conducted this retrospective study to provide clinical evidence.
    METHODS: This is a single-center, retrospective, cohort study. After radical pancreaticoduodenectomy, 32 patients developed metachronous hepatic metastasis with fewer than 3 lesions, the largest of which was less than 3 cm in diameter. These patients underwent combined treatment with chemotherapy and RFA. After 8 weeks of chemotherapy, patients received RFA for hepatic lesions. Additional chemotherapy was administered, and the patients\' tumor status and survival were monitored. The primary endpoint of this study was overall survival (OS). Factors affecting OS were analyzed using the Cox risk model.
    RESULTS: Among the 32 patients, the mean OS was 28.4 months. Univariate and multivariate Cox regression analysis revealed that the time (in months) of liver metastasis (HR = 0.04, 95% CI: 0.01 to 0.19; P < 0.001), the number of liver metastases (HR = 7.08, 95% CI: 1.85 to 27.08, P = 0.004), and PD (progressive disease) response to the second round of chemotherapy (HR = 29.50, 95% CI: 1.46 to 597.27; P = 0.027) were independent predictors of poorer survival.
    CONCLUSIONS: Combined therapy with RFA and chemotherapy is safe in patients with hepatic metastasis after radical pancreaticoduodenectomy. Early recurrence (≤12 months), three liver metastatic lesions, and a poor response to the second round of chemotherapy were associated with poor survival.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:胰腺神经内分泌肿瘤(panNET)患者R0切除术后肝转移对预后有显著影响。结合计算病理学和深度学习影像组学可以增强panNET患者术后肝转移的检测。
    方法:临床数据,病理学幻灯片,收集了复旦大学上海肿瘤中心(FUSCC)和FUSCC病理咨询中心R0切除术后的163例panNET患者的X线图像。数字图像分析和深度学习在Ki67染色的整个载玻片图像(WSI)和增强CT扫描中识别出肝转移相关特征,以创建列线图。该模型的性能在内部和外部测试队列中都得到了验证。
    结果:多因素logistic回归分析确定神经浸润是肝转移的独立危险因素(p<0.05)。Pathomics评分,这是基于热点和Ki67染色的异质性分布,显示肝转移的预测准确性提高(AUC=0.799)。深度学习-影像组学(DLR)评分的AUC为0.875。综合列线图,结合临床,病态,和成像功能,表现突出,训练队列的AUC为0.985,验证队列的AUC为0.961。高危组的中位无复发生存期为28.5个月,而低危组的中位无复发生存期为34.7个月。与预后显著相关(p<0.05)。
    结论:整合了计算病理学评分和深度学习影像组学的新预测模型可以更好地预测panNET患者术后肝转移,帮助临床医生开发个性化治疗。
    BACKGROUND: Postoperative liver metastasis significantly impacts the prognosis of pancreatic neuroendocrine tumor (panNET) patients after R0 resection. Combining computational pathology and deep learning radiomics can enhance the detection of postoperative liver metastasis in panNET patients.
    METHODS: Clinical data, pathology slides, and radiographic images were collected from 163 panNET patients post-R0 resection at Fudan University Shanghai Cancer Center (FUSCC) and FUSCC Pathology Consultation Center. Digital image analysis and deep learning identified liver metastasis-related features in Ki67-stained whole slide images (WSIs) and enhanced CT scans to create a nomogram. The model\'s performance was validated in both internal and external test cohorts.
    RESULTS: Multivariate logistic regression identified nerve infiltration as an independent risk factor for liver metastasis (p < 0.05). The Pathomics score, which was based on a hotspot and the heterogeneous distribution of Ki67 staining, showed improved predictive accuracy for liver metastasis (AUC = 0.799). The deep learning-radiomics (DLR) score achieved an AUC of 0.875. The integrated nomogram, which combines clinical, pathological, and imaging features, demonstrated outstanding performance, with an AUC of 0.985 in the training cohort and 0.961 in the validation cohort. High-risk group had a median recurrence-free survival of 28.5 months compared to 34.7 months for the low-risk group, showing significant correlation with prognosis (p < 0.05).
    CONCLUSIONS: A new predictive model that integrates computational pathologic scores and deep learning-radiomics can better predict postoperative liver metastasis in panNET patients, aiding clinicians in developing personalized treatments.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    多学科技术在内分泌相关癌症管理中的应用对于利用多学科的优势及其在消除肿瘤方面的协调努力至关重要。由于癌细胞的恶性特征,它们具有对化疗和放疗等传统治疗产生抵抗力的能力。然而,尽管努力加强对结果的预测,内分泌相关恶性肿瘤患者的总生存率仍然相当低.因此,研究创新的治疗策略势在必行.治疗策略的最新进展为各种内分泌肿瘤的治疗提供了新的方法。本文研究了纳米药物递送技术的进展以及通过靶向治疗将纳米材料用于精确癌症治疗的应用。这篇综述全面分析了联合药物递送策略在甲状腺癌治疗中的潜力。肾上腺肿瘤,还有胰腺癌.这项研究的目的是更深入地了解当前的治疗方法,刺激新药DDS的开发,并提高这些疾病患者的治疗效果。通过将合成或天然物质植入纳米颗粒中,可以显着提高药物对癌细胞的细胞内摄取,导致内分泌恶性肿瘤的发展大幅减少。
    The application of multidisciplinary techniques in the management of endocrine-related cancers is crucial for harnessing the advantages of multiple disciplines and their coordinated efforts in eliminating tumors. Due to the malignant characteristics of cancer cells, they possess the capacity to develop resistance to traditional treatments such as chemotherapy and radiotherapy. Nevertheless, despite diligent endeavors to enhance the prediction of outcomes, the overall survival rate for individuals afflicted with endocrine-related malignancy remains quite miserable. Hence, it is imperative to investigate innovative therapy strategies. The latest advancements in therapeutic tactics have offered novel approaches for the therapy of various endocrine tumors. This paper examines the advancements in nano-drug delivery techniques and the utilization of nanomaterials for precise cancer cures through targeted therapy. This review provides a thorough analysis of the potential of combined drug delivery strategies in the treatment of thyroid cancer, adrenal gland tumors, and pancreatic cancer. The objective of this study is to gain a deeper understanding of current therapeutic approaches, stimulate the development of new drug DDS, and improve the effectiveness of treatment for patients with these diseases. The intracellular uptake of pharmaceuticals into cancer cells can be significantly improved through the implantation of synthetic or natural substances into nanoparticles, resulting in a substantial reduction in the development of endocrine malignancies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:相关调查显示,胰腺癌患者的精神并发症发生率很高。虽然有研究探讨了胰腺癌患者心理并发症的影响因素,在其他人群中验证的一些因素尚未在胰腺癌人群中得到证实.本研究旨在探讨胰腺癌患者精神并发症的预测因素。
    方法:温州医科大学附属乐清市人民医院收治的胰腺癌患者,从2021年1月至2022年1月进行回顾性分析。护士使用基于精神障碍诊断和统计手册(DSM-IV)的结构化临床访谈(SCID-I)来评估住院期间(基线)和治疗开始后3个月的精神并发症的发生率。二元逻辑回归用于确定精神病并发症的预测因素。
    结果:80例患者纳入本研究,8例患者诊断为精神并发症。在其余72名患者中,8例患者(11.11%)在3个月随访时出现新发精神并发症。性别(赔率比(OR)=1.674,p=0.019),月收入(OR=1.735,p=0.023)和悲伤(MDAnderson症状量表(MDASI))(OR=1.804,p=0.001)均为胰腺癌患者精神并发症的预测因子.
    结论:性别,月收入和MDASI评分是胰腺癌患者精神并发症的预测因素.
    BACKGROUND: The relevant survey has shown a high incidence of psychiatric complications in patients with pancreatic cancer. While some studies have explored the factors influencing psychological complications in pancreatic cancer patients, some factors validated in other populations have not been confirmed in the pancreatic cancer population. This study aims to explore the predictors of psychiatric complications in patients with pancreatic cancer.
    METHODS: Patients with pancreatic cancer admitted to Yueqing People\'s Hospital Affiliated to Wenzhou Medical University, from January 2021 to January 2022 were retrospectively analyzed. The structured clinical interview (SCID-I) based on Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) was used by nurses to assess the incidence of psychiatric complications during hospitalization (baseline) and 3 months after the start of treatment. Binary logistic regression was used to identify predictors of psychiatric complications.
    RESULTS: 80 patients were enrolled in this study and 8 patients were diagnosed with psychiatric complications at base line. Among the rest 72 patients, 8 patients (11.11%) had new-onset psychiatric complications at 3-month follow-up. Gender (Odds Ratio (OR) = 1.674, p = 0.019), monthly income (OR = 1.735, p = 0.023) and sadness (M.D. Anderson Symptom Inventory (MDASI)) (OR = 1.804, p = 0.001) were all predictors for psychiatric complications in patients with pancreatic cancer.
    CONCLUSIONS: Gender, monthly income and MDASI score are predictors of psychiatric complications in patients with pancreatic cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:AGTPBP1是一种胞质羧肽酶,可从α/β微管蛋白的C末端或侧链裂解聚谷氨酸。尽管其表达失调与非小细胞肺癌的发展有关,AGTPBP1在胰腺癌(PC)中的具体作用和机制尚未完全了解。在这项研究中,我们研究了AGTPBP1在体外和体内对PC的作用。
    方法:免疫组化法检测AGTPBP1在PC和非癌组织中的表达。此外,我们在体外和体内评估了siRNA介导的AGTPBP1敲低后PC细胞的恶性行为。进行RNA测序和生物信息学分析以鉴定AGTPBP1调控的差异表达基因。
    结果:我们确定AGTPBP1在PC组织中过度表达,并且AGTPBP1的高表达与肿瘤的位置密切相关。AGTPBP1抑制可显著降低体内和体外细胞进展。此外,AGTPBP1敲低抑制ERK1/2、P-ERK1/2、MYLK的表达,和TUBB4B蛋白通过ERK信号通路。
    结论:我们的研究表明AGTPBP1可能是PC的推定治疗靶点。
    BACKGROUND: AGTPBP1 is a cytosolic carboxypeptidase that cleaves poly-glutamic acids from the C terminus or side chains of α/β tubulins. Although its dysregulated expression has been linked to the development of non-small cell lung cancer, the specific roles and mechanisms of AGTPBP1 in pancreatic cancer (PC) have yet to be fully understood. In this study, we examined the role of AGTPBP1 on PC in vitro and in vivo.
    METHODS: Immunohistochemistry was used to examine the expression of AGTPBP1 in PC and non-cancerous tissues. Additionally, we assessed the malignant behaviors of PC cells following siRNA-mediated AGTPBP1 knockdown both in vitro and in vivo. RNA sequencing and bioinformatics analysis were performed to identify the differentially expressed genes regulated by AGTPBP1.
    RESULTS: We determined that AGTPBP1 was overexpressed in PC tissues and the higher expression of AGTPBP1 was closely related to the location of tumors. AGTPBP1 inhibition can significantly decrease cell progression in vivo and in vitro. Moreover, the knockdown of AGTPBP1 inhibited the expression of ERK1/2, P-ERK1/2, MYLK, and TUBB4B proteins via the ERK signaling pathway.
    CONCLUSIONS: Our research indicates that AGTPBP1 may be a putative therapeutic target for PC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号