Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cancer Lett ; 590: 216840, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38604311

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal malignancies, highlighting the urgent need to elucidate the underlying oncogenic mechanisms. VIRMA is a classic isoform of methyltransferases that participates in epigenetic transcriptomic modification in eukaryotic mRNAs. However, the exact roles of VIRMA in PDAC remain unclear. Here, we identified that VIRMA is highly expressed in PDAC, and histone modifications of the promoter may partly account for this dysregulation. Moreover, VIRMA is closely related to glycolysis and poor prognosis in PDAC. We further determined that STRA6 is a direct downstream target of VIRMA in PDAC by RNA sequencing (RNA-seq) and m6A sequencing (m6A-seq). VIRMA is involved in gene expression regulation via 3' UTR targeting of STRA6 mRNA. Furthermore, the m6A reader IGF2BP2 was shown to critically contribute to the stability of STRA6 mRNA. We describe the role of VIRMA in promoting signaling via the STRA6/STAT3 axis, which results in increased levels of HIF-1α, a key activator of glycolysis. In vivo and in vitro experiments reveal that the VIRMA-STRA6-STAT3-HIF-1α axis plays an instrumental role in glycolysis and tumor progression in PDAC. In conclusion, we demonstrate that VIRMA can increase glycolysis in PDAC by upregulating STRA6, a cell surface membrane protein that stimulates the STAT3 pathway, thereby activating HIF-1α and leading to pancreatic cancer malignancy. Overall, our data strongly suggest that the VIRMA-STRA6-STAT3-HIF-1α axis is a viable therapeutic target in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Gene Expression Regulation, Neoplastic , Glycolysis , Pancreatic Neoplasms , Up-Regulation , Humans , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Glycolysis/genetics , Cell Line, Tumor , Animals , Disease Progression , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Male , Mice, Nude , Signal Transduction
2.
Cancer Lett ; 588: 216784, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38458594

ABSTRACT

Glycolytic metabolism is a hallmark of pancreatic ductal adenocarcinoma (PDAC), and tumor-associated stromal cells play important roles in tumor metabolism. We previously reported that tumor-associated macrophages (TAMs) facilitate PDAC progression. However, little is known about whether TAMs are involved in regulating glycolysis in PDAC. Here, we found a positive correlation between CD68+ TAM infiltration and FDG maximal standardized uptake (FDG SUVmax) on PET-CT images of PDAC. We discovered that the glycolytic gene set was prominently enriched in the high TAM infiltration group through Gene Set Enrichment Analysis using The Cancer Genome Atlas database. Mechanistically, TAMs secreted IL-8 to promote GLUT3 expression in PDAC cells, enhancing tumor glycolysis both in vitro and in vivo, whereas this effect could be blocked by the IL-8 receptor inhibitor reparixin. Furthermore, IL-8 promoted the translocation of phosphorylated STAT3 into the nucleus to activate the GLUT3 promoter. Overall, we demonstrated that TAMs boosted PDAC cell glycolysis through the IL-8/STAT3/GLUT3 signaling pathway. Our cumulative findings suggest that the abrogation of TAM-induced tumor glycolysis by reparixin might exhibit an antitumor impact and offer a potential therapeutic target for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Sulfonamides , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Tumor-Associated Macrophages/metabolism , Fluorodeoxyglucose F18/therapeutic use , Positron Emission Tomography Computed Tomography , Macrophages/metabolism , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Signal Transduction , Glycolysis , Cell Line, Tumor , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
3.
J Transl Med ; 21(1): 43, 2023 01 23.
Article in English | MEDLINE | ID: mdl-36691046

ABSTRACT

BACKGROUND: Intrahepatic cholangiocarcinoma (ICC) is a malignant disease characterized by onset occult, rapid progression, high relapse rate, and high mortality. However, data on how the tumor microenvironment (TME) regulates ICC metastasis at the transcriptomic level remains unclear. This study aimed to explore the mechanisms and interactions between hepatocytes and ICC cells. METHODS: We analyzed the interplay between ICC and liver microenvironment through cytokine antibody array analysis. Then we investigated the role of N6-methyladenosine (m6A) modification and the downstream target in vitro, in vivo experiments, and in clinical specimens. RESULTS: Our study demonstrated that cytokine CCL3, which is secreted by hepatocytes, promotes tumor metastasis by regulating m6A modification via vir-like m6A methyltransferase associated (VIRMA) in ICC cells. Moreover, immunohistochemical analyses showed that VIRMA correlated with poor outcomes in ICC patients. Finally, we confirmed both in vitro and in vivo that CCL3 could activate VIRMA and its critical downstream target SIRT1, which fuels tumor metastasis in ICC. CONCLUSIONS: In conclusion, our results enhanced our understanding of the interaction between hepatocytes and ICC cells, and revealed the molecular mechanism of the CCL3/VIRMA/SIRT1 pathway via m6A-mediated regulation in ICC metastasis. These studies highlight potential targets for the diagnosis, treatment, and prognosis of ICC.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Sirtuin 1 , Neoplasm Recurrence, Local , Cholangiocarcinoma/metabolism , Prognosis , Bile Ducts, Intrahepatic/metabolism , Bile Duct Neoplasms/metabolism , Hepatocytes/pathology , Cytokines , Cell Line, Tumor , Tumor Microenvironment
4.
Cancers (Basel) ; 14(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36230646

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the highest mortalities malignant tumors, which is characterized by difficult diagnosis, rapid progression and high recurrence rate. Nevertheless, PDAC responds poorly to conventional therapies, which highlights the urgency to identify novel prognostic and therapeutic targets. LEMT2 was a newly discovered protein-encoding gene with little cancer research and an unclear mechanism. Thus, this study aimed to illustrate LETM2 as the crucial oncogene for tumor progression in PDAC. In this study, we analyzed the expression level and prognostic value of LETM2 in multiple cancers using pan-cancer analysis. The analyses based on the TCGA-GTEx dataset indicated that the LETM2 expression was obviously elevated in several cancers, and it was the most significantly related to the dismal prognosis of PDAC. Subsequently, we demonstrated the functional role and mechanism of LETM2 by clinical sample evaluation, and in in vitro and in vivo experiments. Immunohistochemical analyses showed that high expression of LETM2 was correlated with poor outcomes of PDAC. Moreover, we demonstrated that LETM2 knockdown significantly inhibited tumor proliferation and metastasis, and promoted cell apoptosis, while LETM2 overexpression exerted the opposite effects. Finally, the impairment caused by LETM2-knockdown could be recovered via excitation of the PI3k-Akt pathway in vitro and in vivo animal models, which suggested that LETM2 could activate the downstream PI3K-Akt pathway to participate in PDAC progression. In conclusion, the study enhanced our understanding of LETM2 as an oncogene hallmark of PDAC. LETM2 may facilitate tumor progression by activating the PI3K-Akt signaling pathway, which provides potential targets for the diagnosis, treatment, and prognosis of pancreatic cancer.

5.
Biomater Sci ; 10(20): 5989-6002, 2022 Oct 11.
Article in English | MEDLINE | ID: mdl-36052559

ABSTRACT

Pancreatic stellate cells (PSCs) constitute the fibrotic tumor microenvironment composed of the stroma matrix, which blocks the penetration of gemcitabine (GEM) in pancreatic adenocarcinoma (PDAC) and results in chemoresistance. We analyzed the expression of α-SMA, collagen type I, and fibronectin by immunohistochemistry of pancreatic cancer tissues and demonstrated that the abundant interstitial stroma is associated with dismal survival. Two desmoplastic pancreatic tumor models are treated with arsenic trioxide (ATO) and GEM to confirm the sensitizing effect of ATO on GEM. RNA-seq was performed to analyze the potential fibrotic genes regulated by ATO. Western blotting, CCK-8 methods, colony formation, and wound healing and transwell assays were utilized to verify that ATO attenuates the tumor-promoting ability of PSCs by inhibiting its activation and decreasing matrix secretion via the PI3K/AKT/AP4/galectin-1 pathway. Furthermore, we developed targeted ATO-loaded nanoparticles self-assembled by poly (D,L-lactide) and poly(ethylene glycol) (PEG-PDLLA) and modified by the single-chain antibody of FAP-α (scAbFAP-α) (scAb-ATO-NPs) to promote the delivery efficiency of ATO to PSCs and enhance anti-tumor effects with gemcitabine. Herein, we elucidate the mechanism of how ATO inhibits the activation of PSCs and enhances the therapeutic effect of GEM. We propose a novel cocktail therapy including scAb-ATO-NPs and GEM, indicating a new perspective in the treatment of PDAC.


Subject(s)
Adenocarcinoma , Nanoparticles , Pancreatic Neoplasms , Adenocarcinoma/pathology , Arsenic Trioxide/therapeutic use , Cell Line, Tumor , Collagen Type I/metabolism , Deoxycytidine/analogs & derivatives , Fibronectins/metabolism , Galectin 1/metabolism , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Phosphatidylinositol 3-Kinases/metabolism , Polyethylene Glycols/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Sincalide/metabolism , Sincalide/pharmacology , Sincalide/therapeutic use , Gemcitabine , Pancreatic Neoplasms
6.
Cancer Med ; 7(11): 5679-5690, 2018 11.
Article in English | MEDLINE | ID: mdl-30311406

ABSTRACT

Tumor-associated macrophages (TAMs) are abundant population of inflammatory cells which play an essential role in remodeling tumor microenvironment and tumor progression. Previously, we found the high density of TAMs was correlated with lymph node metastasis and poor prognosis in pancreatic ductal adenocarcinoma (PDAC). Therefore, this study was designed to investigate the mechanisms of interaction between TAMs and PDAC. THP-1 monocytes were the exposure to conditioned media (CM) produced by PDAC cells; then, monocyte recruitment and macrophage differentiation were assessed. CM from PDAC attracted and polarized THP-1 monocytes to tumor-driven like macrophages. mRNA expression cytokine profiling and ELISA identified the IL-8 secretion was increasing in tumor-driven like macrophages, and STAT3 pathway was involved. Addition of exogenous recombinant human IL-8 promoted PDAC cells motility in vitro and metastasis in vivo via upregulating Twist expression, which mediated epithelial-mesenchymal transition in cancer cells. What is more, IL-8 expression level in tumor stroma by immunohistochemical analysis was related to lymph node metastasis, the number of tumor CD68 but not CD163 positive macrophages and patient outcome. Taken together, these findings shed light on the important interplay between cancer cells and TAMs in tumor microenvironment and suggested that IL-8 signaling might be a potential therapeutic target for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Interleukin-8/genetics , Interleukin-8/metabolism , Macrophages/metabolism , Monocytes/cytology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Culture Media, Conditioned/pharmacology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Macrophages/pathology , Mice , Monocytes/drug effects , Monocytes/metabolism , Neoplasm Metastasis , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , THP-1 Cells , Tumor Microenvironment , Up-Regulation , Pancreatic Neoplasms
7.
Theranostics ; 8(11): 3074-3086, 2018.
Article in English | MEDLINE | ID: mdl-29896303

ABSTRACT

The molecular mechanism of perineural invasion (PNI) is unclear, and insufficient detection during early-stage PNI in vivo hampers its investigation. We aimed to identify a cytokine paracrine loop between pancreatic ductal adenocarcinoma (PDAC) cells and nerves and established a noninvasive method to monitor PNI in vivo. Methods: A Matrigel/ dorsal root ganglia (DRG) system was used to observe PNI in vitro, and a murine sciatic nerve invasion model was established to examine PNI in vivo. PNI was assessed by MRI with iron oxide nanoparticle labeling. We searched publicly available datasets as well as obtained PDAC tissues from 30 patients to examine MMP1 expression in human tumor and non-tumor tissues. Results: Our results showed that matrix metalloproteinase-1 (MMP1) activated AKT and induced protease-activated receptor-1 (PAR1)-expressing DRG to release substance P (SP), which, in turn, activated neurokinin 1 receptor (NK1R)-expressing PDAC cells and enhanced cellular migration, invasion, and PNI via SP/NK1R/ERK. In animals, hind limb paralysis and a decreased hind paw width were observed approximately 20 days after inoculation of cancer cells in the perineurium. MMP1 silencing with shRNA or treatment with either a PAR1 or an NK1R antagonist inhibited PNI. MRI detected PNI as early as 10 days after implantation of PDAC cells. PNI also induced PDAC liver metastasis. Bioinformatic analyses and pathological studies on patient tissues corroborated the clinical relevance of these findings. Conclusion: In this study, we provided evidence that the MMP1/PAR1/SP/NK1R paracrine loop contributes to PNI during the early stage of primary tumor formation. Furthermore, we established a sensitive and non-invasive method to detect nerve invasion using iron oxide nanoparticles and MRI.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Matrix Metalloproteinase 1/metabolism , Pancreatic Neoplasms/pathology , Paracrine Communication , Receptor, PAR-1/metabolism , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Animals , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement , Ferric Compounds/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Humans , Matrix Metalloproteinase 1/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Murinae , Nanoparticles , Neoplasm Metastasis , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Receptor, PAR-1/genetics , Receptors, Neurokinin-1/genetics , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Substance P/genetics
8.
Int J Cancer ; 142(11): 2323-2334, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29315556

ABSTRACT

Oncogenic KRAS plays a crucial role in pancreatic ductal adenocarcinoma (PDAC) development and progression. However, the mechanism has not been clearly elucidated. RKIP is a tumor repressor, and loss of RKIP has been shown in PDAC. Here, we found that KRAS expression was inversely correlated with RKIP expression in PDAC fresh tissue regardless of the KRAS mutant status. The negative correlation between KRAS and RKIP was further confirmed in our PDAC tissue microarray. KRAS overexpression and RKIP downregulation were associated with poor clinical outcomes. Knockdown or overexpression of KRAS in PDAC cell lines robustly increased or decreased, respectively, RKIP protein and mRNA levels. Furthermore, the MAPK-ERK pathway was involved in the regulation of RKIP. KRAS-regulated RKIP expression, which in turn affected the expression of pivotal epithelial-mesenchymal transition (EMT) and apoptosis factors. The biological function of the KRAS-RKIP axis was demonstrated in human pancreatic cancer cells in vitro and in vivo. KRAS knockdown increased RKIP expression and inhibited metastasis and chemoresistance. Moreover, the feature of metastasis and chemoresistance was rescued in the KRAS-knockdown cells through the inhibition of RKIP by RNA interference. In conclusion, our studies demonstrate how KRAS inhibits the tumor suppressor RKIP, thus offering novel justification for targeting RKIP as a strategy to overcome KRAS-induced tumor metastasis and chemoresistance in PDAC.


Subject(s)
Drug Resistance, Neoplasm/genetics , MAP Kinase Signaling System , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phosphatidylethanolamine Binding Protein/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Genotype , Humans , Immunohistochemistry , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Phosphatidylethanolamine Binding Protein/metabolism , Protein Binding , Proto-Oncogene Proteins p21(ras)/metabolism
9.
Cancer Sci ; 108(7): 1493-1503, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28444967

ABSTRACT

Tumor metastasis occurs naturally in pancreatic cancer, and the efficacy of chemotherapy is usually poor. Precision medicine, combining downregulation of target genes with chemotherapy drugs, is expected to improve therapeutic effects. Therefore, we developed a combined therapy of microRNA-21 antisense oligonucleotides (ASO-miR-21) and gemcitabine (Gem) using a targeted co-delivery nanoparticle (NP) carrier and investigated the synergistic inhibitory effects on pancreatic cancer cells metastasis and growth. Polyethylene glycol-polyethylenimine-magnetic iron oxide NPs were used to co-deliver ASO-miR-21 and Gem. An anti-CD44v6 single-chain variable fragment (scFvCD44v6 ) was used to coat the particles to obtain active and targeted delivery. Our results showed that the downregulation of the oncogenic miR-21 by ASO resulted in upregulation of the tumor-suppressor genes PDCD4 and PTEN and the suppression of epithelial-mesenchymal transition, which inhibited the proliferation and induced the clonal formation, migration, and invasion of pancreatic cancer cells in vitro. The co-delivery of ASO-miR-21 and Gem induced more cell apoptosis and inhibited the growth of pancreatic cancer cells to a greater extent than single ASO-miR-21 or Gem treatment in vitro. In animal tests, more scFvCD44v6 -PEG-polyethylenimine/ASO-magnetic iron oxide NP/Gem accumulated at the tumor site than non-targeted NPs and induced a potent inhibition of tumor proliferation and metastasis. Magnetic resonance imaging was used to observed tumor homing of NPs. These results imply that the combination of miR-21 gene silencing and Gem therapy using an scFv-functionalized NP carrier exerted synergistic antitumor effects on pancreatic cancer cells, which is a promising strategy for pancreatic cancer therapy.


Subject(s)
Deoxycytidine/analogs & derivatives , Genetic Therapy/methods , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy/methods , Oligonucleotides, Antisense/administration & dosage , Pancreatic Neoplasms/pathology , Animals , Deoxycytidine/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Female , Ferric Compounds , Humans , Hyaluronan Receptors/administration & dosage , Metal Nanoparticles/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Nanomedicine/methods , Polyethylene Glycols , Polyethyleneimine , Precision Medicine/methods , Single-Chain Antibodies/administration & dosage , Xenograft Model Antitumor Assays , Gemcitabine
10.
Tumour Biol ; 37(8): 11299-309, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26951514

ABSTRACT

It is critical to understand the pathogenesis of preinvasive stages of pancreatic duct adenocarcinoma (PDAC) for developing novel potential diagnostic and therapeutic targets. The polycomb group family member B-lymphoma Moloney murine leukemia virus insertion region-1 (Bmi1) is overexpressed and involved in cancer progression in PDAC; however, its role in the multistep malignant transformation of human pancreatic duct cells has not been directly demonstrated. In this study, we stably expressed Bmi1 in a model of telomerase-immortalized human pancreatic duct-derived cells (HPNE) and showed that Bmi1 promoted HPNE cell proliferation, migration, and invasion but not malignant transformation. We then used mutant KRASG12D as a second oncogene to transform HPNE cells and showed that it further enhanced Bmi1-induced malignant potential. More importantly, coexpression of KRASG12D and Bmi1 caused anchorage-independent growth transformation in vitro but still failed to produce tumors in nude mice. Finally, we found that mutant KRASG12D induced HPNE-Bmi1 cells to undergo partial epithelial-mesenchymal transition (EMT) likely via upregulation of snail. Knockdown of KRASG12D significantly reduced the expression of snail and vimentin at both the messenger RNA (mRNA) and protein level and further impaired the anchorage-independent growth capability of invasive cells. In summary, our findings demonstrate that coexpression of Bmi1 and KRASG12D could lead to transformation of HPNE cells in vitro and suggest potential new targets for diagnosis and treatment of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Pancreatic Neoplasms/pathology , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Heterografts , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Gastric Cancer ; 19(2): 392-402, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25764514

ABSTRACT

BACKGROUND: B-cell-specific Moloney murine leukemia virus integration site 1 (Bmi-1) and Raf kinase inhibitory protein (RKIP) are involved in cancer metastasis and chemotherapeutic resistance, respectively. In this study, we evaluated the association between Bmi-1 and RKIP and outcome of gastric cancer through clinical data analysis and in vitro experiments. METHODS: Bmi-1 expression and RKIP expression were observed in 107 cases of gastric cancer through use of tissue microarray technology to identify their correlations with clinicopathological parameters, patient survival, and susceptibility to chemotherapy. The correlation was confirmed in gastric cancer cell lines, analyzed further by gene overexpression and silencing analysis, a cell invasion assay, and a chemosensitivity test. RESULTS: Positive expression of Bmi-1 was highly correlated with T classification and clinical stage. Diminished or lost expression of RKIP was significantly associated with T classification, lymph node metastasis, distant metastasis, and clinical stage. Bmi-1 is negatively and RKIP is positively related to patient survival. Positive expression of Bmi-1 and negative expression of RKIP are associated with poor patient survival and modest efficacy of postoperative chemotherapy. A meaningfully inverse association between Bmi-1 and RKIP was found in tissue microarray studies, and was verified further in gastric cancer cell lines. Moreover, gene overexpression and silencing analysis indicated that RKIP might be regulated by Bmi-1. Furthermore, the impacts of Bmi-1 on cell invasion and chemotherapy resistance were rescued by knockdown of RKIP. CONCLUSIONS: Our study implies that detection of Bmi-1 and RKIP is valuable in predicting patient survival and therapeutic response in gastric cancer, and the inverse association between Bmi-1 and RKIP reveals the potential molecular mechanisms underlying tumor metastasis and chemotherapy resistance.


Subject(s)
Drug Resistance, Neoplasm , Phosphatidylethanolamine Binding Protein/metabolism , Polycomb Repressive Complex 1/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Aged , Cell Line, Tumor , Female , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis/pathology , Male , Middle Aged , Organoplatinum Compounds/pharmacology , Oxaliplatin , Phosphatidylethanolamine Binding Protein/genetics , Polycomb Repressive Complex 1/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Tissue Array Analysis
12.
J Biomed Nanotechnol ; 12(8): 1654-66, 2016 Aug.
Article in English | MEDLINE | ID: mdl-29342344

ABSTRACT

BACKGROUND: The concept of precision medicine to treat cancer shows promise and a co-delivery carrier for chemotherapy drugs and target genes is the key tool for both basic research and clinical application. To address this, we developed a cancer-targeting nanoparticle vector to transfer gemcitabine (Gem) and small interfering RNA (siRNA) to pancreatic cancer. METHODS: Iron oxide nanoparticles (IONPs) resonant at 15 nm were conjugated with the single chain variable fragment (scFv) against CD44v6 (scFvCD44v6), which has proven pancreatic cancer-targeting specificity as reported in our previous study. Gem was then linked through a lysosomally cleavable tetrapeptide linker, resulting in a scFv-targeted nanoparticle construct, which was subsequently conjugated to siRNA targeting the Bmi-1 oncogene (siBmi-1) to obtain the multifunctional nanoparticle scFv-Gem-siBmi-1-NPs. A series of biological experiments were performed to test its biophysical characterization, gene silencing efficacy and anti-tumor effect in vitro and in vivo. RESULTS: The multifunctional nanoparticle not only possesses an ultra-small size of approximately 80 nm, excellent biocompatibility and biodegradability, but also exerts a synergistic anti-tumor effect both in vitro and in vivo, such as inhibition of tumor cell growth, invasion and migration, reduction of cell cycle progression and promotion of tumor apoptosis. Furthermore, this nanoparticle can efficiently target pancreatic cancer in vivo, resulting in the enhanced bioavailability and efficacy of Gem. CONCLUSION: scFv-Gem-siBmi-1-NPs provide an effective and targeted co-delivery of Gem and siBmi-1 to pancreatic cancer, and exert an efficient and corporate anti-tumor therapeutic effect. This prospective vector shows promise for precise treatment of pancreatic cancer.


Subject(s)
Antineoplastic Agents , Deoxycytidine/analogs & derivatives , Magnetite Nanoparticles/chemistry , Pancreatic Neoplasms/metabolism , RNA, Small Interfering , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Physiological Phenomena/drug effects , Deoxycytidine/chemistry , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Female , Humans , Hyaluronan Receptors , Mice , Mice, Inbred BALB C , Nanoconjugates/chemistry , Particle Size , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Xenograft Model Antitumor Assays , Gemcitabine
13.
Medicine (Baltimore) ; 94(47): e2119, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26632732

ABSTRACT

Primary gastrointestinal lymphoma (PGIL) is a rare malignant tumor without standard diagnosis and treatment methods. This study is aimed to systematically analyze its clinical characteristics and draw out an appropriate flow chart of diagnosis and treatment process for PGIL in China.This study retrospectively analyzed the clinicopathological characteristics, diagnostic approaches, prognostic factors, and therapeutic modalities in 415 cases of PGIL in Chinese province of Guangdong. A systematic review was conducted in 118 studies containing 5075 patients to further identify clinical manifestations and mortalities of therapeutic modalities.The most common clinical presentations were abdominal pain and bloody stools. Endoscopic biopsy was an important diagnostic means, and usually more than once to make a definite diagnosis. Retrospective multicenter clinical study showed that younger onset age (<60 years), female, one region involved, one lesion, early stage, International Prognostic Index (IPI ≤1), normal lactate dehydrogenase (LDH), normal albumin, and nonemergency operation were significant prognostic factors for B-cell lymphoma; non-B symptom, tumor restricted to gastric or ileocecal region, one lesion, performance status (PS ≤1), normal LDH, and nonsurgery alone were significant prognostic factors for T-cell lymphoma. Site of origin and IPI were independent prognostic factors for B-cell lymphoma; PS was the independent prognostic factor for T-cell lymphoma. And T-cell lymphoma had worse overall survival (OS) and progression-free survival (PFS) than B-cell lymphoma. Among different therapeutic modalities, chemotherapy alone or combined with surgery showed better OS and PFS than surgery alone for diffuse large B-cell lymphoma (DLBCL) of stage I/II E and T-cell lymphoma. For DLBCL of stage III E/IV and mucosa-associated lymphoid tissue lymphoma, OS and PFS did not differ among different therapeutic groups. In meta-analysis, surgery plus chemotherapy showed lowest mortality.Chemotherapy alone or combined with surgery may be the first-line treatment for DLBCL of stage I/II E and T-cell lymphoma. A flow chart of diagnosis and treatment process for PGIL was approximately drew out.


Subject(s)
Gastrointestinal Neoplasms/mortality , Gastrointestinal Neoplasms/pathology , Lymphoma/mortality , Lymphoma/pathology , Age Factors , China , Combined Modality Therapy , Endoscopy, Gastrointestinal , Female , Gastrointestinal Neoplasms/therapy , Humans , Kaplan-Meier Estimate , Lymphoma/therapy , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/pathology , Lymphoma, T-Cell/mortality , Lymphoma, T-Cell/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Sex Factors , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...