Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 84(7): 1206-1223.e15, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38423014

ABSTRACT

Appropriate DNA end synapsis, regulated by core components of the synaptic complex including KU70-KU80, LIG4, XRCC4, and XLF, is central to non-homologous end joining (NHEJ) repair of chromatinized DNA double-strand breaks (DSBs). However, it remains enigmatic whether chromatin modifications can influence the formation of NHEJ synaptic complex at DNA ends, and if so, how this is achieved. Here, we report that the mitotic deacetylase complex (MiDAC) serves as a key regulator of DNA end synapsis during NHEJ repair in mammalian cells. Mechanistically, MiDAC removes combinatorial acetyl marks on histone H2A (H2AK5acK9ac) around DSB-proximal chromatin, suppressing hyperaccumulation of bromodomain-containing protein BRD4 that would otherwise undergo liquid-liquid phase separation with KU80 and prevent the proper installation of LIG4-XRCC4-XLF onto DSB ends. This study provides mechanistic insight into the control of NHEJ synaptic complex assembly by a specific chromatin signature and highlights the critical role of H2A hypoacetylation in restraining unscheduled compartmentalization of DNA repair machinery.


Subject(s)
Chromatin , Nuclear Proteins , Animals , Chromatin/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , DNA/genetics , DNA End-Joining Repair , Histones/genetics , Histones/metabolism , Chromosome Pairing , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Mammals/metabolism
2.
Mol Cell ; 82(14): 2571-2587.e9, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35597237

ABSTRACT

The efficiency of homologous recombination (HR) in the repair of DNA double-strand breaks (DSBs) is closely associated with genome stability and tumor response to chemotherapy. While many factors have been functionally characterized in HR, such as TOPBP1, their precise regulation remains unclear. Here, we report that TOPBP1 interacts with the RNA-binding protein HTATSF1 in a cell-cycle- and phosphorylation-dependent manner. Mechanistically, CK2 phosphorylates HTATSF1 to facilitate binding to TOPBP1, which promotes S-phase-specific TOPBP1 recruitment to damaged chromatin and subsequent RPA/RAD51-dependent HR, genome integrity, and cancer-cell viability. The localization of HTATSF1-TOPBP1 to DSBs is potentially independent of the transcription-coupled RNA-binding and processing capacity of HTATSF1 but rather relies on the recognition of poly(ADP-ribosyl)ated RPA by HTATSF1, which can be blunted with PARP inhibitors. Together, our study provides a mechanistic insight into TOPBP1 loading at HR-prone DSB sites via HTATSF1 and reveals how RPA-RAD51 exchange is tuned by a PARylation-phosphorylation cascade.


Subject(s)
Poly ADP Ribosylation , Rad51 Recombinase , DNA Breaks, Double-Stranded , DNA Repair , Homologous Recombination/genetics , Phosphorylation , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
3.
Gastroenterology ; 167(2): 281-297, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38492894

ABSTRACT

BACKGROUND & AIMS: Because pancreatic cancer responds poorly to chemotherapy and immunotherapy, it is necessary to identify novel targets and compounds to overcome resistance to treatment. METHODS: This study analyzed genomic single nucleotide polymorphism sequencing, single-cell RNA sequencing, and spatial transcriptomics. Ehf-knockout mice, KPC (LSL-KrasG12D/+, LSL-Trp53R172H/+ and Pdx1-Cre) mice, CD45.1+ BALB/C nude mice, and CD34+ humanized mice were also used as subjects. Multiplexed immunohistochemistry and flow cytometry were performed to investigate the proportion of tumor-infiltrated C-X-C motif chemokine receptor 2 (CXCR2)+ neutrophils. In addition, multiplexed cytokines assays and chromatin immunoprecipitation assays were used to examine the mechanism. RESULTS: The TP53 mutation-mediated loss of tumoral EHF increased the recruitment of CXCR2+ neutrophils, modulated their spatial distribution, and further induced chemo- and immunotherapy resistance in clinical cohorts and preclinical syngeneic mice models. Mechanistically, EHF deficiency induced C-X-C motif chemokine ligand 1 (CXCL1) transcription to enhance in vitro and in vivo CXCR2+ neutrophils migration. Moreover, CXCL1 or CXCR2 blockade completely abolished the effect, indicating that EHF regulated CXCR2+ neutrophils migration in a CXCL1-CXCR2-dependent manner. The depletion of CXCR2+ neutrophils also blocked the in vivo effects of EHF deficiency on chemotherapy and immunotherapy resistance. The single-cell RNA-sequencing results of PDAC treated with Nifurtimox highlighted the therapeutic significance of Nifurtimox by elevating the expression of tumoral EHF and decreasing the weightage of CXCL1-CXCR2 pathway within the microenvironment. Importantly, by simultaneously inhibiting the JAK1/STAT1 pathway, it could significantly suppress the recruitment and function of CXCR2+ neutrophils, further sensitizing PDAC to chemotherapy and immunotherapies. CONCLUSIONS: The study demonstrated the role of EHF in the recruitment of CXCR2+ neutrophils and the promising role of Nifurtimox in sensitizing pancreatic cancer to chemotherapy and immunotherapy.


Subject(s)
Chemokine CXCL1 , Drug Resistance, Neoplasm , Neutrophil Infiltration , Neutrophils , Pancreatic Neoplasms , Receptors, Interleukin-8B , Animals , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Receptors, Interleukin-8B/antagonists & inhibitors , Humans , Neutrophil Infiltration/drug effects , Drug Resistance, Neoplasm/genetics , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/drug effects , Mice , Chemokine CXCL1/metabolism , Chemokine CXCL1/genetics , Cell Line, Tumor , Mice, Knockout , Tumor Microenvironment , Immunotherapy/methods , Mice, Nude , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mice, Inbred BALB C , Antineoplastic Agents/pharmacology , Signal Transduction , Mutation , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology
4.
Nucleic Acids Res ; 51(21): 11668-11687, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37831098

ABSTRACT

Unscheduled R-loops are a major source of replication stress and DNA damage. R-loop-induced replication defects are sensed and suppressed by ATR kinase, whereas it is not known whether R-loop itself is actively involved in ATR activation and, if so, how this is achieved. Here, we report that the nuclear form of RNA-editing enzyme ADAR1 promotes ATR activation and resolves genome-wide R-loops, a process that requires its double-stranded RNA-binding domains. Mechanistically, ADAR1 interacts with TOPBP1 and facilitates its loading on perturbed replication forks by enhancing the association of TOPBP1 with RAD9 of the 9-1-1 complex. When replication is inhibited, DNA-RNA hybrid competes with TOPBP1 for ADAR1 binding to promote the translocation of ADAR1 from damaged fork to accumulate at R-loop region. There, ADAR1 recruits RNA helicases DHX9 and DDX21 to unwind R-loops, simultaneously allowing TOPBP1 to stimulate ATR more efficiently. Collectively, we propose that the tempo-spatially regulated assembly of ADAR1-nucleated protein complexes link R-loop clearance and ATR activation, while R-loops crosstalk with blocked replication forks by transposing ADAR1 to finetune ATR activity and safeguard the genome.


Subject(s)
DNA-Binding Proteins , R-Loop Structures , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Replication , DNA-Binding Proteins/genetics , RNA/genetics , Humans , Animals , Mice
5.
Nucleic Acids Res ; 50(D1): D1408-D1416, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34570217

ABSTRACT

Interpreting the molecular mechanism of genomic variations and their causal relationship with diseases/traits are important and challenging problems in the human genetic study. To provide comprehensive and context-specific variant annotations for biologists and clinicians, here, by systematically integrating over 4TB genomic/epigenomic profiles and frequently-used annotation databases from various biological domains, we develop a variant annotation database, called VannoPortal. In general, the database has following major features: (i) systematically integrates 40 genome-wide variant annotations and prediction scores regarding allele frequency, linkage disequilibrium, evolutionary signature, disease/trait association, tissue/cell type-specific epigenome, base-wise functional prediction, allelic imbalance and pathogenicity; (ii) equips with our recent novel index system and parallel random-sweep searching algorithms for efficient management of backend databases and information extraction; (iii) greatly expands context-dependent variant annotation to incorporate large-scale epigenomic maps and regulatory profiles (such as EpiMap) across over 33 tissue/cell types; (iv) compiles many genome-scale base-wise prediction scores for regulatory/pathogenic variant classification beyond protein-coding region; (v) enables fast retrieval and direct comparison of functional evidence among linked variants using highly interactive web panel in addition to plain table; (vi) introduces many visualization functions for more efficient identification and interpretation of functional variants in single web page. VannoPortal is freely available at http://mulinlab.org/vportal.


Subject(s)
Databases, Genetic , Genetic Diseases, Inborn/genetics , Genetic Variation/genetics , Molecular Sequence Annotation , Algorithms , Epigenome/genetics , Genetic Diseases, Inborn/classification , Genome, Human/genetics , Genome-Wide Association Study , Genotype , Humans , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Software
6.
Gut ; 72(9): 1722-1737, 2023 09.
Article in English | MEDLINE | ID: mdl-36828627

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal tumour with limited treatment options. Here, we identified syndecan binding protein (SDCBP), also known as syntenin1, as a novel targetable factor in promoting PDAC tumour progression. We also explored a therapeutic strategy for suppressing SDCBP expression. DESIGN: We used samples from patients with PDAC, human organoid models, LSL-KrasG12D/+mice, LSL-Trp53R172H/+ and Pdx1-Cre (KPC) mouse models, and PDX mouse models. Immunostaining, colony formation assay, ethynyl-2-deoxyuridine incorporation assay, real-time cell analysis, cell apoptosis assay, automated cell tracking, invadopodia detection and gelatin degradation assays, coimmunoprecipitation, and pull-down assays were performed in this study. RESULTS: The median overall survival and recurrence-free survival rates in the high-SDCBP group were significantly shorter than those in the low-SDCBP group. In vitro and in vivo studies have demonstrated that SDCBP promotes PDAC proliferation and metastasis. Mechanically, SDCBP inhibits CK1δ/ε-mediated YAP-S384/S387 phosphorylation, which further suppresses ß-TrCP-mediated YAP1 ubiquitination and proteasome degradation by directly interacting with YAP1. SDCBP interacts with the TAD domain of YAP1, mainly through its PDZ1 domain. Preclinical KPC mouse cohorts demonstrated that zinc pyrithione (ZnPT) suppresses PDAC tumour progression by suppressing SDCBP. CONCLUSIONS: SDCBP promotes the proliferation and metastasis of PDAC by preventing YAP1 from ß-TrCP-mediated proteasomal degradation. Therefore, ZnPT could be a promising therapeutic strategy to inhibit PDAC progression by suppressing SDCBP.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Mice , Animals , beta-Transducin Repeat-Containing Proteins/metabolism , Pancreatic Neoplasms/pathology , Pancreas/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Syntenins/metabolism , Pancreatic Neoplasms
7.
Ann Surg ; 278(2): 253-259, 2023 08 01.
Article in English | MEDLINE | ID: mdl-35861061

ABSTRACT

BACKGROUND AND OBJECTIVE: Robotic distal pancreatectomy (DP) is an emerging attractive approach, but its role compared with laparoscopic or open surgery remains unclear. Benchmark values are novel and objective tools for such comparisons. The aim of this study was to identify benchmark cutoffs for many outcome parameters for DP with or without splenectomy beyond the learning curve. METHODS: This study analyzed outcomes from international expert centers from patients undergoing robotic DP for malignant or benign lesions. After excluding the first 10 cases in each center to reduce the effect of the learning curve, consecutive patients were included from the start of robotic DP up to June 2020. Benchmark patients had no significant comorbidities. Benchmark cutoff values were derived from the 75th or the 25th percentile of the median values of all benchmark centers. Benchmark values were compared with a laparoscopic control group from 4 high-volume centers and published open DP landmark series. RESULTS: Sixteen centers contributed 755 cases, whereof 345 benchmark patients (46%) were included the analysis. Benchmark cutoffs included: operation time ≤300 minutes, conversion rate ≤3%, clinically relevant postoperative pancreatic fistula ≤32%, 3 months major complication rate ≤26.7%, and lymph node retrieval ≥9. The comprehensive complication index at 3 months was ≤8.7 without deterioration thereafter. Compared with robotic DP, laparoscopy had significantly higher conversion rates (5×) and overall complications, while open DP was associated with more blood loss and longer hospital stay. CONCLUSION: This first benchmark study demonstrates that robotic DP provides superior postoperative outcomes compared with laparoscopic and open DP. Robotic DP may be expected to become the approach of choice in minimally invasive DP.


Subject(s)
Laparoscopy , Pancreatic Neoplasms , Robotic Surgical Procedures , Humans , Pancreatectomy/adverse effects , Pancreatic Neoplasms/surgery , Benchmarking , Standard of Care , Postoperative Complications/etiology , Laparoscopy/adverse effects , Length of Stay , Treatment Outcome , Retrospective Studies
8.
Gastroenterology ; 162(4): 1256-1271.e14, 2022 04.
Article in English | MEDLINE | ID: mdl-34951995

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is characterized by severe metabolic stress due to fibrosis and poor vascularization. BZW1 is an eIF5-mimic protein involved in tumorigenesis and progression. The aim of this study was to investigate the role of BZW1 in metabolic stress resistance in PDAC. METHODS: BZW1 expression was evaluated in human PDAC tissue microarray and PDAC cells. Glycolysis regulation of BZW1 and its correlation with glycolysis-related genes was analyzed. Tumor growth, cell proliferation, and apoptosis were evaluated in mice xenograft tumors and patient-derived organoids. RESULTS: The results of bioinformatic screening identified that BZW1 was 1 of the top 3 genes favorable for tumor progression in PDAC. The analysis of our cohort confirmed that BZW1 was overexpressed in human PDAC tissues compared with nontumor tissues, and its abnormal expression was correlated with large tumor size and poor prognosis. BZW1 promoted cell proliferation and inhibited apoptosis in both mouse xenograft models and PDAC-derived organoids via facilitating glycolysis in the oxygen-glucose-deprivation condition. Mechanically, BZW1 served as an adaptor for PKR-like endoplasmic reticulum (ER) kinase (PERK), facilitated the phosphorylation of eIF2α, promoted internal ribosome entry site-dependent translation of HIF1α and c-Myc, and thereby boosted the Warburg effect. In organoid-based xenografts with high BZW1 levels, both the PERK/eIF2α phosphorylation inhibitor GSK2606414 and ISRIB significantly suppressed tumor growth and prolonged animal survival. CONCLUSIONS: BZW1 is a key molecule in the internal ribosome entry site-dependent translation of HIF1α/c-Myc and plays crucial roles in the glycolysis of PDAC. BZW1 might serve as a therapeutic target for patients with pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Cell Cycle Proteins , DNA-Binding Proteins , Pancreatic Neoplasms , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Internal Ribosome Entry Sites , Mice , Pancreatic Neoplasms/pathology , Phosphorylation , Prognosis , Pancreatic Neoplasms
9.
Microb Ecol ; 87(1): 5, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38030815

ABSTRACT

Cholangiocarcinoma (CCA) is a serious health problem worldwide. The gut and bile microbiota have not been clearly characterized in patients with CCA, and better noninvasive diagnostic approaches for CCA need to be established. The aim of this study was to investigate the characteristics of the gut and bile microbiota in CCA patients. Forty-two CCA patients and 16 healthy normal controls (HNCs) were enrolled. DNA was extracted from fecal and bile samples and subjected to 16S rRNA gene analysis. We found that there were significant differences in the species diversity, structure, and composition of the microbial communities between the CCA group and the HNC grouAt the phylum level, compared with that in the HNC group, the relative abundance of Firmicutes and Actinobacteriota was significantly decreased in the CCA group, whereas Proteobacteria and Bacteroidota were significantly enriched. The Firmicutes/Bacteroidota (F/B) ratio significantly decreased in the CCA group compared to the HNC grouThe relative abundance of Klebsiella in the CCA group was significantly higher than that in the HNC group, while the relative abundance of Bifidobacterium was significantly decreased. The Bifidobacterium/Klebsiella (B/K) ratio was established as a novel biomarker and was found to be significantly decreased in the CCA group compared with the HNC grouOur findings provide evidence supporting the use of Klebsiella and Bifidobacterium as noninvasive intestinal microbiomarkers for improving the diagnosis of CCA.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Humans , Bifidobacterium/genetics , Klebsiella/genetics , RNA, Ribosomal, 16S/genetics , Bile , Firmicutes/genetics , Bacteroidetes/genetics , Feces/microbiology
10.
Gut ; 71(2): 357-371, 2022 02.
Article in English | MEDLINE | ID: mdl-33674341

ABSTRACT

BACKGROUND AND AIMS: The crosstalk between cancer stem cells (CSCs) and their niche is required for the maintenance of stem cell-like phenotypes of CSCs. Here, we identified E26 transformation-specific homologous factor (EHF) as a key molecule in decreasing the sensitivity of pancreatic cancer (PC) cells to CSCs' niche stimulus. We also explored a therapeutic strategy to restore the expression of EHF. DESIGN: We used a LSL-KrasG12D/+mice, LSL-Trp53R172H/+ and Pdx1-Cre (KPC) mouse model and samples from patients with PC. Immunostaining, flow cytometry, sphere formation assays, anchorage-independent growth assay, in vivo tumourigenicity, reverse transcription PCR, chromatin immunoprecipitation (ChIP) and luciferase analyses were conducted in this study. RESULTS: CXCL12 derived from pancreatic stellate cells (PSCs) mediates the crosstalk between PC cells and PSCs to promote PC stemness. Tumorous EHF suppressed CSC stemness by decreasing the sensitivity of PC to CXCL12 stimulus and inhibiting the crosstalk between PC and CSC-supportive niches. Mechanically, EHF suppressed the transcription of the CXCL12 receptor CXCR4. EHF had a cell autonomous role in suppressing cancer stemness by inhibiting the transcription of Sox9, Sox2, Oct4 and Nanog. Rosiglitazone suppressed PC stemness and inhibited the crosstalk between PC and PSCs by upregulating EHF. Preclinical KPC mouse cohorts demonstrated that rosiglitazone sensitised PDAC to gemcitabine therapy. CONCLUSIONS: EHF decreased the sensitivity of PC to the stimulus from PSC-derived CSC-supportive niche by negatively regulating tumorous CXCR4. Rosiglitazone could be used to target PC stem cells and the crosstalk between CSCs and their niche by upregulating EHF.


Subject(s)
Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Receptors, CXCR4/metabolism , Rosiglitazone/pharmacology , Transcription Factors/metabolism , Animals , Cohort Studies , Disease Models, Animal , Humans , Hypoglycemic Agents/pharmacology , Mice , Mice, Inbred BALB C , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism
11.
Br J Cancer ; 127(8): 1461-1472, 2022 11.
Article in English | MEDLINE | ID: mdl-35986089

ABSTRACT

BACKGROUND: Desmoplastic stroma, a feature of pancreatic ductal adenocarcinoma (PDAC), contains abundant activated pancreatic stellate cells (PSCs). How PSCs promote PDAC progression remains incompletely understood. METHODS: Effect of epithelium-specific E-twenty six factor 3 (ESE3)-positive PSCs on PDAC fibrosis and chemoresistance was examined by western blot, RT-PCR, immunofluorescence, flow cytometry assay, chromatin immunoprecipitation, luciferase assay, immunohistochemistry and subcutaneous pancreatic cancer mouse model. RESULTS: ESE3 expression increased in PSCs in PDAC tissues compared with those in normal PSCs. Clinical data showed that ESE3 upregulation in PSCs was positively correlated with tumour size, pTNM stage, CA19-9, carcinoembryonic antigen and serum CA242 level. ESE3 overexpression in PSCs was an independent negative prognostic factor for disease-free survival and overall survival amongst patients with PDAC. Mechanistically, the conditional medium from the loss and gain of ESE3-expressing PSCs influenced PDAC chemoresistance and tumour growth. ESE3 directly induced the transcription of α-SMA, collagen-I and IL-1ß by binding to ESE3-binding sites on their promoters to activate PSCs. IL-1ß upregulated ESE3 in PSCs through NF-κB activation, and ESE3 was required for PSC activation by tumour cell-derived IL-1ß. CONCLUSION: Inhibiting the IL-1ß/ESE3 (PSCs)/IL-1ß-positive feedback loop is a promising therapeutic strategy to reduce tumour fibrosis and increase chemotherapeutic efficacy in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , CA-19-9 Antigen/metabolism , Carcinoembryonic Antigen/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Collagen/metabolism , Drug Resistance, Neoplasm/genetics , Fibrosis , Interleukin-1beta , NF-kappa B/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Prognosis , Repressor Proteins
12.
Pancreatology ; 21(8): 1516-1523, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34531123

ABSTRACT

BACKGROUND: Extrapancreatic perineural invasion (EPNI) is a risk factor for the prognosis of patients with pancreatic cancer. Few reliable clinical indicators can be used to evaluate EPNI. METHODS: We reviewed clinicopathological information of pancreatic cancer patients received radical surgery in our center from 2014 to 2019. The minimum distance between the tumor boundary and celiac artery (CA), superior mesenteric arteria (SMA) was respectively measured on enhanced-contrast CT images. Receiver Operating Characteristic (ROC) analysis was used to evaluate the diagnostic efficacy, and the optimal cut-off value was determined by Youden index. The latter was used as a diagnostic indicator for imaging perineural invasion (iPNI). K-M method and Cox risk regression model were applied to analyze the prognostic value of iPNI. RESULTS: A total of 384 patients were enrolled in this study. ROC analysis showed the minimum distance is an efficient indicator, and the best cut-off value 6.5 mm provided 71.63% sensitivity and specificity 84.31%. Cox regression model showed that iPNI was an independent risk factor for disease-free survival (DFS) and overall survival (OS). Subgroup analysis indicated that patients with larger tumor size and iPNI positive suggested a worse prognosis. CONCLUSIONS: The minimum distance between tumor boundary and arteries is an efficient imaging indicator for diagnosing EPNI. iPNI is an independent risk factor for DFS and OS. The novel typing method based on plexus pancreaticus capitalis (PLX) potentially invaded may have guiding significance for extent of dissection.


Subject(s)
Pancreatic Neoplasms , Humans , Neoplasm Invasiveness , Pancreatic Neoplasms/diagnostic imaging , Prognosis , Retrospective Studies , Pancreatic Neoplasms
13.
Pancreatology ; 21(4): 763-770, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33712388

ABSTRACT

BACKGROUND: FOLFIRINOX and gemcitabine plus albumin-bound paclitaxel (AG) regimens are recommended as first-line therapy for both locally advanced and metastatic pancreatic cancer. However, there were no specific markers to conduct personalized regimen choice. The research is to assess delta Housfield unit (delta HU), which is the difference in CT attenuation value (in HU) between enhanced and nonenhanced phase of region of interest, as a marker for predicting chemotherapy response of unresectable pancreatic cancer. METHODS: A total of 179 unresectable pancreatic cancer patients were enrolled in the study. Kaplan-Meier analysis and COX regression analysis were performed for progression-free survival (PFS) and overall survival. The differences of clinical characteristics were analyzed by χ 2test. Microvessel density (MVD) was calculated by immunochemistry staining of CD34. RESULTS: Delta HU was an independent risk factor for unresectable pancreatic cancer (P = 0.017, HR 0.672, 95%CI 0.485-0.930). Patients with higher delta HU were associated with better PFS (P = 0.004). For modified FOLFIRINOX (mFOLFIRINOX) group, delta HU was an independent risk factor (P = 0.045, HR 0.571), but not for AG group (P = 0.473, HR 0.855). Delta HU was correlated with stroma MVD (P = 0.000, R = 0.483), not with parenchyma MVD (P = 0.074, R = 0.199). CONCLUSIONS: Delta HU was a marker predicting chemotherapy response for unresectable pancreatic cancer. Higher delta HU was associated with better survival for patients receiving mFOLFIRINOX rather than AG. The delta HU was positively correlated with stroma MVD, explaining the relationship between delta HU and prognosis.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Prognosis , Pancreatic Neoplasms
14.
Gastroenterology ; 154(3): 675-688, 2018 02.
Article in English | MEDLINE | ID: mdl-28989066

ABSTRACT

BACKGROUND & AIMS: Cells of the monocyte lineage contribute to tumor angiogenesis. Interleukin 35 (IL35) is a member of the IL12 family produced by regulatory, but not effector, T cells. IL35 is a dimer comprising the IL12 alpha and IL27 beta chains, encoded by IL12A and EBI3, respectively. Expression of IL35 is increased in pancreatic ductal adenocarcinomas (PDACs) compared with normal pancreatic tissues, and promotes metastasis. We investigated the role of IL35 in monocyte-induced angiogenesis of PDAC in mice. METHODS: We measured levels of IL35 protein, microvessel density, and numbers of monocytes in 123 sequential PDAC tissues from patients who underwent surgery in China in 2010. We performed studies with the human PDAC cell lines CFPAC-1, BxPC-3, Panc-1, MIA-PaCa-2, and mouse PDAC cell line Pan02. Monocyte subsets were isolated by flow cytometry from human peripheral blood mononuclear cells. Fused human or mouse IL12A and EBI3 genes were overexpressed in PDAC cells or knocked down using small hairpin RNAs. Cells were grown as xenograft tumors in SCID mice; some mice were given injections of an IL35-neutralizing antibody and tumor growth was monitored. We performed chemotaxis assays to measure the ability of IL35 to recruit monocytes. We analyzed mRNA sequences of 179 PDACs in the Cancer Genome Atlas to identify correlations between expression of IL12A and EBI3 and monocyte markers. Monocytes incubated with IL35 or PDAC cell supernatants were analyzed in tube formation and endothelial migration assays. RESULTS: In PDAC samples from patients, levels of IL35 mRNA and protein correlated with microvessel density and infiltration of monocyte lineage cells. In cells and mice with xenograft tumors, IL35 increased recruitment of monocytes into PDAC tumors, which required CCL5. Upon exposure to IL35, monocytes increased expression of genes whose products promote angiogenesis (CXCL1 and CXCL8). IL35 activated transcription of CCL5, CXCL1, and CXCL8 by inducing GP130 signaling, via IL12RB2 and phosphorylation of STAT1 and STAT4. A combination of a neutralizing antibody against IL35 and gemcitabine significantly decreased monocyte infiltration, microvessel density, and volume of xenograft tumors grown from PDAC cells in mice. CONCLUSIONS: PDAC cells produce IL35 to recruit monocytes via CCL5 and induce macrophage to promote angiogenesis via expression of CXCL1 and CXCL8. IL35 signaling promotes angiogenesis and growth of xenograft tumors from PDAC cells in mice. IL35 might serve as a therapeutic target for patients with pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Chemotaxis, Leukocyte , Interleukin-12 Subunit p35/metabolism , Interleukins/metabolism , Microvessels/metabolism , Minor Histocompatibility Antigens/metabolism , Monocytes/metabolism , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CCL5/metabolism , Chemokine CXCL1/metabolism , Chemotaxis, Leukocyte/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-12 Subunit p35/antagonists & inhibitors , Interleukin-12 Subunit p35/genetics , Interleukin-8/metabolism , Interleukins/antagonists & inhibitors , Interleukins/genetics , Macrophages/metabolism , Mice, SCID , Microvessels/drug effects , Microvessels/pathology , Minor Histocompatibility Antigens/genetics , Monocytes/drug effects , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Paracrine Communication , RNA Interference , Signal Transduction , Time Factors , Transfection , Tumor Burden , Xenograft Model Antitumor Assays , Gemcitabine
15.
J Biol Chem ; 291(53): 27323-27333, 2016 12 30.
Article in English | MEDLINE | ID: mdl-27879315

ABSTRACT

Fascin is an actin bundling protein that cross-links individual actin filaments into straight, compact, and stiff bundles, which are crucial for the formation of filopodia, stereocillia, and other finger-like membrane protrusions. The dysregulation of fascin has been implicated in cancer metastasis, hearing loss, and blindness. Here we identified monoubiquitination as a novel mechanism that regulates fascin bundling activity and dynamics. The monoubiquitination sites were identified to be Lys247 and Lys250, two residues located in a positive charge patch at the actin binding site 2 of fascin. Using a chemical ubiquitination method, we synthesized chemically monoubiquitinated fascin and determined the effects of monoubiquitination on fascin bundling activity and dynamics. Our data demonstrated that monoubiquitination decreased the fascin bundling EC50, delayed the initiation of bundle assembly, and accelerated the disassembly of existing bundles. By analyzing the electrostatic properties on the solvent-accessible surface of fascin, we proposed that monoubiquitination introduced steric hindrance to interfere with the interaction between actin filaments and the positively charged patch at actin binding site 2. We also identified Smurf1 as a E3 ligase regulating the monoubiquitination of fascin. Our findings revealed a previously unidentified regulatory mechanism for fascin, which will have important implications for the understanding of actin bundle regulation under physiological and pathological conditions.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Microfilament Proteins/metabolism , Ubiquitin/metabolism , Animals , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Rats , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
16.
BMC Cancer ; 17(1): 885, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29273007

ABSTRACT

BACKGROUND: This phase II bridging study assessed the safety and efficacy of nab-paclitaxel/gemcitabine (Metastatic Pancreatic Adenocarcinoma Clinical Trial [MPACT] regimen) in Chinese patients with metastatic pancreatic cancer (MPC). METHODS: This 3-part sequential study evaluated nab-paclitaxel 125 mg/m2 plus gemcitabine 1000 mg/m2 on days 1, 8, and 15 every 4 weeks. Part 1 evaluated safety. Part 2 evaluated efficacy using Simon's optimal 2-stage design: if >2 responses were observed in Stage 1 (n = 28), 54 additional patients would be enrolled in Stage 2. If >9 responses were observed, the study was complete. Otherwise, nab-paclitaxel/gemcitabine would be compared with gemcitabine alone in Part 3. The primary endpoint was overall response rate (ORR). Secondary endpoints included duration of response (DOR), overall survival (OS), and safety. RESULTS: Eighty-three patients were treated. The prespecified primary endpoint was met: the independently assessed ORR in Stages 1 + 2 was 35% (95% CI, 24.8-46.2); therefore, Part 3 was not initiated. The median DOR was 8.9 months (95% CI, 6.01-8.94). The median OS and progression-free survival were 9.2 (95% CI, 7.6-11.1) and 5.5 (95% CI, 5.29-7.16) months, respectively. The 12-month OS rate was 30%. In an updated analysis, the median OS was 9.3 months and the 12-month OS rate was 32%. Longer OS was observed in patients with baseline neutrophil-to-lymphocyte ratio ≤ 5 vs > 5. The most common grade ≥ 3 adverse events were leukopenia (35%), neutropenia (34%), anemia (15%), thrombocytopenia (10%), and fatigue (13%). Grade 3 peripheral neuropathy occurred in 7% of patients (no grade 4 reported). CONCLUSIONS: The MPACT regimen of nab-paclitaxel/gemcitabine is efficacious in Chinese patients with MPC. No new safety signals were observed. TRIAL REGISTRATION: NCT02135822 , May 8, 2014.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/secondary , Adult , Aged , Albumins/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Follow-Up Studies , Humans , Lymphatic Metastasis , Male , Middle Aged , Paclitaxel/administration & dosage , Pancreatic Neoplasms/pathology , Prognosis , Safety , Survival Rate , Gemcitabine , Pancreatic Neoplasms
17.
Int J Mol Sci ; 18(4)2017 Mar 23.
Article in English | MEDLINE | ID: mdl-28333072

ABSTRACT

Pancreatic cancer (PC) is the most lethal type of gastrointestinal cancer, and early detection and monitoring is an urgent problem. Circulating tumor cells (CTCs) are emerging as a non-invasive biomarker for tumor detection. However, the low sensitivity is a main problem in the traditional CellSearch System for detecting CTCs, especially in patients with PC. In this study, we used negative enrichment (NE), immunofluorescence and in situ hybridization (FISH) of chromosome 8 (NE-iFISH) to capture and identify CTCs in PC patients. We showed that the NE-iFISH system exhibited a dramatically high detection rate of CTCs in PC patients (90%). The diagnostic rate of PC reached 97.5% when combining CTCs ≥ 2 and carbohydrate antigen 19-9 (CA19-9) > 37 µmol/L. The 1-year survival in the group of CTCs < 3 was significantly higher than that of CTCs ≥ 3 (p = 0.043). In addition, we analyzed the role of chromosomal instability in CTCs detection. The group of triploid (three hybridization signals of chromosome 8) CTCs ≥ 3 showed a shorter 1-year survival (p = 0.0279) and overall survival (p = 0.0188) than the group with triploid CTCs < 3. Importantly, the triploid CTC number but not the overall CTC counts could be a predictor of chemo-sensitivity. Moreover, circulating tumor microembolus (CTMs) were found in stage IV patients, and were positively related to the poor response to chemotherapy. In conclusion, the NE-iFISH system significantly improved the positive detection rate of CTCs and triploid CTC could be used to predict prognosis or the response to the chemotherapy of PC patients. CTM is a potential indicator of the chemotherapeutic effect in advanced PC patients.


Subject(s)
Early Detection of Cancer/methods , In Situ Hybridization, Fluorescence/methods , Molecular Diagnostic Techniques/methods , Neoplastic Cells, Circulating/pathology , Pancreatic Neoplasms/blood , Adult , Aged , Case-Control Studies , Cell Line, Tumor , Chromosomal Instability , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Ploidies
18.
Cancer Sci ; 106(12): 1678-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26383967

ABSTRACT

Radiotherapy is widely applied for treatment of esophageal squamous cell carcinoma (ESCC). The Rad51-related protein XRCC3 plays roles in the recombinational repair of DNA double-strand breaks to maintain chromosome stability and repair DNA damage. The present study aimed to investigate the effect of XRCC3 on the radiotherapy response of ESCC and the underlying mechanisms of the roles of XRCC3 in ESCC radiosensitivity. XRCC3 expression in ESCC cells and tissues was higher than that in normal esophageal epithelial cells and corresponding adjacent noncancerous esophageal tissue. High XRCC3 expression was positively correlated with resistance to chemoradiotherapy in ESCC and an independent predictor for short disease-specific survival of ESCC patients. Furthermore, the therapeutic efficacy of radiotherapy in vitro and in vivo was substantially increased by knockdown of XRCC3 in ESCC cells. Ectopic overexpression of XRCC3 in both XRCC3-silenced ESCC cells dramatically enhanced ESCC cells' resistance to radiotherapy. Moreover, radiation resistance conferred by XRCC3 was attributed to enhancement of homologous recombination, maintenance of telomere stability, and a reduction of ESCC cell death by radiation-induced apoptosis and mitotic catastrophe. Our data suggest that XRCC3 protects ESCC cells from ionizing radiation-induced death by promoting DNA damage repair and/or enhancing telomere stability. XRCC3 may be a novel radiosensitivity predictor and promising therapeutic target for ESCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins/biosynthesis , Esophageal Neoplasms/genetics , Radiation Tolerance/genetics , Aged , Blotting, Western , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/radiotherapy , DNA Damage , DNA Repair/genetics , DNA-Binding Proteins/genetics , Esophageal Neoplasms/mortality , Esophageal Neoplasms/radiotherapy , Esophageal Squamous Cell Carcinoma , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Immunoprecipitation , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome
19.
Carcinogenesis ; 35(10): 2283-90, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25086759

ABSTRACT

Stem cell factor (SCF), a ligand of c-kit, is a hematopoietic growth factor. Uncontrolled activity of SCF/c-kit signaling pathway contributes to the formation of a variety of human malignancies. In this study, we determined whether SCF expression could risk-stratify patients with hepatocellular carcinoma (HCC) after curative resection. HCC tissues from 160 patients were collected during curative resection and stained with SCF and CD34, a marker for microvessel density (MVD), using immunohistochemistry. Two statistical analyses were performed: an independent continuous and a multivariate categorical analysis, with test/validation set-defined cut points, and Kaplan-Meier estimated outcome measures of overall survival (OS) and relapse-free survival (RFS). We found that higher levels of SCF confer worse OS (continuous P = 0.014; and categorical P = 0.009), and RFS (continuous P = 0.002; categorical P = 0.003) of patients with HCC. SCF varies independently from MVD-CD34, tumor node metastasis, histologic grade, age and gender, and retains prognostic significance when analysed as a categorical variable in a multivariate analysis . We confirmed that MVD-CD34 is also an independent prognostic marker for patients with HCC. The levels of SCF and CD34 showed a positive and significant correlation (P < 0.0001) and double low expression confers superior OS (median = 48 months) and RFS (median = 24 months), whereas double high expression confers shortest RFS (median = 10.5 months) compared with single measurements. The prognostic values of SCF and CD34 were independently determined in this study and we propose that both of them are independent prognostic markers for HCC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/surgery , Liver Neoplasms/surgery , Stem Cell Factor/analysis , Antigens, CD34/analysis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Case-Control Studies , Disease-Free Survival , Humans , Kaplan-Meier Estimate , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Prognosis , Treatment Outcome
20.
Tumour Biol ; 35(11): 11319-28, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25119596

ABSTRACT

The aim of study was to discuss the correlation and regulatory mechanism of HIF-1 and miR-191 expression in pancreatic tumor. The association between the miR-191 and the clinicopathologic characteristics and the prognosis of pancreatic cancer was further explored. After hypoxic cultured for 6 and 12 h, qRT-PCR and Western blot were practiced to analyze the miR-191 and HIF-1 expression of MIA PaCa-2 and Aspac1 cells. We regulated the HIF-1 expression via plasmid and siRNA transfection to observe the alteration of HIF-1 and miR-191 expression. ChIP sequencing identified the binding sites of HIF-1 and miR-191. Dual luciferase assays were practiced to verify the binding sites. Immunohistochemical staining was practiced to analyze the expression of HIF-1, while qRT-PCR were done for investigating miR-191 in tumor tissues. Then, the association between the expression of them and the clinicopathologic characteristics and prognosis of pancreatic cancer were analyzed. After hypoxic cultured 12 h, the expression of HIF-1 protein, HIF-1mRNA and miR-191 of MIA PaCa-2 and AsPC-1 cells increased significantly (P < 0.05). After HIF-1 overexpressing plasmid transfected to the MIA PaCa-2 and AsPC-1 cells for 48 h, the expression of HIF-1 protein, HIF-1mRNA, and miR-191 upregulated significantly (P < 0.05). While after transfected the MIA PaCa-2 cells by HIF-1 siRNA, the significant decreasing of HIF-1 protein, HIF-1mRNA, and miR-191 expression were observed (P < 0.05). ChIP sequencing showed the protein synthesis of HIF-1 increased in hypoxia situation. Only the HRE5 (-1,169 bp, ChIP4) were significantly brighter in hypoxia in comparing with normoxic cells. In dual luciferase assays, after pGL3-miR-191 and HIF-1 overexpressing plasmid co-transfect the MIAPaCa-2 cells for 48 h, its relative expression of bioluminescence was higher than those co-transfected by mutant miR-191 vectors and HIF-1 overexpressing plasmid or by pGL3-miR-191 and HIF-1 empty plasmid. The expression of miR-191 closely associated with the tumor size, pTNM stage, lymph node metastasis, and perineural invasion (P < 0.05). Patients with higher expression of miR-191 were a risk factor for prognosis of pancreatic cancers. Expression of HIF-1 in pancreatic cancer cells increased under the condition of chronic hypoxia, which may bind to HRE2 in 5'flanking region of miR-191 and initiate transcription of miR-191. Expression of miR-191 was significantly higher in pancreatic tumor tissues. The expression of miR-191 closely associated with the tumor size, pTNM stage, lymph node metastasis and perineural invasion and poor prognosis of pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/genetics , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Apoptosis , Blotting, Western , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Cell Hypoxia , Cell Proliferation , Chromatin Immunoprecipitation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Lymphatic Metastasis , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Pancreas/pathology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL