Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nat Commun ; 14(1): 7033, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37923799

ABSTRACT

Bile duct tumor thrombosis (BDTT) is a complication mostly observed in patients with advanced hepatocellular carcinoma (HCC), causing jaundice and associated with poor clinical outcome. However, its underlying molecular mechanism is unclear. Here, we develop spontaneous preclinical HCC animal models with BDTT to identify the role of BMI1 expressing tumor initiating cells (BMI1high TICs) in inducing BDTT. BMI1 overexpression transforms liver progenitor cells into BMI1high TICs, which possess strong tumorigenicity and increased trans-intrahepatic biliary epithelial migration ability by secreting lysosomal cathepsin B (CTSB). Orthotopic liver implantation of BMI1high TICs into mice generates tumors and triggers CTSB mediated bile duct invasion to form tumor thrombus, while CTSB inhibitor treatment prohibits BDTT and extends mouse survival. Clinically, the elevated serum CTSB level determines BDTT incidence in HCC patients. Mechanistically, BMI1 epigenetically up-regulates CTSB secretion in TICs by repressing miR-218-1-3p expression. These findings identify a potential diagnostic and therapeutic target for HCC patients with BDTT.


Subject(s)
Bile Duct Neoplasms , Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Thrombosis , Humans , Animals , Mice , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Cathepsins , Bile Duct Neoplasms/pathology , Thrombosis/pathology , Polycomb Repressive Complex 1/genetics , MicroRNAs/genetics
2.
J Transl Med ; 21(1): 702, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37814317

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is characterised by limited responses to chemoimmunotherapy attributed to highly desmoplastic tumor microenvironment. Disrupting the tumor-stromal cell crosstalk is considered as an improved PDAC treatment strategy, whereas little progress has been made due to poor understanding of its underlying mechanism. Here, we examined the cellular role of melanoma associated antigen A isoforms (MAGEA) in regulating tumor-stromal crosstalk mediated chemoresistance. METHODS: We used clinical samples to explore the correlation between MAGEA expression and patient prognosis in multiple cancers. We utilized cancer cell lines, patient derived organoids and orthotopic PDAC model to examine the function of MAGEA in chemoresistance. We performed biochemical, proteome profiler array and transcriptional analysis to uncover a mechanism that governs tumor-stromal crosstalk. We developed a multi-MAGEA antigen targeted DNA vaccine and tested its effect on PDAC tumor growth. RESULTS: We establish MAGEA as a regulator of the tumor-stromal crosstalk in PDAC. We provide strong clinical evidence indicating that high MAGEA expression, including MAGEA2, MAGEA3 and MAGEA10, correlates with worse chemotherapeutic response and poor prognosis in multiple cancers, while their expression is up-regulated in chemoresistant PDAC patient derived organoids and cancer cell lines. Mechanistically, MAGEA2 prohibits gemcitabine-induced JNK-c-Jun-p53 mediated cancer cell apoptosis, while gemcitabine stimulated pancreatic stellate cells secretes GDF15 to further enhance the gemcitabine resistance of MAGEA2 expressing cells by activating GFRAL-RET mediated Akt and ERK1/2 dependent survival pathway. Strikingly, immunization with a DNA vaccine that targeting multiple MAGEA antigens, including MAGEA2, MAGEA3 and MAGEA10, elicits robust immune responses against the growth of gemcitabine resistant tumors. CONCLUSIONS: These findings suggest that targeting MAGEA-mediated paracrine regulation of chemoresistance by immunotherapy can be an improved pancreatic cancer treatment strategy.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Vaccines, DNA , Humans , Vaccines, DNA/metabolism , Vaccines, DNA/pharmacology , Vaccines, DNA/therapeutic use , Deoxycytidine/pharmacology , Cell Line, Tumor , Pancreatic Neoplasms/pathology , Gemcitabine , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Immunization , Stromal Cells/pathology , Drug Resistance, Neoplasm , Tumor Microenvironment , Pancreatic Neoplasms
3.
NPJ Precis Oncol ; 7(1): 102, 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37821657

ABSTRACT

Conventional chemotherapy targets malignant cells without evaluating counter protection from the tumor microenvironment that often causes treatment failure. Herein, we establish chemoresistant fibroblasts (rCAFs) as regulators of neoadjuvant chemotherapeutic (NACT) response in head and neck squamous cell carcinoma (HNSCC). Clinically, high expression of CAF-related gene signature correlates with worse prognosis and chemotherapeutic response in multiple cancers, while the population of CAFs in the residual tumors of chemoresistant HNSCC patients remains unchanged after NACT treatment, compared to chemosensitive patients. Using a murine cancer model or patient-derived organoid, and primary CAFs isolated from chemo-sensitive (sCAFs) or -resistant patients, we show that rCAFs, but not sCAFs, are resistant to chemotherapy-induced apoptosis while reducing HNSCC cell chemosensitivity via paracrine signals. Combined multi-omics and biochemical analyses indicate an elevated PI3K/AKT/p65 driven cell survival and cytokine production in rCAFs, while rCAF-secreted TGFα promotes cancer cell chemoresistance by activating EGFR/Src/STAT3 survival signaling axis. Treatment with anti-EGFR cetuximab restores the chemosensitivity of tumors derived from co-injection of cancer cells and rCAFs in vivo, while the serum level of TGFα determines NACT response in HNSCC patients. Overall, our findings uncover a novel insight whereby the crosstalk between tumor cell and rCAF determines chemotherapeutic response and prognosis in cancer patients.

5.
J Cancer Res Clin Oncol ; 149(12): 9635-9655, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37227522

ABSTRACT

BACKGROUND: Tumor endothelial cells (TECs) play a significant role in regulating the tumor microenvironment, drug response, and immune cell activities in various cancers. However, the association between TEC gene expression signature and patient prognosis or therapeutic response remains poorly understood. METHODS: We analyzed transcriptomics data of normal and tumor endothelial cells obtained from the GEO database to identify differentially expressed genes (DEGs) associated with TECs. We then compared these DEGs with those commonly found across five different tumor types from the TCGA database to determine their prognostic relevance. Using these genes, we constructed a prognostic risk model integrated with clinical features to develop a nomogram model, which we validated through biological experiments. RESULTS: We identified 12 TEC-related prognostic genes across multiple tumor types, of which five genes were sufficient to construct a prognostic risk model with an AUC of 0.682. The risk scores effectively predicted patient prognosis and immunotherapeutic response. Our newly developed nomogram model provided more accurate prognostic estimates of cancer patients than the TNM staging method (AUC = 0.735) and was validated using external patient cohorts. Finally, RT-PCR and immunohistochemical analyses indicated that the expression of these 5 TEC-related prognostic genes was up-regulated in both patient-derived tumors and cancer cell lines, while depletion of the hub genes reduced cancer cell growth, migration and invasion, and enhanced their sensitivity to gemcitabine or cytarabine. CONCLUSIONS: Our study discovered the first TEC-related gene expression signature that can be used to construct a prognostic risk model for guiding treatment options in multiple cancers.


Subject(s)
Endothelial Cells , Neoplasms , Humans , Transcriptome , Neoplasms/genetics , Neoplasms/therapy , Prognosis , Immunotherapy , Tumor Microenvironment/genetics
6.
Sci Transl Med ; 15(678): eabl7895, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36630483

ABSTRACT

Pancreatic and lung cancers frequently develop resistance to chemotherapy-induced cell apoptosis during the treatment, indicating that targeting nonapoptotic-related pathways, such as pyroptosis, can be an alternative cancer treatment strategy. Pyroptosis is a gasdermin-driven lytic programmed cell death triggered by inflammatory caspases when initiated by canonical or noncanonical pathways that has been recently seen as a potential therapeutic target in cancer treatment. However, overcoming chemoresistance in cancers by modulating pyroptosis has not been explored. Here, we demonstrate that ß5-integrin represses chemotherapy-induced canonical pyroptosis to confer cancer chemoresistance through ASAH2-driven sphingolipid metabolic reprogramming. Clinically, high ß5-integrin expression associates with poor patient prognosis and chemotherapeutic responses in cancers. In addition, chemoresistant cells in vitro fail to undergo chemotherapy-induced pyroptosis, which is controlled by ß5-integrin. Mechanistically, proteomic and lipidomic analyses indicate that ß5-integrin up-regulates sphingolipid metabolic enzyme ceramidase (ASAH2) expression through Src-signal transducer and activator of transcription 3 (STAT3) signaling, which then reduces the metabolite ceramide concentration and subsequent ROS production to prohibit chemotherapy-induced canonical pyroptosis. Using cancer cell lines, patient-derived tumor organoids, and orthotopic lung and pancreatic animal models, we show that administration of a Src or ceramidase inhibitor rescues the response of chemoresistant pancreatic and lung cancer cells to chemotherapy by reactivating pyroptosis in vitro and in vivo. Overall, our results suggest that pyroptosis-based therapy is a means to improve cancer treatment and warrants further investigation.


Subject(s)
Antineoplastic Agents , Drug Resistance, Neoplasm , Pancreatic Neoplasms , Proto-Oncogene Proteins pp60(c-src) , Pyroptosis , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Integrins/metabolism , Lung/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Proteomics , Pyroptosis/drug effects , Proto-Oncogene Proteins pp60(c-src)/drug effects , Proto-Oncogene Proteins pp60(c-src)/metabolism , Humans , Integrin beta Chains/metabolism , STAT3 Transcription Factor/metabolism , Ceramidases/metabolism , Pancreatic Neoplasms
7.
Pharmacol Res ; 187: 106558, 2023 01.
Article in English | MEDLINE | ID: mdl-36410675

ABSTRACT

Dysregulated sphingolipid metabolism contributes to ER+ breast cancer progression and therapeutic response, whereas its underlying mechanism and contribution to tamoxifen resistance (TAMR) is unknown. Here, we establish sphingolipid metabolic enzyme CERK as a regulator of TAMR in breast cancer. Multi-omics analysis reveals an elevated CERK driven sphingolipid metabolic reprogramming in TAMR cells, while high CERK expression associates with worse patient prognosis in ER+ breast cancer. CERK overexpression confers tamoxifen resistance and promotes tumorigenicity in ER+ breast cancer cells. Knocking out CERK inhibits the orthotopic breast tumor growth of TAMR cells while rescuing their tamoxifen sensitivity. Mechanistically, the elevated EHF expression transcriptionally up-regulates CERK expression to prohibit tamoxifen-induced sphingolipid ceramide accumulation, which then inhibits tamoxifen-mediated repression on PI3K/AKT dependent cell proliferation and its driven p53/caspase-3 mediated apoptosis in TAMR cells. This work provides insight into the regulation of sphingolipid metabolism in tamoxifen resistance and identifies a potential therapeutic target for this disease.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Tamoxifen , Female , Humans , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , MCF-7 Cells , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Estrogen/metabolism , Sphingolipids , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
8.
Cancer Med ; 12(5): 6388-6400, 2023 03.
Article in English | MEDLINE | ID: mdl-36404634

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinomas (HNSCC) are the most common type of head and neck cancer with an unimproved prognosis over the past decades. Although the role of cancer-associated-fibroblast (CAF) has been demonstrated in HNSCC, the correlation between CAF-derived gene expression and patient prognosis remains unknown. METHODS: A total of 528 patients from TCGA database and 270 patients from GSE65858 database were contained in this study. After extracting 66 CAF-related gene expression data from TCGA database, consensus clustering was performed to identify different HNSCC subtypes. Limma package was used to distinguish the differentially expression genes (DEGs) between these subtypes, followed by Lasso regression analysis to construct a prognostic model. The model was validated by performing Kaplan-Meier survival, ROC and risk curve, univariate and multivariate COX regression analysis. GO, KEGG, GSEA, ESTIMATE and ssGSEA analyses was performed to explort the potential mechanism leading to different prognosis. RESULTS: Based on the 66 CAF-related gene expression pattern we stratitied HNSCC patients into two previously unreported subtypes with different clinical outcomes. A prognostic model composed of 15 DEGs was constructed and validated. In addition, bioinformatics analysis showed that the prognostic risk of HNSCC patients was also negatively correlated to immune infiltration, implying the role of tumor immune escape in HNSCC prognosis and treatment option. CONCLUSIONS: The study develops a reliable prognostic prediction tool and provides a theoretical treatment guidance for HNSCC patients.


Subject(s)
Cancer-Associated Fibroblasts , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck/genetics , Prognosis , Kaplan-Meier Estimate , Head and Neck Neoplasms/genetics
9.
Cancer Res ; 82(10): 1909-1925, 2022 05 16.
Article in English | MEDLINE | ID: mdl-35350066

ABSTRACT

Despite substantial advances in the treatment of solid cancers, resistance to therapy remains a major obstacle to prolonged progression-free survival. Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers, with a high level of liver metastasis. Primary PDAC is highly hypoxic, and metastases are resistant to first-line treatment, including gemcitabine. Recent studies have indicated that endothelial cell (EC) focal adhesion kinase (FAK) regulates DNA-damaging therapy-induced angiocrine factors and chemosensitivity in primary tumor models. Here, we show that inducible loss of EC-FAK in both orthotopic and spontaneous mouse models of PDAC is not sufficient to affect primary tumor growth but reduces liver and lung metastasis load and improves survival rates in gemcitabine-treated, but not untreated, mice. EC-FAK loss did not affect primary tumor angiogenesis, tumor blood vessel leakage, or early events in metastasis, including the numbers of circulating tumor cells, tumor cell homing, or metastatic seeding. Phosphoproteomics analysis showed a downregulation of the MAPK, RAF, and PAK signaling pathways in gemcitabine-treated FAK-depleted ECs compared with gemcitabine-treated wild-type ECs. Moreover, low levels of EC-FAK correlated with increased survival and reduced relapse in gemcitabine-treated patients with PDAC, supporting the clinical relevance of these findings. Altogether, we have identified a new role of EC-FAK in regulating PDAC metastasis upon gemcitabine treatment that impacts outcome. SIGNIFICANCE: These findings establish the potential utility of combinatorial endothelial cell FAK targeting together with gemcitabine in future clinical applications to control metastasis in patients with pancreatic ductal adenocarcinoma.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Endothelial Cells/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Mice , Neoplasm Recurrence, Local , Pancreatic Neoplasms/pathology , Gemcitabine , Pancreatic Neoplasms
10.
Oncogene ; 41(16): 2340-2356, 2022 04.
Article in English | MEDLINE | ID: mdl-35256782

ABSTRACT

The cellular origin of hepatocellular carcinomas (HCC) and the role of Notch1 signalling in HCC initiation are controversial. Herein, we establish Notch1 as a regulator of HCC development and progression. Clinically, high Notch1 expression correlates with enhanced cancer progression, elevated lung metastasis, increased cancer stem cell (CSC)-like cells' gene signature expression, and poor overall survival in HCC patients. Notch1 intracellular domain (N1ICD) overexpression spontaneously transforms rat liver progenitor cells (LPC) into CSC-like cells (WBN1ICD C5) under a selective growth environment, while orthotopic injection of these cells generates liver tumors and spontaneous pulmonary metastasis in an isogenic rat model. Mechanistically, the elevated Notch1 activity increases c-myc expression, which then transcriptionally upregulates VCAM1 expression to activate macrophage dependent HCC transendothelial migration. In vivo, silencing c-myc prohibits the tumorigenicity of WBN1ICD C5 cells, while depletion of VCAM1 reduces spontaneous lung metastasis without affecting primary WBN1ICD C5 orthotopic liver tumor growth. Importantly, depletion of macrophage or blockade of macrophage VCAM1 binding receptor α4ß1-integrin reduces the number of WBN1ICD C5 lung nodules in an experimental metastasis model. Overall, our work discovers that the Notch1-c-myc-VCAM1 signaling axis initiates LPC-driven hepatocarcinogenesis and metastasis, providing a preclinical model for HCC study and therapeutic targets for an improved HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Lung Neoplasms , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Humans , Liver Neoplasms/pathology , Lung Neoplasms/genetics , Rats , Receptor, Notch1/metabolism , Stem Cells/metabolism
11.
Nat Commun ; 12(1): 6011, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34650057

ABSTRACT

Defective pericyte-endothelial cell interaction in tumors leads to a chaotic, poorly organized and dysfunctional vasculature. However, the underlying mechanism behind this is poorly studied. Herein, we develop a method that combines magnetic beads and flow cytometry cell sorting to isolate pericytes from tumors and normal adjacent tissues from patients with non-small cell lung cancer (NSCLC) and hepatocellular carcinoma (HCC). Pericytes from tumors show defective blood vessel supporting functions when comparing to those obtained from normal tissues. Mechanistically, combined proteomics and metabolic flux analysis reveals elevated hexokinase 2(HK2)-driven glycolysis in tumor pericytes, which up-regulates their ROCK2-MLC2 mediated contractility leading to impaired blood vessel supporting function. Clinically, high percentage of HK2 positive pericytes in blood vessels correlates with poor patient overall survival in NSCLC and HCC. Administration of a HK2 inhibitor induces pericyte-MLC2 driven tumor vasculature remodeling leading to enhanced drug delivery and efficacy against tumor growth. Overall, these data suggest that glycolysis in tumor pericytes regulates their blood vessel supporting role.


Subject(s)
Blood Vessels/abnormalities , Glycolysis , Hexokinase/metabolism , Neoplasms, Vascular Tissue/metabolism , Pericytes/metabolism , A549 Cells , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Hexokinase/genetics , Humans , Mice , Mice, Inbred C57BL , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Neoplasms/metabolism , Neoplasms, Vascular Tissue/drug therapy , Neoplasms, Vascular Tissue/genetics , Neoplasms, Vascular Tissue/pathology , Tumor Microenvironment/physiology , Up-Regulation , rho-Associated Kinases
12.
Front Cell Dev Biol ; 9: 676342, 2021.
Article in English | MEDLINE | ID: mdl-34179005

ABSTRACT

Pericytes (PCs), known as mural cells, play an important blood vessel (BV) supporting role in regulating vascular stabilization, permeability and blood flow in microcirculation as well as blood brain barrier. In carcinogenesis, defective interaction between PCs and endothelial cells (ECs) contributes to the formation of leaky, chaotic and dysfunctional vasculature in tumors. However, recent works from other laboratories and our own demonstrate that the direct interaction between PCs and other stromal cells/cancer cells can modulate tumor microenvironment (TME) to favor cancer growth and progression, independent of its BV supporting role. Furthermore, accumulating evidence suggests that PCs have an immunomodulatory role. In the current review, we focus on recent advancement in understanding PC's regulatory role in the TME by communicating with ECs, immune cells, and tumor cells, and discuss how we can target PC's functions to re-model TME for an improved cancer treatment strategy.

13.
EBioMedicine ; 66: 103301, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33813137

ABSTRACT

BACKGROUND: Sphingolipid metabolism is among the top dysregulated pathways in non-small cell lung carcinomas (NSCLC). However, the molecular control of sphingolipid metabolic reprogramming in cancer progression remains unclear. METHODS: We first determined the correlation between sphingolipid metabolic gene expression and patient prognosis. We then carried out sphingolipidomics analysis of health individual and NSCLC patient sera as well as B3GNT5 and GAL3ST1 genetically perturbed NSCLC cell lines. We used these cell lines to perform tumorigenesis study to determine the cellular role of B3GNT5 and GAL3ST1 in cancer growth and progression. FINDINGS: The expression of B3GNT5 and GAL3ST1 among sphingolipid metabolic enzymes is most significantly associated with patient prognosis, whilst sphingolipidomics analysis of healthy individual and NSCLC patient sera identifies their metabolites, lacto/neolacto-series glycosphingolipid and sulfatide species, as potential biomarkers that were more effective than current clinical biomarkers for staging patients. Further network analysis of the sphingolipidomes reveals a circular network of coregulated sphingolipids, indicating that the lacto/neolacto-series glycosphingolipid/sulfatide balance functions as a checkpoint to determine sphingolipid metabolic reprograming during patient progression. Sphingolipidomics analysis of B3GNT5/GAL3ST1 genetically perturbed NSCLC cell lines confirms their key regulatory role in sphingolipid metabolism, while B3GNT5 and GAL3ST1 expression has an opposite role on tumorigenesis. INTERPRETATION: Our results provide new insights whereby B3GNT5 and GAL3ST1 differentially regulate sphingolipid metabolism in lung cancer growth and progression. FUNDING: This work was supported by the Natural Science Foundation of China (81872142, 81920108028); Guangzhou Science and Technology Program (201904020008); Guangdong Science and Technology Department (2020A0505100029, 2019A1515011802, 2020A1515011280, 2020B1212060018, 2020B1212030004); China Postdoctoral Science Foundation (2019M650226, 2019M650227).


Subject(s)
Lipid Metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Sphingolipids/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Computational Biology/methods , Databases, Genetic , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Lipidomics/methods , Lung Neoplasms/etiology , Lung Neoplasms/mortality , Mice , Neoplasm Staging , Prognosis
14.
Hum Genet ; 140(4): 609-623, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33140241

ABSTRACT

Alzheimer's disease (AD) is one of the most common neurodegeneration diseases caused by multiple factors. The mechanistic insight of AD remains limited. To disclose molecular mechanisms of AD, many studies have been proposed from transcriptome analyses. However, no analysis across multiple levels of transcription has been conducted to discover co-expression networks of AD. We performed gene-level and isoform-level analyses of RNA sequencing (RNA-seq) data from 544 brain tissues of AD patients, mild cognitive impaired (MCI) patients, and healthy controls. Gene and isoform levels of co-expression modules were constructed by RNA-seq data. The associations of modules with AD were evaluated by integrating cognitive scores of patients, Genome-wide association studies (GWAS), alternative splicing analysis, and dementia-related genes expressed in brain tissues. Totally, 29 co-expression modules were found with expressions significantly correlated with the cognitive scores. Among them, two isoform modules were enriched with AD-associated SNPs and genes whose mRNA splicing displayed significant alteration in relation to AD disease. These two modules were further found enriched with dementia-related genes expressed in four brain regions of 125 AD patients. Analyzing expressions of these two modules revealed expressions of 39 isoforms (corresponding to 35 genes) significantly correlated with cognitive scores of AD patients, in which 38 isoforms were significantly up-regulated in AD patients comparing to controls, and 33 isoforms (corresponding to 29 genes) were not reported as AD-related previously. Employing the co-expression modules and the drug-induced gene expression data from Connectivity Map (CMAP), 12 drugs were predicted as significant in restoring the gene expression of AD patients towards health, which include nine drugs reported for relieving AD. In comparison, four of the top 12 significant drugs were known for relieving AD if the drug prediction was performed by the genes expressed significantly different in AD and healthy controls. Analysis of multiple levels of the transcriptomic organization is useful in suggesting AD-related co-expression networks and discovering drugs.


Subject(s)
Alzheimer Disease/genetics , Gene Expression Regulation , Gene Regulatory Networks , Protein Isoforms/genetics , Transcriptome , Alternative Splicing , Alzheimer Disease/drug therapy , Datasets as Topic , Female , Genome-Wide Association Study , Humans , Male , Polymorphism, Single Nucleotide , Prefrontal Cortex/metabolism , RNA Splicing , RNA-Seq , Tacrolimus/therapeutic use , Vorinostat/therapeutic use
15.
Cancer Lett ; 500: 228-243, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33309857

ABSTRACT

Aberrant glycosylation in pancreatic cancer has been linked to cancer development, progression and chemoresistance. However, the role of glycogene, such as galactosyltransferase, in pancreatic cancer remains unknown. Herein, we establish beta-1.4-galactosyltransferase 1 (B4GALT1) as a clinical marker and regulator of chemoresistance. Clinically, high B4GALT1 expression correlates with poor survival, enhanced tumor size, increased lymph node metastasis, elevated cancer progression and enhanced incidence of relapse in PDAC patients. Expression of B4GALT1 is up-regulated in gemcitabine resistant patient derived organoids as well as chemoresistant cancer cell lines, while genetic perturbation of its expression in PDAC cell lines regulates cancer progression and chemoresistance. Mechanistically, we show that elevated p65 activity transcriptionally up-regulates B4GALT1 expression, which then interacts with and stabilizes cyclin dependent kinase 11 isomer CDK11p110 protein via N-linked glycosylation, in order to promote cancer progression and chemoresistance. Finally, depletion of B4GALT1 rescues the response of chemoresistant cells to gemcitabine in an orthotopic PDAC model. Overall, our data uncovers a mechanism by which p65-B4GALT1-CDK11p110 signalling axis determines cancer progression and chemoresistance, providing a new therapeutic target for an improved pancreatic cancer treatment.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Cyclin-Dependent Kinases/genetics , Galactosyltransferases/genetics , Transcription Factor RelA/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glycosylation/drug effects , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Organoids/drug effects , Gemcitabine
16.
Oncogenesis ; 9(11): 101, 2020 Nov 09.
Article in English | MEDLINE | ID: mdl-33168810

ABSTRACT

EZH2, a histone methyltransferase, has been shown to involve in cancer development and progression via epigenetic regulation of tumor suppressor microRNAs, whereas BMI1, a driver of hepatocellular carcinoma (HCC), is a downstream target of these microRNAs. However, it remains unclear whether EZH2 can epigenetically regulate microRNA expression to modulate BMI1-dependent hepatocarcinogenesis. Here, we established that high EZH2 expression correlated with enhanced tumor size, elevated metastasis, increased relapse, and poor prognosis in HCC patients. Further clinical studies revealed that EZH2 overexpression was positively correlated to its gene copy number gain/amplification in HCC. Mechanistically, EZH2 epigenetically suppressed miR-200c expression both in vitro and in vivo, and more importantly, miR-200c post-transcriptionally regulated BMI1 expression by binding to the 3'-UTR region of its mRNA. Furthermore, miR-200c overexpression inhibits the growth of HCC cells in vivo. Silencing miR-200c rescued the tumorigenicity of EZH2-depleted HCC cells, whereas knocking down BMI1 reduced the promoting effect of miR-200c depletion on HCC cell migration. Finally, combination treatment of EZH2 and BMI1 inhibitors further inhibited the viability of HCC cells compared with the cells treated with EZH2 or BMI1 inhibitor alone. Our findings demonstrated that alteration of EZH2 gene copy number status induced BMI1-mediated hepatocarcinogenesis via epigenetically silencing miR-200c, providing novel therapeutic targets for HCC treatment.

17.
Nat Commun ; 11(1): 2810, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32499572

ABSTRACT

The overexpression of the protein tyrosine kinase, Focal adhesion kinase (FAK), in endothelial cells has implicated its requirement in angiogenesis and tumour growth, but how pericyte FAK regulates tumour angiogenesis is unknown. We show that pericyte FAK regulates tumour growth and angiogenesis in multiple mouse models of melanoma, lung carcinoma and pancreatic B-cell insulinoma and provide evidence that loss of pericyte FAK enhances Gas6-stimulated phosphorylation of the receptor tyrosine kinase, Axl with an upregulation of Cyr61, driving enhanced tumour growth. We further show that pericyte derived Cyr61 instructs tumour cells to elevate expression of the proangiogenic/protumourigenic transmembrane receptor Tissue Factor. Finally, in human melanoma we show that when 50% or more tumour blood vessels are pericyte-FAK negative, melanoma patients are stratified into those with increased tumour size, enhanced blood vessel density and metastasis. Overall our data uncover a previously unknown mechanism of tumour growth by pericytes that is controlled by pericyte FAK.


Subject(s)
Cysteine-Rich Protein 61/metabolism , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Neovascularization, Pathologic , Pericytes/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Aorta, Thoracic/pathology , Carcinoma, Lewis Lung/metabolism , Cell Adhesion , Cell Proliferation , Female , Focal Adhesion Kinase 1/genetics , Humans , Lymphokines/metabolism , Male , Melanoma/blood supply , Melanoma/metabolism , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/pathology , Placenta Growth Factor/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Signal Transduction , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism , Axl Receptor Tyrosine Kinase
18.
Cell ; 181(6): 1346-1363.e21, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32473126

ABSTRACT

Enhanced blood vessel (BV) formation is thought to drive tumor growth through elevated nutrient delivery. However, this observation has overlooked potential roles for mural cells in directly affecting tumor growth independent of BV function. Here we provide clinical data correlating high percentages of mural-ß3-integrin-negative tumor BVs with increased tumor sizes but no effect on BV numbers. Mural-ß3-integrin loss also enhances tumor growth in implanted and autochthonous mouse tumor models with no detectable effects on BV numbers or function. At a molecular level, mural-cell ß3-integrin loss enhances signaling via FAK-p-HGFR-p-Akt-p-p65, driving CXCL1, CCL2, and TIMP-1 production. In particular, mural-cell-derived CCL2 stimulates tumor cell MEK1-ERK1/2-ROCK2-dependent signaling and enhances tumor cell survival and tumor growth. Overall, our data indicate that mural cells can control tumor growth via paracrine signals regulated by ß3-integrin, providing a previously unrecognized mechanism of cancer growth control.


Subject(s)
Integrin beta3/metabolism , Neoplasms/metabolism , Tumor Burden/physiology , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Female , Humans , Male , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
19.
J Chem Inf Model ; 60(1): 400-409, 2020 01 27.
Article in English | MEDLINE | ID: mdl-31833767

ABSTRACT

Motivation: Identification of ligand-binding proteins is an important issue for drug development. Most of the current computational approach is developed only utilizing ligand structure similarity. However, the ligand structure similarity has failed to reflect the binding quality between the ligand and the target protein, which limited the performance of current methods. Results: The present study integrated two-dimensional (2D) and three-dimensional (3D) ligand structure similarity between query ligand and template with known ligand-protein binding affinity (BA) to identify proteins binding with the query ligand. This method is named as DStruBTarget. The performance of DStruBTarget was evaluated by 10-fold cross-validation in a dataset containing 9197 ligands and 1111 ligand-binding proteins (DBD dataset). This dataset was constructed by excluding the ligands with similar structures and the proteins with high sequence identity. The DStruBTarget achieved a hit rate of 77% in top 1 prediction, which is 4.80 and 3.00% better than the methods only using 2D structure similarity, and the method integrating 2D and 3D structure similarity (2D + 3D), respectively. An independent test of DStruBTarget was performed in a publicly available dataset constructed by SwissTargetPrediction. In this dataset, the top 1 hit rate of DStruBTarget reached 44.02%, which was better than the SwissTargetPrediction, and also outstands other methods, such as 2D, 3D, 2D + 3D, 2D integrating binding affinity (2D + BA), and 3D integrating binding affinity (3D + BA). DStruBTarget was compared to another newly published method HitPickV2 and achieved 52.17% hit rate of the top 1 prediction, which was significantly better than the result of HitpickV2 (30.43%). Finally, DStruBTarget was integrated with protein BLAST to predict the ligand-binding proteins not limited in a certain database. DStruBTarget with BLAST was tested in the DBD dataset. Its top 1 hit rate was 51.15%, which is lower than DStruBTarget without BLAST. Further comparison was on the ligands that bind to multiple numbers of proteins, which illustrated that DStruBTarget with BLAST performed better than without BLAST when the number of binding proteins of the query ligands is larger than six. Meanwhile, the prediction power of the DStruBTarget with BLAST in top 1 prediction was found to be positively correlated with the number of proteins binding with the query ligands, while the top 1 prediction power of DStruBTarget without BLAST was negatively correlated with the number of binding proteins for query ligands. Thus, DStruBTarget with BLAST is a potentially useful approach for predicting novel proteins for ligands that bind to multiple proteins.


Subject(s)
Proteins/metabolism , Crystallography, X-Ray , Datasets as Topic , Ligands , Molecular Docking Simulation , Protein Binding , Protein Conformation , Proteins/chemistry
20.
Nat Cell Biol ; 21(4): 498-510, 2019 04.
Article in English | MEDLINE | ID: mdl-30936474

ABSTRACT

Metabolic reprogramming is a hallmark of cancer. Here, we demonstrate that tumour-associated macrophages (TAMs) enhance the aerobic glycolysis and apoptotic resistance of breast cancer cells via the extracellular vesicle (EV) transmission of a myeloid-specific lncRNA, HIF-1α-stabilizing long noncoding RNA (HISLA). Mechanistically, HISLA blocks the interaction of PHD2 and HIF-1α to inhibit the hydroxylation and degradation of HIF-1α. Reciprocally, lactate released from glycolytic tumour cells upregulates HISLA in macrophages, constituting a feed-forward loop between TAMs and tumour cells. Blocking EV-transmitted HISLA inhibits the glycolysis and chemoresistance of breast cancer in vivo. Clinically, HISLA expression in TAMs is associated with glycolysis, poor chemotherapeutic response and shorter survival of patients with breast cancer. Our study highlights the potential of lncRNAs as signal transducers that are transmitted between immune and tumour cells via EVs to promote cancer aerobic glycolysis.


Subject(s)
Breast Neoplasms/metabolism , Extracellular Vesicles/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/metabolism , RNA, Long Noncoding/metabolism , Aerobiosis , Apoptosis , Breast Neoplasms/genetics , Cell Line, Tumor , Cells, Cultured , Extracellular Vesicles/genetics , Female , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Lactic Acid/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...