Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Immunol Immunother ; 72(2): 351-369, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35895109

ABSTRACT

BACKGROUND: Immunotherapy is an emerging cancer therapy with potential great success; however, immune checkpoint inhibitor (e.g., anti-PD-1) has response rates of only 10-30% in solid tumor because of the immunosuppressive tumor microenvironment (TME). This affliction can be solved by vascular normalization and TME reprogramming. METHODS: By using the single-cell RNA sequencing (scRNAseq) approach, we tried to find out the reprogramming mechanism that the Fc-VEGF chimeric antibody drug (Fc-VFD) enhances immune cell infiltration in the TME. RESULTS: In this work, we showed that Fc-VEGF121-VEGF165 (Fc-VEGF chimeric antibody drug, Fc-VFD) arrests excess angiogenesis and tumor growth through vascular normalization using in vitro and in vivo studies. The results confirmed that the treatment of Fc-VFD increases immune cell infiltration including cytotoxic T, NK, and M1-macrophages cells. Indeed, Fc-VFD inhibits Lon-induced M2 macrophages polarization that induces angiogenesis. Furthermore, Fc-VFD inhibits the secretion of VEGF-A, IL-6, TGF-ß, or IL-10 from endothelial, cancer cells, and M2 macrophage, which reprograms immunosuppressive TME. Importantly, Fc-VFD enhances the synergistic effect on the combination immunotherapy with anti-PD-L1 in vivo. CONCLUSIONS: In short, Fc-VFD fusion normalizes intratumor vasculature to reprogram the immunosuppressive TME and enhance cancer immunotherapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Tumor Microenvironment , Vascular Endothelial Growth Factor A , Immunotherapy , Antineoplastic Agents/pharmacology , Immunosuppressive Agents/pharmacology
2.
Hematol Oncol ; 41(3): 499-509, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36790759

ABSTRACT

Our previous study demonstrated that myc, mitochondrial oxidative phosphorylation, mTOR, and stemness are independently responsible for chemoresistance in acute myeloid leukemia (AML) cells. This study aimed to identify potential mechanisms of chemoresistance of the "7 + 3" induction in AML by using a single-cell RNA sequencing (scRNA-seq) approach. In the present study, 13 untreated patients with de novo AML were enrolled and stratified into two groups: complete remission (CR; n = 8) and non-CR (n = 5). Single-cell RNA sequencing was used to analyze genetic profiles of 28,950 AML cells from these patients; results were validated using a previously published bulk RNA-seq dataset. Our study results showed chemoresistant AML cells had premature accumulation during early hematopoiesis. Hematopoietic stem cell-like cells from the non-CR group expressed more leukemic stem cell markers (CD9, CD82, IL3RA, and IL1RAP) than those from the CR group. Chemoresistant progenitor cells had impaired myeloid differentiation owing to early arrest of hematopoiesis. Notably, AML cells analyzed by scRNA-seq and bulk RNA-seq harbored a comparable myeloid lineage cell fraction, which internally validated our results. Using the TCGA database, our analysis demonstrated that patients with AML with higher expression of chemoresistant genetic markers (IL3RA and IL1RAP) had a worse overall survival (p < 0.01 for IL3RA; p < 0.05 for IL1RAP). In conclusion, AML cells responsive and resistant to the "7 + 3" induction were derived from a diverse cancerous hematopoietic stem cell population, as indicated by the specific genetic biomarkers obtained using scRNA-seq approach. Furthermore, arrest of hematopoiesis was shown to occur earlier in chemoresistant AML cells, furthering the current understanding of chemoresistance in AML.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute , Humans , Drug Resistance, Neoplasm/genetics , Prognosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Hematopoietic Stem Cells , Sequence Analysis, RNA
3.
J Clin Lab Anal ; 34(4): e23110, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31733007

ABSTRACT

BACKGROUND: Despite having chronic gastritis, most people infected by Helicobacter pylori (H. pylori) are asymptomatic and have no specific clinical signs and symptoms. H. pylori infection can be diagnosed by several detection methods. Giemsa stain and rapid urease test (CLO test) are the most performed tests of H. pylori infection at first-line clinical examination because of their simplicity and reliability. However, the sensitivity of CLO test is significantly reduced in patients with atrophic gastritis and intestinal metaplasia, and the weaknesses of Giemsa stain are higher cost and time-consuming. METHODS: The Giemsa stain was modified in several staining solutions and procedures based on the simplified Giemsa technique described by Gray, Wyatt, & Rathbone (1986). The modified Giemsa stain is examined its efficacy and compared with the CLO test using 233 H. pylori-infected patients with gastric disease. RESULTS: The modified Giemsa stain is comparable to the traditional one. Statistical analysis indicated that the modified Giemsa stain obtains greater accuracy in H. pylori-infected patients with gastritis and ulcer than the CLO test (48.1% vs. 43.7%). Moreover, considering the prognosis of different symptoms of gastric diseases, the modified Giemsa stain has a more accurate prognosis than combination symptoms (P = 1.8E-05 vs. P = 5.49E-05). The modified Giemsa stain is confirmed to be better than CLO test using 233 H. pylori-infected patients with gastric disease. CONCLUSIONS: The modified Giemsa stain is more simplified and time-saving than traditional Giemsa stain, which is comparable to the traditional one and is confirmed to be better than CLO test using 233 H. pylori-infected patients with gastric disease. In clinical examination, this modified Giemsa stain can be applied to routine examination and provides quick and accurate diagnosis and prognosis to H. pylori-infected patients with gastric diseases.


Subject(s)
Azure Stains , Helicobacter Infections/diagnosis , Helicobacter Infections/pathology , Urease , Biopsy , Gastritis/microbiology , Humans , Stomach Ulcer/microbiology , Urease/metabolism
4.
Environ Res ; 162: 261-270, 2018 04.
Article in English | MEDLINE | ID: mdl-29367177

ABSTRACT

BACKGROUND: Phthalic acid esters are ubiquitous and antiandrogenic, and may cause systemic effects in humans, particularly with in utero exposure. Epigenetic modification, such as DNA methylation, has been hypothesized to be an important mechanism that mediates certain biological processes and pathogenic effects of in utero phthalate exposure. OBJECTIVE: The aim of this study was to examine the association between genome-wide DNA methylation at birth and prenatal exposure to phthalate. METHODS: We studied 64 infant-mother pairs included in TMICS (Taiwan Maternal and Infant Cohort Study), a long-term follow-up birth cohort from the general population. DNA methylation levels at more than 450,000 CpG sites were measured in cord blood samples using Illumina Infinium HumanMethylation450 BeadChips. The concentrations of three metabolites of di-(2-ethylhexyl) phthalate (DEHP) were measured using liquid chromatography tandem-mass spectrometry (LC-MS/MS) in urine samples collected from the pregnant women during 28-36 weeks gestation. RESULTS: We identified 25 CpG sites whose methylation levels in cord blood were significantly correlated with prenatal DEHP exposure using a false discovery rate (FDR) of 5% (q-value < 0.05). Via gene-set enrichment analysis (GSEA), we also found that there was significant enrichment of genes involved in the androgen response, estrogen response, and spermatogenesis within those genes showing DNA methylation changes in response to exposure. Specifically, PA2G4, HMGCR, and XRCC6 genes were involved in genes in response to androgen. CONCLUSIONS: Phthalate exposure in utero may cause significant alterations in the DNA methylation in cord blood. These changes in DNA methylation might serve as biomarkers of maternal exposure to phthalate in infancy and potential candidates for studying mechanisms via which phthalate may impact on health in later life. Future investigations are warranted.


Subject(s)
DNA Methylation , Diethylhexyl Phthalate , Endocrine Disruptors , Phthalic Acids , Prenatal Exposure Delayed Effects , Adaptor Proteins, Signal Transducing/metabolism , Chromatography, Liquid , Cohort Studies , Endocrine Disruptors/toxicity , Female , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Infant, Newborn , Ku Autoantigen/metabolism , Male , Maternal Exposure , Phthalic Acids/toxicity , Pregnancy , RNA-Binding Proteins/metabolism , Taiwan , Tandem Mass Spectrometry
5.
Am J Respir Crit Care Med ; 195(5): 663-673, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27669169

ABSTRACT

RATIONALE: Patients with non-small cell lung cancer (NSCLC) with mutated epidermal growth factor receptor (EGFR) are relatively sensitive to EGFR-tyrosine kinase inhibitor (TKI) treatment and have longer progression-free survival (PFS) when treated with EGFR-TKI compared with platinum-based chemotherapy. However, many patients with advanced NSCLC who have mutated EGFR do not respond to first-line EGFR-TKI treatment and still have shorter PFS. OBJECTIVES: The aim of this study was to identify genetic variants associated with PFS among patients with lung adenocarcinoma who were treated with first-line EGFR-TKIs. METHODS: A genome-wide association study on PFS was performed in never-smoking women diagnosed with lung adenocarcinoma and who were treated with first-line EGFR-TKIs (n = 128). Significant single-nucleotide polymorphisms (SNPs) were selected for follow-up association analysis (n = 198) and for replication assay in another independent cohort (n = 153). MEASUREMENTS AND MAIN RESULTS: We identified SNPs at 4q12 associated with PFS at genome-wide significance (P < 10-8) and with an estimated hazard ratio of more than 4. This association was also replicated in a larger but similar cohort and in an independent NSCLC cohort. Follow-up functional analyses showed that these SNPs were associated with the expression of EGFR, which encodes the TKI target, and with a nearby gene neuromedin-U, which encodes a G protein-coupled receptor ligand known to be involved in the progression of NSCLC. Considering these as possible prognostic biomarkers for the treatment of patients with late-stage lung cancer, we found that these SNPs were not associated with EGFR mutation status or with polymorphism of the Bcl2-interacting mediator of cell death gene. CONCLUSIONS: Genetic variants in 4q12 merit further investigation to assess their potential as pharmacogenomic predictors for and to understand the biology underlying its influence on PFS in patients treated with TKI therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Follow-Up Studies , Genome-Wide Association Study/methods , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Young Adult
6.
Mol Carcinog ; 55(5): 499-513, 2016 May.
Article in English | MEDLINE | ID: mdl-25728212

ABSTRACT

Growth differentiation factor-10 (GDF10), commonly referred as BMP3b, is a member of the transforming growth factor-ß (TGF-ß) superfamily. GDF10/BMP3b has been considered as a tumor suppressor, however, little is known about the molecular mechanism of its roles in tumor suppression in oral cancer. Clinical significance of GDF10 downregulation in oral squamous cell carcinoma (OSCC) was evaluated using three independent cohorts of OSCC patients. The molecular mechanisms of GDF10 in the suppression of cell survival, cell migration/invasion and epithelial-mesenchymal transition (EMT) were investigated by using oral cancer cell lines. The present study shows that GDF10 is downregulated during oral carcinogenesis, and GDF10 expression is also an independent risk factor for overall survival of OSCC patients. Overexpression of GDF10 attenuates cell proliferation, transformation, migration/invasion, and EMT. GDF10-inhibited EMT is mediated by ERK signaling but not by typical TGF-ß signaling. In addition, overexpression of GDF10 promotes DNA damage-induced apoptosis and sensitizes the response to all-trans retinoic acid (ATRA) and camptothecin (CPT). Intriguingly, the expression of GDF10 is induced by type III TGF-ß receptor (TGFBR3) through TGF-ß-SMAD2/3 signaling. Our findings suggest that TGFBR3 is an upstream activator of GDF10 expression and they share the same signaling to inhibit EMT and migration/invasion. These results support that GDF10 acts as a hinge to collaborate with TGFBR3 in the transition of EMT-MET program. Taken together, we illustrated the clinical significance and the molecular mechanisms of tumor-suppressive GDF10 in OSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Growth Differentiation Factor 10/metabolism , Mouth Neoplasms/pathology , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Growth Differentiation Factor 10/genetics , Humans , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Prognosis , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/genetics , Survival Analysis
7.
PLoS Genet ; 9(7): e1003652, 2013.
Article in English | MEDLINE | ID: mdl-23874238

ABSTRACT

Coronary heart disease (CHD) is the leading cause of mortality in both developed and developing countries worldwide. Genome-wide association studies (GWAS) have now identified 46 independent susceptibility loci for CHD, however, the biological and disease-relevant mechanisms for these associations remain elusive. The large-scale meta-analysis of GWAS recently identified in Caucasians a CHD-associated locus at chromosome 6q23.2, a region containing the transcription factor TCF21 gene. TCF21 (Capsulin/Pod1/Epicardin) is a member of the basic-helix-loop-helix (bHLH) transcription factor family, and regulates cell fate decisions and differentiation in the developing coronary vasculature. Herein, we characterize a cis-regulatory mechanism by which the lead polymorphism rs12190287 disrupts an atypical activator protein 1 (AP-1) element, as demonstrated by allele-specific transcriptional regulation, transcription factor binding, and chromatin organization, leading to altered TCF21 expression. Further, this element is shown to mediate signaling through platelet-derived growth factor receptor beta (PDGFR-ß) and Wilms tumor 1 (WT1) pathways. A second disease allele identified in East Asians also appears to disrupt an AP-1-like element. Thus, both disease-related growth factor and embryonic signaling pathways may regulate CHD risk through two independent alleles at TCF21.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Coronary Disease/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Transcription Factor AP-1/genetics , Alleles , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cells, Cultured , Coronary Disease/pathology , Coronary Vessels/cytology , Coronary Vessels/growth & development , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Genome-Wide Association Study , Haplotypes , Humans , Myocytes, Smooth Muscle/cytology , Polymorphism, Single Nucleotide , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Wilms Tumor/genetics , Wilms Tumor/metabolism
8.
Hum Genet ; 134(3): 333-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25566987

ABSTRACT

We previously carried out a multi-stage genome-wide association study (GWAS) on lung cancer among never smokers in the Female Lung Cancer Consortium in Asia (FLCCA) (6,609 cases, 7,457 controls) that identified novel susceptibility loci at 10q25.2, 6q22.2, and 6p21.32, and confirmed two previously identified loci at 5p15.33 and 3q28. Household air pollution (HAP) attributed to solid fuel burning for heating and cooking, is the leading cause of the overall disease burden in Southeast Asia, and is known to contain lung carcinogens. To evaluate the gene-HAP interactions associated with lung cancer in loci independent of smoking, we analyzed data from studies participating in FLCCA with fuel use information available (n = 3; 1,731 cases; 1,349 controls). Coal use was associated with a 30% increased risk of lung cancer (OR 1.3, 95% CI 1.0-1.6). Among the five a priori SNPs identified by our GWAS, two showed a significant interaction with coal use (HLA Class II rs2395185, p = 0.02; TP63 rs4488809 (rs4600802), p = 0.04). The risk of lung cancer associated with coal exposure varied with the respective alleles for these two SNPs. Our observations provide evidence that genetic variation in HLA Class II and TP63 may modify the association between HAP and lung cancer risk. The roles played in the cell cycle and inflammation pathways by the proteins encoded by these two genes provide biological plausibility for these interactions; however, additional replication studies are needed in other non-smoking populations.


Subject(s)
Adenocarcinoma/genetics , Air Pollutants/toxicity , Lung Neoplasms/genetics , Adenocarcinoma/chemically induced , Adult , Aged , Air Pollution, Indoor , Case-Control Studies , Female , Gene-Environment Interaction , Genetic Markers , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Lung Neoplasms/chemically induced , Middle Aged , Polymorphism, Single Nucleotide , Risk
9.
Ann Surg Oncol ; 22(3): 889-98, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25212833

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is defined by reduced expression of the estrogen receptor, progesterone receptor, and HER2. TNBC is an especially aggressive group of breast cancers with poor prognosis. There are currently no validated molecular targets to effectively treat this disease. Thus, it is necessary to identify effective molecular targets and therapeutic strategies for TNBC patients. METHODS: The expression of HSPA5 in patients with breast cancer was examined by immunohistochemistry. The association of HSPA5 expression with tumor grade and metastatic events in TNBC patients was analyzed using the Oncomine database. The knockdown and overexpression of HSPA5 protein were performed to investigate the effects on E1A-suppressed cell migration/invasion of TNBC using in vitro transwell assays and tumor growth/experimental metastasis studies in animal models. RESULTS: The expression of HSPA5 was positively correlated with high-grade tumors, metastatic events, and poor overall survival in breast cancer patients with TNBC. E1A-inhibited HSPA5 expression suppressed cell migration/invasive ability of TNBC cell lines. Moreover, E1A significantly abolished lung metastases from breast cancer cells by inhibiting HSPA5 expression in a xenograft tumor model. CONCLUSIONS: The overexpression of HSPA5 is critical for high-risk metastasis of breast cancer and TNBC. The results of our study suggest that HSPA5 may be a crucial mediator of E1A-suppressed metastatic ability of breast cancer cells. Thus, E1A may be a potential target for diagnosis and individualized treatment in clinical practice.


Subject(s)
Adenovirus E1A Proteins/genetics , Cell Movement , Cell Proliferation , Heat-Shock Proteins/antagonists & inhibitors , Lung Neoplasms/prevention & control , Triple Negative Breast Neoplasms/prevention & control , Animals , Apoptosis , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Mice , Mice, SCID , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Survival Rate , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Int J Cancer ; 135(4): 809-19, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24477565

ABSTRACT

SOX2 is a transcription factor essential for self-renewal and pluripotency of embryonic stem cells. Recently, SOX2 was found overexpressed in the majority of the lung squamous cell carcinoma (SQC), in which it acts as a lineage-survival oncogene. However, downstream targets/pathways of SOX2 in lung SQC cells remain to be identified. Here, we show that BMP4 is a downstream target of SOX2 in lung SQC. We found that SOX2-silencing-mediated inhibition of cell growth was accompanied by upregulation of BMP4 mRNA and its protein expression. Meta-analysis with 293 samples and qRT-PCR validation with 73 clinical samples revealed an inversely correlated relationship between levels of SOX2 and BMP4 mRNA, and significantly lower mRNA levels in tumor than in adjacent normal tissues. This was corroborated by immunohistochemistry analysis of 35 lung SQC samples showing lower BMP4 protein expression in tumor tissues. Cell-based experiments including siRNA transfection, growth assay and flow cytometry assay, further combined with a xenograft tumor model in mice, revealed that reactivation of BMP4 signaling could partially account for growth inhibition and cell cycle arrest in lung SQC cells upon silencing SOX2. Finally, chromatin immunoprecipitation analysis and luciferase reporter assay revealed that SOX2 could negatively regulate BMP4 promoter activity, possibly through binding to the promoter located in the first intron region of BMP4. Collectively, our findings suggest that BMP4 could act as a tumor suppressor and its downregulation by elevated SOX2 resulting in enhanced growth of lung SQC cells.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/genetics , Databases, Genetic , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Lung/metabolism , Lung Neoplasms/genetics , Mice , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Signal Transduction
11.
Gastroenterology ; 145(5): 1110-20, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23896173

ABSTRACT

BACKGROUND & AIMS: Many patients with pancreatic ductal adenocarcinoma (PDAC) develop recurrent or metastatic diseases after surgery, so it is important to identify those most likely to benefit from aggressive therapy. Disruption of tissue microarchitecture is an early step in pancreatic tumorigenesis and a parameter used in pathology grading of glandular tumors. We investigated whether changes in gene expression during pancreatic epithelial morphogenesis were associated with outcomes of patients with PDAC after surgery. METHODS: We generated architectures of human pancreatic duct epithelial cells in a 3-dimensional basement membrane matrix. We identified gene expression profiles of the cells during different stages of tubular morphogenesis (tubulogenesis) and of PANC-1 cells during spheroid formation. Differential expression of genes was confirmed by immunoblot analysis. We compared the gene expression profile associated with pancreatic epithelial tubulogenesis with that of PDAC samples from 27 patients, as well as with their outcomes after surgery. RESULTS: We identified a gene expression profile associated with tubulogenesis that resembled the profile of human pancreatic tissue with differentiated morphology and exocrine function. Patients with PDACs with this profile fared well after surgery. Based on this profile, we established a 6-28 gene tubulogenesis-specific signature that accurately determined the prognosis of independent cohorts of patients with PDAC (total n = 128; accuracy = 81.2%-95.0%). One gene, ASPM, was down-regulated during tubulogenesis but up-regulated in human PDAC cell lines and tumor samples; up-regulation correlated with patient outcomes (Cox regression P = .0028). Bioinformatic, genetic, biochemical, functional, and clinical correlative studies showed that ASPM promotes aggressiveness of PDAC by maintaining Wnt-ß-catenin signaling and stem cell features of PDAC cells. CONCLUSIONS: We identified a gene expression profile associated with pancreatic epithelial tubulogenesis and a tissue architecture-specific signature of PDAC cells that is associated with patient outcomes after surgery.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Nerve Tissue Proteins/physiology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Transcriptome/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/physiology , Carcinoma, Pancreatic Ductal/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Disease Models, Animal , Epithelium/pathology , Follow-Up Studies , Gene Expression Regulation, Neoplastic/physiology , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/genetics , Pancreatic Neoplasms/genetics , Prognosis , Retrospective Studies , Signal Transduction/genetics , Signal Transduction/physiology , Transcriptome/physiology , Wnt Proteins/physiology , beta Catenin/physiology
12.
Am J Cancer Res ; 14(6): 3171-3185, 2024.
Article in English | MEDLINE | ID: mdl-39005677

ABSTRACT

Early detection of cancer recurrence using specific biomarkers remains a clinically unmet need, although methodologies for monitoring tumor markers, cell-free DNA, and circulating tumor cells have been established for decades. Tumor recurrence develops in metastatic or dormant cancer cells under continuous immune surveillance. Alterations in the population and function of immune cells may contribute to cancer recurrence. Here, we utilized an animal model to imitate breast tumor recurrence after surgical resection and investigated the abundance and gene expression profiles of immune cells using NanoString analysis. Bioinformatic analysis of a published single-cell RNA sequencing database of myeloid-derived suppressor cells (MDSCs) was performed to identify common targets between the two studies. Identified biomarkers were validated using human peripheral blood mononuclear cell (PBMC) datasets. The inhibitory effect of MDSCs on T-cell proliferation was assessed in vitro. Our data demonstrated that the number of MDSCs significantly increased during recurrence. Comparison of our NanoString data with a single-cell RNA sequencing dataset of MDSCs in another spontaneous breast cancer model identified colony-stimulating factor 3 receptor (Csf3r)-positive MDSCs as a potential marker for predicting tumor relapse. We validated our findings using two previously published PBMC databases of patients with breast cancer with or without recurrence and confirmed the elevated MDSC gene signature and CSF3R expression in patients with tumor recurrence. 35 patients with breast cancer were also included in our study, that patients with higher levels of CSF3R had worse survival. In vitro experiments demonstrated that Csf3r + MDSCs exhibited enhanced reactive oxygen species (ROS) levels and robust T-cell suppression ability. We conclude that an increase in CSF3R + MDSCs is a potential biomarker for early detection of tumor recurrence in patients with breast cancer.

13.
Aging (Albany NY) ; 16(9): 7889-7901, 2024 05 03.
Article in English | MEDLINE | ID: mdl-38709264

ABSTRACT

Despite neoadjuvant chemoradiotherapy (CRT) being the established standard for treating advanced rectal cancer, clinical outcomes remain suboptimal, necessitating the identification of predictive biomarkers for improved treatment decisions. Previous studies have hinted at the oncogenic properties of the Fc fragment of IgG binding protein (FCGBP) in various cancers; however, its clinical significance in rectal cancer remains unclear. In this study, we first conducted an analysis of a public transcriptome comprising 46 rectal cancer patients. Focusing on cell adhesion during data mining, we identified FCGBP as the most upregulated gene associated with CRT resistance. Subsequently, we assessed FCGBP immunointensity using immunohistochemical staining on 343 rectal cancer tissue blocks. Elevated FCGBP immunointensity correlated with lymph node involvement before treatment (p = 0.001), tumor invasion, and lymph node involvement after treatment (both p < 0.001), vascular invasion (p = 0.001), perineural invasion (p = 0.041), and reduced tumor regression (p < 0.001). Univariate analysis revealed a significant association between high FCGBP immunoexpression and inferior disease-specific survival, local recurrence-free survival, and metastasis-free survival (all p ≤ 0.0002). Furthermore, high FCGBP immunoexpression independently emerged as an unfavorable prognostic factor for all three survival outcomes in the multivariate analysis (all p ≤ 0.025). Enriched pathway analysis substantiated the role of FCGBP in conferring resistance to radiation. In summary, our findings suggest that elevated FCGBP immunoexpression in rectal cancer significantly correlates with a poor response to CRT and diminished patient survival. FCGBP holds promise as a valuable prognostic biomarker for rectal cancer patients undergoing CRT.


Subject(s)
Chemoradiotherapy , Rectal Neoplasms , Humans , Rectal Neoplasms/therapy , Rectal Neoplasms/metabolism , Rectal Neoplasms/pathology , Rectal Neoplasms/genetics , Rectal Neoplasms/mortality , Female , Male , Middle Aged , Chemoradiotherapy/methods , Aged , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Prognosis , Treatment Outcome , Neoadjuvant Therapy/methods , Adult
14.
Cells ; 13(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38994997

ABSTRACT

Extracellular HSP90α (eHSP90α) is a promoter of tumor development and malignant progression. Patients with malignancies, including pancreatic ductal adenocarcinoma (PDAC), have generally shown 5~10-fold increases in serum/plasma eHSP90α levels. In this study, we developed a humanized antibody HH01 to target eHSP90α and evaluated its anticancer efficacy. HH01, with novel complementarity-determining regions, exhibits high binding affinity toward HSP90α. It recognizes HSP90α epitope sites 235AEEKEDKEEE244 and 251ESEDKPEIED260, with critical amino acid residues E237, E239, D240, K241, E253, and K255. HH01 effectively suppressed eHSP90α-induced invasive and spheroid-forming activities of colorectal cancer and PDAC cell lines by blocking eHSP90α's ligation with the cell-surface receptor CD91. In mouse models, HH01 potently inhibited the tumor growth of PDAC cell grafts/xenografts promoted by endothelial-mesenchymal transition-derived cancer-associated fibroblasts while also reducing serum eHSP90α levels, reflecting its anticancer efficacy. HH01 also modulated tumor immunity by reducing M2 macrophages and reinvigorating immune T-cells. Additionally, HH01 showed low aggregation propensity, high water solubility, and a half-life time of >18 days in mouse blood. It was not cytotoxic to retinal pigmented epithelial cells and showed no obvious toxicity in mouse organs. Our data suggest that targeting eHSP90α with HH01 antibody can be a promising novel strategy for PDAC therapy.


Subject(s)
Antibodies, Monoclonal, Humanized , HSP90 Heat-Shock Proteins , Pancreatic Neoplasms , Humans , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Animals , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Mice , Cell Line, Tumor , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Xenograft Model Antitumor Assays , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Endothelial-Mesenchymal Transition
15.
Cell Death Dis ; 14(3): 199, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36927870

ABSTRACT

During hypoxia, FUNDC1 acts as a mitophagy receptor and accumulates at the ER (endoplasmic reticulum)-mitochondria contact sites (EMC), also called mitochondria-associated membranes (MAM). In mitophagy, the ULK1 complex phosphorylates FUNDC1(S17) at the EMC site. However, how mitochondria sense the stress and send the signal from the inside to the outside of mitochondria to trigger mitophagy is still unclear. Mitochondrial Lon was reported to be localized at the EMC under stress although the function remained unknown. In this study, we explored the mechanism of how mitochondrial sensors of hypoxia trigger and stabilize the FUNDC1-ULK1 complex by Lon in the EMC for cell survival and cancer progression. We demonstrated that Lon is accumulated in the EMC and associated with FUNDC1-ULK1 complex to induce mitophagy via chaperone activity under hypoxia. Intriguingly, we found that Lon-induced mitophagy is through binding with mitochondrial Na+/Ca2+ exchanger (NCLX) to promote FUNDC1-ULK1-mediated mitophagy at the EMC site in vitro and in vivo. Accordingly, our findings highlight a novel mechanism responsible for mitophagy initiation under hypoxia by chaperone Lon in mitochondria through the interaction with FUNDC1-ULK1 complex at the EMC site. These findings provide a direct correlation between Lon and mitophagy on cell survival and cancer progression.


Subject(s)
Membrane Proteins , Mitophagy , Humans , Phosphorylation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Hypoxia/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
16.
Sci Rep ; 13(1): 6727, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185775

ABSTRACT

Because of the cancer incidence increase and population aging in Taiwan, we aimed to assess the cancer prevalence, to summarize the comorbidities of older patients with the five most common cancers (i.e., breast, colorectal, liver, lung, and oral), and to develop a Taiwan cancer comorbidity index (TCCI) for studying their actual prognosis. The linkage of the Taiwan Cancer Registry, Cause of Death Database, and National Health Insurance Research Database was used. We followed the standard statistical learning steps to obtain a survival model with good discriminatory accuracy in predicting death due to noncancer causes, from which we obtained the TCCI and defined comorbidity levels. We reported the actual prognosis by age, stage, and comorbidity level. In Taiwan, cancer prevalence nearly doubled in 2004-2014, and comorbidities were common among older patients. Stage was the major predictor of patients' actual prognoses. For localized and regional breast, colorectal, and oral cancers, comorbidities correlated with noncancer-related deaths. Compared with the US, the chances of dying from comorbidities in Taiwan were lower and the chances of dying from cancer were higher for breast, colorectal, and male lung cancers. These actual prognoses could help clinicians and patients in treatment decision-making and help policymakers in resource planning.


Subject(s)
Colorectal Neoplasms , Lung Neoplasms , Humans , Male , Aged , Prevalence , Taiwan/epidemiology , Comorbidity , Lung Neoplasms/epidemiology , Colorectal Neoplasms/epidemiology
18.
Cancer Epidemiol Biomarkers Prev ; 31(12): 2208-2218, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36129788

ABSTRACT

BACKGROUND: Methods synthesizing multiple data sources without prospective datasets have been proposed for absolute risk model development. This study proposed methods for adapting risk models for another population without prospective cohorts, which would help alleviate the health disparities caused by advances in absolute risk models. To exemplify, we adapted the lung cancer risk model PLCOM2012, well studied in the west, for Taiwan. METHODS: Using Taiwanese multiple data sources, we formed an age-matched case-control study of ever-smokers (AMCCSE), estimated the number of ever-smoking lung cancer patients in 2011-2016 (NESLP2011), and synthesized a dataset resembling the population of cancer-free ever-smokers in 2010 regarding the PLCOM2012 risk factors (SPES2010). The AMCCSE was used to estimate the overall calibration slope, and the requirement that NESLP2011 equals the estimated total risk of individuals in SPES2010 was used to handle the calibration-in-the-large problem. RESULTS: The adapted model PLCOT-1 (PLCOT-2) had an AUC of 0.78 (0.75). They had high performance in calibration and clinical usefulness on subgroups of SPES2010 defined by age and smoking experience. Selecting the same number of individuals for low-dose computed tomography screening using PLCOT-1 (PLCOT-2) would have identified approximately 6% (8%) more lung cancers than the US Preventive Services Task Forces 2021 criteria. Smokers having 40+ pack-years had an average PLCOT-1 (PLCOT-2) risk of 3.8% (2.6%). CONCLUSIONS: The adapted PLCOT models had high predictive performance. IMPACT: The PLCOT models could be used to design lung cancer screening programs in Taiwan. The methods could be applicable to other cancer models.


Subject(s)
Lung Neoplasms , Humans , Lung Neoplasms/epidemiology , Early Detection of Cancer , Taiwan/epidemiology , Case-Control Studies , Smokers
19.
Sci Rep ; 11(1): 644, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436772

ABSTRACT

Ephrin type-A receptor 10 (EPHA10) has been implicated as a potential target for breast and prostate cancer therapy. However, its involvement in oral squamous cell carcinoma (OSCC) remains unclear. We demonstrated that EPHA10 supports in vivo tumor growth and lymphatic metastasis of OSCC cells. OSCC cell migration, epithelial mesenchymal transition (EMT), and sphere formation were found to be regulated by EPHA10, and EPHA10 was found to drive expression of some EMT- and stemness-associated transcription factors. Among EPHA10 ligands, exogenous ephrin A4 (EFNA4) induced the most OSCC cell migration and sphere formation, as well as up-regulation of SNAIL, NANOG, and OCT4. These effects were abolished by extracellular signal-regulated kinase (ERK) inhibition and NANOG knockdown. Also, EPHA10 was required for EFNA4-induced cell migration, sphere formation, and expression of NANOG and OCT4 mRNA. Our microarray dataset revealed that EFNA4 mRNA expression was associated with expression of NANOG and OCT4 mRNA, and OSCC patients showing high co-expression of EFNA4 with NANOG or OCT4 mRNA demonstrated poor recurrence-free survival rates. Targeting forward signaling of the EFNA4-EPHA10 axis may be a promising therapeutic approach for oral malignancies, and the combination of EFNA4 mRNA and downstream gene expression may be a useful prognostic biomarker for OSCC.


Subject(s)
Cell Movement , Ephrin-A4/metabolism , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/pathology , Nanog Homeobox Protein/metabolism , Receptors, Eph Family/metabolism , Spheroids, Cellular/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Ephrin-A4/genetics , Epithelial-Mesenchymal Transition , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Nanog Homeobox Protein/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Receptors, Eph Family/genetics , Spheroids, Cellular/metabolism , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Environ Int ; 147: 105975, 2021 02.
Article in English | MEDLINE | ID: mdl-33385923

ABSTRACT

We previously identified 10 lung adenocarcinoma susceptibility loci in a genome-wide association study (GWAS) conducted in the Female Lung Cancer Consortium in Asia (FLCCA), the largest genomic study of lung cancer among never-smoking women to date. Furthermore, household coal use for cooking and heating has been linked to lung cancer in Asia, especially in Xuanwei, China. We investigated the potential interaction between genetic susceptibility and coal use in FLCCA. We analyzed GWAS-data from Taiwan, Shanghai, and Shenyang (1472 cases; 1497 controls), as well as a separate study conducted in Xuanwei (152 cases; 522 controls) for additional analyses. We summarized genetic susceptibility using a polygenic risk score (PRS), which was the weighted sum of the risk-alleles from the 10 previously identified loci. We estimated associations between a PRS, coal use (ever/never), and lung adenocarcinoma with multivariable logistic regression models, and evaluated potential gene-environment interactions using likelihood ratio tests. There was a strong association between continuous PRS and lung adenocarcinoma among never coal users (Odds Ratio (OR) = 1.69 (95% Confidence Interval (CI) = 1.53, 1.87), p=1 × 10-26). This effect was attenuated among ever coal users (OR = 1.24 (95% CI: 1.03, 1.50), p = 0.02, p-interaction = 6 × 10-3). We observed similar attenuation among coal users from Xuanwei. Our study provides evidence that genetic susceptibility to lung adenocarcinoma among never-smoking Asian women is weaker among coal users. These results suggest that lung cancer pathogenesis may differ, at least partially, depending on exposure to coal combustion products. Notably, these novel findings are among the few instances of sub-multiplicative gene-environment interactions in the cancer literature.


Subject(s)
Adenocarcinoma of Lung , Air Pollution, Indoor , Lung Neoplasms , Adenocarcinoma of Lung/epidemiology , Adenocarcinoma of Lung/genetics , Asia , Case-Control Studies , China/epidemiology , Coal , Female , Genome-Wide Association Study , Humans , Lung Neoplasms/epidemiology , Lung Neoplasms/genetics , Risk Factors , Smoking , Taiwan
SELECTION OF CITATIONS
SEARCH DETAIL