Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 526
Filter
1.
PeerJ ; 12: e17602, 2024.
Article in English | MEDLINE | ID: mdl-38952968

ABSTRACT

Background: Peritoneal metastasis (PM) is the most prevalent type of metastasis in patients with gastric cancer (GC) and has an extremely poor prognosis. The detection of free cancer cells (FCCs) in the peritoneal cavity has been demonstrated to be one of the worst prognostic factors for GC. However, there is a lack of sensitive detection methods for FCCs in the peritoneal cavity. This study aimed to use a new peritoneal lavage fluid cytology examination to detect FCCs in patients with GC, and to explore its clinical significance on diagnosing of occult peritoneal metastasis (OPM) and prognosis. Methods: Peritoneal lavage fluid from 50 patients with GC was obtained and processed via the isolation by size of epithelial tumor cells (ISET) method. Immunofluorescence and fluorescence in situ hybridization (FISH) were used to identify FCCs expressing chromosome 8 (CEP8), chromosome 17 (CEP17), and epithelial cell adhesion molecule (EpCAM). Results: Using a combination of the ISET platform and immunofluorescence-FISH, the detection of FCCs was higher than that by light microscopy (24.0% vs. 2.0%). Samples were categorized into positive and negative groups, based on the expressions of CEP8, CEP17, and EpCAM. Statistically significant relationships were demonstrated between age (P = 0.029), sex (P = 0.002), lymphatic invasion (P = 0.001), pTNM stage (P = 0.001), and positivity for FCCs. After adjusting for covariates, patients with positive FCCs had lower progression-free survival than patients with negative FCCs. Conclusion: The ISET platform highly enriched nucleated cells from peritoneal lavage fluid, and indicators comprising EpCAM, CEP8, and CEP17 confirmed the diagnosis of FCCs. As a potential detection method, it offers an opportunity for early intervention of OPM and an extension of patient survival.


Subject(s)
In Situ Hybridization, Fluorescence , Peritoneal Lavage , Peritoneal Neoplasms , Stomach Neoplasms , Humans , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/diagnosis , Male , Female , Middle Aged , Stomach Neoplasms/pathology , Stomach Neoplasms/diagnosis , Aged , Ascitic Fluid/pathology , Ascitic Fluid/cytology , Prognosis , Epithelial Cell Adhesion Molecule/metabolism , Epithelial Cell Adhesion Molecule/genetics , Adult , Cytodiagnosis/methods , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism , Cytology
2.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928484

ABSTRACT

Platinum-resistant high-grade serous carcinoma (HGSC) is an incurable disease, so biomarkers that could help with timely treatment adjustments and personalized approach are extensively being sought. Tumor-derived extracellular vesicles (EVs) that can be isolated from ascites and blood of HGSC patients are such promising biomarkers. Epithelial cell adhesion molecule (EpCAM) expression is upregulated in most epithelium-derived tumors; however, studies on prognostic value of EpCAM overexpression in ovarian carcinoma have shown contradictory results. The aim of our study was to evaluate the potential of total and EpCAM-positive EVs as prognostic and predictive biomarkers for advanced HGSC. Flow cytometry was used to determine the concentration of total and EpCAM-positive EVs in paired pretreatment ascites and plasma samples of 37 patients with advanced HGSC who underwent different first-line therapy. We found that higher EpCAM-positive EVs concentration in ascites is associated with shorter progression-free survival (PFS) regardless of treatment strategy. We also found a strong correlation of EpCAM-positive EVs concentration between ascites and plasma. Our findings indicate that EpCAM-positive EVs in ascites of patients with advanced HGSC have the potential to serve as prognostic biomarkers for predicting early recurrence and thereby likelihood of more aggressive tumor biology and development of chemoresistance.


Subject(s)
Ascites , Biomarkers, Tumor , Cystadenocarcinoma, Serous , Epithelial Cell Adhesion Molecule , Extracellular Vesicles , Ovarian Neoplasms , Progression-Free Survival , Humans , Epithelial Cell Adhesion Molecule/metabolism , Extracellular Vesicles/metabolism , Female , Ascites/metabolism , Ascites/pathology , Middle Aged , Aged , Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/mortality , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/mortality , Prognosis , Adult , Neoplasm Grading
3.
Sci Rep ; 14(1): 14273, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38902362

ABSTRACT

Tumor-derived extracellular vesicles (EVs) show great potential as biomarkers for several diseases, including pancreatic cancer, due to their roles in cancer development and progression. However, the challenge of utilizing EVs as biomarkers lies in their inherent heterogeneity in terms of size and concentration, making accurate quantification difficult, which is highly dependent on the isolation and quantification methods used. In our study, we compared three EV isolation techniques and two EV quantification methods. We observed variations in EV concentration, with approximately 1.5-fold differences depending on the quantification method used. Interestingly, all EV isolation techniques consistently yielded similar EV quantities, overall size distribution, and modal sizes. In contrast, we found a notable increase in total EV amounts in samples from pancreatic cancer cell lines, mouse models, and patient plasma, compared to non-cancerous conditions. Moreover, individual tumor-derived EVs exhibited at least a 3-fold increase in several EV biomarkers. Our data, obtained from EVs isolated using various techniques and quantified through different methods, as well as originating from various pancreatic cancer models, suggests that EV profiling holds promise for the identification of unique and cancer-specific biomarkers in pancreatic cancer.


Subject(s)
Biomarkers, Tumor , Epithelial Cell Adhesion Molecule , Extracellular Vesicles , Glypicans , Pancreatic Neoplasms , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Extracellular Vesicles/metabolism , Humans , Biomarkers, Tumor/metabolism , Animals , Mice , Cell Line, Tumor , Epithelial Cell Adhesion Molecule/metabolism , Glypicans/metabolism , Integrin alphaV/metabolism
4.
Anal Chem ; 96(26): 10800-10808, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38904228

ABSTRACT

Tumor-derived extracellular vesicles (TEVs) are rich in cellular information and hold great promise as a biomarker for noninvasive cancer diagnosis. However, accurate measurement of TEVs presents challenges due to their low abundance and potential interference from a high number of EVs derived from normal cells. Herein, an aptamer-proximity-ligation-activated rolling circle amplification (RCA) method for EV membrane recognition, coupled with single particle inductively coupled plasma mass spectrometry (sp-ICP-MS) for the quantification of TEVs, is developed. When DNA-labeled ultrasmall gold nanoparticle (AuNP) probes bind to the long chains formed by RCA, they aggregate to form large particles. Notably, small AuNPs scarcely produce pulse signals in sp-ICP-MS, thereby detecting TEVs in a wash-free manner. By leveraging the strong binding affinity of aptamers, dual aptamers for EpCAM and PD-L1 recognition, and the sp-ICP-MS technique, this method offers remarkable sensitivity and selectivity in tracing TEVs. Under optimized conditions, the present method shows a favorable linear relationship between the pulse signal frequency of sp-ICP-MS and TEV concentration within the range of 105-107 particles/mL, along with a detection limit of 1.1 × 104 particles/mL. The pulse signals from sp-ICP-MS combined with machine learning algorithms are used to discriminate cancer patients from healthy donors with 100% accuracy. Due to its simple and fast operation and excellent sensitivity and accuracy, this approach holds significant potential for diverse applications in life sciences and personalized medicine.


Subject(s)
Aptamers, Nucleotide , Extracellular Vesicles , Gold , Mass Spectrometry , Metal Nanoparticles , Nucleic Acid Amplification Techniques , Humans , Aptamers, Nucleotide/chemistry , Extracellular Vesicles/chemistry , Nucleic Acid Amplification Techniques/methods , Metal Nanoparticles/chemistry , Gold/chemistry , Mass Spectrometry/methods , Neoplasms , Epithelial Cell Adhesion Molecule/metabolism , Limit of Detection
5.
Anal Chem ; 96(23): 9585-9592, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38816678

ABSTRACT

The PD-L1 protein on extracellular vesicles (EVs) is a promising biomarker for tumor immunotherapy. However, PD-L1+ EVs have various cell origins, so further analysis of the subpopulations is essential to help understand better their relationship with tumor immunotherapy. Different from the previous work which focus on the level of total PD-L1+ EVs expression, we, herein, report a dual-recognition mediated autocatalytic amplification (DRMAA) assay to detect the PD-L1 derived from tumors (EpCAM+), immune T cells (CD3+), and total (Lipids) EVs, respectively. The DRMAA assay employed proximity hybridization to construct a complete trigger sequence and then catalyzed the cross-hybridization of three hairpin probes, producing a three-way DNA junction (3-WJ) structure carrying the newly exposed trigger sequence. The 3-WJ complex subsequently initiated an autocatalytic amplification reaction and higher sensitivity than the traditional catalytic hairpin assembly assay was obtained. It was found that the EpCAM+ and PD-L1+ EVs were more effective than others in distinguishing lung cancer patients from healthy people. Surprisingly, the CD3+ and PD-L1+ EVs in lung cancer patients were also upregulated, indicating that immune cell-derived PD-L1+ EVs are also non-negligible marker in a tumor microenvironment. Our results suggested that the DRMAA assay would improve the study of subpopulations of PD-L1+ EVs to provide new insights for cancer immunotherapies.


Subject(s)
B7-H1 Antigen , Extracellular Vesicles , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Lung Neoplasms/immunology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Catalysis , Epithelial Cell Adhesion Molecule/metabolism , Nucleic Acid Amplification Techniques , Biomarkers, Tumor , Nucleic Acid Hybridization
6.
ACS Appl Mater Interfaces ; 16(23): 29760-29769, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38813974

ABSTRACT

Multivalent receptor-ligand interactions (RLIs) exhibit excellent affinity for binding when targeting cell membrane receptors with low expression. However, existing strategies only allow for limited control of the valency and spacing of ligands for a certain receptor, lacking recognition patterns for multiple interested receptors with complex spatial distributions. Here, we developed flexible DNA nanoclaws with multivalent aptamers to achieve powerful cell recognition by controlling the spacing of aptamers to match the spatial patterns of receptors. The DNA nanoclaw with spacing-controllable binding sites was constructed via hybrid chain reaction (HCR), enabling dual targeting of HER2 and EpCAM molecules. The results demonstrate that the binding affinity of multivalent DNA nanoclaws to tumor cells is enhanced. We speculate that the flexible structure may conform better to irregularly shaped membrane surfaces, increasing the probability of intermolecular contact. The capture efficiency of circulating tumor cells successfully verified the high affinity and selectivity of this spatial pattern. This strategy will further promote the potential application of DNA frameworks in future disease diagnosis and treatment.


Subject(s)
Aptamers, Nucleotide , DNA , Epithelial Cell Adhesion Molecule , Receptor, ErbB-2 , Humans , Aptamers, Nucleotide/chemistry , Epithelial Cell Adhesion Molecule/metabolism , Receptor, ErbB-2/metabolism , DNA/chemistry , Cell Line, Tumor , Nanostructures/chemistry , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism
7.
J Nanobiotechnology ; 22(1): 231, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720360

ABSTRACT

BACKGROUND: Circulating tumor cells (CTCs) are considered as a useful biomarker for early cancer diagnosis, which play a crucial role in metastatic process. Unfortunately, the tumor heterogeneity and extremely rare occurrence rate of CTCs among billions of interfering leukocytes seriously hamper the sensitivity and purity of CTCs isolation. METHODS: To address these, we firstly used microfluidic chips to detect the broad-spectrum of triple target combination biomarkers in CTCs of 10 types of cancer patients, including EpCAM, EGFR and Her2. Then, we constructed hybrid engineered cell membrane-camouflaged magnetic nanoparticles (HE-CM-MNs) for efficient capture of heterogeneous CTCs with high-purity, which was enabled by inheriting the recognition ability of HE-CM for various CTCs and reducing homologous cell interaction with leukocytes. Compared with single E-CM-MNs, HE-CM-MNs showed a significant improvement in the capture efficiency for a cell mixture, with an efficiency of 90%. And the capture efficiency of HE-CM-MNs toward 12 subpopulations of tumor cells was ranged from 70 to 85%. Furthermore, by using HE-CM-MNs, we successfully isolated heterogeneous CTCs with high purity from clinical blood samples. Finally, the captured CTCs by HE-CM-MNs could be used for gene mutation analysis. CONCLUSIONS: This study demonstrated the promising potential of HE-CM-MNs for heterogeneous CTCs detection and downstream analysis.


Subject(s)
Biomarkers, Tumor , Cell Membrane , Cell Separation , Magnetite Nanoparticles , Neoplastic Cells, Circulating , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism , Humans , Magnetite Nanoparticles/chemistry , Cell Separation/methods , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/chemistry , Biomarkers, Tumor/blood , Receptor, ErbB-2 , Epithelial Cell Adhesion Molecule/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Neoplasms
8.
Sci Rep ; 14(1): 12245, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806508

ABSTRACT

Following the discovery of circulating tumor cells (CTCs) in the peripheral blood of cancer patients, CTCs were initially postulated to hold promise as a valuable prognostic tool through liquid biopsy. However, a decade and a half of accumulated data have revealed significant complexities in the investigation of CTCs. A challenging aspect lies in the reduced expression or complete loss of key epithelial markers during the epithelial-mesenchymal transition (EMT). This likely hampers the identification of a pathogenetically significant subset of CTCs. Nevertheless, there is a growing body of evidence regarding the prognostic value of such molecules as CD24 expressing in the primary breast tumor. Herewith, the exact relevance of CD24 expression on CTCs remains unclear. We used two epithelial markers (EpCAM and cytokeratin 7/8) to assess the count of CTCs in 57 breast cancer patients, both with (M0mts) and without metastasis (M0) during the follow-up period, as well as in M1 breast cancer patients. However, the investigation of these epithelial markers proved ineffective in identifying cell population expressing different combinations of EpCAM and cytokeratin 7/8 with prognostic significance for breast cancer metastases. Surprisingly, we found CD24+ circulating cells (CCs) in peripheral blood of breast cancer patients which have no epithelial markers (EpCAM and cytokeratin 7/8) but was strongly associated with distant metastasis. Namely, the count of CD45-EpCAM-CK7/8-CD24+ N-cadherin-CCs was elevated in both groups of patients, those with existing metastasis and those who developed metastases during the follow-up period. Simultaneously, an elevation in these cell counts beyond the established threshold of 218.3 cells per 1 mL of blood in patients prior to any treatment predicted a 12-fold risk of metastases, along with a threefold decrease in distant metastasis-free survival over a 90-month follow-up period. The origin of CD45-EpCAM-CK7/8-CD24+ N-cadherin-CCs remains unclear. In our opinion their existence can be explained by two most probable hypotheses. These cells could exhibit a terminal EMT phenotype, or it might be immature cells originating from the bone marrow. Nonetheless, if this hypothesis holds true, it's worth noting that the mentioned CCs do not align with any of the recognized stages of monocyte or neutrophil maturation, primarily due to the presence of CD45 expression in the myeloid cells. The results suggest the presence in the peripheral blood of patients with metastasis (both during the follow-up period and prior to inclusion in the study) of a cell population with a currently unspecified origin, possibly arising from both myeloid and tumor sources, as confirmed by the presence of aneuploidy.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms , CD24 Antigen , Epithelial Cell Adhesion Molecule , Neoplastic Cells, Circulating , Humans , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Epithelial Cell Adhesion Molecule/metabolism , CD24 Antigen/metabolism , Female , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/blood , Breast Neoplasms/mortality , Prognosis , Middle Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Aged , Adult , Epithelial-Mesenchymal Transition , Keratin-7/metabolism , Keratin-8/metabolism
9.
Anal Chem ; 96(19): 7747-7755, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38691774

ABSTRACT

Accurate classification of tumor cells is of importance for cancer diagnosis and further therapy. In this study, we develop multimolecular marker-activated transmembrane DNA computing systems (MTD). Employing the cell membrane as a native gate, the MTD system enables direct signal output following simple spatial events of "transmembrane" and "in-cell target encounter", bypassing the need of multistep signal conversion. The MTD system comprises two intelligent nanorobots capable of independently sensing three molecular markers (MUC1, EpCAM, and miR-21), resulting in comprehensive analysis. Our AND-AND logic-gated system (MTDAND-AND) demonstrates exceptional specificity, allowing targeted release of drug-DNA specifically in MCF-7 cells. Furthermore, the transformed OR-AND logic-gated system (MTDOR-AND) exhibits broader adaptability, facilitating the release of drug-DNA in three positive cancer cell lines (MCF-7, HeLa, and HepG2). Importantly, MTDAND-AND and MTDOR-AND, while possessing distinct personalized therapeutic potential, share the ability of outputting three imaging signals without any intermediate conversion steps. This feature ensures precise classification cross diverse cells (MCF-7, HeLa, HepG2, and MCF-10A), even in mixed populations. This study provides a straightforward yet effective solution to augment the versatility and precision of DNA computing systems, advancing their potential applications in biomedical diagnostic and therapeutic research.


Subject(s)
DNA , Epithelial Cell Adhesion Molecule , MicroRNAs , Humans , Epithelial Cell Adhesion Molecule/metabolism , DNA/chemistry , MicroRNAs/analysis , MicroRNAs/metabolism , Mucin-1/metabolism , Mucin-1/analysis , Computers, Molecular , MCF-7 Cells , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis , Cell Membrane/metabolism , Cell Membrane/chemistry , Hep G2 Cells
10.
Hum Cell ; 37(4): 986-996, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38615309

ABSTRACT

We aimed to elucidate the mechanism underlying carcinogenesis by comparing normal and BRCA1/2-mutated ovarian epithelial cells established via Sendai virus-based immortalization. Ovarian epithelial cells (normal epithelium: Ovn; with germline BRCA1 mutation: OvBRCA1; with germline BRCA2 mutation: OvBRCA2) were infected with Sendai virus vectors carrying three immortalization genes (Bmi-1, hTERT, and SV40T). The immunoreactivity to anti-epithelial cellular adhesion molecule (EpCAM) antibodies in each cell line and cells after 25 passages was confirmed using flow cytometry. Chromosomes were identified and karyotyped to detect numerical and structural abnormalities. Total RNA extracted from the cells was subjected to human transcriptome sequencing. Highly expressed genes in each cell line were confirmed using real-time polymerase chain reaction. Immortalization techniques allowed 25 or more passages of Ovn, OvBRCA1, and OvBRCA2 cells. No anti-EpCAM antibody reactions were observed in primary cultures or after long-term passages of each cell line. Structural abnormalities in the chromosomes were observed in each cell line; however, the abnormal chromosomes were successfully separated from the normal structures via cloning. Only normal cells from each cell line were cloned. MMP1, CCL2, and PAPPA were more predominantly expressed in OvBRCA1 and OvBRCA2 cells than in Ovn cells. Immortalized ovarian cells derived from patients with germline BRCA1 or BRCA2 mutations showed substantially higher MMP1 expression than normal ovarian cells. However, the findings need to be validated in the future.


Subject(s)
BRCA1 Protein , BRCA2 Protein , Epithelial Cells , Ovary , Humans , Female , Epithelial Cells/metabolism , Ovary/cytology , Ovary/metabolism , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Gene Expression/genetics , Mutation/genetics , Cell Line, Transformed , Germ-Line Mutation/genetics , Telomerase/genetics , Genes, BRCA1 , Carcinogenesis/genetics
11.
Pathol Res Pract ; 257: 155317, 2024 May.
Article in English | MEDLINE | ID: mdl-38657558

ABSTRACT

Lung cancer (LC) remains a leading cause of cancer-related mortality worldwide, necessitating the exploration of innovative therapeutic strategies. This study delves into the in vitro potential of liposomal therapeutics utilizing Curcumin-loaded PlexoZome® (CUR-PLXZ) in targeting EpCAM/TROP1 and Estrogen Receptor Alpha (ERα) signalling pathways for LC management. The prevalence of LC, particularly non-small cell lung cancer (NSCLC), underscores the urgent need for effective treatments. Biomarkers like EpCAM/TROP1 and ERα/NR3A1 play crucial roles in guiding targeted therapies and influencing prognosis. EpCAM plays a key role in cell-cell adhesion and signalling along with ERα which is a nuclear receptor that binds estrogen and regulates gene expression in response to hormonal signals. In LC, both often get overexpressed and are associated with tumour progression, metastasis, and poor prognosis. Curcumin, a phytochemical with diverse therapeutic properties, holds promise in targeting these pathways. However, its limited solubility and bioavailability necessitate advanced formulations like CUR-PLXZ. Our study investigates the biological significance of these biomarkers in the A549 cell line and explores the therapeutic potential of CUR-PLXZ, which modulates the expression of these two markers. An in vitro analysis of the A549 human lung adenocarcinoma cell line identified that CUR-PLXZ at a dose of 5 µM effectively inhibited the expression of EpCAM and ERα. This finding paves the way for targeted intervention strategies in LC management.


Subject(s)
Curcumin , Epithelial Cell Adhesion Molecule , Estrogen Receptor alpha , Liposomes , Lung Neoplasms , Humans , Epithelial Cell Adhesion Molecule/metabolism , Curcumin/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , A549 Cells , Antineoplastic Agents/pharmacology
12.
J Colloid Interface Sci ; 668: 335-342, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38678888

ABSTRACT

Membrane receptors perform a diverse range of cellular functions, accounting for more than half of all drug targets. The mechanical microenvironment regulates cell behaviors and phenotype. However, conventional analysis methods of membrane receptors often ignore the effects of the extracellular matrix stiffness, failing to reveal the heterogeneity of cell membrane receptors expression. Herein, we developed an in-situ surface-enhanced Raman scattering (SERS) imaging method to visualize single-cell membrane receptors on substrates with different stiffness. Two SERS substrates, Au@4-mercaptobenzonitrile@Ag@Sgc8c and Au@4-pethynylaniline@Ag@SYL3c, were employed to specifically target protein tyrosine kinase-7 (PTK7) and epithelial cell adhesion molecule (EpCAM), respectively. The polyacrylamide (PA) gels with tunable stiffness (2.5-25 kPa) were constructed to mimic extracellular matrix. The simultaneous SERS imaging of dual membrane receptors on single cancer cells on substrates with different stiffness was achieved. Our findings reveal decreased expression of PTK7 and EpCAM on cells cultured on stiffer substrates and higher migration ability of the cells. The results elucidate the heterogeneity of membrane receptors expression of cells cultured on the substrates with different stiffness. This single-cell analysis method offers an in-situ platform for investigating the impacts of extracellular matrix stiffness on the expression of membrane receptors, providing insights into the role of cell membrane receptors in cancer metastasis.


Subject(s)
Epithelial Cell Adhesion Molecule , Extracellular Matrix , Single-Cell Analysis , Spectrum Analysis, Raman , Extracellular Matrix/metabolism , Humans , Epithelial Cell Adhesion Molecule/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Gold/chemistry , Acrylic Resins/chemistry , Silver/chemistry , Surface Properties , Cell Line, Tumor , Aniline Compounds/chemistry , Particle Size , Cell Adhesion Molecules
13.
Cancer Sci ; 115(5): 1646-1655, 2024 May.
Article in English | MEDLINE | ID: mdl-38433331

ABSTRACT

The clinical features of sporadic mismatch repair deficiency (MMRd) and Lynch syndrome (LS) in Japanese patients with endometrial cancer (EC) were examined by evaluating the prevalence and prognostic factors of LS and sporadic MMRd in patients with EC. Targeted sequencing of five LS susceptibility genes (MLH1, MSH2, MSH6, PMS2, and EPCAM) was carried out in 443 patients with EC who were pathologically diagnosed with EC at the National Cancer Center Hospital between 2011 and 2018. Pathogenic variants in these genes were detected in 16 patients (3.7%). Immunohistochemistry for MMR proteins was undertaken in 337 of the 433 (77.9%) EC patients, and 91 patients (27.0%) showed absent expression of at least one MMR protein. The 13 cases of LS with MMR protein loss (93.8%) showed a favorable prognosis with a 5-year overall survival (OS) rate of 100%, although there was no statistically significant difference between this group and the sporadic MMRd group (p = 0.27). In the MMRd without LS group, the 5-year OS rate was significantly worse in seven patients with an aberrant p53 expression pattern than in those with p53 WT (53.6% vs. 93.9%, log-rank test; p = 0.0016). These results suggest that p53 abnormalities and pathogenic germline variants in MMR genes could be potential biomarkers for the molecular classification of EC with MMRd.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , DNA Mismatch Repair , Endometrial Neoplasms , Tumor Suppressor Protein p53 , Uterine Neoplasms , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Mismatch Repair/genetics , DNA-Binding Proteins/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Japan , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , Prognosis , Tumor Suppressor Protein p53/genetics , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
14.
ACS Sens ; 9(4): 2043-2049, 2024 04 26.
Article in English | MEDLINE | ID: mdl-38520356

ABSTRACT

Extracellular vesicles, especially exosomes, have attracted attention in the last few decades as novel cancer biomarkers. Exosomal membrane proteins provide easy-to-reach targets and can be utilized as information sources of their parent cells. In this study, a MagLev-based, highly sensitive, and versatile biosensor platform for detecting minor differences in the density of suspended objects is proposed for exosome detection. The developed platform utilizes antibody-functionalized microspheres to capture exosomal membrane proteins (ExoMPs) EpCAM, CD81, and CD151 as markers for cancerous exosomes, exosomes, and non-small cell lung cancer (NSCLC)-derived exosomes, respectively. Initially, the platform was utilized for protein detection and quantification by targeting solubilized ExoMPs, and a dynamic range of 1-100 nM, with LoD values of 1.324, 0.638, and 0.722 nM for EpCAM, CD81, and CD151, were observed, respectively. Then, the sensor platform was tested using exosome isolates derived from NSCLC cell line A549 and MRC5 healthy lung fibroblast cell line. It was shown that the sensor platform is able to detect and differentiate exosomal biomarkers derived from cancerous and non-cancerous cell lines. Overall, this innovative, simple, and rapid method shows great potential for the early diagnosis of lung cancer through exosomal biomarker detection.


Subject(s)
Epithelial Cell Adhesion Molecule , Exosomes , Lung Neoplasms , Exosomes/chemistry , Humans , Lung Neoplasms/pathology , Epithelial Cell Adhesion Molecule/metabolism , Tetraspanin 28/metabolism , Tetraspanin 28/analysis , Biosensing Techniques/methods , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/diagnosis , Biomarkers, Tumor/analysis , Tetraspanin 24 , A549 Cells
15.
J Cell Biochem ; 125(4): e30541, 2024 04.
Article in English | MEDLINE | ID: mdl-38372186

ABSTRACT

Epithelial cells (ECs) have been proposed to contribute to myofibroblasts or fibroblasts through epithelial-mesenchymal transition (EMT) during renal fibrosis. However, since EMT may occur dynamically, transiently, and reversibly during kidney fibrosis, conventional lineage tracing based on Cre-loxP recombination in renal ECs could hardly capture the transient EMT activity, yielding inconsistent results. Moreover, previous EMT research has primarily focused on renal proximal tubule ECs, with few reports of distal tubules and collecting ducts. Here, we generated dual recombinases-mediated genetic lineage tracing systems for continuous monitoring of transient mesenchymal gene expression in E-cadherin+ and EpCAM+ ECs of distal tubules and collecting ducts during renal fibrosis. Activation of key EMT-inducing transcription factor (EMT-TF) Zeb1 and mesenchymal markers αSMA, vimentin, and N-cadherin, were investigated following unilateral ureteral obstruction (UUO). Our data revealed that E-cadherin+ and EpCAM+ ECs did not transdifferentiate into myofibroblasts, nor transiently expressed these mesenchymal genes during renal fibrosis. In contrast, in vitro a large amount of cultured renal ECs upregulated mesenchymal genes in response to TGF-ß, a major inducer of EMT.


Subject(s)
Epithelial-Mesenchymal Transition , Kidney Diseases , Humans , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Fibrosis , Kidney Diseases/metabolism , Epithelial Cells/metabolism , Cadherins/genetics , Cadherins/metabolism , Transforming Growth Factor beta1/metabolism
16.
Cancer Cytopathol ; 132(5): 297-308, 2024 May.
Article in English | MEDLINE | ID: mdl-38373107

ABSTRACT

BACKGROUND: Circulating tumor cells (CTCs) shed into blood provide prognostic and/or predictive information. Previously, the authors established an assay to detect carcinoma cells from pleural fluid, termed effusion tumor cells (ETCs), by employing an immunofluorescence-based CTC-identification platform (RareCyte) on air-dried unstained ThinPrep (TP) slides. To facilitate clinical integration, they evaluated different slide processing and storage conditions, hypothesizing that alternative comparable conditions for ETC detection exist. METHODS: The authors enumerated ETCs on RareCyte, using morphology and mean fluorescence intensity (MFI) cutoffs of >100 arbitrary units (a.u.) for epithelial cellular adhesion molecule (EpCAM) and <100 a.u. for CD45. They analyzed malignant pleural fluid from three patients under seven processing and/or staining conditions, three patients after short-term storage under three conditions, and seven samples following long-term storage at -80°C. MFI values of 4',6-diamidino-2-phenylindol, cytokeratin, CD45, and EpCAM were compared. RESULTS: ETCs were detected in all conditions. Among the different processing conditions tested, the ethanol-fixed, unstained TP was most similar to the previously established air-dried, unstained TP protocol. All smears and Pap-stained TPs had significantly different marker MFIs from the established condition. After short-term storage, the established condition showed comparable results, but ethanol-fixed and Pap-stained slides showed significant differences. ETCs were detectable after long-term storage at -80°C in comparable numbers to freshly prepared slides, but most marker MFIs were significantly different. CONCLUSIONS: It is possible to detect ETCs under different processing and storage conditions, lending promise to the application of this method in broader settings. Because of decreased immunofluorescence-signature distinctions between cells, morphology may need to play a larger role.


Subject(s)
Epithelial Cell Adhesion Molecule , Neoplastic Cells, Circulating , Pleural Effusion, Malignant , Humans , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism , Pleural Effusion, Malignant/pathology , Pleural Effusion, Malignant/diagnosis , Epithelial Cell Adhesion Molecule/metabolism , Specimen Handling/methods , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis , Leukocyte Common Antigens/metabolism , Leukocyte Common Antigens/analysis , Fluorescent Antibody Technique/methods
17.
Life Sci Alliance ; 7(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38171596

ABSTRACT

The mouse cortical collecting duct cell line presents a tight epithelium with regulated ion and water transport. The epithelial sodium channel (ENaC) is localized in the apical membrane and constitutes the rate-limiting step for sodium entry, thereby enabling transepithelial transport of sodium ions. The membrane-bound serine protease Tmprss2 is co-expressed with the alpha subunit of ENaC. αENaC gene expression followed the Tmprss2 expression, and the absence of Tmprss2 resulted not only in down-regulation of αENaC gene and protein expression but also in abolished transepithelial sodium transport. In addition, RNA-sequencing analyses unveiled drastic down-regulation of the membrane-bound protease CAP3/St14, the epithelial adhesion molecule EpCAM, and the tight junction proteins claudin-7 and claudin-3 as also confirmed by immunohistochemistry. In summary, our data clearly demonstrate a dual role of Tmprss2 in maintaining not only ENaC-mediated transepithelial but also EpCAM/claudin-7-mediated paracellular barrier; the tight epithelium of the mouse renal mCCD cells becomes leaky. Our working model proposes that Tmprss2 acts via CAP3/St14 on EpCAM/claudin-7 tight junction complexes and through regulating transcription of αENaC on ENaC-mediated sodium transport.


Subject(s)
Claudins , Sodium , Animals , Mice , Biological Transport/physiology , Claudins/genetics , Claudins/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Ion Transport , Sodium/metabolism
18.
Biochem Biophys Res Commun ; 696: 149512, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38224664

ABSTRACT

Epithelial cell adhesion molecules (EpCAMs) have been identified as surface markers of proliferating ductal cells, which are referred to as liver progenitor cells (LPCs), during liver regeneration and correspond to malignancies. These cells can differentiate into hepatocytes and biliary epithelial cells (BECs) in vitro. EpCAM-positive LPCs are involved in liver regeneration following severe liver injury; however, the in vivo function of EpCAMs in the regenerating liver remains unclear. In the present study, we used a zebrafish model of LPC-driven liver regeneration to elucidate the function of EpCAMs in the regenerating liver in vivo. Proliferating ductal cells were observed after severe hepatocyte loss in the zebrafish model. Analyses of the liver size as well as hepatocyte and BEC markers revealed successful conversion of LPCs to hepatocytes and BECs in epcam mutants. Notably, epcam mutants exhibited severe defects in intrahepatic duct maturation and bile acid secretion in regenerating hepatocytes, suggesting that epcam plays a critical role in intrahepatic duct reconstruction during LPC-driven liver regeneration. Our findings provide insights into human diseases involving non-parenchymal cells, such as primary biliary cholangitis, by highlighting the regulatory effect of epcam on intrahepatic duct reconstruction.


Subject(s)
Cholangitis , Zebrafish , Animals , Humans , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Liver/metabolism , Bile Ducts, Intrahepatic/metabolism , Hepatocytes/metabolism , Epithelial Cells/metabolism , Cholangitis/pathology , Liver Regeneration
19.
Microb Pathog ; 188: 106549, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38281605

ABSTRACT

The five epidermal growth factor-like domains (EGF) of Eimeria tenella microneme protein 8 (EtMIC8) (EtMIC8-EGF) plays a vital role in host cell attachment and invasion. These processes require interactions between parasite proteins and receptors on the surface of host cells. In this study, five chicken membrane proteins potentially interacting with EtMIC8-EGF were identified using the GST pull-down assay and mass spectrometry analysis, and only chicken (Gallus gallus) epithelial cell adhesion molecule (EPCAM) could bind to EtMIC8-EGF. EPCAM-specific antibody and recombinant EPCAM protein (rEPCAM) inhibited the EtMIC8-EGF binding to host cells in a concentration-dependent manner. Furthermore, the rEPCAM protein showed a binding activity to sporozoites in vitro, and a significant reduction of E. tenella invasion in DF-1 cells was further observed after pre-incubation of sporozoites with rEPCAM. The specific anti-EPCAM antibody further significantly decreased weight loss, lesion score and oocyst output during E. tenella infection, displaying partial inhibition of E. tenella infection. These results indicate that chicken EPCAM is an important EtMIC8-interacting host protein involved in E. tenella-host cell adhesion and invasion. The findings will contribute to a better understanding of the role of adhesion-associated microneme proteins in E. tenella.


Subject(s)
Coccidiosis , Eimeria tenella , Poultry Diseases , Animals , Eimeria tenella/chemistry , Eimeria tenella/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Chickens , Protozoan Proteins , Epidermal Growth Factor/metabolism , Recombinant Proteins , Sporozoites/metabolism , Coccidiosis/veterinary , Coccidiosis/parasitology , Poultry Diseases/parasitology
20.
Clin Chim Acta ; 552: 117651, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37980974

ABSTRACT

PURPOSE: Despite its limitations, the cytology of body fluids is widely used in diagnosing neoplastic cells. Flow cytometry detects and identifies individual cells, enabling the detection of circulating tumor cells and facilitating diagnosis. This study compared the diagnostic utility of flow cytometry and cytology for detecting cancer cells in peritoneal and pleural fluids. METHODOLOGY: We used flow cytometry and cytology to examine 119 pleural and peritoneal effusions received for routine screening. Antibodies against clusters of differentiation 45 (CD45), 14 (CD14), and Epithelial cell adhesion molecule (EpCAM) were used to detect malignant cells. Based on combined clinical and diagnostic information, 37 fluid specimens were malignant, and 77 were benign. RESULTS: Flow cytometry correctly identified 34 cancers, while cytology identified 26 cancers (sensitivity 91.89 % vs. 70.27, respectively). Both methods had equal specificity (98.7 %). At a cut-off of > 0.29 % EpCAM(+) cells to all cells in the samples, flow cytometry accurately detected cancer cells, achieving 89.2 % sensitivity, 90.9 % specificity, and an AUC of 0.959 (p < 0.001). CONCLUSION: Flow cytometry improves the detection of epithelial cancer cells in peritoneal and pleural fluids compared to conventional cytology. Due to similar specificity and higher sensitivity, flow cytometry offers a promising alternative to cytology for patient screening.


Subject(s)
Neoplastic Cells, Circulating , Pleural Effusion, Malignant , Humans , Epithelial Cell Adhesion Molecule/metabolism , Neoplastic Cells, Circulating/pathology , Flow Cytometry/methods , Ascitic Fluid , Pleural Effusion, Malignant/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...