Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cancer Cell ; 41(9): 1650-1661.e4, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37652006

ABSTRACT

Deep learning (DL) can accelerate the prediction of prognostic biomarkers from routine pathology slides in colorectal cancer (CRC). However, current approaches rely on convolutional neural networks (CNNs) and have mostly been validated on small patient cohorts. Here, we develop a new transformer-based pipeline for end-to-end biomarker prediction from pathology slides by combining a pre-trained transformer encoder with a transformer network for patch aggregation. Our transformer-based approach substantially improves the performance, generalizability, data efficiency, and interpretability as compared with current state-of-the-art algorithms. After training and evaluating on a large multicenter cohort of over 13,000 patients from 16 colorectal cancer cohorts, we achieve a sensitivity of 0.99 with a negative predictive value of over 0.99 for prediction of microsatellite instability (MSI) on surgical resection specimens. We demonstrate that resection specimen-only training reaches clinical-grade performance on endoscopic biopsy tissue, solving a long-standing diagnostic problem.


Subject(s)
Algorithms , Colorectal Neoplasms , Humans , Biomarkers , Biopsy , Microsatellite Instability , Colorectal Neoplasms/genetics
2.
Molecules ; 27(6)2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35335261

ABSTRACT

Here, a series of half-sandwich arene Ru(II) complexes with difluorinated ligands [Ru(η6-arene)(L)Cl] (L1 = 2-(2,3-difluorophenyl)imidazole[4,5f][1,10]-phenanthroline; L2 = 2-(2,4-difluorophenyl)imidazole[4,5f][1,10]-phenanthroline; arene = benzene, toluene, and p-cymene) were synthesized and characterized. Molecular docking analysis showed that these complexes bind to c-myc G-quadruplex DNA through either groove binding or π-π stacking, and the relative difluorinated site in the main ligand plays a role in regulating the binding mode. The binding behavior of these complexes with c-myc G-quadruplex DNA was evaluated using ultraviolet-visible spectroscopy, fluorescence intercalator displacement assay, fluorescence resonance energy transfer melting assay, and polymerase chain reaction. The comprehensive analysis indicated that complex 1 exhibited a better affinity and stability in relation to c-myc G-quadruplex DNA with a DC50 of 6.6 µM and ΔTm values of 13.09 °C, than other molecules. Further activity evaluation results displayed that this class of complexes can also inhibit the growth of various tumor cells, especially complexes 3 and 6, which exhibited a better inhibitory effect against human U87 glioblastoma cells (51.61 and 23.75 µM) than other complexes, even superior to cisplatin (32.59 µM). Owing to a befitting lipophilicity associated with the high intake of drugs by tumor cells, complexes 3 and 6 had favorable lipid-water partition coefficients of -0.6615 and -0.8077, respectively. Moreover, it was found that complex 6 suppressed the proliferation of U87 cells mainly through an induced obvious S phase arrest and slight apoptosis, which may have resulted from the stabilization of c-myc G-quadruplex DNA to block the transcription and expression of c-myc. In brief, these types of arene Ru(II) complexes with difluorinated ligands can be developed as potential inducers of S-phase arrest and apoptosis through the binding and stabilization of c-myc G-quadruplex DNA, and could be used in clinical applications in the future.


Subject(s)
G-Quadruplexes , Ruthenium , DNA/chemistry , Humans , Ligands , Molecular Docking Simulation , Ruthenium/chemistry , Ruthenium/pharmacology
4.
Stem Cell Reports ; 16(6): 1468-1477, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33961792

ABSTRACT

Induced pluripotent stem cells (iPSCs) and cancer cells share cellular similarities and transcriptomic profiles. Here, we show that an iPSC-based cancer vaccine, comprised of autologous iPSCs and CpG, stimulated cytotoxic antitumor CD8+ T cell effector and memory responses, induced cancer-specific humoral immune responses, reduced immunosuppressive CD4+ T regulatory cells, and prevented tumor formation in 75% of pancreatic ductal adenocarcinoma (PDAC) mice. We demonstrate that shared gene expression profiles of "iPSC-cancer signature genes" and others are overexpressed in mouse and human iPSC lines, PDAC cells, and multiple human solid tumor types compared with normal tissues. These results support further studies of iPSC vaccination in PDAC in preclinical and clinical models and in other cancer types that have low mutational burdens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Carcinoma, Pancreatic Ductal/immunology , Induced Pluripotent Stem Cells/immunology , Pancreatic Neoplasms/immunology , T-Lymphocytopenia, Idiopathic CD4-Positive/immunology , Animals , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/metabolism , Cancer Vaccines/therapeutic use , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunologic Memory , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/therapy , T-Lymphocytopenia, Idiopathic CD4-Positive/metabolism , Transcriptome , Xenograft Model Antitumor Assays
5.
Article in English | MEDLINE | ID: mdl-32843430

ABSTRACT

Although cutaneous squamous cell carcinoma (cSCC) is treatable in the majority of cases, deadly invasive and metastatic cases do occur. To date there are neither reliable predictive biomarkers of disease progression nor FDA-approved targeted therapies as standard of care. To address these issues, we screened patient-derived primary cultured cells from invasive/metastatic cSCC with 107 small-molecule inhibitors. In-house bioinformatics tools were used to cross-analyze drug responses and DNA mutations in tumors detected by whole-exome sequencing (WES). Aberrations in molecular pathways with evidence of potential drug targets were identified, including the Eph-ephrin and neutrophil degranulation signaling pathways. Using a screening panel of siRNAs, we identified EPHA6 and EPHA7 as targets within the Eph-ephrin pathway responsible for mitigating decreased cell viability. These studies form a plausible foundation for detecting biomarkers of high-risk progressive disease applicable in dermatopathology and for patient-specific therapeutic options for invasive/metastatic cSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Cell Survival/genetics , Disease Progression , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Genomics/methods , Humans , Male , Middle Aged , Mutation/genetics , Receptor, EphA6/antagonists & inhibitors , Receptor, EphA6/metabolism , Receptor, EphA7/antagonists & inhibitors , Receptor, EphA7/metabolism , Signal Transduction/genetics , Skin Neoplasms/genetics , Small Molecule Libraries/pharmacology , Exome Sequencing/methods
6.
Immunobiology ; 225(4): 151982, 2020 07.
Article in English | MEDLINE | ID: mdl-32747027

ABSTRACT

Neural cell adhesion molecule 1 (NCAM1/CD56) is expressed on immune cells, myoblasts, and malignant cells, and there is a growing demand for the genetic detection of CD56 and CD56-targeted therapy. In the present study, we developed a novel peptide ligand (designated Natein) that binds to human CD56 by using T7 phage display technology. Natein recognized the extracellular region of CD56 and could bind to natural killer (NK) cells and CD56-positive (CD56+) cancer cells. CD56+ cells enriched from human peripheral blood mononuclear cells (PBMCs) using biotinylated Natein-conjugated microbeads, similarly to CD56 antibody-isolated cells, demonstrated functional cytotoxicity against K562 cells. In addition, Natein could be used to stain CD56+ lymphoma cells in nasal-type extranodal NK/T-cell lymphoma tissues similarly to a CD56 antibody. These findings suggest that Natein has the potential to be alternative to CD56 antibody that could be used for peptide-based cell isolation and diagnosis.


Subject(s)
CD56 Antigen/antagonists & inhibitors , Drug Discovery , Peptides/pharmacology , Cell Surface Display Techniques , Drug Discovery/methods , Humans , Immunoconjugates/pharmacology , K562 Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Peptide Library , Peptides/chemistry , Protein Binding
7.
Front Immunol ; 10: 1510, 2019.
Article in English | MEDLINE | ID: mdl-31338094

ABSTRACT

Over a century ago, it was reported that immunization with embryonic/fetal tissue could lead to the rejection of transplanted tumors in animals. Subsequent studies demonstrated that vaccination of embryonic materials in animals induced cellular and humoral immunity against transplantable tumors and carcinogen-induced tumors. Therefore, it has been hypothesized that the shared antigens between tumors and embryonic/fetal tissues (oncofetal antigens) are the key to anti-tumor immune responses in these studies. However, early oncofetal antigen-based cancer vaccines usually utilize xenogeneic or allogeneic embryonic stem cells or tissues, making it difficult to tease apart the anti-tumor immunity elicited by the oncofetal antigens vs. graft-vs.-host responses. Recently, one oncofetal antigen-based cancer vaccine using autologous induced pluripotent stem cells (iPSCs) demonstrated marked prophylactic and therapeutic potential, suggesting critical roles of oncofetal antigens in inducing anti-tumor immunity. In this review, we present an overview of recent studies in the field of oncofetal antigen-based cancer vaccines, including single peptide-based cancer vaccines, embryonic stem cell (ESC)- and iPSC-based whole-cell vaccines, and provide insights on future directions.


Subject(s)
Cancer Vaccines/immunology , Induced Pluripotent Stem Cells/immunology , Animals , Antigens, Neoplasm/immunology , Embryonic Stem Cells/immunology , Humans , Vaccination/methods
8.
Cancer Biol Ther ; 19(10): 921-933, 2018.
Article in English | MEDLINE | ID: mdl-29856687

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) currently only has one FDA-approved cancer intrinsic targeted therapy, the epidermal growth factor receptor (EGFR) inhibitor cetuximab, to which only approximately 10% of tumors are sensitive. In order to extend therapy options, we subjected patient-derived HNSCC cells to small-molecule inhibitor and siRNA screens, first, to find effective combination therapies with an EGFR inhibitor, and second, to determine a potential mechanistic basis for repurposing the FDA approved agents for HNSCC. The combinations of EGFR inhibitor with anaplastic lymphoma kinase (ALK) inhibitors demonstrated synergy at the highest ratio in our cohort, 4/8 HNSCC patients' derived tumor cells, and this corresponded with an effectiveness of siRNA targeting ALK combined with the EGFR inhibitor gefitinib. Co-targeting EGFR and ALK decreased HNSCC cell number and colony formation ability and increased annexin V staining. Because ALK expression is low and ALK fusions are infrequent in HNSCC, we hypothesized that gefitinib treatment could induce ALK expression. We show that ALK expression was induced in HNSCC patient-derived cells both in 2D and 3D patient-derived cell culture models, and in patient-derived xenografts in mice. Four different ALK inhibitors, including two (ceritinib and brigatinib) FDA approved for lung cancer, were effective in combination with gefitinib. Together, we identified induction of ALK by EGFR inhibitor as a novel mechanism potentially relevant to resistance to EGFR inhibitor, a high ratio of response of HNSCC patient-derived tumor cells to a combination of ALK and EGFR inhibitors, and applicability of repurposing ALK inhibitors to HNSCC that lack ALK aberrations.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Squamous Cell Carcinoma of Head and Neck/genetics , Anaplastic Lymphoma Kinase/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Xenograft Model Antitumor Assays
9.
Oncol Lett ; 15(6): 8825-8832, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29805622

ABSTRACT

The aim of the present study was to investigate magnetic resonance (MR) features of meningeal solitary fibrous tumors (SFTs) in order to improve their recognition. The study retrospectively analyzed MR manifestations in 12 cases of meningeal SFTs confirmed by surgery. The lesions were analyzed in terms of the site of their growth, growth mode, morphology, size, changes in the MR signal and the edge of the focus. The tumors were malignant in 3 cases, benign in 7 cases and borderline in 2 cases. A total of 3 cases spanned the tentorium cerebelli, 3 spanned the falx and another case was close to the falx. Overall, 4 superficial tumors were accompanied by adjacent bony destruction. One case was located at the triangular area created by the right lateral ventricles. The tumors were all lobular, with a large diameter and more cystic areas in the center of the focus. The signals were often heterogeneous. The parenchyma in the tumors was significantly enhanced. For certain tumors, the signal was usually inhomogeneous and not uniform. The MR features of a SFT were often similar to other intracranial tumors and easily misdiagnosed. There were certain innate characteristics, for example, the tumor was often a solitary large soft-tissue mass with an irregular edge and a clear boundary, with a lobulated contour, and was widely involved with other tissues. The tumors often grew across either the falx or tentorium cerebelli. A significantly inhomogeneous signal was found and either a low or low-high mixed signal on T2-weighted images in particular. These features are useful for the differential diagnosis of SFTs and other tumors.

10.
Oncol Lett ; 13(4): 2274-2280, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28454391

ABSTRACT

Kin17 DNA and RNA binding protein (Kin17) is a highly conserved protein that participates in DNA replication, DNA repair and cell cycle progression. Recently, the tumor-promoting function of Kin17 has been demonstrated and increasingly studied. In the present study, the role of Kin17 in the invasion and metastasis of non-small cells lung cancer (NSCLC) was investigated. Elevated Kin17 mRNA and protein expression was identified in a total of 97 NSCLC and benign lung lesion tissue specimens. Kin17 overexpression was significantly correlated with high tumor grade and lymph node metastasis, indicating poor patient prognosis. Scratch and Transwell assays demonstrated that the knockdown of KIN17 inhibited the ability of NSCLC cells to migrate and invade. Furthermore, reverse transcription-quantitative polymerase chain reaction and western blot analyses confirmed that knockdown of KIN17 decreased the expression of matrix metalloproteinase 7, epidermal growth factor receptor and v-myc avian myelocytomatosis viral oncogene homolog. The results of the present study indicate that Kin17 is markedly overexpressed in NSCLC tissues compared with benign lung lesion and peritumoral tissue. The upregulation of KIN17 may serve an important role in the metastasis of NSCLC cells. These results indicate that Kin17 is a novel diagnostic and prognostic biomarker of NSCLC, in addition to being a potential therapeutic target for the treatment of patients with NSCLC.

11.
Neurol Res ; 39(5): 448-452, 2017 May.
Article in English | MEDLINE | ID: mdl-28262035

ABSTRACT

Posterior limb of the internal capsule lesions (PLICL) are one of the MRI features of neuromyelitis optica spectrum disorder (NMOSD). However, there is no evidence that such lesions are pathogenically related to NMOSD. We retrospectively analyzed features of PLICL in NMOSD, and other central nervous system inflammatory disorders, in 561 patients. We also examined the pathological samples of six patients. Of the 561 patients investigated, PLICL were found in 65 patients (11.6%). Lesions were bilateral in 26 cases (40%) and unilateral in 39 cases (60%). Unilateral lesions were mainly located on the left side (74.3%, 29/39). Of the 65 patients with PLICL, 46 patients had NMOSD (70.8%) and were positive for anti-aquaporin (AQP4-IgG), four had NMOSD (6.2%) and were AQP4-IgG negative, 10 patients had multiple sclerosis (MS), three patients had NMDAR encephalitis, and two had autoimmune meningoencephalitis. Of the six patients whose pathological samples were evaluated, all had PLICL and were negative for AQP4-IgG, and none had pathological NMOSD lesion features. These cases included three patients with multiple sclerosis, one with anti-N-methyl-D-aspartate receptor encephalitis, and two with autoimmune meningoencephalitis. In conclusion, PLICL are found not only in patients with NMOSD, but also in MS and other disorders.


Subject(s)
Internal Capsule/pathology , Neuromyelitis Optica/pathology , Adult , Aged , Antibodies/blood , Antibodies/cerebrospinal fluid , Antigens, CD/metabolism , Aquaporin 4/immunology , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Internal Capsule/diagnostic imaging , Internal Capsule/metabolism , Magnetic Resonance Imaging , Male , Neuromyelitis Optica/blood , Neuromyelitis Optica/cerebrospinal fluid , Neuromyelitis Optica/diagnostic imaging , Receptors, N-Methyl-D-Aspartate/immunology , Retrospective Studies , Young Adult
12.
Food Chem Toxicol ; 50(8): 2648-55, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22613217

ABSTRACT

Scolopendra subspinipes mutilans L. Koch has been used for cancer treatment in traditional Chinese medicine for hundreds of years. In this study, the effects of a polysaccharide-protein complex from Scolopendra subspinipes mutilans L. Koch (SPPC) on the tumor growth and immune function were assessed in sarcoma S180 and hepatoma H22 bearing mice. Results showed that SPPC significantly inhibited the growth of S180 transplanted in mice and prolonged the survival time of H22- bearing mice. In S180-bearing mice, it promoted specific and nonspecific immune response as evidenced by enhancing the activities of natural killer (NK) cells, cytotoxic T lymphocytes (CTL) and the ratio of Th1/Th2 cytokines, and increasing the percentages of CD4(+) T cells, B cells and NK cells. Furthermore, SPPC not only significantly inhibited mRNA expression and production of the immunosuppressive cytokines (IL-10 and TGF-ß), but also diminished arachidonic acid (AA)-metabolizing enzymes (COX-2 and CYP4A) and their products (PGE(2) and 20-HETE) in tumor-associated macrophages (TAMs). Taken together, our results indicate that SPPC inhibits tumor growth in vivo by improving antitumor immune responses at least partly via downregulating AA-metabolic pathways in TAMs, and could act as an anti-tumor agent with immunomodulatory activity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Drugs, Chinese Herbal/chemistry , Neoplasms, Experimental/drug therapy , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Catalysis , Cell Line, Tumor , Cell Proliferation/drug effects , Diterpene Alkaloids , Drug Screening Assays, Antitumor , Interleukin-10/metabolism , Lymphocyte Subsets , Mice , Neoplasms, Experimental/pathology , Plant Extracts/therapeutic use , Plant Proteins/pharmacology , Polysaccharides/pharmacology , Transforming Growth Factor beta/metabolism
13.
PLoS One ; 6(9): e25343, 2011.
Article in English | MEDLINE | ID: mdl-21980430

ABSTRACT

BACKGROUND: Kin17 is ubiquitously expressed at low levels in human tissue and participates in DNA replication, DNA repair and cell cycle control. Breast cancer cells are characterized by enabling replicative immortality and accumulated DNA damage. However, whether kin17 contributes to breast carcinogenesis remains unknown. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we show for the first time that kin17 is an important molecule related to breast cancer. Our results show that kin17 expression was markedly increased in clinical breast tumors and was associated with tumor grade, Ki-67 expression, p53 mutation status and progesterone receptor expression, which were assessed in a clinicopathologic characteristics review. Knockdown of kin17 inhibited DNA replication and repair, blocked cell cycle progression and inhibited anchorage-independent growth, while increasing sensitivity to chemotherapy in breast cancer cells. Moreover, kin17 silencing decreased EGF-stimulated cell growth. Furthermore, overexpression of kin17 promoted DNA replication and cell proliferation in MCF-10A. CONCLUSIONS/SIGNIFICANCE: Our findings indicate that up-regulation of kin17 is strongly associated with cellular proliferation, DNA replication, DNA damage response and breast cancer development. The increased level of kin17 was not only a consequence of immortalization but also associated with tumorigenesis. Therefore, kin17 could be a novel therapeutic target for inhibiting cell growth in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , DNA-Binding Proteins/metabolism , RNA-Binding Proteins/metabolism , Up-Regulation , Adult , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Repair/drug effects , DNA Repair/genetics , DNA Replication/drug effects , DNA Replication/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Progression , Epidermal Growth Factor/pharmacology , Female , Gene Knockdown Techniques , Humans , RNA-Binding Proteins/genetics
14.
World J Gastroenterol ; 11(14): 2157-61, 2005 Apr 14.
Article in English | MEDLINE | ID: mdl-15810084

ABSTRACT

AIM: To study the effect of hepatitis C virus nonstructural region 3 (HCV NS3) protein on proliferation and transformation of normal human liver cell line. METHODS: QSG7701 cells were transfected with pRcHCNS3-5', pRcHCNS3-3' and pRcCMV using lipofectamine transfecting technique and selected with G418 method. Expression of HCV NS3 protein was determined by immunohistochemistry. Biologic characteristics of transfected cells were evaluated by population doubling time and soft agar assays. Activation of MAPK was analyzed using Western blot with phosphospecific monoclonal antibody against dually phosphorylated MAPK. RESULTS: QSG7701 cells transfected with pRcHCNS3-5' showed strong intracellular expression of HCVNS3 protein, and the positive signal was localized in cytoplasm. The expressing strength of HCVNS3 protein in pRcHCNS3-3'-transfected cells was weaker than that in pRcHCNS3-5'-transfected cells. The population doubling time in the transfected cells with pRcHCNS3-5' (12 h) was much shorter than those with pRcHCNS3-3', pRcCMV and normal cells (24, 26, 28 h, respectively) (P<0.01). The transfected cells with pRcHCNS3-5' showed much more anchorage independent colonies than that in those with pRcHCNS3-3' and pRcCMV (P<0.01). The cloning efficiencies of transfected cells with pRcHCNS3-5', pRcHCNS3-3', pRcCMV and controls were 33%, 1.33%, 1.46%, 1.11% respectively. The level of phosphorylated MAPK in the cells with pRcHCNS3-5' was much higher than that in those with pRcHCNS3-3'and pRcCMV and normal cells (P<0.01). CONCLUSION: The results suggest that (1) QSG7701 cells are a better human liver cell line for investigating the pathogenesis of HCV NS3 protein. (2) 5' region of the HCV genome segment encoding HCV NS3 is involved in cell growth and cell phenotype. (3) HCV NS3 N-terminal peptide may up-regulate the activation of MAPK, but not affect the expression of MAPK.


Subject(s)
Hepatocytes/virology , Mitogen-Activated Protein Kinase Kinases/metabolism , Viral Nonstructural Proteins/genetics , Cell Division/physiology , Cell Line , Cell Transformation, Viral/physiology , Hepatocytes/cytology , Hepatocytes/enzymology , Humans , MAP Kinase Signaling System/physiology , Phosphorylation , Plasmids
15.
Chin Med J (Engl) ; 115(4): 597-602, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12133307

ABSTRACT

OBJECTIVE: To study the effect of hepatitis C virus nonstructural protein NS(3) (HCV NS3) on telomerase activity and carcinogenesis. METHODS: Streptavidin-peroxidase (SP) conjugated method was used to detect the expression of HCV NS(3) protein in NIH3T3 cells transfected with plasmid pRcHCNS(3)-5' and pRcHCNS(3)-3'. Telomerase activity was detected by an in situ telomerase activity labeling method, telomeric repeat amplification protocol polymerase chain reaction (TRAP-PCR) and telomerase PCR enzyme linked immunosorbent assay (ELISA) technology in the transfected and non-transfected NIH3T3 cells. RESULTS: HCV NS(3) protein was expressed in the NIH3T3 cells transfected with plasmid pRcHCNS(3)-5' expressing HCV NS(3) C-terminal deleted protein or with plasmid pRcHCNS(3)-3' expressing HCV NS(3) N-terminal deleted protein. The positive signal of HCV NS(3) protein was localized in the cytoplasm of NIH3T3 cells, and the signal intensity of the former was stronger. Telomerase activity in NIH3T3 cells transfected with plasmid pRcHCNS(3)-5' was stronger than that in NIH3T3 cells transfected with plasmid pRcHCNS(3)-3' (P < 0.01), whereas telomerase activity in NIH3T3 cells transfected with plasmid pRcCMV or untreated NIH3T3 cells was weaker than that in NIH3T3 cells transfected with plasmid pRcHCNS(3)-3' (P < 0.05). The expression level of HCV NS(3) protein was significantly correlated with the strength of telomerase activity (P < 0.05). The results obtained by in situ telomerase activity labeling corresponded to the results by telomerase PCR ELISA technology. CONCLUSIONS: HCV NS(3) protein may activate telomerase through endogenous mechanism to induce host cell transformation. The effect of HCV NS(3) C-terminal deleted protein on telomerase activity in the host cell may be stronger than that of HCV NS(3) N-terminal deleted protein. In situ telomerase activity labeling was a reliable technology for studying pathological morphology and telomerase activity in tissues and cells.


Subject(s)
Telomerase/metabolism , Viral Nonstructural Proteins/physiology , 3T3 Cells , Animals , Enzyme-Linked Immunosorbent Assay/methods , Mice , Plasmids/genetics , Polymerase Chain Reaction/methods , Telomerase/genetics , Transfection , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...