Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cancer Immunol Immunother ; 71(4): 851-863, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34463793

ABSTRACT

Cancer immunotherapy using immune checkpoint inhibitors (ICIs) has been recognized as a novel therapeutic option for head and neck squamous cell carcinoma (HNSCC). However, only approximately 20-30% of patients with recurrent/metastatic (R/M) HNSCC benefit. Moreover, the mechanisms underlying the response to ICIs remain unclear. We investigated the proportion, activation status, and expression level of immune checkpoint molecules in circulating T cell subsets in R/M HNSCC patients treated with nivolumab using flow cytometry and mass cytometry, and then determined whether treatment response was associated with these values. We also assessed the changes in the frequency of tumor-associated antigens, MAGE-A4 and p53, -specific T cells prior to and after nivolumab treatment using the IFN-γ ELISPOT assay. The proportion of activated CD4+ and CD8+ TEMRA cells significantly increased in the disease-controlled patients but not in disease-progressed patients. As expected, the expression of PD-1 in T cells markedly decreased regardless of the therapeutic response. Meanwhile, T cell immunoglobulin mucin-3 expression on CD8+ T cells was significantly higher in patients with disease progression than in disease-controlled patients after treatment. The frequency of the tumor-associated antigens, MAGE-A4- and p53-specific T cells, was not correlated with clinical responses; however, in the disease-controlled patients, the frequency of MAGE-A4-specific T cells was significantly augmented. We concluded that in R/M HNSCC patients treated with nivolumab, circulating T cells show dynamic alterations depending on treatment efficacy. An analysis of the immunokinetics of circulating T cells could thus provide new insights into rational therapeutic strategies in cancer immunotherapy for HNSCC.


Subject(s)
Head and Neck Neoplasms , Nivolumab , Head and Neck Neoplasms/drug therapy , Humans , Neoplasm Recurrence, Local/drug therapy , Nivolumab/pharmacology , Squamous Cell Carcinoma of Head and Neck/drug therapy , T-Lymphocyte Subsets
2.
J Mater Sci Mater Med ; 32(6): 58, 2021 May 17.
Article in English | MEDLINE | ID: mdl-33999320

ABSTRACT

The evaluation of antitumor immune responses is essential for immune monitoring to predict clinical outcomes as well as treatment efficacies in cancer patients. In this study, we produced two tumor antigen (TA) proteins, melanoma antigen family A4 and wild type p53, using TG silkworm systems and evaluated anti-TA-specific immune responses by enzyme-linked immunosorbent spot assays in patients with head and neck cancer. Eleven (61.1%) of 18 patients showed significant IFN-γ production in response to at least one TA; however, the presence of TA-specific immune responses did not significantly contribute to better prognosis (overall survival, p = 0.1768; progression-free survival, p = 0.4507). Further studies will need to be performed on a larger scale to better assess the clinical significance of these systems. The production of multiple TA proteins may provide new avenues for the development of immunotherapeutic strategies to stimulate a potent and specific immune response against tumor cells as well as precise assessment of antitumor immune responses in cancer patients.


Subject(s)
Antigens, Neoplasm/chemistry , Head and Neck Neoplasms/immunology , Immune System , Immunotherapy/methods , Adult , Aged , Animals , Animals, Genetically Modified , Antigens, Neoplasm/biosynthesis , Bombyx , Female , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/therapy , Humans , Immunity , Immunohistochemistry , In Vitro Techniques , Kaplan-Meier Estimate , Leukocytes, Mononuclear/cytology , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Progression-Free Survival , Tumor Suppressor Protein p53/biosynthesis
3.
Anticancer Res ; 39(11): 5983-5990, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31704823

ABSTRACT

BACKGROUND/AIM: DJ-1, an oncogenic molecule, helps to maintain somatic stem cells by reducing the intracellular level of reactive oxygen species (ROS). This study investigated the role of DJ-1 in glioma stem cells (GSCs). MATERIALS AND METHODS: U87-MG (U87) and U251-MG (U251) glioblastoma cell lines that express wild-type and mutant p53, respectively, were used. These were cultured with DJ-1-targeting siRNA and subjected to a variety of in vitro experiments or intracranial transplantation into nude mice. RESULTS: Knockdown of DJ-1 reduced clonogenicity only in U87 cells, which was rescued by p53 depletion. ROS accumulated in DJ-1-depleted cells, although treatment with N-acetyl cysteine, which quenches ROS, did not affect exhaustion of CSCs among U87 cells by DJ-1 knockdown. In a serial transplantation study, DJ-1 knockdown prolonged the survival of mice in secondary transplantation. CONCLUSION: DJ-1 plays a pivotal role in maintenance of stem cell self-renewal in the U87 cell line.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Cell Self Renewal , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Protein Deglycase DJ-1/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Prognosis , Protein Deglycase DJ-1/antagonists & inhibitors , Protein Deglycase DJ-1/genetics , RNA, Small Interfering/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Genome Biol Evol ; 10(10): 2686-2696, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30239708

ABSTRACT

Endogenous viral sequences in eukaryotic genomes, such as those derived from plant pararetroviruses (PRVs), can serve as genomic fossils to study viral macroevolution. Many aspects of viral evolutionary rates are heterogeneous, including substitution rate differences between genes. However, the evolutionary dynamics of this viral gene rate heterogeneity (GRH) have been rarely examined. Characterizing such GRH may help to elucidate viral adaptive evolution. In this study, based on robust phylogenetic analysis, we determined an ancient endogenous PRV group in Oryza genomes in the range of being 2.41-15.00 Myr old. We subsequently used this ancient endogenous PRV group and three younger groups to estimate the GRH of PRVs. Long-term substitution rates for the most conserved gene and a divergent gene were 2.69 × 10-8 to 8.07 × 10-8 and 4.72 × 10-8 to 1.42 × 10-7 substitutions/site/year, respectively. On the basis of a direct comparison, a long-term GRH of 1.83-fold was identified between these two genes, which is unexpectedly low and lower than the short-term GRH (>3.40-fold) of PRVs calculated using published data. The lower long-term GRH of PRVs was due to the slightly faster rate decay of divergent genes than of conserved genes during evolution. To the best of our knowledge, we quantified for the first time the long-term GRH of viral genes using paleovirological analyses, and proposed that the GRH of PRVs might be heterogeneous on time scales (time-dependent GRH). Our findings provide special insights into viral gene macroevolution and should encourage a more detailed examination of the viral GRH.


Subject(s)
Biological Evolution , Oryza/virology , Tungrovirus/genetics , Genome, Plant
5.
Oncol Rep ; 23(2): 551-61, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20043121

ABSTRACT

Most tumors of patients with Lynch syndrome and a fraction of sporadic colorectal cancers (CRCs) exhibit high levels of microsatellite instability (MSI) at mono- and dinucleotide repeat loci. A different type of instability, elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) has been found in non-colonic cancers. Our previous study demonstrated that EMAST is common in sporadic CRC. Here, we focused on the relationships between EMAST and other genomic instability parameters or clinicopathological features in an unselected series of 88 sporadic CRCs. Of the tumors in the sample, 4 (4.5%) were MSI-high (MSI-H), 9 (10.2%) were MSI-low (MSI-L) and 75 (85.2%) were microsatellite stable. EMAST status was determined using 7 EMAST markers. Fifty-three (60.2%) tumors without MSI-H showed instability at >or=1 EMAST loci. All 4 MSI-H tumors showed instability at several EMAST loci. Instability profiles of MSI-H tumors at EMAST loci were more complex than those of non-MSI-H tumors. A tendency of positive association was observed between MSI-L and EMAST (P=0.023). The frequency of loss of heterozygosity (LOH) for the 14 loci in EMAST-positive tumors was significantly higher than negative tumors (P=0.048). Among the clinicopathological parameters, only tumor location at the distal colon was associated with EMAST-negative tumors (P=0.0084, one-tailed). A relatively higher frequency of well-differentiated adenocarcinomas was observed in EMAST tumors as opposed to non-EMAST tumors, though the survival rate was similar. These results suggest that overlapping mechanisms that cause MSI-L, EMAST and LOH in CRCs may exist.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Microsatellite Instability , Microsatellite Repeats/genetics , Cohort Studies , Gene Frequency , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Japan , Loss of Heterozygosity , Microsatellite Repeats/physiology
6.
Am J Nephrol ; 30(3): 295-302, 2009.
Article in English | MEDLINE | ID: mdl-19546527

ABSTRACT

BACKGROUND: Transforming growth factor beta1 (TGF-beta1) and vascular endothelial growth factor (VEGF) are involved in peritoneal deterioration in continuous ambulatory peritoneal dialysis. The present study was designed to determine whether new peritoneal dialysis solutions (PDS), pyridoxamine (advanced glycation end products (AGE) inhibitor) or AT1 receptor blocker (ARB), affect the expression of VEGF and TGF-beta1 in rat peritoneal mesothelial cells (RPMC). METHODS: RPMC were stimulated by phosphate-buffered saline (PBS) as control, Dianeal 1.5% (D 1.5%), Dianeal 2.5% (D 2.5%), Dianeal 4.25% (D 4.25%), Dianeal N 1.5% (N 1.5%), Dianeal N 2.5% (N 2.5%) or Extraneal (Ex). In co-incubation experiments, RPMC were stimulated with N 2.5% including pyridoxamine or olmesartan (ARB). VEGF and TGF-beta1 protein and mRNA expression in RPMC were analyzed by ELISA and RT-PCR. RESULTS: Glucose-containing PDS, even N 2.5% diluted twofold with M199 (which contains 1.25% glucose), increased VEGF and TGF-beta1 expression in RPMC (p < 0.05). Ex did not inhibit VEGF expression and did not inhibit TGF- beta1 expression after 24 h in RPMC. Pyridoxamine and ARB significantly reduced N 2.5%-induced VEGF and TGF-beta1 protein and mRNA expression in RPMC (p < 0.01). CONCLUSIONS: Neither new pH-neutral, lactate-buffered, low-GDP, two-chamber bag PDS, nor 7.5% icodextrin PDS alone satisfactorily inhibited VEGF and TGF-beta1 overproduction in RPMC, but ARB or pyridoxamine effectively inhibited glucose-containing PDS (N 2.5%)-induced overproduction.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hemodialysis Solutions , Peritoneal Dialysis , Peritoneum/drug effects , Peritoneum/metabolism , Pyridoxamine/pharmacology , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/drug effects , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/drug effects , Animals , Cells, Cultured , Male , Peritoneum/cytology , Rats , Rats, Sprague-Dawley
7.
Anticancer Res ; 29(6): 2009-14, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19528459

ABSTRACT

BACKGROUND: A major factor that impedes the clinical success of cisplatin-based chemotherapy for cancer is cisplatin resistance by cancer cells. MATERIALS AND METHODS: The sensitivity of parental HCT116 human colon cancer cell line and its isogenic gene-knockout sub-lines to cisplatin was determined by clonogenicity assay; furthermore, p53 activation, p21 expression, cell cycle arrest and senescence in these cells after cisplatin treatment were investigated. RESULTS: Parental cells were six times more resistant than 14-3-3sigma-knockout (sigma-KO) cells to cisplatin. Moreover, activation of p53, p53-dependent expression of p21 and p21-dependent senescence were observed in sigma-KO, but not parental cells after a treatment with a low cisplatin dose. CONCLUSION: A 14-3-3sigma-dependent mechanism inhibits p53 activation in parental cells treated with a low cisplatin dose, thereby blocking p21 expression that is essential for senescence and consequently conferring to the parental cells a significant degree of resistance to cisplatin.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Cisplatin/pharmacology , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , Exonucleases/metabolism , Neoplasm Proteins/metabolism , 14-3-3 Proteins , Biomarkers, Tumor/genetics , Blotting, Western , Cellular Senescence/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Exonucleases/genetics , Exoribonucleases , Flow Cytometry , Gene Knockout Techniques , Humans , Neoplasm Proteins/genetics , Tumor Cells, Cultured , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/physiology
8.
Cancer Res ; 68(20): 8465-72, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18922920

ABSTRACT

Microsatellite instability (MSI) is a hallmark of mismatch repair (MMR) deficiency. High levels of MSI at mononucleotide and dinucleotide repeats in colorectal cancer (CRC) are attributed to inactivation of the MMR genes, hMLH1 and hMSH2. CRC with low levels of MSI (MSI-L) exists; however, its molecular basis is unclear. There is another type of MSI--elevated microsatellite alterations at selected tetranucleotide repeats (EMAST)--where loci containing [AAAG](n) or [ATAG](n) repeats are unstable. EMAST is frequent in non-CRCs; however, the incidence of EMAST and its cause in CRC is not known. Here, we report that MutS homologue 3 (MSH3) knockdown or MSH3-deficient cells exhibit the EMAST phenotype and low levels of mutations at dinucleotide repeats. About 60% of 117 sporadic CRC cases exhibit EMAST. All of the cases defined as MSI-H (16 cases) exhibited high levels of EMAST. Among 101 non-MSI-H cases, all 19 cases of MSI-L and 35 of 82 cases of MSS exhibited EMAST. Although non-MSI-H CRC tissues contained MSH3-negative tumor cells ranging from 2% to 50% of the total tumor cell population, the tissues exhibiting EMAST contained more MSH3-negative cells (average, 31.5%) than did the tissues not exhibiting EMAST (8.4%). Taken together, our results support the concept that MSH3 deficiency causes EMAST or EMAST with low levels of MSI at loci with dinucleotide repeats in CRC.


Subject(s)
Colorectal Neoplasms/genetics , DNA-Binding Proteins/physiology , Microsatellite Instability , Adaptor Proteins, Signal Transducing/deficiency , DNA-Binding Proteins/analysis , DNA-Binding Proteins/deficiency , HCT116 Cells , Humans , Microsatellite Repeats , MutL Protein Homolog 1 , MutS Homolog 3 Protein , Nuclear Proteins/deficiency
9.
Biochem Genet ; 45(9-10): 671-81, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17690979

ABSTRACT

Inactivation of the DNA mismatch repair gene hMLH1 predisposes one to colorectal cancer. We have identified a C to G nucleotide substitution at position -107 relative to the hMLH1 gene translation initiation site in three of 163 colorectal cancer patients with an allele frequency of 0.0092 (3/326). One of the three -107G alleles occurred in one patient out of five with reduced hMLH1 expression in the tumor tissue. The -107G was not found in 63 healthy individuals. This substitution reduced transcriptional activity by 51% compared with -107C (P<0.01) and impeded the promoter-binding capacity of nuclear proteins. Although the small number of identified -107G alleles is insufficient to evaluate the contribution to the carcinogenesis and clinicopathological properties of the tumors, the effects of -107G on hMLH1 gene transcription and nuclear protein binding to the promoter sequence implicate the site, including -107C, as a crucial element interacting with the activator that maintains hMLH1 gene expression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Colorectal Neoplasms/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Adaptor Proteins, Signal Transducing/metabolism , Base Sequence , Case-Control Studies , Colorectal Neoplasms/metabolism , DNA Primers/genetics , DNA Repair/genetics , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Humans , MutL Protein Homolog 1 , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Protein Binding , Transcription, Genetic
10.
Anal Bioanal Chem ; 382(7): 1702-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15959768

ABSTRACT

Platinum (Pt)-DNA adducts formed by the anti-tumor agent cisplatin are recognized by the DNA mismatch repair (MMR) system. To investigate the involvement of MMR proteins including hMLH1 in the removal of these adducts, we developed a mL-scale wet-digestion method for inductively coupled plasma mass spectrometry (ICP-MS). The detection limit was 0.01 ng mL(-1) Pt, which corresponded to 2 pg Pt/microg DNA when 10 microg of DNA was used. The mean relative errors were 5.4% or better for a dynamic range of 0.01-10 ng mL(-1) Pt. DNA (approximately 500 microg) had no matrix effect. To improve the accuracy, DNA preparations were treated with ribonuclease and the apparent reduction in the concentration of Pt was corrected using cellular DNA levels, which were determined with Hoechst 33258. No significant differences were observed, in terms of the formation of Pt-DNA adducts or their removal over 6 h, between hMLH1-deficient HCT116 cells, a human colorectal cancer cell line, and hMLH1-complemented HCT116+ch3 cells (n=5; P>0.05), indicating that the hMLH1-dependent DNA repair systems contribute to neither the formation nor the removal of the adducts at detectable levels. In addition, approximately 19% of the adducts were removed within 6 h in both cell lines. A time course analysis (~24 h) suggested that the removal of cisplatin-generated Pt-DNA adducts follows first-order kinetics (t(1/2)=32 h). The amount of Pt-DNA adduct formed by oxaliplatin in 1 h was 56% (ratio of means) of that generated by an equimolar concentration of cisplatin in HCT116. The proposed procedure could be useful for determining Pt-DNA adducts formed by Pt(II) complexes.


Subject(s)
Antineoplastic Agents/metabolism , Carrier Proteins/metabolism , Cisplatin/analysis , DNA Adducts/analysis , DNA Repair , DNA, Neoplasm/metabolism , Nuclear Proteins/metabolism , Organoplatinum Compounds/metabolism , Platinum/analysis , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Mass Spectrometry , MutL Protein Homolog 1 , Organoplatinum Compounds/pharmacology , Oxaliplatin , Sensitivity and Specificity , Spectrophotometry, Atomic
12.
Oncol Rep ; 10(4): 859-66, 2003.
Article in English | MEDLINE | ID: mdl-12792735

ABSTRACT

The deficiency of the DNA mismatch repair (MMR) system is involved in tumorigenesis of either familial or sporadic colorectal cancers showing microsatellite instability (MSI). To investigate the involvement of the mutated hMSH2 gene in carcinogenesis, we searched for alteration of the gene in 15 MSI tumors of Japanese patients with sporadic colorectal cancer by a polymerase chain reaction (PCR)-single strand conformation polymorphism (SSCP) and DNA sequencing analyses. We found 20 alterations including 7 novel mutations, 6 germline and one somatic. To assume an oncogenic pathway of tumor of two patients carrying germline missense mutations, G40S located in an evolutionarily conserved amino-terminal motif and Y619C in a domain interacting with either hMSH3 or hMSH6, somatic mutations in 9 target genes of the MMR defect and in the p53 and K-ras genes and loss of heterozygosity (LOH) at the hMLH1 and p53 gene loci were then studied. In the tumor carrying G40S, other somatic hMSH2 mutations, G203R and 687delA in the (A)(7) repeat, and 5 one-bp deletions in the target genes were found, while no mutation in the p53 and K-ras genes. These results indicate that G40S may affect the hMSH2 function and the tumor may be developed by a typical MSI pathway. In another tumor with Y619C, LOH at the hMLH1 gene locus, no mutation in MMR target genes, and two-hit inactivation of the p53 gene were detected. This MSI tumor seems to be developed by another than MSI pathway. These results indicate that there are different oncogenic pathways in the MSI sporadic colorectal cancers with germline missense mutations in the hMSH2 gene. We conclude that familial colorectal cancer-suspected cases exist in a small population of sporadic colorectal cancers.


Subject(s)
Colorectal Neoplasms/genetics , Germ-Line Mutation/genetics , Microsatellite Repeats/genetics , Mutation, Missense/genetics , Proto-Oncogene Proteins/genetics , Amino Acid Sequence , Base Pair Mismatch , Case-Control Studies , DNA Primers/chemistry , DNA, Neoplasm , DNA-Binding Proteins/genetics , Genes, p53/physiology , Genes, ras/physiology , Humans , Loss of Heterozygosity , Molecular Sequence Data , MutS Homolog 2 Protein , MutS Homolog 3 Protein , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL