Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Gynecol Oncol ; 185: 202-211, 2024 06.
Article in English | MEDLINE | ID: mdl-38834399

ABSTRACT

OBJECTIVE: To report long-term efficacy and safety of selinexor maintenance therapy in adults with TP53 wild-type (TP53wt) stage IV or recurrent endometrial cancer (EC) who achieved partial remission (PR) or complete remission (CR) following chemotherapy. METHODS: Analysis of the prespecified, exploratory subgroup of patients with TP53wt EC from the phase 3 SIENDO study was performed. Progression-free survival (PFS) benefit in patients with TP53wt EC and across other patient subgroups were exploratory endpoints. Safety and tolerability were also assessed. RESULTS: Of the 263 patients enrolled in the SIENDO trial, 113 patients had TP53wt EC; 70/113 (61.9%) had TP53wt/proficient mismatch repair (pMMR) EC, and 29/113 (25.7%) had TP53wt/deficient mismatch repair (dMMR) EC. As of April 1, 2024, the median PFS (mPFS) for TP53wt patients who received selinexor compared with placebo was 28.4 versus 5.2 months (36.8-month follow-up, HR 0.44; 95% CI 0.27-0.73). A benefit in mPFS was seen with selinexor versus placebo regardless of MMR status (patients with TP53wt/pMMR EC: 39.5 vs 4.9 months, HR 0.36; 95% CI 0.19-0.71; patients with TP53wt/dMMR EC: 13.1 vs 3.7 months, HR 0.49; 95% CI 0.18-1.34). Selinexor treatment was generally manageable, with no new safety signals identified. CONCLUSION: In the phase 3 SIENDO study, selinexor maintenance therapy showed a promising efficacy signal and a manageable safety profile in the prespecified subgroup of patients with TP53wt EC who achieved a PR or CR following chemotherapy. These results are being further evaluated in an ongoing randomized phase 3 trial (NCT05611931).


Subject(s)
Endometrial Neoplasms , Hydrazines , Neoplasm Recurrence, Local , Triazoles , Tumor Suppressor Protein p53 , Humans , Female , Triazoles/administration & dosage , Triazoles/adverse effects , Triazoles/therapeutic use , Middle Aged , Hydrazines/adverse effects , Hydrazines/administration & dosage , Hydrazines/therapeutic use , Aged , Tumor Suppressor Protein p53/genetics , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Neoplasm Recurrence, Local/drug therapy , Adult , Follow-Up Studies , Progression-Free Survival , Aged, 80 and over , Maintenance Chemotherapy/methods , Neoplasm Staging
2.
J Thromb Haemost ; 22(6): 1569-1582, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38382738

ABSTRACT

BACKGROUND: Patients with cancer are at an increased risk of developing coagulation complications, and chemotherapy treatment increases the risk. Tumor progression is closely linked to the hemostatic system. Breast cancer tumors express coagulation factor V (FV), an essential factor in blood coagulation. The functional role of FV during treatment with chemotherapy is poorly understood and was explored in this study. OBJECTIVES: We aimed to investigate the role of FV in breast cancer progression by exploring associations with treatment response, gene regulation, and the functional effects of FV. METHODS: The receiver operating characteristic plotter was used to explore the predictive value of FV mRNA (F5) expression for treatment with FEC (5-fluorouracil, anthracycline, and cyclophosphamide). Breast cancer cohorts were analyzed to study treatment response to FEC. The effect of chemotherapy on F5 expression, the regulation of F5, and the functional effects of FV dependent and independent of chemotherapy were studied in breast cancer cell lines. RESULTS: F5 tumor expression was significantly higher in responders to FEC than in nonresponders. In vitro experiments revealed that anthracycline treatment increased the expression of F5. Inhibition and knockdown of p53 reduced the anthracycline-induced F5 expression. Mutation of a p53 half-site (c.158+1541/158+1564) in a luciferase plasmid reduced luciferase activity, suggesting that p53 plays a role in regulating F5. FV overexpression increased apoptosis and reduced proliferation slightly during anthracycline treatment. CONCLUSION: Our study identified F5 as a p53-regulated tumor suppressor candidate and a promising marker for response to chemotherapy. FV may have functional effects that are therapeutically relevant in breast cancer.


Subject(s)
Breast Neoplasms , Factor V , Gene Expression Regulation, Neoplastic , Tumor Suppressor Protein p53 , Female , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blood Coagulation/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclophosphamide/therapeutic use , Epirubicin/pharmacology , Epirubicin/therapeutic use , Factor V/genetics , Factor V/metabolism , Fluorouracil/therapeutic use , Fluorouracil/pharmacology , MCF-7 Cells , Mutation , RNA, Messenger/metabolism , RNA, Messenger/genetics , Treatment Outcome , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics
3.
Clin Transl Radiat Oncol ; 36: 106-112, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35993091

ABSTRACT

Background: The microscopic tumor extension before, during or after radiochemotherapy (RCHT) and its correlation with the tumor microenvironment (TME) are presently unknown. This information is, however, crucial in the era of image-guided, adaptive high-precision photon or particle therapy. Materials and methods: In this pilot study, we analyzed formalin-fixed paraffin-embedded (FFPE) tumor resection specimen from patients with histologically confirmed squamous cell carcinoma (SCC; n = 10) or adenocarcinoma (A; n = 10) of the esophagus, having undergone neoadjuvant radiochemotherapy followed by resection (NRCHT + R) or resection (R)]. FFPE tissue sections were analyzed by immunohistochemistry regarding tumor hypoxia (HIF-1α), proliferation (Ki67), immune status (PD1), cancer cell stemness (CXCR4), and p53 mutation status. Marker expression in HIF-1α subvolumes was part of a sub-analysis. Statistical analyses were performed using one-sided Mann-Whitney tests and Bland-Altman analysis. Results: In both SCC and AC patients, the overall percentages of positive tumor cells among the five TME markers, namely HIF-1α, Ki67, p53, CXCR4 and PD1 after NRCHT were lower than in the R cohort. However, only PD1 in SCC and Ki67 in AC showed significant association (Ki67: p = 0.03, PD1: p = 0.02). In the sub-analysis of hypoxic subvolumes among the AC patients, the percentage of positive tumor cells within hypoxic regions were statistically significantly lower in the NRCHT than in the R cohort across all the markers except for PD1. Conclusion: In this pilot study, we showed changes in the TME induced by NRCHT in both SCC and AC. These findings will be correlated with microscopic tumor extension measurements in a subsequent cohort of patients.

4.
Saudi J Biol Sci ; 28(12): 7257-7268, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34867030

ABSTRACT

INTRODUCTION: Piper crocatum Ruiz & Pav (P. crocatum) has been reported to accelerate the diabetic wound healing process empirically. Some studies showed the benefits of P. crocatum in treating various diseases but its mechanisms in diabetic wound healing have never been reported. In the present study we investigated the diabetic wound healing activity of the active fraction of P. crocatum on wounded hyperglycemia fibroblasts (wHFs). METHODS: Bioassay-guided fractionation was performed to get the most active fraction. The selected active fraction was applied to wHFs within 72 h incubation. Mimicking a diabetic condition was done using basal glucose media containing an additional 17 mMol/L D-glucose. A wound was simulated via the scratch assay. The collagen deposition was measured using Picro-Sirius Red and wound closure was measured using scratch wound assay. Underlying mechanisms through p53, αSMA, SOD1 and E-cadherin were measured using western blotting. RESULTS: We reported that FIV is the most active fraction of P. crocatum. We confirmed that FIV \(7.81 µg/ml, 15.62 µg/ml, 31.25 µg/ml, 62.5 µg/ml, and 125 µg/ml) induced the collagen deposition and wound closure of wHFs. Furthermore, FIV treatment (7.81 µg/ml, 15.62 µg/ml, 31.25 µg/ml) down-regulated the protein expression level of p53 and up-regulated the protein expression levels of αSMA, E-cadherin, and SOD1. DISCUSSION/CONCLUSIONS: Our findings suggest that ameliorating collagen deposition and wound closure through protein regulation of p53, αSMA, E-cadherin, and SOD1 are some of the mechanisms by which FIV of P. crocatum is involved in diabetic wound healing therapy.

5.
BMC Cancer ; 21(1): 295, 2021 Mar 20.
Article in English | MEDLINE | ID: mdl-33743635

ABSTRACT

BACKGROUND: PCNA-associated factor, the protein encoded by the KIAA0101/PCLAF gene, is a cell-cycle regulated oncoprotein that regulates DNA synthesis, maintenance of DNA methylation, and DNA-damage bypass, through the interaction with the human sliding clamp PCNA. KIAA0101/PCLAF is overexpressed in various cancers, including hepatocellular carcinoma (HCC). However, it remains unknown whether KIAA0101/PCLAF overexpression is coupled to gene amplification in HCC. METHODS: KIAA0101/PCLAF mRNA expression levels were assessed by quantitative real-time PCR (qRT-PCR) in 40 pairs of snap-frozen HCC and matched-non-cancerous tissues. KIAA0101/PCLAF gene copy numbers were evaluated by droplet digital PCR (ddPCR) in 36 pairs of the tissues, and protein expression was detected by immunohistochemistry (IHC) in 81 pairs of formalin-fixed paraffin-embedded (FFPE) tissues. The KIAA0101/PCLAF gene copy number alteration and RNA expression was compared by Spearman correlation. The relationships between KIAA0101 protein expression and other clinicopathological parameters, including Ki-67, p53, and HBsAg protein expression in HCC tissues, were evaluated using Chi-square test. RESULTS: Our results demonstrated that KIAA0101/PCLAF mRNA levels were significantly higher in HCC than in the matched-non-cancerous tissues (p < 0.0001). The high KIAA0101/PCLAF mRNA levels in HCC were associated with poor patient survival. The KIAA0101/PCLAF gene was not amplified in HCC, and KIAA0101/PCLAF gene copy numbers were not associated with KIAA0101/PCLAF transcript levels. KIAA0101 protein was overexpressed in the majority of HCC tissues (77.8%) but was not detectable in matched-non-cancerous tissues. Significant correlations between the expression of KIAA0101 protein in HCC tissues and p53 tumor suppressor protein (p = 0.002) and Ki-67 proliferation marker protein (p = 0.017) were found. However, KIAA0101 protein levels in HCC tissues were not correlated with patient age, tumor size, serum AFP level, or the HBsAg expression. CONCLUSIONS: KIAA0101/PCLAF mRNA and protein overexpression is frequently observed in HCC but without concurrent KIAA0101/PCLAF gene amplification. Significant correlations between the expression of KIAA0101 protein and p53 and Ki-67 proteins were observed in this study. Thus, detection of KIAA0101/PCLAF mRNA/protein might be used, along with the detection of p53 and Ki-67 proteins, as potential biomarkers to select candidate patients for further studies of novel HCC treatment related to these targets.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , DNA-Binding Proteins/genetics , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/pathology , DNA Copy Number Variations , Female , Gene Amplification , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Ki-67 Antigen/genetics , Liver/pathology , Liver Neoplasms/pathology , Male , Middle Aged , RNA, Messenger/metabolism , Retrospective Studies , Tumor Suppressor Protein p53/genetics
6.
Metabol Open ; 9: 100075, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33409483

ABSTRACT

Ethylene glycol monomethyl ether (EGME) has been used in many products usually handled by humans including inks, paints, polishes, brake fluids and so on. This present study therefore, investigated its effect on lung, in a time-course study in male Wistar rats. Animals were orally administered 50 mg/kg body weight of EGME for a period of 7, 14, and 21 days. Following 7 days of oral exposure to EGME, activities of GPx and SOD were significantly increased, as well as levels of K-Ras, c-Myc, p53, caspase-3, TNF-α and, IL-6, while NO level and GST activity were significantly reduced compared with control. At the end of 14 days exposure, GSH level was significantly decreased, while levels of K-Ras, c-Myc, p53, caspase-3, TNF-α, IL-6, NO and the activities of SOD and GPx were significantly elevated with respect to control. After 21 days of EGME administration, levels of Bcl-2, IL-10, GSH and NO as well as GST activity were significantly decreased, while levels of K-Ras, c-Myc, p53, Bax, caspase-3, IL-6, IL-1ß, TNF-α, as well as GPx, CAT, and SOD activities were significantly elevated compared with control. Lung histopathology revealed chronic disseminated alveolar inflammation, bronchiolitis, severe alveolar and bronchi hyperplasia, severe disseminated inflammation, thrombosis, and thickened vessels as a result of EGME exposures. Exposures to EGME could trigger lung damage via the disorganization of the antioxidant system, eliciting the up-regulation of inflammatory, apoptotic, and oncogenic markers in rats.

7.
Metabol Open ; 7: 100051, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32924002

ABSTRACT

Ethylene glycol monomethyl ether (EGME) is a major component of paints, lacquers, inks, and automobile brake fluids. As a result, exposures to humans are inevitable. We therefore, investigated in this study, its effect on testicular cells in a time-course manner in male Wistar rats. Animals were orally administered 50 mg/kg body weight of EGME for duration of 7, 14, and 21 days. Following 7 days of the administration, levels of NO and GSH were significantly reduced, while levels of c-Myc, K-Ras, caspase-3, IL-6, TNF-α, and IL-1ß were significantly increased compared with control. At the end of 14 days exposure, GPx, and SOD activities, as well as IL-10 level were significantly decreased, while levels of c-Myc, K-Ras, p53, Bax, caspase-3, IL-6, TNF-α, IL-1ß, and GST activity were significantly elevated compared with control. After 21 days of EGME administration, Bcl-2, IL-10, and NO levels were significantly decreased, while levels of c-Myc, K-Ras, p53, Bax, caspase-3, IL-6, TNF-α, IL-1ß, MDA and GST activity were significantly increased compared with control. After 7, 14, and 21 days of EGME administrations, testis histopathology showed severe loss of seminiferous tubules, the seminiferous epithelium revealed very few spermatocytes, spermatids, spermatogonia, spermatozoa, and Sertoli cells, while the interstitial tissue is eroded, with scanty abnormal Leydig cells, compared with the control that appeared normal. We therefore, concluded that EGME-induced testicular toxicity as a result of EGME administration could be via the disorganization of the endogenous antioxidant systems as well as up-regulation of pro-inflammatory, apoptotic and oncogenic mediators in rats.

8.
Biochem Biophys Rep ; 24: 100806, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32913901

ABSTRACT

2-methoxyethanol (2-ME) is an organic solvent widely used in the manufacture of brake fluids, paints, resins, varnish, nail polish, acetate cellulose, wood coloring, and as a plasticizer in plastics manufacturing. We therefore, investigated its effect on the liver, in a time-course study in male Wistar rats. Animals were orally administered 50 mg/kg body weight of 2-ME for a period of 7, 14, and 21 days. Following 7 days of administration of 2-ME, there was a significant increase in the level of Bax, c-Myc, K-Ras, TNF-α, IL-1ß, IL-6, MDA and GPx activity, while the levels of Bcl-2, NO and GSH were significantly reduced compared with control. At the end of 14 days exposure, Bcl-2, and GSH levels, as well as GST activity, were significantly decreased, while levels of Bax, c-Myc, K-Ras, caspase-3, TNF-α, IL-1ß, IL-6, MDA and NO were significantly increased compared with control. After 21 days of 2-ME administration, Bcl-2, IL-10, and GSH levels, as well as SOD and GST activities, were significantly decreased, while levels of Bax, c-Myc, K-Ras, caspase-3, p53, TNF-α, IL-1ß, IL-6, MDA and NO were significantly increased compared with control. Lastly, liver histopathology confirmed and corroborated the biochemical findings reported above. We therefore, advised that exposures to 2-ME should be strictly avoided as it could trigger hepatic damage through the disorganization of the antioxidant system, up-regulation of inflammatory, apoptotic, and oncogenic markers in rats.

9.
Toxicol Rep ; 7: 779-787, 2020.
Article in English | MEDLINE | ID: mdl-32642444

ABSTRACT

Methyl cellosolve (MC) is used in production of textile, paints, stains, inks, surface coatings, and anti-icing additive in hydraulic fluids and jet fuel. Consequently, the present study investigated its effect on renal cells, in a time-course study in male Wistar rats. Animals were orally administered 50 mg/kg body weight of MC for a period of 7, 14, and 21 days. Following 7 days of administration of MC, there was a significant increase in the levels of K-Ras, c-Myc, TNF-α, IL-6 and NO, while GSH level and SOD activity were significantly reduced compared with control. At the end of 14 days exposure, RKW, GSH, NO, and Bcl-2 levels were significantly decreased, while levels of K-Ras, c-Myc, p53, Bax, caspase-3, TNF-α, IL-1ß, IL-6, MDA and GPx activity were significantly increased compared with control. After 21 days of MC administration, RKW, GSH, NO, IL-10 and Bcl-2 levels were significantly decreased, while levels of K-Ras, c-Myc, p53, Bax, caspase-3, TNF-α, IL-1ß, IL-6, MDA and GST activity were significantly increased compared with control. Exposures to MC in any way should be strictly avoided as it could trigger renal damage through the disorganization of the antioxidant system, up-regulation of inflammatory, apoptotic, and oncogenic markers in rats.

10.
Curr Med Chem ; 27(22): 3706-3734, 2020.
Article in English | MEDLINE | ID: mdl-31223076

ABSTRACT

Cancer is a leading cause of death worldwide. It initiates when cell cycle regulatory genes lose their function either by environmental and/or by internal factors. Tumor suppressor protein p53, known as "Guardian of genome", plays a central role in maintaining genomic stability of the cell. Mutation of TP53 is documented in more than 50% of human cancers, usually by overexpression of negative regulator protein MDM2. Hence, reactivation of p53 by blocking the protein-protein interaction between the murine double minute 2 (MDM2) and the tumor suppressor protein p53 has become the most promising therapeutic strategy in oncology. Several classes of small molecules have been identified as potent, selective and efficient p53-MDM2 inhibitors. Herein, we review the druggability of p53-MDM2 inhibitors and their optimization approaches as well as clinical candidates categorized by scaffold type.


Subject(s)
Neoplasms , Animals , Antineoplastic Agents , Humans , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53
11.
Int J Mol Sci ; 18(2)2017 Feb 17.
Article in English | MEDLINE | ID: mdl-28218667

ABSTRACT

The ubiquitination pathway and proteasomal degradation machinery dominantly regulate p53 tumor suppressor protein stability, localization, and functions in both normal and cancerous cells. Selective E3 ubiquitin ligases dominantly regulate protein levels and activities of p53 in a large range of physiological conditions and in response to cellular changes induced by exogenous and endogenous stresses. The regulation of p53's functions by E3 ubiquitin ligases is a complex process that can lead to positive or negative regulation of p53 protein in a context- and cell type-dependent manner. Accessory proteins bind and modulate E3 ubiquitin ligases, adding yet another layer of regulatory control for p53 and its downstream functions. This review provides a comprehensive understanding of p53 regulation by selective E3 ubiquitin ligases and their potential to be considered as a new class of biomarkers and therapeutic targets in diverse types of cancers.


Subject(s)
Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Humans , Models, Biological , Protein Binding
12.
Natal; s.n; 23 fev 2017. 122 p. ilus, tab, graf.
Thesis in Portuguese | LILACS, BBO - Dentistry | ID: biblio-1427266

ABSTRACT

O câncer na cavidade oral é uma das lesões malignas mais frequentes na população mundial. Como o processo de desenvolvimento das neoplasias malignas remete a danos que promovem um desequilíbrio na regulação da divisão e morte celular, nos últimos anos, diversos estudos foram realizados com o intuito de verificar a influência desses danos no comportamento global das células e na evolução da doença. Nesse contexto, pesquisas recentes mostraram que alterações na expressão de REG podem desempenhar um importante papel na progressão tumoral de várias neoplasias malignas, por interferir na regulação da apoptose. Diante disso, o objetivo desse trabalho foi investigar a expressão imunoistoquímica dos marcadores REG, p53, MDM-2, Bcl-2 e Bax em carcinomas epidermoides de língua (CELs) oral, com a finalidade de promover uma análise comparativa da imunoexpressão destas proteínas com os parâmetros clínico-patológicos de agressividade da lesão, no intuito de identificar se o REG contribui para a progressão do tumor e se interfere na expressão das proteínas relacionadas a apoptose. Para tanto, foram coletadas informações clínicas de 58 pacientes acometidos por CELs. Em seguida, foi realizada análise histopatológica e imunoistoquímica dos marcadores supracitados, em amostras de material biológico parafinado da lesão. Os resultados mostraram que os tumores com metástase nodal e de alto grau histopatológico de malignidade apresentavam percentuais significativamente menores de REG (p<0,05). Em adição, a análise da expressão de p53, MDM-2 e Bax nos diferentes parâmetros clínico-patológicos avaliados nesse trabalho, não revelou diferenças significativas nos percentuais de imunopositividade desses marcadores. Com relação ao Bcl-2, foi visto que tumores de alto grau de malignidade e com óbitos relacionados a doença apresentavam percentual significativamente menor de positividade dessa proteína (p<0,05). Por fim, o teste de correlação de Spearman demonstrou existir fraca correlação positiva, estatisticamente significativa, entre os percentuais de REG e das proteínas MDM-2 e Bcl-2. Baseado nesses achados, pode-se concluir que a redução da expressão de REG parece contribuir para a progressão do CEL oral e pode influenciar na expressão das proteínas relacionadas a regulação da apoptose (AU).


Cancer of the oral cavity is one of the most common malignancies in the world. Since the development of malignant neoplasms is related to damage that causes an imbalance in the regulation of cell division and cell death, many studies have been conducted in recent years to verify the influence of this damage on overall cell behavior and on the progression of the disease. Within this context, recent studies suggest that changes in the expression of REG play an important role in the progression of different malignant tumors by interfering with the regulation of apoptosis. Therefore, the objective of the present study was to investigate the immunohistochemical expression of REG, p53, MDM-2, Bcl-2 and Bax in oral tongue squamous cell carcinoma (OTSCC), correlating the immunoexpression of these proteins with clinicalpathological parameters of tumor aggressiveness, in order to determine whether REG contributes to tumor progression and interferes with the expression of apoptosisrelated proteins. For this purpose, the clinical data of 58 patients with OTSCC were collected and paraffin-embedded tumor specimens were submitted to histopathological analysis and immunohistochemistry using the markers cited above. The results showed significantly higher expression of REG (p<0.05) in low-grade tumors and without lymph node metastases. In addition, comparison of the expression patterns of p53, MDM-2 and Bax according to the clinical-pathological parameters studied revealed no significant differences in the percentage of immunostaining for these markers. Regarding Bcl-2, a significantly lower percentage of immunostaining for this protein was observed in tumors with a high grade of malignancy and tumors associated with deaths related to the disease (p<0.05). Finally, Spearman's correlation test demonstrated a significant weak positive correlation between the percentages of REG and of MDM-2 and Bcl-2. These findings suggest that the reduced expression of REG contributes to the progression of OTSCC and may influence the expression of proteins related to the regulation of apoptosis (AU).


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Aged , Aged, 80 and over , Immunohistochemistry/methods , Carcinoma, Squamous Cell/pathology , Tumor Suppressor Protein p53 , Apoptosis , Neoplasms/pathology , Photomicrography/methods , Chi-Square Distribution , Statistics, Nonparametric
13.
Front Oncol ; 6: 247, 2016.
Article in English | MEDLINE | ID: mdl-27933271

ABSTRACT

Aurora kinases play critical roles in regulating spindle assembly, chromosome segregation, and cytokinesis to ensure faithful segregation of chromosomes during mitotic cell division cycle. Molecular and cell biological studies have revealed that Aurora kinases, at physiological levels, orchestrate complex sequential cellular processes at distinct subcellular locations through functional interactions with its various substrates. Aberrant expression of Aurora kinases, on the other hand, cause defects in mitotic spindle assembly, checkpoint response activation, and chromosome segregation leading to chromosomal instability. Elevated expression of Aurora kinases correlating with chromosomal instability is frequently detected in human cancers. Recent genomic profiling of about 3000 human cancer tissue specimens to identify various oncogenic signatures in The Cancer Genome Atlas project has reported that recurrent amplification and overexpression of Aurora kinase-A characterize distinct subsets of human tumors across multiple cancer types. Besides the well-characterized canonical pathway interactions of Aurora kinases in regulating assembly of the mitotic apparatus and chromosome segregation, growing evidence also supports the notion that deregulated expression of Aurora kinases in non-canonical pathways drive transformation and genomic instability by antagonizing tumor suppressor and exacerbating oncogenic signaling through direct interactions with critical proteins. Aberrant expression of the Aurora kinases-p53 protein family signaling axes appears to be critical in the abrogation of p53 protein family mediated tumor suppressor pathways frequently deregulated during oncogenic transformation process. Recent findings reveal the existence of feedback regulatory loops in mRNA expression and protein stability of these protein families and their consequences on downstream effectors involved in diverse physiological functions, such as mitotic progression, checkpoint response pathways, as well as self-renewal and pluripotency in embryonic stem cells. While these investigations have focused on the functional consequences of Aurora kinase protein family interactions with wild-type p53 family proteins, those involving Aurora kinases and mutant p53 remain to be elucidated. This article presents a comprehensive review of studies on Aurora kinases-p53 protein family interactions along with a prospective view on the possible functional consequences of Aurora kinase-mutant p53 signaling pathways in tumor cells. Additionally, we also discuss therapeutic implications of these findings in Aurora kinases overexpressing subsets of human tumors.

14.
Oncoimmunology ; 5(3): e1112941, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27141366

ABSTRACT

Activated antigen-presenting cells (APC) deliver the three signals cytotoxic T cells require to differentiate into effector cells that destroy the tumor. These comprise antigen, co-stimulatory signals and cytokines. Once these cells have carried out their function, they apoptose. We hypothesized that the tumor suppressor protein, p53, played an important role in generating the antitumor response facilitated by APC. CD11c+ APC derived from p53 wild-type (wt) mouse (wt p53) GM-CSF bone marrow cultures (BMAPC) and activated had reduced survival compared to BMAPC from p53 null consistent with p53-mediated apoptosis following activation. There was a lower percentage of antigenic peptide/MHC I complexes on antigen-pulsed p53 null cells suggesting p53 played a role in antigen processing but there was no difference in antigen-specific T cell proliferative responses to these cells in vivo. In contrast, antigen-specific cytotoxicity in vivo was markedly reduced in response to p53 null BMAPC. When these cells were pulsed with a model tumor antigen and delivered as a prophylactic vaccination, they provided no protection against melanoma cell growth whereas wt BMAPC were very effective. This suggested that p53 might regulate the requisite third signal and, indeed, we found that p53 null BMAPC produced less IL-12 than wt p53 BMAPC and that p53 bound to the promoter region of IL-12. This work suggests that p53 in activated BMAPC is associated with the generation of IL-12 required for the differentiation of cytotoxic immune responses and an effective antitumor response. This is a completely new role for this protein that has implications for BMAPC-mediated immunotherapy.

15.
Cell Cycle ; 14(14): 2251-64, 2015.
Article in English | MEDLINE | ID: mdl-26030591

ABSTRACT

Although every organism shares some common features of replication, this process varies greatly among eukaryotic species. Current data show that mathematical models of the organization of origins based on possibility theory may be applied (and remain accurate) in every model organism i.e. from yeast to humans. The major differences lie within the dynamics of origin firing and the regulation mechanisms that have evolved to meet new challenges throughout the evolution of the organism. This article elaborates on the relations between chromatin structure, organization of origins, their firing times and the impact that these features can have on genome stability, showing both differences and parallels inside the eukaryotic domain.


Subject(s)
DNA Replication , Replication Origin/genetics , Cell Cycle Proteins/metabolism , Chromatin/chemistry , Chromatin/metabolism , Humans , Minichromosome Maintenance Proteins/metabolism , Models, Theoretical , Nuclear Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
16.
J Laryngol Otol ; 129(1): 86-92, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25482503

ABSTRACT

OBJECTIVE: To identify clinical factors that can explain the differences in treatment outcome, and examine the value of human papillomavirus infection as a prognostic biomarker in stage IVa tonsillar carcinomas. METHODS: Fifty-nine patients with tonsillar carcinoma classified as stage IVa were retrospectively analysed for survival outcomes according to various clinical factors. Human papillomavirus infection was evaluated using a human papillomavirus DNA chip test and immunohistochemical staining for p16 and p53. RESULTS: Lower disease-free survival rates were associated with increasing local invasiveness and nodal status. Although human papillomavirus positivity and p16 expression was more common in locally advanced tonsillar carcinomas with advanced nodal status, the overall survival rate was better for patients with human papillomavirus positive, p16-positive tumours. CONCLUSION: The disease-free survival rate may differ according to local tumour invasiveness and nodal status, even for stage IVa tonsillar cancers. Human papillomavirus infection may be a useful biomarker for predicting treatment outcomes for stage VIa tumours.


Subject(s)
Carcinoma/mortality , Carcinoma/pathology , Papillomavirus Infections/mortality , Papillomavirus Infections/pathology , Tonsillar Neoplasms/mortality , Tonsillar Neoplasms/pathology , Adult , Age of Onset , Aged , Biomarkers, Tumor/analysis , Carcinoma/chemistry , Carcinoma/virology , Cyclin-Dependent Kinase Inhibitor p16 , Disease Progression , Disease-Free Survival , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Proteins/metabolism , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Papillomaviridae/isolation & purification , Retrospective Studies , Smoking , Tonsillar Neoplasms/chemistry , Tonsillar Neoplasms/virology , Tumor Suppressor Protein p53/metabolism
17.
Biophys Chem ; 183: 9-18, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23849959

ABSTRACT

High hydrostatic pressure (HHP) is a valuable tool to study processes such as protein folding, protein hydration and protein-protein interactions. HHP is a nondestructive technique because it reversibly affects internal cavities excluded from the solvent present in the hydrophobic core of proteins. HHP allows the solvation of buried amino acid side chains, thus shifting the equilibrium towards states of the studied molecule or molecular ensemble that occupy smaller volumes. HHP has long been used to dissociate multimeric proteins and protein aggregates and allows investigation of intermediate folding states, some of which are formed by proteins involved in human degenerative diseases, such as spongiform encephalopathies and Parkinson's disease, as well as cancer. When coupled with nuclear magnetic resonance and spectroscopic methods such as infrared and fluorescence spectroscopy, HHP treatment facilitates the understanding of protein folding and misfolding processes; the latter is related to protein aggregation into amyloid or amorphous species. In this review, we will address how HHP provides information about intermediate folding states and the aggregation processes of p53, which is related to cancer, and prion proteins, transthyretin and α-synuclein, which are related to human degenerative diseases.


Subject(s)
Amyloid/chemistry , Hydrostatic Pressure , Prealbumin/chemistry , Prions/chemistry , Tumor Suppressor Protein p53/chemistry , alpha-Synuclein/chemistry , Animals , Humans , Neoplasms/physiopathology , Neurodegenerative Diseases , Nuclear Magnetic Resonance, Biomolecular , Prealbumin/genetics , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Quaternary , Thermodynamics , Tumor Suppressor Protein p53/genetics , alpha-Synuclein/genetics
18.
Hepat Mon ; 11(5): 364-7, 2011 May.
Article in English | MEDLINE | ID: mdl-22087162

ABSTRACT

BACKGROUND: Previous studies suggest that annonaceous may cause permeability glycoprotein (P-gp) function to abate, leading to cell apoptosis. It has also been reported that annonaceous acetogenins affect hepatocellular carcinoma (HCC) cells in the G1 phase, leading to apoptosis. Desacetyluvaricin (Des), a new type of annonaceous acetogenin monomer, has a significant effect on HCC, with few side effects. OBJECTIVES: To investigate the effect of Des on the expression of Toll-like receptor 4 (TLR4) and P53 protein in HCC. MATERIALS AND METHODS: HCC HepG2.2.15 cell was cultured by routine method. HepG2.2.15 cells were divided into three groups: control group, treated with Des and DDP (cisplatin) which were examined by immunofluorescence flow cytometry for expression of TLR4 and P53. RESULTS: TLR4 was expressed by more cells in the Des group than in the cisplatin or serum-only groups (71.94%, 42.64%, and 37.16%, respectively; Des vs.cisplatin: p < 0.05; Des vs. serum only: p < 0.05), with no difference between the cisplatin and serum-only groups (p > 0.05). P53 was expressed by more cells in the Des and cisplatin groups than in the serum-only group (32.6%, 31.5% and 3.3%, respectively; Des vs. serum only, p < 0.05; cisplatin vs. serum only, p < 0.05), with no difference between the Des and cisplatin groups (p > 0.05). CONCLUSIONS: Des increases TLR4 and P53 expression in HCC cells. Improved immune recognition by the former effect and induction of apoptosis by the latter could be the mechanisms of Des's clinical effects on HCC.

SELECTION OF CITATIONS
SEARCH DETAIL