Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Dent Res ; 103(6): 585-595, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38722077

RÉSUMÉ

The prognosis and survival rate of head and neck squamous cell carcinoma (HNSCC) have remained unchanged for years, and the pathogenesis of HNSCC is still not fully understood, necessitating further research. An ideal animal model that accurately replicates the complex microenvironment of HNSCC is urgently needed. Among all the animal models for preclinical cancer research, tumor-bearing mouse models are the best known and widely used due to their high similarity to humans. Currently, mouse models for HNSCC can be broadly categorized into chemical-induced models, genetically engineered mouse models (GEMMs), and transplanted mouse models, each with its distinct advantages and limitations. In chemical-induced models, the carcinogen spontaneously initiates tumor formation through a multistep process. The resemblance of this model to human carcinogenesis renders it an ideal preclinical platform for studying HNSCC initiation and progression from precancerous lesions. The major drawback is that these models are time-consuming and, like human cancer, unpredictable in terms of timing, location, and number of lesions. GEMMs involve transgenic and knockout mice with gene modifications, leading to malignant transformation within a tumor microenvironment that recapitulates tumorigenesis in vivo, including their interaction with the immune system. However, most HNSCC GEMMs exhibit low tumor incidence and limited prognostic significance when translated to clinical studies. Transplanted mouse models are the most widely used in cancer research due to their consistency, availability, and efficiency. Based on the donor and recipient species matching, transplanted mouse models can be divided into xenografts and syngeneic models. In the latter, transplanted cells and host are from the same strain, making syngeneic models relevant to study functional immune system. In this review, we provide a comprehensive summary of the characteristics, establishment methods, and potential applications of these different HNSCC mouse models, aiming to assist researchers in choosing suitable animal models for their research.


Sujet(s)
Modèles animaux de maladie humaine , Tumeurs de la tête et du cou , Animaux , Souris , Tumeurs de la tête et du cou/génétique , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Humains , Microenvironnement tumoral , Cancérogènes
2.
Cancer Lett ; 518: 102-114, 2021 10 10.
Article de Anglais | MEDLINE | ID: mdl-34139286

RÉSUMÉ

This study reviews the molecular landscape of oral potentially malignant disorders (OPMD). We examine the impact of tumour heterogeneity, the spectrum of driver mutations (TP53, CDKN2A, TERT, NOTCH1, AJUBA, PIK3CA, CASP8) and gene transcription on tumour progression. We comment on how some of these mutations impact cellular senescence, field cancerization and cancer stem cells. We propose that OPMD can be monitored more closely and more dynamically through the use of liquid biopsies using an appropriate biomarker of transformation. We describe new gene interactions through the use of a systems biology approach and we highlight some of the first studies to identify functional genes using CRISPR-Cas9 technology. We believe that this information has translational implications for the use of re-purposed existing drugs and/or new drug development. Further, we argue that the use of digital technology encompassing clinical and laboratory-based data will create relevant datasets for machine learning/artificial intelligence. We believe that therapeutic intervention at an early molecular premalignant stage should be an important preventative strategy to inhibit the development of oral squamous cell carcinoma and that this approach is applicable to other aerodigestive tract cancers.


Sujet(s)
Tumeurs de la bouche/génétique , Animaux , Intelligence artificielle , Vieillissement de la cellule/génétique , Humains , Apprentissage machine , Tumeurs de la bouche/anatomopathologie , Cellules souches tumorales/anatomopathologie , États précancéreux/génétique , États précancéreux/anatomopathologie , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie
3.
Clin Radiol ; 74(12): 903-911, 2019 Dec.
Article de Anglais | MEDLINE | ID: mdl-31471062

RÉSUMÉ

Splenic injury is commonly encountered in severe blunt abdominal trauma. Technological improvements and the increasing availability of both diagnostic computed tomography (CT) and therapeutic splenic artery embolisation (SAE) are key factors in defining the high success rate of modern-day non-operative management (NOM) for blunt splenic injuries (BSIs). The Association for Surgery for Trauma (AAST) Organ Injury Scale (OIS) is commonly used by both radiologists and clinicians to stratify injury severity, traditionally based on the degree of parenchymal disruption seen on CT, and guide management. Its recent 2018 update takes splenic vascular injuries (i.e., active bleed, pseudoaneurysm, and traumatic arteriovenous fistulae) into consideration, the presence of which will indicate at least a grade IV (i.e., high-grade) injury. This is a reflection of the paradigm shift towards spleen conservation with regular use of SAE as the current standard of treatment. Prompted by the latest AAST OIS revision, which represents a more complete and current grading system, we present the spectrum of pertinent CT findings that the diagnostic radiologist should accurately identify and convey to the multidisciplinary trauma team (including the interventional radiologist). This review divides imaging findings based on the AAST OIS definitions and categorises them into (1) parenchymal and (2) vascular injuries. Features that may help in the detection of subtle BSIs are also described. Lastly, it touches on the key changes made to the new AAST OIS, substantiated by case illustrations.


Sujet(s)
Rate/traumatismes , Plaies non pénétrantes/imagerie diagnostique , Humains , Rate/imagerie diagnostique , Rate/anatomopathologie , Tomodensitométrie , Centres de traumatologie , Plaies non pénétrantes/anatomopathologie
4.
J Dent Res ; 97(8): 909-916, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-29512401

RÉSUMÉ

Head and neck cancer (HNC)-derived cell lines represent fundamental models for studying the biological mechanisms underlying cancer development and precision therapies. However, mining the genomic information of HNC cells from available databases requires knowledge on bioinformatics and computational skill sets. Here, we developed a user-friendly web resource for exploring, visualizing, and analyzing genomics information of commonly used HNC cell lines. We populated the current version of GENIPAC with 44 HNC cell lines from 3 studies: ORL Series, OPC-22, and H Series. Specifically, the mRNA expressions for all the 3 studies were derived with RNA-seq. The copy number alterations analysis of ORL Series was performed on the Genome Wide Human Cytoscan HD array, while copy number alterations for OPC-22 were derived from whole exome sequencing. Mutations from ORL Series and H Series were derived from RNA-seq information, while OPC-22 was based on whole exome sequencing. All genomic information was preprocessed with customized scripts and underwent data validation and correction through data set validator tools provided by cBioPortal. The clinical and genomic information of 44 HNC cell lines are easily assessable in GENIPAC. The functional utility of GENIPAC was demonstrated with some of the genomic alterations that are commonly reported in HNC, such as TP53, EGFR, CCND1, and PIK3CA. We showed that these genomic alterations as reported in The Cancer Genome Atlas database were recapitulated in the HNC cell lines in GENIPAC. Importantly, genomic alterations within pathways could be simultaneously visualized. We developed GENIPAC to create access to genomic information on HNC cell lines. This cancer omics initiative will help the research community to accelerate better understanding of HNC and the development of new precision therapeutic options for HNC treatment. GENIPAC is freely available at http://genipac.cancerresearch.my/ .


Sujet(s)
Lignée cellulaire tumorale , Bases de données génétiques , Génomique/méthodes , Tumeurs de la tête et du cou/génétique , Internet , Variations de nombre de copies de segment d'ADN , Analyse de profil d'expression de gènes , Génome humain , Humains , Mutation , ARN messager/analyse ,
5.
Biotech Histochem ; 91(4): 283-95, 2016.
Article de Anglais | MEDLINE | ID: mdl-27008034

RÉSUMÉ

We describe a new scaffold-free three-dimensional (3D) cell culture model using cholesteryl ester based lyotropic liquid crystal (LC) substrates. Keratinocytes were deposited randomly on the LC surface where they self-assembled into 3D microtissues or keratinospheroids. The cell density required to form spheroids was optimized. We investigated cell viability using dead/live cell assays. The adhesion characteristics of cells within the microtissues were determined using histological sectioning and immunofluorescence staining. Fourier transform infrared spectroscopy (FTIR) was used to characterize the biochemistry of the keratinospheroids. We found that both cells and microtissues could migrate on the LC surface. The viability study indicated approximately 80% viability of cells in the microtissues up to 20 days of culture. Strong intercellular adhesion was observed in the stratification of the multi-layered microspheroids using field emission-scanning electron microscopy (FE-SEM) and histochemical staining. The cytoskeleton and vinculins of the cells in the microtissues were expressed diffusely, but the microtissues were enriched with lipids and nucleic acids, which indicates close resemblance to the conditions in vivo. The basic 3D culture model based on LC may be used for cell and microtissue migration studies in response to cytochemical treatment.


Sujet(s)
Techniques de culture cellulaire/méthodes , Kératinocytes/cytologie , Cristaux liquides , Ingénierie tissulaire , Lignée cellulaire , Survie cellulaire , Technique d'immunofluorescence , Humains , Spectroscopie infrarouge à transformée de Fourier
6.
Oral Dis ; 21(1): e62-9, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-24930489

RÉSUMÉ

Periodontal bio-repositories, which allow banking of clinically validated human data and biological samples, provide an opportunity to derive biomarkers for periodontal diagnosis, prognosis and therapeutic activities which are expected to improve patient management. This article presents the establishing of the Malaysian Periodontal Database and Biobank System (MPDBS) which was initiated in 2011 with the aim to facilitate periodontal research. Partnerships were established with collaborating centres. Policies on specimen access, authorship and acknowledgement policies were agreed upon by all participating centres before the initiation of the periodontal biobank. Ethical approval for the collection of samples and data were obtained from institutional ethics review boards. A broad-based approach for informed consent was used, which covered areas related to quality of life impacts, genetics and molecular aspects of periodontal disease. Sample collection and processing was performed using a standardized protocol. Biobanking resources such as equipment and freezers were shared with the Malaysian Oral Cancer Database and Tissue Bank System (MOCDTBS). In the development of the MPDBS, challenges that were previously faced by the MOCDTBS were considered. Future challenges in terms of ethical and legal issues will be faced when international collaborations necessitate the transportation of specimens across borders.


Sujet(s)
Biobanques , Recherche biomédicale , Parodonte/anatomie et histologie , Biobanques/éthique , Biobanques/organisation et administration , Recherche biomédicale/éthique , Recherche biomédicale/méthodes , Comportement coopératif , Humains , Maladies parodontales/anatomopathologie
7.
J Periodontal Res ; 49(6): 683-95, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-24528298

RÉSUMÉ

Studies to elucidate the role of genetics as a risk factor for periodontal disease have gone through various phases. In the majority of cases, the initial 'hypothesis-dependent' candidate-gene polymorphism studies did not report valid genetic risk loci. Following a large-scale replication study, these initially positive results are believed to be caused by type 1 errors. However, susceptibility genes, such as CDKN2BAS (Cyclin Dependend KiNase 2B AntiSense RNA; alias ANRIL [ANtisense Rna In the Ink locus]), glycosyltransferase 6 domain containing 1 (GLT6D1) and cyclooxygenase 2 (COX2), have been reported as conclusive risk loci of periodontitis. The search for genetic risk factors accelerated with the advent of 'hypothesis-free' genome-wide association studies (GWAS). However, despite many different GWAS being performed for almost all human diseases, only three GWAS on periodontitis have been published - one reported genome-wide association of GLT6D1 with aggressive periodontitis (a severe phenotype of periodontitis), whereas the remaining two, which were performed on patients with chronic periodontitis, were not able to find significant associations. This review discusses the problems faced and the lessons learned from the search for genetic risk variants of periodontitis. Current and future strategies for identifying genetic variance in periodontitis, and the importance of planning a well-designed genetic study with large and sufficiently powered case-control samples of severe phenotypes, are also discussed.


Sujet(s)
Variation génétique/génétique , Étude d'association pangénomique , Parodontite/génétique , Études cas-témoins , Prédisposition génétique à une maladie/génétique , Humains , Parodontite/classification , Phénotype , Polymorphisme génétique/génétique
8.
Oral Dis ; 20(8): 762-7, 2014 Nov.
Article de Anglais | MEDLINE | ID: mdl-24320099

RÉSUMÉ

OBJECTIVES: The objective of the study was to determine the prevalence of HPV seropositivity among patients with oral squamous cell carcinoma (OSCC) and healthy individuals and to correlate the association between HPV 16 seropositivity and risk of OSCC. MATERIALS AND METHODS: HPV 16 E6 and E7 plasmids were constructed for the production of recombinant protein, which was used as the antigen in ELISA. HPV ELISA was performed on serum samples from 50 healthy individuals and 50 patients with OSCC. RESULTS: Using the HPV ELISA, 30% (OR = 2.25, 95% CI = 0.85-5.93) and 18% (OR = 1.61, 95% CI = 0.53-4.92) of patients with oral cancer were found to be HPV 16 E6 and E7 seropositive, respectively. Significant association was found between HPV 16 seropositivity and increased risk of OSCC in men, but not in male subjects. A similar trend was observed in non-betel quid chewers. CONCLUSIONS: Potential associations between HPV 16 E6/E7 seropositivity and oral cancer were revealed in men and non-betel quid chewer subjects, suggesting a possible etiological role of HPV 16 in subgroup of patients with OSCC in Malaysia.


Sujet(s)
Alphapapillomavirus/isolement et purification , Carcinome épidermoïde/virologie , Tumeurs de la bouche/virologie , Protéines des oncogènes viraux/sang , Protéines E7 de papillomavirus/sang , Protéines de répression/sang , Adulte , Études cas-témoins , Humains , Adulte d'âge moyen , Facteurs de risque
9.
Oral Dis ; 20(4): 345-51, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-23651447

RÉSUMÉ

OBJECTIVES: To identify differentially expressed miRNA between oral squamous cell carcinoma (OSCC) and non-cancer (NC) and to associate these with clinico-pathological parameters. MATERIALS AND METHODS: miRNA microarray profiling was utilized to obtain the expression profile of miRNAs in four OSCC and four NC samples. The expression of miR-31 and miR-375 was further validated in 26 OSCC and three NC samples using real-time-PCR. The association between miRNA expression and clinico-pathological parameters was tested by univariate and multivariate analyses. RESULTS: Microarray profiling demonstrated that 15 and four miRNAs were up-regulated and down-regulated, respectively, in OSCC as compared with NC. miR-31 and miR-375 were validated as up- and down-regulated miRNAs, respectively. In univariate analyses, expression of miR-31 was significantly elevated in early stage, tumours with no metastatic nodes and those from the buccal mucosa. By contrast, low miR-375 expression was significantly associated with late stage disease, larger tumour size and the non-cohesive type of pattern of invasion in OSCC. The association between miR-31 expression with tumour staging and site and miR-375 with tumour staging remained significant in multivariate analyses. CONCLUSIONS: This study has identified 19 miRNAs significantly associated with OSCC, and expressions of miR-31 and miR-375 were significantly related with clinico-pathological parameters suggesting they could be important in driving oral tumourigenesis.


Sujet(s)
Carcinome épidermoïde/génétique , microARN/génétique , Tumeurs de la bouche/génétique , Adulte , Carcinome épidermoïde/anatomopathologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Adulte d'âge moyen , Tumeurs de la bouche/anatomopathologie
10.
Oral Oncol ; 46(5): 379-86, 2010 May.
Article de Anglais | MEDLINE | ID: mdl-20371203

RÉSUMÉ

Despite the advances in cancer treatment, the 5-year survival rate for oral cancer has not changed significantly for the past 40 years and still remains among the worst of all anatomic sites. Gene expression microarrays have been used successfully in the identification of genetic alterations in cancer development, however, these have hitherto been limited by the need for specimens with good quality intact RNA. Here, we demonstrated the use of formalin-fixed paraffin-embedded tissues in microarray experiments to identify genes differentially expressed between cancerous and normal oral tissues. Forty-three tissue samples were macrodissected and gene expression analyses were conducted using the Illumina DASL assay. We report RNA yield of 2.4 and 0.8 microg/mm(3) from tumour and normal tissues, respectively and this correlated directly with the tissue volume used for RNA extraction. Using unsupervised hierarchical clustering, distinct gene expression profiles for tumour and normal samples could be generated, and differentially expressed genes could be identified. The majority of these genes were involved in regulation of apoptosis and cell cycle, metastasis and cell adhesion including BCL2A1, BIRC5, MMP1, MMP9 and ITGB4. Representative genes were further validated in independent samples suggesting that these genes may be directly associated with oral cancer development. The ability to conduct microarrays on formalin-fixed paraffin-embedded specimens represents a significant advancement that could open up avenues for finding genes that could be used as prognostication and predictive tools for cancer.


Sujet(s)
Carcinome épidermoïde/génétique , Analyse de profil d'expression de gènes/méthodes , Tumeurs de la bouche/génétique , Carcinome épidermoïde/mortalité , Carcinome épidermoïde/anatomopathologie , Formaldéhyde , Régulation de l'expression des gènes tumoraux , Humains , Bouche/cytologie , Tumeurs de la bouche/mortalité , Tumeurs de la bouche/anatomopathologie , Inclusion en paraffine , ARN tumoral/génétique
11.
Oral Oncol ; 45(8): 712-9, 2009 Aug.
Article de Anglais | MEDLINE | ID: mdl-19147396

RÉSUMÉ

Oral squamous cell carcinoma (OSCC) is a world health problem and is associated with exposure to different risk factors. In the west, smoking and alcohol consumption are considered to be the main risk factors whilst in India and southeast Asia, betel quid (BQ) chewing is predominant. In this study, we compared the gene expression patterns of oral cancers associated with BQ chewing to those caused by smoking using Affymetrix microarrays. We found that 281 genes were differentially expressed between OSCC and normal oral mucosa regardless of aetiological factors including MMP1, PLAU, MAGE-D4, GNA12, IFITM3 and NMU. Further, we identified 168 genes that were differentially expressed between the BQ and smoking groups including CXCL-9, TMPRSS2, CA12 and RNF24. The expression of these genes was validated using qPCR using independent tissue samples. The results demonstrate that whilst common genes/pathways contribute to the development of oral cancer, there are also other gene expression changes that are specific to certain risk factors. The findings suggest that different carcinogens activate or inhibit specific pathways during cancer development and progression. These unique gene expression profiles should be taken into consideration when developing biomarkers for future use in prognostic or therapeutic applications.


Sujet(s)
Areca/effets indésirables , Carcinome épidermoïde/génétique , Régulation de l'expression des gènes tumoraux/physiologie , Tumeurs de la bouche/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Carcinome épidermoïde/induit chimiquement , Femelle , Analyse de profil d'expression de gènes , Humains , Malaisie , Mâle , Analyse sur microréseau , Adulte d'âge moyen , Tumeurs de la bouche/induit chimiquement , Réaction de polymérisation en chaîne , Facteurs de risque , Fumer/effets indésirables
12.
Cancer Gene Ther ; 15(1): 40-50, 2008 Jan.
Article de Anglais | MEDLINE | ID: mdl-18034197

RÉSUMÉ

The majority of clinical trials evaluating replication-selective oncolytic adenoviruses utilized mutants with immunomodulatory E3B genes deleted, likely contributing to the attenuated efficacy. We investigated whether an intact immune response could contribute to the observed improved efficacy in response to combinations with chemotherapeutics. Seven carcinoma cell lines were evaluated by combining viral mutants; dl309 (DeltaE3B), dl704 (DeltaE3gp19K), dl312 (DeltaE1A) or wild-type Ad5 with the commonly used clinical drugs cisplatin and paclitaxel. Synergistic effects on cell death were determined by generation of combination indexes in cultured cells. In vivo tumor growth inhibition was achieved by virotherapy alone and was most efficacious with wild-type virus and least with the DeltaE3B mutant. Significantly higher efficacy was observed when the viruses were combined with drugs. The greatest enhancement of tumor inhibition was in combination with the DeltaE3B mutant restoring potency to that of Ad5 wild-type levels, observed only in animals with intact immune response. Increases in infectivity, viral gene expression and replication were identified as potential mechanisms contributing to the synergistic effects. Our results suggest that the attenuation of DeltaE3B mutants can be overcome by low doses of chemotherapeutics only in the presence of an intact immune response indicating a role for T-cell-mediated functions.


Sujet(s)
Adenoviridae/métabolisme , Protéines E1A d'adénovirus/métabolisme , Antinéoplasiques d'origine végétale/pharmacologie , Cisplatine/pharmacologie , Tumeurs expérimentales/thérapie , Thérapie virale de cancers , Virus oncolytiques/métabolisme , Paclitaxel/pharmacologie , Adenoviridae/génétique , Adenoviridae/immunologie , Protéines E1A d'adénovirus/génétique , Protéines E1A d'adénovirus/immunologie , Protéines E3 d'adénovirus/génétique , Animaux , Antinéoplasiques d'origine végétale/agonistes , Mort cellulaire/effets des médicaments et des substances chimiques , Mort cellulaire/génétique , Mort cellulaire/immunologie , Lignée cellulaire tumorale , Cisplatine/agonistes , Délétion de gène , Humains , Immunité cellulaire , Souris , Tumeurs expérimentales/génétique , Tumeurs expérimentales/immunologie , Tumeurs expérimentales/métabolisme , Virus oncolytiques/génétique , Virus oncolytiques/immunologie , Paclitaxel/agonistes , Lymphocytes T/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Br J Cancer ; 95(4): 496-505, 2006 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-16880792

RÉSUMÉ

Cellular senescence, the irreversible proliferative arrest seen in somatic cells after a limited number of divisions, is considered a crucial barrier to cancer, but direct evidence for this in vivo was lacking until recently. The best-known form of human cell senescence is attributed to telomere shortening and a DNA-damage response through p53 and p21. There is also a more rapid form of senescence, dependent on the p16-retinoblastoma pathway. p16 (CDKN2A) is a known melanoma susceptibility gene. Here, we use retrovirally mediated gene transfer to confirm that the normal form of senescence in cultured human melanocytes involves p16, since disruption of the p16/retinoblastoma pathway is required as well as telomerase activation for immortalisation. Expression (immunostaining) patterns of senescence mediators and markers in melanocytic lesions provide strong evidence that cell senescence occurs in benign melanocytic naevi (moles) in vivo and does not involve p53 or p21 upregulation, although p16 is widely expressed. In comparison, dysplastic naevi and early (radial growth-phase, RGP) melanomas show less p16 and some p53 and p21 immunostaining. All RGP melanomas expressed p21, suggesting areas of p53-mediated senescence, while most areas of advanced (vertical growth-phase) melanomas lacked both p16 and p21, implying escape from both forms of senescence (immortalisation). Moreover, nuclear p16 but not p21 expression can be induced in human melanocytes by oncogenic BRAF, as found in around 80% of naevi. We conclude that cell senescence can form a barrier to melanoma development. This also provides a potential explanation of why p16 is a melanoma suppressor gene.


Sujet(s)
Vieillissement de la cellule , Inhibiteur p16 de kinase cycline-dépendante/physiologie , Mélanome/anatomopathologie , Naevus/anatomopathologie , Tumeurs cutanées/anatomopathologie , Survie cellulaire , Cellules cultivées , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Évolution de la maladie , Humains , Mélanocytes/métabolisme , Protéines proto-oncogènes B-raf/pharmacologie , Protéine p53 suppresseur de tumeur/métabolisme
14.
J Exp Clin Cancer Res ; 24(4): 639-46, 2005 Dec.
Article de Anglais | MEDLINE | ID: mdl-16471328

RÉSUMÉ

Human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase, is strongly associated with telomerase activity implicated in cellular immortalization and tumorigenesis. In situ detection of hTERT will aid in determining the localization of telomerase positive cells. The aim of this study was to detect hTERT protein expression in multistep oral carcinogenesis using paraffin embedded tissue samples, and to study the relationship of hTERT expression with different histological stages in oral carcinogenesis. Normal (n = 4), hyperplastic (n = 4), dysplastic (n = 4) and neoplastic (n = 10) oral epithelia representing different histological stages in oral carcinogenesis were included in the study. hTERT protein detection was done by immunohistochemistry (IHC) technique. Nuclear staining intensities were noted and the hTERT-labelling index was determined. Dysplastic and neoplastic oral epithelia showed an increased percentage of hTERT positive cells (Grade 4: > 50% positive staining nuclei) with intense staining in the basal, parabasal and superficial layers of the epithelia, unlike normal oral mucosa which showed intense staining only in the basal and parabasal cell layers, which are the normal proliferative progenitor compartments. hTERT protein expression was elevated with the corresponding advancement of the histological stages of oral carcinogenesis, from normal to hyperplasia to dysplasia to carcinoma. There seems to be an upregulation of hTERT protein expression during the progression of oral cancer, therefore, this may indicate the feasibility of IHC detection of hTERT protein in oral carcinogenesis as a potential diagnostic or prognostic marker.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Transformation cellulaire néoplasique/métabolisme , Protéines de liaison à l'ADN/biosynthèse , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/anatomopathologie , Telomerase/biosynthèse , Lignée cellulaire tumorale , Transformation cellulaire néoplasique/anatomopathologie , Humains , Hyperplasie/métabolisme , Hyperplasie/anatomopathologie , Immunohistochimie , États précancéreux/métabolisme , États précancéreux/anatomopathologie , Régulation positive
15.
Gene Ther ; 6(5): 873-81, 1999 May.
Article de Anglais | MEDLINE | ID: mdl-10505113

RÉSUMÉ

Recombinant adenovirus vectors are popular tools for gene transfer and gene therapy. However biosafety constraints require that all handling of the vectors and vector-containing samples is restricted to dedicated containment laboratories, unless they had undergone a validated virus-inactivation procedure, which decontaminates the samples from any active virus. In this study we evaluated the feasibility of photodynamic treatment (PDT) with visible light to inactivate recombinant adenovirus vectors in biological samples, with minimum associated effects on other biological activities. Several photosensitizers were tested for their capacity to inactivate a model human adenovirus vector, AdCMVLuc, upon illumination. Four photosensitizers (methylene blue (MB), rose bengal (RB), uroporphyrin (UP) and aluminum phthalocynine tetrasulphonate (AIPcS4)) could inactivate the adenovirus, as measured by expression of the luciferase reporter gene and by plaque assay. Of these, MB demonstrated to be the most effective sensitizer in phosphate-buffered saline (PBS), giving > 7 log10 inactivation of the adenovirus. DNA isolated from MB- and light-treated virions was inefficient as a template for transcription. Furthermore, Southern blot analysis revealed fragmentation of the viral DNA. Based on its preference for DNA, MB is suited for adenovirus inactivation in blood plasma. Spiking experiments in which AdCMVLuc was added to plasma samples demonstrated a reduction (> 4 log10-fold) of reporter gene expression to almost background levels. In contrast to MB, photodynamic treatment with RB, UP or AIPcS4 did not lead to DNA damage. Although alterations of the viral capsid could not be detected, the binding pattern of the particles to target cells was significantly changed. Taken together, our data demonstrate that PDT is an efficient, convenient and useful method for the inactivation of adenovirus vectors in biological samples.


Sujet(s)
Adenoviridae/génétique , ADN viral/effets des radiations , Thérapie génétique/méthodes , Vecteurs génétiques , Lumière , Activation virale/effets des radiations , Adenoviridae/ultrastructure , Fragmentation de l'ADN , Études de faisabilité , Humains , Bleu de méthylène , Microscopie électronique , Photosensibilisants
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...