Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtrer
1.
J Clin Invest ; 2024 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-39325547

RÉSUMÉ

RNA N6-methyladenosine (m6A) reader YTHDF1 is implicated in cancer etiology and progression. We discovered that radiotherapy (RT) increased YTHDF1 expression in dendritic cells (DCs) of PBMCs from cancer patients, but not in other immune cells tested. Elevated YTHDF1 expression of DCs was associated with poor outcomes in patients receiving RT. We found that loss of Ythdf1 in DCs enhanced the antitumor effects of ionizing radiation (IR) via increasing the cross-priming capacity of DCs across multiple murine cancer models. Mechanistically, IR upregulated YTHDF1 expression in DCs through STING-IFN-I signaling. YTHDF1 in turn triggered STING degradation by increasing lysosomal cathepsins, thereby reducing IFN-I production. We created a YTHDF1 deletion/inhibition prototype DC vaccine, significantly improving the therapeutic effect of RT and radio-immunotherapy in a murine melanoma model. Our findings reveal a new layer of regulation between YTHDF1/m6A and STING in response to IR, which opens new paths for the development of YTHDF1-targeting therapies.

2.
Food Chem ; 461: 140906, 2024 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-39173262

RÉSUMÉ

A novel sensitive and reusable electrochemical biosensor for Listeria monocytegenes DNA has been constructed based on the recognition of water-soluble hydroxylated fullerene (fullerol) to single- and double-stranded DNA. First, the fullerol was electrodeposited on glassy carbon electrode (GCE), acting as a matrix for non-covalent adsorption of single-stranded probe DNA. Upon hybridization with the target DNA, the double helix structure was formed and desorbed from the electrode surface, driving synchronous regeneration of the biosensing interfaces. The biosensor showed a probe DNA loading density of 144 pmol∙cm-2 with the hybridization efficiency of 72.2%. The biosensor is applicable for the analysis of target DNA in actual milk samples with recoveries between 101.0% and 104.0%. This sensing platform provides a simple method for the construction of sensitive and reusable biosensor to monitor Listeria monocytogenes-related food pollution.


Sujet(s)
Techniques de biocapteur , Listeria monocytogenes , Listeria monocytogenes/isolement et purification , Listeria monocytogenes/génétique , Lait/microbiologie , Lait/composition chimique , Fullerènes/composition chimique , Techniques électrochimiques/instrumentation , Animaux , Hybridation d'acides nucléiques , ADN bactérien/génétique , ADN simple brin/composition chimique , Électrodes , Contamination des aliments/analyse
3.
Clin Cancer Res ; 30(19): 4450-4463, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38691100

RÉSUMÉ

PURPOSE: Radiation-mediated immune suppression limits efficacy and is a barrier in cancer therapy. Radiation induces negative regulators of tumor immunity including regulatory T cells (Treg). Mechanisms underlying Treg infiltration after radiotherapy (RT) are poorly defined. Given that conventional dendritic cells (cDC) maintain Treg, we sought to identify and target cDC signaling to block Treg infiltration after radiation. EXPERIMENTAL DESIGN: Transcriptomics and high dimensional flow cytometry revealed changes in murine tumor cDC that not only mediate Treg infiltration after RT but also associate with worse survival in human cancer datasets. Antibodies perturbing a cDC-CCL22-Treg axis were tested in syngeneic murine tumors. A prototype interferon-anti-epidermal growth factor receptor fusion protein (αEGFR-IFNα) was examined to block Treg infiltration and promote a CD8+ T cell response after RT. RESULTS: Radiation expands a population of mature cDC1 enriched in immunoregulatory markers that mediates Treg infiltration via the Treg-recruiting chemokine CCL22. Blocking CCL22 or Treg depletion both enhanced RT efficacy. αEGFR-IFNα blocked cDC1 CCL22 production while simultaneously inducing an antitumor CD8+ T cell response to enhance RT efficacy in multiple EGFR-expressing murine tumor models, including following systemic administration. CONCLUSIONS: We identify a previously unappreciated cDC mechanism mediating Treg tumor infiltration after RT. Our findings suggest blocking the cDC1-CCL22-Treg axis augments RT efficacy. αEGFR-IFNα added to RT provided robust antitumor responses better than systemic free interferon administration and may overcome clinical limitations to interferon therapy. Our findings highlight the complex behavior of cDC after RT and provide novel therapeutic strategies for overcoming RT-driven immunosuppression to improve RT efficacy. See related commentary by Kalinski et al., p. 4260.


Sujet(s)
Chimiokine CCL22 , Cellules dendritiques , Lymphocytes T régulateurs , Animaux , Cellules dendritiques/immunologie , Chimiokine CCL22/métabolisme , Chimiokine CCL22/génétique , Souris , Lymphocytes T régulateurs/immunologie , Humains , Radiotolérance , Lignée cellulaire tumorale , Lymphocytes T CD8+/immunologie , Récepteurs ErbB/antagonistes et inhibiteurs , Tumeurs/immunologie , Tumeurs/radiothérapie , Tumeurs/anatomopathologie , Femelle , Modèles animaux de maladie humaine
4.
Clin Cancer Res ; 30(9): 1945-1958, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38427437

RÉSUMÉ

PURPOSE: Radiotherapy (RT) is a widely employed anticancer treatment. Emerging evidence suggests that RT can elicit both tumor-inhibiting and tumor-promoting immune effects. The purpose of this study is to investigate immune suppressive factors of radiotherapy. EXPERIMENTAL DESIGN: We used a heterologous two-tumor model in which adaptive concomitant immunity was eliminated. RESULTS: Through analysis of PD-L1 expression and myeloid-derived suppressor cells (MDSC) frequencies using patient peripheral blood mononuclear cells and murine two-tumor and metastasis models, we report that local irradiation can induce a systemic increase in MDSC, as well as PD-L1 expression on dendritic cells and myeloid cells, and thereby increase the potential for metastatic dissemination in distal, nonirradiated tissue. In a mouse model using two distinct tumors, we found that PD-L1 induction by ionizing radiation was dependent on elevated chemokine CXCL10 signaling. Inhibiting PD-L1 or MDSC can potentially abrogate RT-induced metastasis and improve clinical outcomes for patients receiving RT. CONCLUSIONS: Blockade of PD-L1/CXCL10 axis or MDSC infiltration during irradiation can enhance abscopal tumor control and reduce metastasis.


Sujet(s)
Antigène CD274 , Cellules myéloïdes suppressives , Animaux , Antigène CD274/métabolisme , Souris , Cellules myéloïdes suppressives/immunologie , Cellules myéloïdes suppressives/métabolisme , Humains , Métastase tumorale , Lignée cellulaire tumorale , Femelle , Modèles animaux de maladie humaine , Chimiokine CXCL10/métabolisme
5.
J Clin Invest ; 133(24)2023 12 15.
Article de Anglais | MEDLINE | ID: mdl-38099498

RÉSUMÉ

Activation of TGF-ß signaling serves as an extrinsic resistance mechanism that limits the potential for radiotherapy. Bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) antagonizes TGF-ß signaling and is implicated in cancer progression. However, the molecular mechanisms of BAMBI regulation in immune cells and its impact on antitumor immunity after radiation have not been established. Here, we show that ionizing radiation (IR) specifically reduces BAMBI expression in immunosuppressive myeloid-derived suppressor cells (MDSCs) in both murine models and humans. Mechanistically, YTH N6-methyladenosine RNA-binding protein F2 (YTHDF2) directly binds and degrades Bambi transcripts in an N6-methyladenosine-dependent (m6A-dependent) manner, and this relies on NF-κB signaling. BAMBI suppresses the tumor-infiltrating capacity and suppression function of MDSCs via inhibiting TGF-ß signaling. Adeno-associated viral delivery of Bambi (AAV-Bambi) to the tumor microenvironment boosts the antitumor effects of radiotherapy and radioimmunotherapy combinations. Intriguingly, combination of AAV-Bambi and IR not only improves local tumor control, but also suppresses distant metastasis, further supporting its clinical translation potential. Our findings uncover a surprising role of BAMBI in myeloid cells, unveiling a potential therapeutic strategy for overcoming extrinsic radioresistance.


Sujet(s)
Tumeurs , Facteur de croissance transformant bêta , Animaux , Humains , Souris , Protéines membranaires/métabolisme , Tumeurs/génétique , Tumeurs/radiothérapie , Protéines de liaison à l'ARN/génétique , Facteurs de transcription/métabolisme , Facteur de croissance transformant bêta/génétique , Facteur de croissance transformant bêta/métabolisme , Microenvironnement tumoral
6.
Cancer Cell ; 41(7): 1294-1308.e8, 2023 07 10.
Article de Anglais | MEDLINE | ID: mdl-37236197

RÉSUMÉ

RNA N6-methyladenosine (m6A) modification is implicated in cancer progression. However, the impact of m6A on the antitumor effects of radiotherapy and the related mechanisms are unknown. Here we show that ionizing radiation (IR) induces immunosuppressive myeloid-derived suppressor cell (MDSC) expansion and YTHDF2 expression in both murine models and humans. Following IR, loss of Ythdf2 in myeloid cells augments antitumor immunity and overcomes tumor radioresistance by altering MDSC differentiation and inhibiting MDSC infiltration and suppressive function. The remodeling of the landscape of MDSC populations by local IR is reversed by Ythdf2 deficiency. IR-induced YTHDF2 expression relies on NF-κB signaling; YTHDF2 in turn leads to NF-κB activation by directly binding and degrading transcripts encoding negative regulators of NF-κB signaling, resulting in an IR-YTHDF2-NF-κB circuit. Pharmacological inhibition of YTHDF2 overcomes MDSC-induced immunosuppression and improves combined IR and/or anti-PD-L1 treatment. Thus, YTHDF2 is a promising target to improve radiotherapy (RT) and RT/immunotherapy combinations.


Sujet(s)
Facteur de transcription NF-kappa B , Tumeurs , Animaux , Humains , Souris , Régulation de l'expression des gènes , Cellules myéloïdes/métabolisme , Tumeurs/génétique , Tumeurs/radiothérapie , Facteur de transcription NF-kappa B/métabolisme , Protéines de liaison à l'ARN/métabolisme , Transduction du signal
7.
Nat Nanotechnol ; 17(12): 1322-1331, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-36302963

RÉSUMÉ

The clinical utility of stimulator of interferon genes (STING) agonists has been limited due to poor tumour-targeting and unwanted toxicity following systemic delivery. Here we describe a robust tumour-targeted STING agonist, ZnCDA, formed by the encapsulation of bacterial-derived cyclic dimeric adenosine monophosphate (CDA) in nanoscale coordination polymers. Intravenously injected ZnCDA prolongs CDA circulation and efficiently targets tumours, mediating robust anti-tumour effects in a diverse set of preclinical cancer models at a single dose. Our findings reveal that ZnCDA enhances tumour accumulation by disrupting endothelial cells in the tumour vasculature. ZnCDA preferentially targets tumour-associated macrophages to modulate antigen processing and presentation and subsequent priming of an anti-tumour T-cell response. ZnCDA reinvigorates the anti-tumour activity of both radiotherapy and immune checkpoint inhibitors in immunologically 'cold' pancreatic and glioma tumour models, offering a promising combination strategy for the treatment of intractable human cancers.


Sujet(s)
Nanoparticules , Tumeurs , Humains , AMP cyclique , Macrophages associés aux tumeurs , Zinc/pharmacologie , Cellules endothéliales , Protéines membranaires , Tumeurs/traitement médicamenteux , Nanoparticules/usage thérapeutique , AMP
8.
Sci Immunol ; 6(60)2021 06 15.
Article de Anglais | MEDLINE | ID: mdl-34723044

RÉSUMÉ

Radiotherapy (RT) is an important anti-cancer treatment modality that activates innate and adaptive immune responses. When all-trans retinoic acid (RA) was administered with radiation, we observed superior antitumor responses compared to ionizing radiation (IR) alone or RA alone. The superior antitumor effects of combination treatment were accompanied by a dramatic increase of TNF-α- and inducible nitric oxide synthase (iNOS)-producing inflammatory macrophages in local and distal non-irradiated (distal) tumors. Inflammatory macrophages are essential for the therapeutic efficacy of combination treatment by inducing effector T cell infiltration and enhancing the effector T cell to regulatory T cell ratio in local and distal tumors. T cells and T cell-derived IFN-γ are crucial for increasing inflammatory macrophage levels in IR and RA treated tumors. Notably, whereas CD8+ T cells are required for the antitumor response to IR, CD4+ T cells are required for the effectiveness of the IR and RA combination. Combination treatment with RA enhanced the abscopal response when radiation and PD-L1 blockade were used together. The synergistic positive feedback loop of inflammatory macrophages and adaptive immunity is required for the antitumor efficacy of IR plus RA combination treatment. Our findings provide a translational and relatively nontoxic strategy for enhancing the local and systemic antitumor effects of IR.


Sujet(s)
Chimioradiothérapie/méthodes , Macrophages/effets des médicaments et des substances chimiques , Tumeurs/thérapie , Trétinoïne/pharmacologie , Animaux , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/effets des radiations , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des radiations , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Humains , Interféron gamma/génétique , Interféron gamma/métabolisme , Macrophages/immunologie , Souris , Souris knockout , Tumeurs/immunologie , Tumeurs/anatomopathologie , Radiotolérance/effets des médicaments et des substances chimiques , Radiotolérance/immunologie , Récepteurs CCR2/génétique , Récepteurs CCR2/métabolisme , Trétinoïne/usage thérapeutique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des radiations
9.
Dig Dis Sci ; 56(5): 1309-14, 2011 May.
Article de Anglais | MEDLINE | ID: mdl-21053077

RÉSUMÉ

BACKGROUND: The role of proton pump inhibitors in Barrett's metaplasia and esophageal adenocarcinoma has been an area of controversy. AIMS: We evaluated the effectiveness of the proton pump inhibitor rabeprazole as a chemoprevention agent in a surgical rat reflux model of esophageal cancer. METHODS: The rat reflux model was created by performing a jejuno-esophagostomy on Sprague-Dawley rats. The surgery promoted the reflux of gastro-duodenal contents into the esophagus. Rabeprazole sodium (Eisai, Tokyo, Japan) was dissolved in 0.9% physiological saline to a desired concentration of 1.5% (W/V). Beginning 4 weeks post-surgery, all animals were administered either 0.2 ml per 100 g body weight injections of rabeprazole or equivalent injections of saline 3 days per week into the subcutaneous tissue of the back. Forty animals were killed 40 weeks after surgery and their esophagi were examined. Of these, 23 were included in the control group, while the remaining 17 were subjected to rabeprazole. RESULTS: While 74% (17/23) of the controls developed esophageal cancer, animals administered rabeprazole had an incidence of cancer of 29% (5/17) (p < 0.05, Fisher's exact test). Barrett's metaplasia was found on 100% (23/23) of the rats in the placebo group, but there was a protective effect in the rabeprazole group with 65% (11/17) of the rats displaying signs of Barrett's metaplasia (p < 0.05, Fisher's exact test). All of the rats developed proliferative hyperplasia. CONCLUSIONS: Rabeprazole protected against the development of esophageal cancer in a clinically relevant surgical reflux model. Rabeprazole warrants further investigation for potential clinical use as a chemoprevention agent.


Sujet(s)
(Pyridin-2-ylméthyl)sulfinyl-1H-benzimidazoles/pharmacologie , Antiulcéreux/pharmacologie , Tumeurs de l'oesophage/étiologie , Tumeurs de l'oesophage/prévention et contrôle , Reflux gastro-oesophagien/complications , Animaux , Tumeurs de l'oesophage/mortalité , Mâle , Rabéprazole , Rats , Rat Sprague-Dawley
10.
Dig Dis Sci ; 53(11): 2858-67, 2008 Nov.
Article de Anglais | MEDLINE | ID: mdl-18343998

RÉSUMÉ

PURPOSE: We developed and evaluated a GM-CSF whole-cell tumor vaccine for esophageal cancer. EXPERIMENTAL DESIGN: Cell lines derived from surgically induced rat reflux esophageal tumors were passaged in vitro and transfected with GM-CSF. First, the GM-CSF whole cell vaccine was evaluated against subcutaneously transplanted esophageal tumor cells. In a subsequent study, the vaccine was tested to see if it could reduce the incidence of cancer in the surgical reflux model. RESULTS: While subcutaneously transplanted tumor cells produced lasting tumors in PBS non-vaccinated placebo rats, transplanted tumors regressed and were immunologically rejected in animals vaccinated prior to implantation. In the surgical reflux model, the vaccine reduced the incidence of cancer from 17/23 (74%) in the controls to 6/16 (38%) in the vaccinated animals (P = 0.046). CONCLUSIONS: The GM-CSF whole cell tumor vaccine effectively promoted a strong immune response against subcutaneously transplanted tumors and protected animals from developing esophageal cancer in the reflux model.


Sujet(s)
Adénocarcinome/prévention et contrôle , Oesophage de Barrett/complications , Vaccins anticancéreux/usage thérapeutique , Tumeurs de l'oesophage/prévention et contrôle , Facteur de stimulation des colonies de granulocytes et de macrophages/usage thérapeutique , Adénocarcinome/étiologie , Adénocarcinome/anatomopathologie , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Tumeurs de l'oesophage/étiologie , Tumeurs de l'oesophage/anatomopathologie , Oesophagite peptique/complications , Oesophagostomie , Rats , Rat Sprague-Dawley , Transfection , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Methods Mol Biol ; 423: 383-91, 2008.
Article de Anglais | MEDLINE | ID: mdl-18370216

RÉSUMÉ

Keratinocyte growth factor-1 (KGF-1) is a member of the fibroblast growth factor (FGF) family FGF7 and is expressed in normal and wounded skin. KGF-1 is massively produced in the early stages of the wound healing process as well as during the later remodeling process (1, 2). We have studied the effects of the electroporation of a KGF-1 plasmid into excisional wounds of different rodent models mimicking diseases known to impair the normal wound healing process. We have used a genetically diabetic mouse model and a septic rat model in our experiments, and we have shown improvement of the healing rate (92% of the wounds are healed at day 12 vs. 40% of the control), the quality of epithelialization (histological score of 3.3 vs. 1.5), and the density of new blood vessels (85% more new blood vessels in the superficial layers than that of the control) (3, 4). Considering these results, we believe we can further explore the treatment modalities for using the electroporation-assisted transfection of DNA plasmid expression vectors of growth factors to enhance cutaneous wound healing.


Sujet(s)
Électrochimiothérapie/méthodes , Facteur de croissance fibroblastique de type 7/génétique , Thérapie génétique/méthodes , Cicatrisation de plaie/génétique , Animaux , Séquence nucléotidique , Amorces ADN/génétique , ADN recombiné/administration et posologie , ADN recombiné/génétique , Femelle , Expression des gènes , Humains , Souris , Souches mutantes de souris , Plasmides/administration et posologie , Plasmides/génétique , Rats , Rat Sprague-Dawley , Protéines recombinantes/génétique , Sepsie/anatomopathologie , Peau/traumatismes
12.
J Gastrointest Surg ; 12(1): 2-7; discussion 7-9, 2008 Jan.
Article de Anglais | MEDLINE | ID: mdl-17957441

RÉSUMÉ

BACKGROUND & AIMS: We developed a granulocyte-macrophage-colony-stimulating factor (GM-CSF) tumor vaccine for esophageal cancer. We evaluated the effectiveness of the vaccine as a prevention option in a surgical reflux rat model of esophageal cancer. METHODS: A surgical model involving a jejuno-esophagostomy was used to create Barrett's esophagus and esophageal cancer in rats. No carcinogen exposure was utilized. Cell lines derived from these tumors were stably passaged in vitro. GM-CSF-secreting tumor cells were generated using stable transfection. All rats underwent a total gastrectomy, followed by a jejuno-esophagostomy. The surgery promoted the reflux of duodenal contents into the esophagus. All animals were administered either a GM-CSF secreting whole cell vaccine or a phosphate-buffered saline (PBS) placebo injection 4, 6, 14, and 16 weeks post-surgery. RESULTS: While 15 of 16 animals in the non-vaccinated placebo group developed esophageal cancer, 94% (15 of 16), animals in the vaccine group had an incidence of cancer of 25% (4 of 16) (p<0.05). Barrett's esophagus was seen in 100% (16 of 16) of the controls and 83% (13 of 16) of the vaccinated animals. CONCLUSIONS: A GM-CSF-secreting whole cell tumor vaccine impeded esophageal tumor growth, but not the development of Barrett's esophagus, in a clinically relevant surgical reflux model.


Sujet(s)
Oesophage de Barrett/traitement médicamenteux , Vaccins anticancéreux/usage thérapeutique , Tumeurs de l'oesophage/prévention et contrôle , Oesophagite peptique/traitement médicamenteux , Oesophagostomie/effets indésirables , Facteur de stimulation des colonies de granulocytes et de macrophages/immunologie , Iléostomie/effets indésirables , Adénocarcinome/étiologie , Adénocarcinome/anatomopathologie , Adénocarcinome/prévention et contrôle , Animaux , Oesophage de Barrett/complications , Oesophage de Barrett/anatomopathologie , Carcinome épidermoïde/étiologie , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/prévention et contrôle , Lignée cellulaire tumorale , Tumeurs de l'oesophage/étiologie , Tumeurs de l'oesophage/anatomopathologie , Oesophagite peptique/complications , Oesophagite peptique/anatomopathologie , Études de suivi , Gastrectomie/effets indésirables , Tumeurs expérimentales , Rats , Rat Sprague-Dawley , Résultat thérapeutique
13.
J Thorac Cardiovasc Surg ; 133(3): 763-9, 2007 Mar.
Article de Anglais | MEDLINE | ID: mdl-17320581

RÉSUMÉ

OBJECTIVES: The reasons for the increasing incidence of esophageal adenocarcinoma are not clear. A causal relation between gastroesophageal reflux disease and esophageal adenocarcinoma has been suggested. Support for this comes from the development of esophageal adenocarcinoma in the rat reflux model. However, to date, no systematic characterization of the tumors derived from this model has been reported. METHODS: We induced biliary reflux by creating esophagojejunal anastomoses in 12 Sprague-Dawley rats. The experiment was terminated at 9 months, and rat esophagi were harvested for histopathologic documentation of reflux-associated changes and evidence of tumor formation. Three cell lines were established from 2 of the reflux-associated tumors. We tested the ability of these cells to grow in vitro in tissue culture and in vivo as xenografts in an orthotopic location at the gastroesophageal junction. Furthermore, we performed a cytogenetic analysis and determined the array-based gene expression profiles of these 3 rodent carcinoma lines compared with normal esophageal mucosa. RESULTS: At 9 months, 12 of 12 rodents had histologic features of metaplastic columnar epithelium in the esophagus, with 7 having invasive carcinomas with glandular differentiation (either adenocarcinomas or adenosquamous carcinomas). The 3 cell lines established from 2 reflux-associated tumors were capable of sustained in vitro propagation and grew successfully as xenografts in both subcutaneous and orthotopic locations, confirming the tumorigenic nature of these lines. Despite their establishment from primary tumors with glandular features, the histology of the xenografts was that of well-differentiated squamous carcinomas. Karyotype analyses demonstrated cytogenetic heterogeneity and aneuploidy; furthermore, translocation (7:11) was present in all 3 lines. Array-based gene expression profiling confirmed upregulation of several cancer-related genes important in human esophageal cancer. Quantitative reverse transcription-polymerase chain reaction was used to confirm the differential expression of selected transcripts (vascular endothelial growth factor [VEGF], polo-like kinases [PLK], cyclin dependent kinase 4 [CDK4], hypoxia-inducible factor 1alpha [HIF1alpha], and insulin-like growth factor 1 [IGF-1]) in comparison with nonneoplastic esophageal mucosal scrapings. CONCLUSIONS: The rodent reflux model is capable of inducing metaplastic epithelial changes simulating Barrett esophagus, as well as subsequent neoplastic transformation, at a high frequency. Cell lines have been established from these tumors that are capable of in vitro and in vivo passaging. The rodent reflux model should be a valuable model for studying therapy and chemoprevention efforts for Barrett esophagus, whereas the established cell lines provide a useful resource for drug discovery and other high-throughput studies.


Sujet(s)
Adénocarcinome/génétique , Transformation cellulaire néoplasique/anatomopathologie , Tumeurs de l'oesophage/génétique , Reflux gastro-oesophagien/génétique , Adénocarcinome/anatomopathologie , Animaux , Lignée cellulaire tumorale , Analyse cytogénétique , ADN complémentaire/analyse , Modèles animaux de maladie humaine , Tumeurs de l'oesophage/anatomopathologie , Reflux gastro-oesophagien/anatomopathologie , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Techniques in vitro , Mâle , Souris , Souris nude , Tumeurs expérimentales , Rats , Rat Sprague-Dawley , RT-PCR , Sensibilité et spécificité , Transplantation hétérologue
14.
Wound Repair Regen ; 14(5): 618-24, 2006.
Article de Anglais | MEDLINE | ID: mdl-17014675

RÉSUMÉ

We have previously shown that wound healing was improved in a diabetic mouse model of impaired wound healing following transfection with keratinocyte growth factor-1 (KGF-1) cDNA. We now extend these findings to the characterization of the effects of DNA plasmid vectors delivered to rats using electroporation (EP) in vivo in a sepsis-based model of impaired wound healing. To assess plasmid transfection and wound healing, gWIZ luciferase and PCDNA3.1/KGF-1 expression vectors were used, respectively. Cutaneous wounds were produced using an 8 mm-punch biopsy in Sprague-Dawley rats in which healing was impaired by cecal ligation-induced sepsis. We used National Institutes of Health image analysis software and histologic assessment to analyze wound closure and found that EP increased expression of gWIZ luciferase vector up to 53-fold compared with transfection without EP (p < 0.001). EP-assisted plasmid transfection was found to be localized to skin. Septic rats had a 4.7 times larger average wound area on day 9 compared with control (p < 0.001). Rats that underwent PCDNA3.1/KGF-1 transfection with EP had 60% smaller wounds on day 12 compared with vector without EP (p < 0.009). Quality of healing with KGF-1 vector plus EP scored 3.0 +/- 0.3 and was significantly better than that of 1.8 +/- 0.3 for treatment with vector alone (p < 0.05). We conclude that both the rate and quality of healing were improved with DNA plasmid expression vector for growth factor delivered with EP to septic rats.


Sujet(s)
Électroporation , Facteur de croissance fibroblastique de type 7/génétique , Thérapie génétique/méthodes , Sepsie/thérapie , Cicatrisation de plaie/génétique , Analyse de variance , Animaux , Cellules cultivées , Amorces ADN , Modèles animaux de maladie humaine , Facteur de croissance fibroblastique de type 7/usage thérapeutique , Plasmides/génétique , Plasmides/usage thérapeutique , Réaction de polymérisation en chaîne , Rats , Rat Sprague-Dawley , Sepsie/physiopathologie , Peau , Transfection , Cicatrisation de plaie/physiologie
15.
J Surg Res ; 134(1): 1-9, 2006 Jul.
Article de Anglais | MEDLINE | ID: mdl-16488438

RÉSUMÉ

BACKGROUND: Advancements in experimental therapeutics for esophageal cancers have been hampered by the lack of a reliable preclinical model that recapitulates the biology of human cancer, including in vivo growth in an animal model. METHODS: Bilious reflux was induced by esophago-jejunostomy in Sprague-Dawley rats. Nine of 12 (75%) Sprague-Dawley rats developed squamous or adenosquamous cancers, and three cell lines were created by in vitro propagation of freshly resected tumors, JA and JB lines from one cancer, and the AMY cell line from another. We subsequently tested the ability of these cell lines to propagate long-term in vitro and form xenografts in vivo, both hallmarks of transformed cells. In addition, we determined the effects of small molecule inhibitors of two important oncogenic pathways-the epidermal growth factor receptor (EGFR) and Hedgehog (Hh) signaling pathways, in vitro, as a "proof of principle" of using these unique cell lines for developing targeted therapies for esophageal cancer. Mechanism-based growth inhibition was assessed by down-regulation of activated downstream targets of EGFR in the case of Iressa, and by Hh luciferase reporter activity with cyclopamine. RESULTS: JA, JB, and AMY cell lines were able to grow continuously in vitro and consistently form xenografts in vivo in athymic mice, both subcutaneously, as well as in the "orthotopic" location at the gastroesophageal serosal junction (n = 2 mice per line, six of six engrafted). By histology, the tumors grow in vivo as well-differentiated keratinizing squamous cell carcinomas. JB cells had the highest expression of EGFR protein and also the most profound response to Iressa (gefitinib), an EGFR inhibitor (IC50 < 1 microm). Growth inhibition by Iressa was mirrored functionally by down-regulation of activated targets of the EGFR pathway, phospho-ERK1/2 and phospho-MEK levels. AMY cells expressed approximately 900-fold elevation of the Hh ligand, Indian Hh (Ihh), compared with normal esophageal epithelium, whereas expression of another Hh ligand, Sonic Hh (Shh), was not detected. On treatment with the specific Hh small molecule inhibitor cyclopamine, AMY cells demonstrated growth inhibition, which was accompanied by significant down-regulation of endogenous Hh luciferase reporter activity at 24 h and increased apoptosis in treated cells. CONCLUSIONS: We have established a model of esophageal carcinogenesis, capable of long-term in vitro and in vivo passage, and demonstrated therapeutic potential of targeting the EGFR and Hh pathways in the cell lines created from the rodent cancers. These unique cell lines should provide a platform for rapid preclinical validation of novel therapeutics for esophageal cancers.


Sujet(s)
Récepteurs ErbB/physiologie , Tumeurs de l'oesophage/métabolisme , Transduction du signal/physiologie , Transactivateurs/physiologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Géfitinib , Protéines Hedgehog , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mâle , Quinazolines/pharmacologie , Rats , Rat Sprague-Dawley , Alcaloïdes de Veratrum/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE