Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22
Filtrer
Plus de filtres











Base de données
Gamme d'année
2.
Nanotheranostics ; 3(4): 299-310, 2019.
Article de Anglais | MEDLINE | ID: mdl-31723547

RÉSUMÉ

Despite considerable progress with our understanding of glioblastoma multiforme (GBM) and the precise delivery of radiotherapy, the prognosis for GBM patients is still unfavorable with tumor recurrence due to radioresistance being a major concern. We recently developed a cross-linked iron oxide nanoparticle conjugated to azademethylcolchicine (CLIO-ICT) to target and eradicate a subpopulation of quiescent cells, glioblastoma initiating cells (GICs), which could be a reason for radioresistance and tumor relapse. The purpose of our study was to investigate if CLIO-ICT has an additive therapeutic effect to enhance the response of GBMs to ionizing radiation. Methods: NSG™ mice bearing human GBMs and C57BL/6J mice bearing murine GBMs received CLIO-ICT, radiation, or combination treatment. The mice underwent pre- and post-treatment magnetic resonance imaging (MRI) scans, bioluminescence imaging (BLI), and histological analysis. Tumor nanoparticle enhancement, tumor flux, microvessel density, GIC, and apoptosis markers were compared between different groups using a one-way ANOVA and two-tailed Mann-Whitney test. Additional NSG™ mice underwent survival analyses with Kaplan-Meier curves and a log rank (Mantel-Cox) test. Results: At 2 weeks post-treatment, BLI and MRI scans revealed significant reduction in tumor size for CLIO-ICT plus radiation treated tumors compared to monotherapy or vehicle-treated tumors. Combining CLIO-ICT with radiation therapy significantly decreased microvessel density, decreased GICs, increased caspase-3 expression, and prolonged the survival of GBM-bearing mice. CLIO-ICT delivery to GBM could be monitored with MRI. and was not significantly different before and after radiation. There was no significant caspase-3 expression in normal brain at therapeutic doses of CLIO-ICT administered. Conclusion: Our data shows additive anti-tumor effects of CLIO-ICT nanoparticles in combination with radiotherapy. The combination therapy proposed here could potentially be a clinically translatable strategy for treating GBMs.


Sujet(s)
Tumeurs du cerveau/traitement médicamenteux , Désoxyadénosine/usage thérapeutique , Glioblastome/traitement médicamenteux , Nanomédecine théranostique , Animaux , Encéphale/imagerie diagnostique , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/radiothérapie , Caspase-3/métabolisme , Lignée cellulaire tumorale , Association thérapeutique , Désoxyadénosine/composition chimique , Désoxyadénosine/pharmacologie , Vecteurs de médicaments/composition chimique , Femelle , Composés du fer III/composition chimique , Glioblastome/mortalité , Glioblastome/radiothérapie , Humains , Estimation de Kaplan-Meier , Imagerie par résonance magnétique , Nanoparticules métalliques/composition chimique , Souris , Souris de lignée C57BL , Souris transgéniques , Microvaisseaux/physiologie
3.
Mol Oncol ; 13(10): 2049-2061, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31376208

RÉSUMÉ

The long-term survival of osteosarcoma patients with metastatic or recurrent disease remains dismal, and new therapeutic options are urgently needed. The purpose of our study was to compare the efficacy of CD47 mAb plus doxorubicin combination therapy in mouse models of osteosarcoma with CD47 mAb and doxorubicin monotherapy. Forty-eight NOD scid gamma (NSG) mice with intratibial MNNG/HOS tumors received CD47 mAb, doxorubicin, combination therapy, or control IgG treatment. Twenty-four mice (n = 6 per group) underwent pre- and post-treatment magnetic resonance imaging (MRI) scans with the macrophage marker ferumoxytol, bioluminescence imaging, and histological analysis. Tumor ferumoxytol enhancement, tumor flux, and tumor-associated macrophages (TAM) density were compared between different groups using a one-way ANOVA. Twenty-four additional NSG mice underwent survival analyses with Kaplan-Meier curves and a log-rank (Mantel-Cox) test. Intratibial osteosarcomas demonstrated significantly stronger ferumoxytol enhancement and significantly increased TAM quantities after CD47 mAb plus doxorubicin combination therapy compared to CD47 mAb (P = 0.02) and doxorubicin monotherapy (P = 0.001). Tumor-bearing mice treated with CD47 mAb plus doxorubicin combination therapy demonstrated significantly reduced tumor size and prolonged survival compared to control groups that received CD47 mAb (P = 0.03), doxorubicin monotherapy (P = 0.01), and control IgG (P = 0.001). In conclusion, CD47 mAb plus doxorubicin therapy demonstrates an additive therapeutic effect in mouse models of osteosarcomas, which can be monitored with an immediately clinically applicable MRI technique.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Tumeurs osseuses/traitement médicamenteux , Antigènes CD47/immunologie , Doxorubicine/usage thérapeutique , Ostéosarcome/traitement médicamenteux , Animaux , Tumeurs osseuses/imagerie diagnostique , Tumeurs osseuses/immunologie , Lignée cellulaire tumorale , Oxyde ferrosoferrique/analyse , Humains , Luminescence , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Imagerie par résonance magnétique , Souris de lignée NOD , Imagerie optique , Ostéosarcome/imagerie diagnostique , Ostéosarcome/immunologie , Phagocytose/effets des médicaments et des substances chimiques
4.
Cell Death Dis ; 10(2): 36, 2019 01 15.
Article de Anglais | MEDLINE | ID: mdl-30674867

RÉSUMÉ

CD47 monoclonal antibodies (mAbs) activate tumor-associated macrophages (TAMs) in sarcomas to phagocytose and eliminate cancer cells. Though CD47 mAbs have entered clinical trials, diagnostic tests for monitoring therapy response in vivo are currently lacking. Ferumoxytol is an FDA-approved iron supplement which can be used "off label" as a contrast agent: the nanoparticle-based drug is phagocytosed by TAM and can be detected with magnetic resonance imaging (MRI). We evaluated if ferumoxytol-enhanced MRI can monitor TAM response to CD47 mAb therapy in osteosarcomas. Forty-eight osteosarcoma-bearing mice were treated with CD47 mAb or control IgG and underwent pre- and post-treatment ferumoxytol-MRI scans. Tumor enhancement, quantified as T2 relaxation times, was compared with the quantity of TAMs as determined by immunofluorescence microscopy and flow cytometry. Quantitative data were compared between experimental groups using exact two-sided Wilcoxon rank-sum tests. Compared to IgG-treated controls, CD47 mAb-treated tumors demonstrated significantly shortened T2 relaxation times on ferumoxytol-MRI scans (p < 0.01) and significantly increased F4/80+CD80+ M1 macrophages on histopathology (p < 0.01). CD47 mAb-treated F4/80+ macrophages demonstrated significantly augmented phagocytosis of ferumoxytol nanoparticles (p < 0.01). Thus, we conclude that ferumoxytol-MRI can detect TAM response to CD47 mAb in mouse models of osteosarcoma. The ferumoxytol-MRI imaging test could be immediately applied to monitor CD47 mAb therapies in clinical trials.


Sujet(s)
Anticorps monoclonaux/métabolisme , Antigènes CD47/génétique , Immunothérapie/méthodes , Macrophages/métabolisme , Imagerie par résonance magnétique/méthodes , Nanoparticules/métabolisme , Ostéosarcome/génétique , Animaux , Humains , Souris , Ostéosarcome/anatomopathologie
5.
Mol Cancer Ther ; 16(9): 1909-1921, 2017 09.
Article de Anglais | MEDLINE | ID: mdl-28659432

RÉSUMÉ

Glioblastoma (GBM) has a dismal prognosis. Evidence from preclinical tumor models and human trials indicates the role of GBM-initiating cells (GIC) in GBM drug resistance. Here, we propose a new treatment option with tumor enzyme-activatable, combined therapeutic and diagnostic (theranostic) nanoparticles, which caused specific toxicity against GBM tumor cells and GICs. The theranostic cross-linked iron oxide nanoparticles (CLIO) were conjugated to a highly potent vascular disrupting agent (ICT) and secured with a matrix-metalloproteinase (MMP-14) cleavable peptide. Treatment with CLIO-ICT disrupted tumor vasculature of MMP-14-expressing GBM, induced GIC apoptosis, and significantly impaired tumor growth. In addition, the iron core of CLIO-ICT enabled in vivo drug tracking with MR imaging. Treatment with CLIO-ICT plus temozolomide achieved tumor remission and significantly increased survival of human GBM-bearing mice by more than 2-fold compared with treatment with temozolomide alone. Thus, we present a novel therapeutic strategy with significant impact on survival and great potential for clinical translation. Mol Cancer Ther; 16(9); 1909-21. ©2017 AACR.


Sujet(s)
Tumeurs du cerveau/génétique , Expression des gènes , Glioblastome/génétique , Matrix metalloproteinase 14/génétique , Nanomédecine théranostique , Animaux , Antinéoplasiques/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du cerveau/diagnostic , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/thérapie , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Dacarbazine/administration et posologie , Dacarbazine/analogues et dérivés , Modèles animaux de maladie humaine , Composés du fer III/composition chimique , Cytométrie en flux , Glioblastome/diagnostic , Glioblastome/mortalité , Glioblastome/thérapie , Humains , Imagerie par résonance magnétique , Spectrométrie de masse , Matrix metalloproteinase 14/métabolisme , Souris , Nanoparticules/composition chimique , Témozolomide , Nanomédecine théranostique/méthodes , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Leuk Lymphoma ; 58(3): 676-688, 2017 03.
Article de Anglais | MEDLINE | ID: mdl-27338091

RÉSUMÉ

Elevated cyclin D1 (CCND1) expression levels in mantle cell lymphoma (MCL) are associated with aggressive clinical manifestations related to chemoresistance, but little is known about how this important proto-oncogene contributes to the resistance of MCL. Here, we showed that RNA interference-mediated depletion of CCND1 increased caspase-3 activities and induced apoptosis in the human MCL lines UPN-1 and JEKO-1. In vitro and xenotransplant studies revealed that the toxic effect of CCND1 depletion in MCL cells was likely due to increase in histone H2AX phosphorylation, a DNA damage marker. DNA fiber analysis suggested deregulated replication initiation after CCND1 depletion as a potential cause of DNA damage. Finally, in contrast to depletion or inhibition of cyclin-dependent kinase 4, CCND1 depletion increased chemosensitivity of MCL cells to replication inhibitors hydroxyurea and cytarabine. Our findings have an important implication for CCND1 as a potential therapeutic target in MCL patients who are refractory to standard chemotherapy.


Sujet(s)
Cycline D1/métabolisme , Altération de l'ADN , Lymphome à cellules du manteau/génétique , Lymphome à cellules du manteau/métabolisme , Animaux , Apoptose/génétique , Cycle cellulaire/génétique , Lignée cellulaire tumorale , Survie cellulaire/génétique , Réplication de l'ADN , Modèles animaux de maladie humaine , Hétérogreffes , Humains , Lymphome à cellules du manteau/anatomopathologie , Souris , Proto-oncogène Mas , Interférence par ARN , Petit ARN interférent/génétique
7.
JCI Insight ; 1(6)2016 May 06.
Article de Anglais | MEDLINE | ID: mdl-27182558

RÉSUMÉ

Limited transendothelial permeability across tumor microvessels represents a significant bottleneck in the development of tumor-specific diagnostic agents and theranostic drugs. Here, we show an approach to increase transendothelial permeability of macromolecular and nanoparticle-based contrast agents via inhibition of the type I TGF-ß receptor, activin-like kinase 5 (Alk5), in tumors. Alk5 inhibition significantly increased tumor contrast agent delivery and enhancement on imaging studies, while healthy organs remained relatively unaffected. Imaging data correlated with significantly decreased tumor interstitial fluid pressure, while tumor vascular density remained unchanged. This immediately clinically translatable concept involving Alk5 inhibitor pretreatment prior to an imaging study could be leveraged for improved tumor delivery of macromolecular and nanoparticle-based imaging probes and, thereby, facilitate development of more sensitive imaging tests for cancer diagnosis, enhanced tumor characterization, and personalized, image-guided therapies.

8.
BMC Cancer ; 16: 39, 2016 Jan 26.
Article de Anglais | MEDLINE | ID: mdl-26810856

RÉSUMÉ

BACKGROUND: Cancer metastasis is one of the most common causes of treatment failure and death in cancer patients. It has been acknowledged that aberrant activation of epithelial-to-mesenchymal transition (EMT) program, endows cancer cells with metastatic competence for which E-cadherin switch is a well-established hallmark. Suppression of E-cadherin by its transcriptional repressor Slug is thus a determining factor for EMT. Here, we aimed at discerning (i) the molecular mechanisms that regulate Slug/E-cadherin axis in oncogenic K-ras-expressing non-small cell lung carcinoma (NSCLC) cells, and (ii) the effect of aspirin in modulating the same. METHODS: The migratory behaviour of NSCLC cell line A549 were deciphered by wound healing assay. Further assessment of the molecular mechanisms was done by western blotting, RT-PCR, confocal microscopy, chromatin immunoprecipitation and small interfering RNA (siRNA)-mediated gene silencing. RESULTS: Here we report that in oncogenic K-ras-expressing A549 cells, Ras/ERK downstream Elk-1 forms p-Elk-1-p300 complex that being directly recruited to SLUG promoter acetylates the same to ensure p65NFκB binding for transcriptional up-regulation of Slug, a transcriptional repressor of E-cadherin. Aspirin inhibits EMT and decelerates the migratory potential of A549 cells by down-regulating Slug and thereby up-regulating E-cadherin. Aspirin impedes activation and nuclear translocation of p65NFκB, essential for this transcription factor being available for SLUG promoter binding. As a consequence, Slug transcription is down-regulated relieving A549 cells from Slug-mediated repression of E-cadherin transcription, thereby diminishing the metastatic potential of these oncogenic Ras-expressing NSCLC cells. CONCLUSIONS: Cumulatively, these results signify a crucial role of the anti-inflammatory agent aspirin as a novel negative regulator of epithelial-to-mesenchymal transition thereby suggesting its candidature as a promising tool for deterring metastasis of highly invasive K-ras-expressing NSCLC cells.


Sujet(s)
Acide acétylsalicylique/administration et posologie , Cadhérines/biosynthèse , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Protéines proto-oncogènes p21(ras)/biosynthèse , Facteurs de transcription/biosynthèse , Cadhérines/génétique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Protéines proto-oncogènes p21(ras)/génétique , Facteurs de transcription de la famille Snail , Facteur de transcription RelA/biosynthèse , Facteur de transcription RelA/génétique , Facteurs de transcription/génétique
9.
Immunology ; 144(4): 561-73, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25284464

RÉSUMÉ

Tumour progression is associated with immune-suppressive conditions that facilitate the escape of tumour cells from the regimen of immune cells, subsequently paralysing the host defence mechanisms. Induction of CD4(+)  CD25(+)  FoxP3(+) T regulatory (Treg) cells has been implicated in the tumour immune escape mechanism, although the novel anti-cancer treatment strategies targeting Treg cells remain unknown. The focus of this study is to define the interaction between tumour and immune system, i.e. how immune tolerance starts and gradually leads to the induction of adaptive Treg cells in the tumour microenvironment. Our study identified hyperactivated mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) -signalling as a potential target for reversing Treg cell augmentation in breast cancer patients. In more mechanistic detail, pharmacological inhibitors of MEK/ERK signalling inhibited transforming growth factor-ß (TGF-ß) production in tumour cells that essentially blocked TGF-ß-SMAD3/SMAD4-mediated induction of CD25/interleukin-2 receptor α on CD4(+) T-cell surface. As a result high-affinity binding of interleukin-2 on those cells was prohibited, causing lack of Janus kinase 1 (JAK1)/JAK3-mediated signal transducer and activator of transcription 3 (STAT3)/STAT5 activation required for FoxP3 expression. Finally, for a more radical approach towards a safe MEK inhibitor, we validate the potential of multi-kinase inhibitor curcumin, especially the nano-curcumin made out of pure curcumin with greater bioavailability; in repealing tumour-shed TGF-ß-induced Treg cell augmentation.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Tumeurs du sein/traitement médicamenteux , Curcumine/pharmacologie , MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , Communication paracrine/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme , Microenvironnement tumoral , Animaux , Tumeurs du sein/enzymologie , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Études cas-témoins , Chimie pharmaceutique , Techniques de coculture , Relation dose-effet des médicaments , Femelle , Facteurs de transcription Forkhead/métabolisme , Humains , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , MAP Kinase Kinase Kinases/génétique , MAP Kinase Kinase Kinases/métabolisme , Souris de lignée BALB C , Nanoparticules , Facteurs de transcription STAT/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéines Smad/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Facteurs temps , Facteur de croissance transformant bêta/génétique , Cellules cancéreuses en culture , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques
10.
J Biol Chem ; 289(42): 29074-85, 2014 Oct 17.
Article de Anglais | MEDLINE | ID: mdl-25157104

RÉSUMÉ

Matrix attachment region (MAR)-binding proteins have been implicated in the transcriptional regulation of host as well as viral genes, but their precise role in HPV-infected cervical cancer remains unclear. Here we show that HPV18 promoter contains consensus MAR element in the LCR and E6 sequences where SMAR1 binds and reinforces HPV18 E6 transcriptional silencing. In fact, curcumin-induced up-regulation of SMAR1 ensures recruitment of SMAR1-HDAC1 repressor complex at the LCR and E6 MAR sequences, thereby decreasing histone acetylation at H3K9 and H3K18, leading to reorientation of the chromatin. As a consequence, c-Fos binding at the putative AP-1 sites on E6 promoter is inhibited. E6 depletion interrupts degradation of E6-mediated p53 and lysine acetyl transferase, Tip60. Tip60, in turn, acetylates p53, thereby restoring p53-mediated transactivation of proapoptotic genes to ensure apoptosis. This hitherto unexplained function of SMAR1 signifies the potential of this unique scaffold matrix-associated region-binding protein as a critical regulator of E6-mediated anti-apoptotic network in HPV18-infected cervical adenocarcinoma. These results also justify the candidature of curcumin for the treatment of HPV18-infected cervical carcinoma.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Chromatine/métabolisme , Protéines de liaison à l'ADN/métabolisme , Régulation de l'expression des gènes viraux , Protéines nucléaires/métabolisme , Protéines des oncogènes viraux/métabolisme , Protéines proto-oncogènes c-fos/métabolisme , Transcription génétique , Acétylation , Apoptose , Cellules HeLa , Histone/métabolisme , Humains , Régions promotrices (génétique) , Liaison aux protéines , Facteur de transcription AP-1/métabolisme
11.
J Biol Chem ; 289(37): 25431-44, 2014 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-25086032

RÉSUMÉ

The evolution of the cancer cell into a metastatic entity is the major cause of death in patients with cancer. It has been acknowledged that aberrant activation of a latent embryonic program, known as the epithelial-mesenchymal transition (EMT), can endow cancer cells with the migratory and invasive capabilities associated with metastatic competence for which E-cadherin switch is a well-established hallmark. Discerning the molecular mechanisms that regulate E-cadherin expression is therefore critical for understanding tumor invasiveness and metastasis. Here we report that SMAR1 overexpression inhibits EMT and decelerates the migratory potential of breast cancer cells by up-regulating E-cadherin in a bidirectional manner. While SMAR1-dependent transcriptional repression of Slug by direct recruitment of SMAR1/HDAC1 complex to the matrix attachment region site present in the Slug promoter restores E-cadherin expression, SMAR1 also hinders E-cadherin-MDM2 interaction thereby reducing ubiquitination and degradation of E-cadherin protein. Consistently, siRNA knockdown of SMAR1 expression in these breast cancer cells results in a coordinative action of Slug-mediated repression of E-cadherin transcription, as well as degradation of E-cadherin protein through MDM2, up-regulating breast cancer cell migration. These results indicate a crucial role for SMAR1 in restraining breast cancer cell migration and suggest the candidature of this scaffold matrix-associated region-binding protein as a tumor suppressor.


Sujet(s)
Tumeurs du sein/génétique , Cadhérines/biosynthèse , Protéines du cycle cellulaire/biosynthèse , Protéines de liaison à l'ADN/biosynthèse , Transition épithélio-mésenchymateuse/génétique , Protéines nucléaires/biosynthèse , Tumeurs du sein/anatomopathologie , Cadhérines/génétique , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Femelle , Régulation de l'expression des gènes tumoraux , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme , Humains , Métastase tumorale , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Protéolyse , Protéines proto-oncogènes c-mdm2/génétique , Protéines proto-oncogènes c-mdm2/métabolisme , Facteurs de transcription de la famille Snail , Facteurs de transcription/génétique
12.
Immunity ; 39(6): 1057-69, 2013 12 12.
Article de Anglais | MEDLINE | ID: mdl-24315995

RÉSUMÉ

This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the authors. This study provided an explanation for why loss of FoxP3 in inducible regulatory T cells results in reduced expression of interleukin (IL)-10 despite the absence of FoxP3 binding sites in the IL-10 promoter. STAT3 binding sites do exist in the promoter, and evidence for a direct molecular interaction between FoxP3 and STAT3 proteins was provided as an explanation of the effect of loss of FoxP3. As supporting evidence, we reported modeling of a structural interaction between these two transcription factors in Figure 4D. As the N-terminal region of FoxP3, which consists of the Exon-2 region, had not been solved at structural resolution, we mistakenly used what we deduced to be a FoxP3 related transcription factor, NFAT, in the modeling. The model depicted in Figure 4D therefore did not represent a putative interaction between FoxP3 and STAT3 as labeled, but rather a putative interaction between NFAT and STAT3. Given the incorrect labeling of Figure 4D, the lack of documentation in the paper describing exactly how the modeling was performed, the lack of evidence shown in the paper for the choice of NFAT as the modeling partner, and the limited supporting evidence for a cooperative interaction between FoxP3 and STAT3, the editors have concluded with the corresponding author that the appropriate course of action is to retract the paper. We apologize for any confusion and inconvenience caused to readers.


Sujet(s)
Tumeurs du sein/physiopathologie , Facteurs de transcription Forkhead/métabolisme , Régulation de l'expression des gènes tumoraux , Facteur de transcription STAT-3/métabolisme , Lymphocytes T régulateurs/métabolisme , Lignée cellulaire tumorale , Femelle , Humains , Modèles moléculaires , Facteurs de transcription
13.
BMC Complement Altern Med ; 13: 230, 2013 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-24053127

RÉSUMÉ

BACKGROUND: Complementary medicines, including homeopathy, are used by many patients with cancer, usually alongside with conventional treatment. However, the molecular mechanisms underneath the anti-cancer effect, if any, of these medicines have still remained unexplored. To this end we attempted to evaluate the efficacy of calcarea carbonica, a homeopathic medicine, as an anti-cancer agent and to delineate the detail molecular mechanism(s) underlying calcerea carbonica-induced tumor regression. METHODS: To investigate and delineate the underlying mechanisms of calcarea carbonica-induced tumor regression, Trypan blue dye-exclusion test, flow cytometric, Western blot and reverse transcriptase-PCR techniques were employed. Further, siRNA transfections and inhibitor studies were used to validate the involvement of p53 pathway in calcarea carbonica-induced apoptosis in cancer cells. RESULTS: Interestingly, although calcarea carbonica administration to Ehrlich's ascites carcinoma (EAC)- and Sarcoma-180 (S-180)-bearing Swiss albino mice resulted in 30-35% tumor cell apoptosis, it failed to induce any significant cell death in ex vivo conditions. These results prompted us to examine whether calcarea carbonica employs the immuno-modulatory circuit in asserting its anti-tumor effects. Calcarea carbonica prevented tumor-induced loss of effector T cell repertoire, reversed type-2 cytokine bias and attenuated tumor-induced inhibition of T cell proliferation in tumor-bearing host. To confirm the role of immune system in calcarea carbonica-induced cancer cell death, a battery of cancer cells were co-cultured with calcarea carbonica-primed T cells. Our results indicated a "two-step" mechanism of the induction of apoptosis in tumor cells by calcarea carbonica i.e., (1) activation of the immune system of the host; and (2) induction of cancer cell apoptosis via immuno-modulatory circuit in p53-dependent manner by down-regulating Bcl-2:Bax ratio. Bax up-regulation resulted in mitochondrial transmembrane potential loss and cytochrome c release followed by activation of caspase cascade. Knocking out of p53 by RNA-interference inhibited calcarea carbonica-induced apoptosis thereby confirming the contribution of p53. CONCLUSION: These observations delineate the significance of immuno-modulatory circuit during calcarea carbonica-mediated tumor apoptosis. The molecular mechanism identified may serve as a platform for involving calcarea carbonica into immunotherapeutic strategies for effective tumor regression.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Carbonate de calcium/pharmacologie , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Antinéoplasiques/composition chimique , Tumeurs du sein , Carbonate de calcium/composition chimique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Femelle , Humains , Souris , Mitochondries/effets des médicaments et des substances chimiques , Lymphocytes T auxiliaires/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Apoptosis ; 18(5): 589-604, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23329180

RÉSUMÉ

Mutations in REarranged during Transfection (RET) receptor tyrosine, followed by the oncogenic activation of RET kinase is responsible for the development of medullary thyroid carcinoma (MTC) that responds poorly to conventional chemotherapy. Targeting RET, therefore, might be useful in tailoring surveillance of MTC patients. Here we showed that theaflavins, the bioactive components of black tea, successfully induced apoptosis in human MTC cell line, TT, by inversely modulating two molecular pathways: (i) stalling PI3K/Akt/Bad pathway that resulted in mitochondrial transmembrane potential (MTP) loss, cytochrome-c release and activation of the executioner caspases-9 and -3, and (ii) upholding p38MAPK/caspase-8/caspase-3 pathway via inhibition of Ras/Raf/ERK. Over-expression of either constitutively active myristoylated-Akt-cDNA (Myr-Akt-cDNA) or dominant-negative-caspase-8-cDNA (Dn-caspase-8-cDNA) partially blocked theaflavin-induced apoptosis, while co-transfection of Myr-Akt-cDNA and Dn-caspase-8-cDNA completely eradicated the effect of theaflavins thereby negating the possibility of existence of other pathways. A search for the upstream signaling revealed that theaflavin-induced disruption of lipid raft caused interference in anchorage of RET in lipid raft that in turn stalled phosphorylation of Ras and PI3Kinase. In such anti-survival cellular micro-environment, pro-apoptotic signals were triggered to culminate into programmed death of MTC cell. These findings not only unveil a hitherto unexplained mechanism underlying theaflavin-induced MTC death, but also validate RET as a promising and potential target for MTC therapy.


Sujet(s)
Caspase 8/génétique , Phosphatidylinositol 3-kinases/génétique , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-ret/génétique , Tumeurs de la thyroïde/génétique , p38 Mitogen-Activated Protein Kinases/génétique , Antinéoplasiques d'origine végétale/pharmacologie , Apoptose , Biflavonoïdes/pharmacologie , Carcinome neuroendocrine , Caspase 8/métabolisme , Catéchine/pharmacologie , Lignée cellulaire tumorale , Cytochromes c/métabolisme , ADN complémentaire , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Microdomaines membranaires/effets des médicaments et des substances chimiques , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Protéine oncogène p21(ras)/génétique , Protéine oncogène p21(ras)/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-ret/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs de la thyroïde/métabolisme , Tumeurs de la thyroïde/anatomopathologie , Transfection , Protéine Bad/génétique , Protéine Bad/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme
15.
J Biol Chem ; 287(39): 32881-96, 2012 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-22851172

RÉSUMÉ

Multiple mechanisms have been proposed by which tumors induce T cell apoptosis to circumvent tumor immune-surveillance. Although sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) have long been known to regulate intracellular Ca(2+) homeostasis, few studies have examined the role of SERCA in processes of T lymphocyte survival and activation. In this context it remains largely unexplored as to how tumors jeopardize SERCA function to disable T cell-mediated anti-tumor immunity. Here, we show that human CD4(+) T cells in the presence of tumor conditions manifested an up-regulation of SERCA3 expression that resulted in development of endoplasmic reticulum stress leading to CD4(+) T cell apoptosis. Prostaglandin E(2) produced by the tumor cell plays a critical role in up-regulating SERCA3 by enhancing the binding of its transcription factor Sp1. Gene manipulation and pharmacological approaches further established that an increase in SERCA expression also resulted in subsequent inhibition of PKCα and -θ and retention of NFκB in the cytosol; however, down-modulation of SERCA3 expression by a dihydropyrimidone derivative, ethyl-4-(3-nitro)-phenyl-6-methyl-2-oxo-1,2,3,4-tetrahydropyrimidine-5 carboxylate (nifetepimine), protected the CD4(+) T cells from tumor-induced apoptosis. In fact, nifetepimine-mediated restoration of PKC activity resulted in nuclear translocation of p65NFκB, thereby ensuring its survival. Studies further undertaken in a tumor-bearing mice model revalidated the immunoprotective role of nifetepimine. Our present study thus strongly suggests that imbalance in cellular calcium homeostasis is an important factor leading to CD4(+) T cell death during cancer and holds promise that nifetepimine may have the potential to be used as an immunorestoring agent in cancer bearers.


Sujet(s)
Tumeurs du sein/enzymologie , Lymphocytes T CD4+/métabolisme , Calcium/métabolisme , Facteurs immunologiques/pharmacologie , Protéines tumorales/métabolisme , Pyrimidinones/pharmacologie , Sarcoplasmic Reticulum Calcium-Transporting ATPases/biosynthèse , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Animaux , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Lymphocytes T CD4+/immunologie , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Survie cellulaire/immunologie , Dinoprostone/génétique , Dinoprostone/immunologie , Dinoprostone/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/génétique , Stress du réticulum endoplasmique/immunologie , Femelle , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes codant pour des enzymes/génétique , Régulation de l'expression des gènes codant pour des enzymes/immunologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/génétique , Régulation de l'expression des gènes tumoraux/immunologie , Humains , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/génétique , Activation des lymphocytes/immunologie , Mâle , Souris , Protéines tumorales/génétique , Protéines tumorales/immunologie , Transplantation tumorale , Protein kinase C-alpha/génétique , Protein kinase C-alpha/immunologie , Protein kinase C-alpha/métabolisme , Sarcoplasmic Reticulum Calcium-Transporting ATPases/génétique , Sarcoplasmic Reticulum Calcium-Transporting ATPases/immunologie , Facteur de transcription Sp1/génétique , Facteur de transcription Sp1/immunologie , Facteur de transcription Sp1/métabolisme , Transplantation hétérologue , Microenvironnement tumoral/génétique , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/génétique , Régulation positive/immunologie
16.
Front Biosci (Schol Ed) ; 4(1): 300-20, 2012 01 01.
Article de Anglais | MEDLINE | ID: mdl-22202062

RÉSUMÉ

With phytochemicals executing a plethora of anti-tumor mechanisms, targeting the 'guardian angel' p53 appears to be a critical strategy to energize the process of cancer therapeutics. Regulation of anti-tumor p53 functions by dietary plant polyphenols particularly black tea and its active component theaflavins has gained immense recognition from the point of view of both efficacy and safety. This review highlights the complexities of p53 functions, molecular mechanisms of its inactivation in cancer, and therapeutic strategies for rescuing p53 dysfunction in tumors using theaflavins. It describes how theaflavins, by steering a single molecular target - p53, regulate multiple hallmarks of carcinogenesis i.e., tumor glycolysis, angiogenesis, metastasis, apoptosis and drug resistance. Additionally, considering the rising of the current concept of cancer stem cells (CSCs), the sole participant in tumor evolution, the review discusses about the possible role of theaflavin-p53 cross talk in targeting CSCs. Such attempts to target the complexities of p53 functions during neogenesis will be of immense help in developing a "new" strategy for successful cancer prevention and therapy by theaflavins.


Sujet(s)
Biflavonoïdes/pharmacologie , Biflavonoïdes/usage thérapeutique , Catéchine/pharmacologie , Catéchine/usage thérapeutique , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Antioxydants/pharmacologie , Antioxydants/usage thérapeutique , Humains , Tumeurs/anatomopathologie
17.
Front Biosci (Schol Ed) ; 4(2): 502-19, 2012 01 01.
Article de Anglais | MEDLINE | ID: mdl-22202073

RÉSUMÉ

Despite major advances in understanding the mechanisms of tumor immunity, its successful translation into effective tumor immunotherapy is hindered by the ability of tumors to foster a tolerant microenvironment and to activate a plethora of immunosuppressive mechanisms. Among different strategies employed by tumors to thwart immune responses, shedding of immunosuppressive molecules, such as sialic acid-containing glycosphingolipids, gangliosides, by the tumor is one important strategy. Aberrant and elevated expression of gangliosides has been demonstrated on the surface of cancer cells. Here we discuss about the molecular mechanisms underneath the contribution of tumor gangliosides in targeting multiple steps of T cell response. We shall also underscore the contribution of T-regulatory, NK and dendritic cells in this immunosuppressive network. Inhibitory effects of gangliosides ultimately converge to T cell apoptosis in receptor-dependent and -independent manners via IL-2 deprivation, ROS production, cytochrome c release, NFkappaB inhibition and caspase activation. Current wealth of information promises a future scenario in which synchronized blockade of immunosuppressive mechanisms and removal of inhibitory signals might be effective in overcoming immunological tolerance and promoting tumor regression.


Sujet(s)
Gangliosides/immunologie , Tumeurs/immunologie , Lymphocytes T/immunologie , Animaux , Communication cellulaire/immunologie , Humains , Tolérance immunitaire , Immunothérapie , Tumeurs/thérapie
18.
J Biol Chem ; 286(49): 42232-42247, 2011 Dec 09.
Article de Anglais | MEDLINE | ID: mdl-22013068

RÉSUMÉ

Breast cancer cells often develop multiple mechanisms of drug resistance during tumor progression, which is the major reason for the failure of breast cancer therapy. High constitutive activation of NFκB has been found in different cancers, creating an environment conducive for chemotherapeutic resistance. Here we report that doxorubicin-induced SMAR1-dependent transcriptional repression and SMAR1-independent degradation of IkBα resulted in nuclear translocation of p65NFκB and its association with p300 histone acetylase and subsequent transcription of Bcl-2 to impart protective response in drug-resistant cells. Consistently SMAR1-silenced drug-resistant cells exhibited IkBα-mediated inhibition of p65NFκB and induction of p53-dependent apoptosis. Interestingly, curcumin pretreatment of drug-resistant cells alleviated SMAR1-mediated p65NFκB activation and hence restored doxorubicin sensitivity. Under such anti-survival condition, induction of p53-p300 cross-talk enhanced the transcriptional activity of p53 and intrinsic death cascade. Importantly, promyelocyte leukemia-mediated SMAR1 sequestration that relieved the repression of apoptosis-inducing genes was indispensable for such chemo-sensitizing ability of curcumin. A simultaneous decrease in drug-induced systemic toxicity by curcumin might also have enhanced the efficacy of doxorubicin by improving the intrinsic defense machineries of the tumor-bearer. Overall, the findings of this preclinical study clearly demonstrate the effectiveness of curcumin to combat doxorubicin-resistance. We, therefore, suggest curcumin as a potent chemo-sensitizer to improve the therapeutic index of this widely used anti-cancer drug. Taken together, these results suggest that curcumin can be developed into an adjuvant chemotherapeutic drug.


Sujet(s)
Antinéoplasiques/pharmacologie , Curcumine/métabolisme , Protéine p300-E1A/métabolisme , Tumeurs mammaires de l'animal/traitement médicamenteux , Tumeurs mammaires de l'animal/génétique , Facteur de transcription RelA/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Transport nucléaire actif , Animaux , Apoptose , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Altération de l'ADN , Protéines de liaison à l'ADN/métabolisme , Doxorubicine/pharmacologie , Résistance aux médicaments antinéoplasiques , Femelle , Souris , Transplantation tumorale , Protéines nucléaires/métabolisme , Petit ARN interférent/métabolisme
19.
Cell Mol Immunol ; 7(4): 306-15, 2010 Jul.
Article de Anglais | MEDLINE | ID: mdl-20305684

RÉSUMÉ

Immune dysfunction is well documented during tumor progression and likely contributes to tumor immune evasion. CD8(+) cytotoxic T lymphocytes (CTLs) are involved in antigen-specific tumor destruction and CD4(+) T cells are essential for helping this CD8(+) T cell-dependent tumor eradication. Tumors often target and inhibit T-cell function to escape from immune surveillance. This dysfunction includes loss of effector and memory T cells, bias towards type 2 cytokines and expansion of T regulatory (Treg) cells. Curcumin has previously been shown to have antitumor activity and some research has addressed the immunoprotective potential of this plant-derived polyphenol in tumor-bearing hosts. Here we examined the role of curcumin in the prevention of tumor-induced dysfunction of T cell-based immune responses. We observed severe loss of both effector and memory T-cell populations, downregulation of type 1 and upregulation of type 2 immune responses and decreased proliferation of effector T cells in the presence of tumors. Curcumin, in turn, prevented this loss of T cells, expanded central memory T cell (T(CM))/effector memory T cell (T(EM)) populations, reversed the type 2 immune bias and attenuated the tumor-induced inhibition of T-cell proliferation in tumor-bearing hosts. Further investigation revealed that tumor burden upregulated Treg cell populations and stimulated the production of the immunosuppressive cytokines transforming growth factor (TGF)-beta and IL-10 in these cells. Curcumin, however, inhibited the suppressive activity of Treg cells by downregulating the production of TGF-beta and IL-10 in these cells. More importantly, curcumin treatment enhanced the ability of effector T cells to kill cancer cells. Overall, our observations suggest that the unique properties of curcumin may be exploited for successful attenuation of tumor-induced suppression of cell-mediated immune responses.


Sujet(s)
Immunité acquise/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Curcumine/pharmacologie , Tumeurs/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Animaux , Antigènes CD4/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytotoxicité immunologique/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Tests de criblage d'agents antitumoraux , Facteurs de transcription Forkhead/métabolisme , Mémoire immunologique/effets des médicaments et des substances chimiques , Interleukine-10/métabolisme , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Déplétion lymphocytaire , Souris , Tumeurs/anatomopathologie , Analyse de survie , Lymphocytes T auxiliaires/effets des médicaments et des substances chimiques , Lymphocytes T auxiliaires/immunologie , Lymphocytes T auxiliaires/anatomopathologie , Lymphocytes T régulateurs/cytologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Facteur de croissance transformant bêta/métabolisme
20.
FEBS Lett ; 584(1): 7-14, 2010 Jan 04.
Article de Anglais | MEDLINE | ID: mdl-19883646

RÉSUMÉ

The present study demonstrates that theaflavins exploit p53 to impede metastasis in human breast cancer cells. Our data suggest that p53-dependent reactive oxygen species (ROS) induce p53-phosphorylation via p38MAPK in a feedback loop to inhibit IkappaBalpha-phosphorylation and NF-kappaB/p65 nuclear translocation, thereby down-regulating the metastatic proteins metalloproteinase (MMP)-2 and MMP-9. When wild-type p53-expressing MCF-7 cells are transfected with p53 short-interfering RNA, or treated with a pharmacological inhibitor of ROS, theaflavins fail to inhibit NF-kappaB-mediated cell migration. On the other hand, NF-kappaB over-expression bestows MCF-7 cells with resistance to the anti-migratory effect of theaflavins. These results indicate that inhibition of NF-kappaB via p53-ROS crosstalk is a pre-requisite for theaflavins to accomplish the anti-migratory effect in breast cancer cells.


Sujet(s)
Antioxydants/pharmacologie , Biflavonoïdes/pharmacologie , Tumeurs du sein/anatomopathologie , Carcinomes/secondaire , Catéchine/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Protéine p53 suppresseur de tumeur/antagonistes et inhibiteurs , Transport nucléaire actif/effets des médicaments et des substances chimiques , Tumeurs du sein/métabolisme , Carcinomes/métabolisme , Lignée cellulaire tumorale , Noyau de la cellule/métabolisme , Femelle , Humains , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 9/métabolisme , Inhibiteurs de métalloprotéinases matricielles , Facteur de transcription NF-kappa B/métabolisme , Métastase tumorale , Espèces réactives de l'oxygène/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE