Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Int J Mol Sci ; 24(8)2023 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-37108498

RÉSUMÉ

A highly stereo-selective, one-pot, multicomponent method was chosen to synthesize the novel functionalized 1, 3-cycloaddition spirooxindoles (SOXs) (4a-4h). Synthesized SOXs were analyzed for their drug-likeness and ADME parameters and screened for their anticancer activity. Our molecular docking analysis revealed that among all derivatives of SOXs (4a-4h), 4a has a substantial binding affinity (∆G) -6.65, -6.55, -8.73, and -7.27 Kcal/mol with CD-44, EGFR, AKR1D1, and HER-2, respectively. A functional study demonstrated that SOX 4a has a substantial impact on human cancer cell phenotypes exhibiting abnormality in cytoplasmic and nuclear architecture as well as granule formation leading to cell death. SOX 4a treatment robustly induced reactive oxygen species (ROS) generation in cancer cells as observed by enhanced DCFH-DA signals. Overall, our results suggest that SOX (4a) targets CD-44, EGFR, AKR1D1, and HER-2 and induces ROS generation in cancer cells. We conclude that SOX (4a) could be explored as a potential chemotherapeutic molecule against various cancers in appropriate pre-clinical in vitro and in vivo model systems.


Sujet(s)
Antinéoplasiques , Humains , Structure moléculaire , Relation structure-activité , Antinéoplasiques/composition chimique , Simulation de docking moléculaire , Espèces réactives de l'oxygène , Récepteurs ErbB/métabolisme
2.
Photochem Photobiol ; 99(2): 874-877, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36151964

RÉSUMÉ

The UVB irradiation is well known for its impact on the development of skin cancer. However, low UVB irradiation plays a protective role against various human diseases including cancer through its effect on tumor suppression. This article summarizes the key findings of the paper by Park et al., which describes a novel molecular mechanism of moderate UVB irradiation in suppressing the growth of melanoma and colorectal cancer. Key observations in this article are that moderate UVB irradiation can enhance tumor immunity by (1) increased infiltration of CD4+ and CD8+ T cells; (2) increased infiltration of CD103+ conventional type 1 dendritic cells (cDC1); and (3) a significant decrease of M2 tumor associate macrophages (TAMs) into the tumor. The authors further identified the role of Batf3 transcription factor in moderate UVB irradiation-mediated anti-tumor immune response. Deletion of Batf3 transcription factor reversed the tumor suppressive effect with decreased CD103+ cDC1 cell infiltration. This pre-clinical study provides a very novel mechanistic insight into the utilization of moderate UVB irradiation for the management of melanoma and colorectal cancer. This study further provides the direction of new future research to explore moderate UVB irradiation in combination with checkpoint blockade antibodies to enhance immunotherapeutic response against various solid tumors.


Sujet(s)
Tumeurs colorectales , Mélanome , Tumeurs cutanées , Humains , Lymphocytes T CD8+/anatomopathologie , Tumeurs cutanées/anatomopathologie , Rayons ultraviolets , Facteurs de transcription , Tumeurs colorectales/radiothérapie
3.
Photochem Photobiol ; 99(2): 344-355, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36029171

RÉSUMÉ

Sunlight exposure is a significant risk factor for UV-induced deteriorating transformations of epidermal homeostasis leading to skin carcinogenesis. The ability of UVB radiation to cause melanoma, as well as basal and squamous cell carcinomas, makes UVB the most harmful among the three known UV ranges. UVB-induced DNA mutations and dysregulation of signaling pathways contribute to skin cancer formation. Among various signaling pathways modulated by UVB, tyrosine phosphorylation signaling which is mediated by the action of protein tyrosine kinases (PTKs) on specific tyrosine residues is highly implicated in photocarcinogenesis. Following UVB irradiation, PTKs get activated and their downstream signaling pathways contribute to photocarcinogenesis by promoting the survival of damaged keratinocytes and increasing cell proliferation. While UVB activates oncogenic signaling pathways, it can also activate tumor suppressive signaling pathways as initial protective mechanisms to maintain epidermal homeostasis. Tyrosine dephosphorylation is one of the protective mechanisms and is mediated by the action of protein tyrosine phosphatases (PTPs). PTP can counteract UVB-mediated PTK activation and downregulate oncogenic signaling pathways. However, PTPs have not been studied extensively in photocarcinogenesis with previous studies regarding their inactivation induced by UVB. This current review will summarize the recent progress in the protective function of PTPs in epidermal photocarcinogenesis.


Sujet(s)
Tumeurs cutanées , Rayons ultraviolets , Humains , Phosphorylation , Kératinocytes/effets des radiations , Protein Tyrosine Phosphatases/métabolisme , Carcinogenèse , Tumeurs cutanées/étiologie , Tumeurs cutanées/métabolisme , Tyrosine/métabolisme
4.
Life (Basel) ; 12(3)2022 Mar 07.
Article de Anglais | MEDLINE | ID: mdl-35330134

RÉSUMÉ

Multidrug resistance (MDR) is one of the major therapeutic challenges that limits the efficacy of chemotherapeutic response resulting in poor prognosis of ovarian cancer (OC). The multidrug resistance protein 1 (MRP1) is a membrane-bound ABC transporter involved in cross resistance to many structurally and functionally diverse classes of anticancer drugs including doxorubicin, taxane, and platinum. In this study, we utilize homology modelling and molecular docking analysis to determine the binding affinity and the potential interaction sites of MRP1 with Carboplatin, Gemcitabine, Doxorubicin, Paclitaxel, and Topotecan. We used AutoDock Vina scores to compare the binding affinities of the anticancer drugs against MRP1. Our results depicted Carboplatin < Gemcitabine < Topotecan < Doxorubicin < Paclitaxel as the order of binding affinities. Paclitaxel has shown the highest binding affinity whereas Carboplatin displayed the lowest affinity to MRP1. Interestingly, our data showed that Carboplatin, Paclitaxel, and Topotecan bind specifically to Asn510 residue in the transmembrane domains 1 of the MRP1. Our results suggest that Carboplatin could be an appropriate therapeutic choice against MRP1 in OC as it couples weakly with Carboplatin. Further, our findings also recommend opting Carboplatin with Gemcitabine as a combinatorial chemotherapeutic approach to overcome MDR phenotype associated with recurrent OC.

5.
Semin Cancer Biol ; 86(Pt 3): 1175-1185, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35189322

RÉSUMÉ

Tumor heterogeneity is a predominant feature of hepatocellular carcinoma (HCC) that plays a crucial role in chemoresistance and limits the efficacy of available chemo/immunotherapy regimens. Thus, a better understanding regarding the molecular determinants of tumor heterogeneity will help in developing newer strategies for effective HCC management. Chemokines, a sub-family of cytokines are one of the key molecular determinants of tumor heterogeneity in HCC and are involved in cell survival, growth, migration, and angiogenesis. Herein, we provide a panoramic insight into the role of chemokines in HCC heterogeneity at genetic, epigenetic, metabolic, immune cell composition, and tumor microenvironment levels and its impact on clinical outcomes. Interestingly, our in-silico analysis data showed that expression of chemokine receptors impacts infiltration of various immune cell populations into the liver tumor and leads to heterogeneity. Thus, it is evident that aberrant chemokines clouding impacts HCC tumor heterogeneity and understanding this phenomenon in depth could be harnessed for the development of personalized medicine strategies in future.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Humains , Tumeurs du foie/génétique , Tumeurs du foie/thérapie , Tumeurs du foie/métabolisme , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/métabolisme , Chimiokines/métabolisme , Microenvironnement tumoral/génétique , Néovascularisation pathologique
6.
Nanomedicine ; 20: 102027, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31170509

RÉSUMÉ

Pancreatic cancer (PanCa) is a major cause of cancer-related death due to limited therapeutic options. As pancreatic tumors are highly desmoplastic, they prevent appropriate uptake of therapeutic payloads. Thus, our objective is to develop a next-generation nanoparticle system for treating PanCa. We generated a multi-layered Pluronic F127 and polyvinyl alcohol stabilized and poly-L-lysine coated paclitaxel loaded poly(lactic-co-glycolic acid) nanoparticle formulation (PPNPs). This formulation exhibited optimal size (~160 nm) and negative Zeta potential (-6.02 mV), efficient lipid raft mediated internalization, pronounced inhibition in growth and metastasis in vitro, and in chemo-naïve and chemo-exposed orthotopic xenograft mouse models. Additionally, PPNPs altered nanomechanical properties of PanCa cells as suggested by the increased elastic modulus in nanoindentation analyses. Immunohistochemistry of orthotopic tumors demonstrated decreased expression of tumorigenic and metastasis associated proteins (ki67, vimentin and slug) in PPNPs treated mice. These results suggest that PPNPs represent a viable and robust platform for (PanCa).


Sujet(s)
Nanoparticules/composition chimique , Paclitaxel/usage thérapeutique , Tumeurs du pancréas/traitement médicamenteux , Animaux , Apoptose/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Endocytose , Humains , Microdomaines membranaires/métabolisme , Souris nude , Invasion tumorale , Métastase tumorale , Paclitaxel/pharmacologie , Tumeurs du pancréas/anatomopathologie , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Biomaterials ; 208: 83-97, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-30999154

RÉSUMÉ

Pancreatic cancer is a complex disease accounting for fibrotic tumors and an aggressive phenotype. Gemcitabine (GEM) is used as a standard therapy, which develops chemoresistance leading to poor patient outcome. We have recently developed a superparamagnetic iron oxide nanoparticle (SPION) formulation of curcumin (SP-CUR), which is a nontoxic, bioactive anti-inflammatory/anti-cancer agent for its enhanced delivery in tumors. In this study, we demonstrate that SP-CUR effectively delivers bioactive curcumin to pancreatic tumors, simultaneously enhances GEM uptake and its efficacy. Mechanistic revelations suggest that SP-CUR targets tumor microenvironment via suppression of sonic hedgehog (SHH) pathway and an oncogenic CXCR4/CXCL12 signaling axis that inhibits bidirectional tumor-stromal cells interaction. Increased GEM uptake was observed due to upregulation of the human nucleoside transporter genes (DCK, hCNT) and blocking ribonucleotide reductase subunits (RRM1/RRM2). Additionally, co-treatment of SP-CUR and GEM targets cancer stem cells by regulating pluripotency maintaining stemness factors (Nanog, Sox2, c-Myc and Oct-4), and restricting tumor sphere formation. In an orthotopic mouse model, an enhanced accumulation of SP-CUR was found in pancreas, which potentiated GEM to reduce tumor growth and metastasis. Analysis of tumor tissues suggest that the treatment inhibits tumor stroma (α-SMA, Desmin and Hyluronic Acid) and induces changes in cell stiffness, as measured via Atomic Force Microscopy. This was accompanied by alteration of key cellular proteins of SHH signaling such as SHH, Gli-1, Gli-2, Sufu, and NFĸB-65 as indicated by Immunoblotting and Immunohistochemistry. These results suggest that SP-CUR has a great potential for future clinical use in the management of pancreatic cancer.


Sujet(s)
Curcumine/composition chimique , Curcumine/usage thérapeutique , Désoxycytidine/analogues et dérivés , Composés du fer III/composition chimique , Nanoparticules de magnétite/composition chimique , Nanoparticules/composition chimique , Tumeurs du pancréas/traitement médicamenteux , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Chimiokine CXCL12/métabolisme , Désoxycytidine/composition chimique , Désoxycytidine/usage thérapeutique , Test ELISA , Humains , Souris , Microscopie confocale , Tumeurs du pancréas/métabolisme , Récepteurs CXCR4/métabolisme ,
8.
J Exp Clin Cancer Res ; 38(1): 29, 2019 Jan 23.
Article de Anglais | MEDLINE | ID: mdl-30674344

RÉSUMÉ

BACKGROUND: The management of pancreatic cancer (PanCa) is exceptionally difficult due to poor response to available therapeutic modalities. Tubulins play a major role in cell dynamics, thus are important molecular targets for cancer therapy. Among various tubulins, ßIII and ßIV-tubulin isoforms have been primarily implicated in PanCa progression, metastasis and chemo-resistance. However, specific inhibitors of these isoforms that have potent anti-cancer activity with low toxicity are not readily available. METHODS: We determined anti-cancer molecular mechanisms and therapeutic efficacy of a novel small molecule inhibitor (VERU-111) using in vitro (MTS, wound healing, Boyden chamber and real-time xCELLigence assays) and in vivo (xenograft studies) models of PanCa. The effects of VERU-111 treatment on the expression of ß-tubulin isoforms, apoptosis, cancer markers and microRNAs were determined by Western blot, immunohistochemistry (IHC), confocal microscopy, qRT-PCR and in situ hybridization (ISH) analyses. RESULTS: We have identified a novel small molecule inhibitor (VERU-111), which preferentially represses clinically important, ßIII and ßIV tubulin isoforms via restoring the expression of miR-200c. As a result, VERU-111 efficiently inhibited tumorigenic and metastatic characteristics of PanCa cells. VERU-111 arrested the cell cycle in the G2/M phase and induced apoptosis in PanCa cell lines via modulation of cell cycle regulatory (Cdc2, Cdc25c, and Cyclin B1) and apoptosis - associated (Bax, Bad, Bcl-2, and Bcl-xl) proteins. VERU-111 treatment also inhibited tumor growth (P < 0.01) in a PanCa xenograft mouse model. CONCLUSIONS: This study has identified an inhibitor of ßIII/ßIV tubulins, which appears to have excellent potential as monotherapy or in combination with conventional therapeutic regimens for PanCa treatment.


Sujet(s)
Carcinogenèse/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs du pancréas/traitement médicamenteux , Modulateurs de la polymérisation de la tubuline/administration et posologie , Tubuline/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Humains , Souris , Métastase tumorale , Paclitaxel/administration et posologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tubuline/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Br J Cancer ; 118(4): 587-599, 2018 02 20.
Article de Anglais | MEDLINE | ID: mdl-29465084

RÉSUMÉ

BACKGROUND: Cancer progression and metastasis is profoundly influenced by protein kinase D1 (PKD1) and metastasis-associated protein 1 (MTA1) in addition to other pathways. However, the nature of regulatory relationship between the PKD1 and MTA1, and its resulting impact on cancer metastasis remains unknown. Here we present evidence to establish that PKD1 is an upstream regulatory kinase of MTA1. METHODS: Protein and mRNA expression of MTA1 in PKD1-overexpressing cells were determined using western blotting and reverse-transcription quantitative real-time PCR. Immunoprecipitation and proximity ligation assay (PLA) were used to determine the interaction between PKD1 and MTA1. PKD1-mediated nucleo-cytoplasmic export and polyubiquitin-dependent proteosomal degradation was determined using immunostaining. The correlation between PKD1 and MTA1 was determined using intra-tibial, subcutaneous xenograft, PTEN-knockout (PTEN-KO) and transgenic adenocarcinoma of mouse prostate (TRAMP) mouse models, as well as human cancer tissues. RESULTS: We found that MTA1 is a PKD1-interacting substrate, and that PKD1 phosphorylates MTA1, supports its nucleus-to-cytoplasmic redistribution and utilises its N-terminal and kinase domains to effectively inhibit the levels of MTA1 via polyubiquitin-dependent proteosomal degradation. PKD1-mediated downregulation of MTA1 was accompanied by a significant suppression of prostate cancer progression and metastasis in physiologically relevant spontaneous tumour models. Accordingly, progression of human prostate tumours to increased invasiveness was also accompanied by decreased and increased levels of PKD1 and MTA1, respectively. CONCLUSIONS: Overall, this study, for the first time, establishes that PKD1 is an upstream regulatory kinase of MTA1 status and its associated metastatic activity, and that the PKD1-MTA1 axis could be targeted for anti-cancer strategies.


Sujet(s)
Histone deacetylases/génétique , Histone deacetylases/métabolisme , Tumeurs de la prostate/anatomopathologie , Protéines de répression/génétique , Protéines de répression/métabolisme , Canaux cationiques TRPP/génétique , Canaux cationiques TRPP/métabolisme , Animaux , Lignée cellulaire tumorale , Noyau de la cellule/métabolisme , Cytoplasme/métabolisme , Techniques de knock-out de gènes , Humains , Cellules MCF-7 , Mâle , Souris , Métastase tumorale , Transplantation tumorale , Phosphohydrolase PTEN/génétique , Phosphorylation , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Transactivateurs
10.
Mol Cancer Ther ; 16(10): 2267-2280, 2017 10.
Article de Anglais | MEDLINE | ID: mdl-28615299

RÉSUMÉ

Ormeloxifene is a clinically approved selective estrogen receptor modulator, which has also shown excellent anticancer activity, thus it can be an ideal repurposing pharmacophore. Herein, we report therapeutic effects of ormeloxifene on prostate cancer and elucidate a novel molecular mechanism of its anticancer activity. Ormeloxifene treatment inhibited epithelial-to-mesenchymal transition (EMT) process as evident by repression of N-cadherin, Slug, Snail, vimentin, MMPs (MMP2 and MMP3), ß-catenin/TCF-4 transcriptional activity, and induced the expression of pGSK3ß. In molecular docking analysis, ormeloxifene showed proficient docking with ß-catenin and GSK3ß. In addition, ormeloxifene induced apoptosis, inhibited growth and metastatic potential of prostate cancer cells and arrested cell cycle in G0-G1 phase via modulation of cell-cycle regulatory proteins (inhibition of Mcl-1, cyclin D1, and CDK4 and induction of p21 and p27). In functional assays, ormeloxifene remarkably reduced tumorigenic, migratory, and invasive potential of prostate cancer cells. In addition, ormeloxifene treatment significantly (P < 0.01) regressed the prostate tumor growth in the xenograft mouse model while administered through intraperitoneal route (250 µg/mouse, three times a week). These molecular effects of ormeloxifene were also observed in excised tumor tissues as shown by immunohistochemistry analysis. Our results, for the first time, demonstrate repurposing potential of ormeloxifene as an anticancer drug for the treatment of advanced stage metastatic prostate cancer through a novel molecular mechanism involving ß-catenin and EMT pathway. Mol Cancer Ther; 16(10); 2267-80. ©2017 AACR.


Sujet(s)
Benzopyranes/administration et posologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs de la prostate/traitement médicamenteux , bêta-Caténine/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Benzopyranes/effets indésirables , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Humains , Mâle , Souris , Simulation de docking moléculaire , Métastase tumorale , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , bêta-Caténine/composition chimique
11.
Sci Rep ; 6: 36594, 2016 11 08.
Article de Anglais | MEDLINE | ID: mdl-27824155

RÉSUMÉ

In this study, we for the first time, investigated the potential anti-cancer effects of a novel analogue of cucurbitacin (Cucurbitacin D) against cervical cancer in vitro and in vivo. Cucurbitacin D inhibited viability and growth of cervical cancer cells (CaSki and SiHa) in a dose-dependent manner. IC50 of Cucurbitacin D was recorded at 400 nM and 250 nM in CaSki and SiHa cells, respectively. Induction of apoptosis was observed in Cucurbitacin D treated cervical cancer cells as measured by enhanced Annexin V staining and cleavage in PARP protein. Cucurbitacin D treatment of cervical cancer cells arrested the cell cycle in G1/S phase, inhibited constitutive expression of E6, Cyclin D1, CDK4, pRb, and Rb and induced the protein levels of p21 and p27. Cucurbitacin D also inhibited phosphorylation of STAT3 at Ser727 and Tyr705 residues as well as its downstream target genes c-Myc, and MMP9. Cucurbitacin D enhanced the expression of tumor suppressor microRNAs (miR-145, miRNA-143, and miRNA34a) in cervical cancer cells. Cucurbitacin D treatment (1 mg/kg body weight) effectively inhibited growth of cervical cancer cells derived orthotopic xenograft tumors in athymic nude mice. These results demonstrate the potential therapeutic efficacy of Cucurbitacin D against cervical cancer.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Points de contrôle de la phase G1 du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle de la phase S du cycle cellulaire/effets des médicaments et des substances chimiques , Triterpènes/pharmacologie , Tumeurs du col de l'utérus/traitement médicamenteux , Animaux , Lignée cellulaire tumorale , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Souris nude , Protéines tumorales/biosynthèse , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Oncotarget ; 7(22): 33069-80, 2016 May 31.
Article de Anglais | MEDLINE | ID: mdl-27102301

RÉSUMÉ

PKCε is a transforming oncogene and a predictive biomarker of various human cancers. However, a precise in vivo link of PKCε to cancer induction, progression and metastasis remain undefined. To achieve these goals, we generated tissue specific conditional PKCε knockout mice (PKCε-CKO) using cre-lox technology. Homozygous PKCε(LoxP/LoxP) mice have normal body weight and phenotype. To determine what effect loss of PKCε would have on the prostate, the PKCε(LoxP/LoxP) mice were bred to probasin cre (PB-Cre4+) mice which express cre specifically in the prostate epithelium of postnatal mice. Western blot and immunohistochemical analyses showed reduced levels of PKCε specifically in the prostate of PKCε-CKO mice. Histopathological analyses of prostate from both PKCε(LoxP/LoxP) and prostate PKCε-CKO mice showed normal pathology. To determine the functional impact of prostate specific deletion of PKCε on prostate tumor growth, we performed an orthotopic xenograft study. Transgenic adenocarcinoma of the mouse prostate (TRAMP) cells (TRAMPC1, 2×106) were implanted in the prostate of PKCε-CKO mice. Mice were sacrificed at 6th week post-implantation. Results demonstrated a significant (P<0.05) decrease in the growth of TRAMPC1 cells-derived xenograft tumors in PKCε-CKO mice compared to wild type. To determine a link of PKCε to ultraviolet radiation (UVR) exposure-induced epidermal Stat3 phosphorylation, PKCε(LoxP/LoxP) mice were bred to tamoxifen-inducible K14 Cre mice. PKCε deletion in the epidermis resulted in inhibition of UVR-induced Stat3 phosphorylation. In summary, our novel PKCε(LoxP/LoxP) mice will be useful for defining the link of PKCε to various cancers in specific organ, tissue, or cells.


Sujet(s)
Tumeurs de la prostate/génétique , Protein kinase C-epsilon/métabolisme , Animaux , Évolution de la maladie , Femelle , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Tumeurs de la prostate/anatomopathologie , Protein kinase C-epsilon/déficit , Protein kinase C-epsilon/génétique
13.
Oncotarget ; 7(14): 17945-56, 2016 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-26918454

RÉSUMÉ

Chronic exposure to ultraviolet radiation (UVR) is linked to the development of cutaneous squamous cell carcinoma (SCC), a non-melanoma form of skin cancer that can metastasize. Tumor necrosis factor-alpha (TNFα), a pro-inflammatory cytokine, is linked to UVR-induced development of SCC. To find clues about the mechanisms by which TNFα may promote UVR-induced development of SCC, we investigated changes in the expression profiling of microRNAs (miRNA), a novel class of short noncoding RNAs, which affects translation and stability of mRNAs. In this experiment, TNFα knockout (TNFα KO) mice and their wild type (WT) littermates were exposed to acute UVR (2.0 kJ/m2) and the expression profiling of epidermal miRNA was determined 4hr post UVR exposure. TNFα deletion in untreated WT mice resulted in differential expression (log fold change>1) of seventeen miRNA. UVR exposure in WT mice induced differential expression of 22 miRNA. However, UVR exposure in TNFα KO mice altered only two miRNAs. Four miRNA, were differentially expressed between WT+UVR and TNFα KO+UVR groups. Differentially expressed selected miRNAs were further validated using real time PCR. Few of the differentially expressed miRNAs (miR-31-5p, miR-196a-5p, miR-127-3p, miR-206-3p, miR-411-5p, miR-709, and miR-322-5p) were also observed in UVR-induced SCC. Finally, bio-informatics analysis using DIANA, MIRANDA, Target Scan, and miRDB algorithms revealed a link with major UVR-induced pathways (MAPK, PI3K-Akt, transcriptional mis-regulation, Wnt, and TGF-beta).


Sujet(s)
Carcinome épidermoïde/étiologie , Épiderme/métabolisme , microARN/biosynthèse , Lésions radiques expérimentales/étiologie , Lésions radiques expérimentales/métabolisme , Tumeurs cutanées/étiologie , Facteur de nécrose tumorale alpha/biosynthèse , Rayons ultraviolets/effets indésirables , Animaux , Carcinome épidermoïde/génétique , Carcinome épidermoïde/métabolisme , Épiderme/effets des radiations , Souris , Souris knockout , microARN/génétique , Lésions radiques expérimentales/génétique , Tumeurs cutanées/génétique , Tumeurs cutanées/métabolisme
14.
Cancer Prev Res (Phila) ; 8(5): 375-86, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25627799

RÉSUMÉ

Prostate cancer continues to remain the most common cancer and the second leading cause of cancer-related deaths in American males. The Pten deletions and/or mutations are frequently observed in both primary prostate cancers and metastatic prostate tissue samples. Pten deletion in prostate epithelium in mice results in prostatic intraepithelial neoplasia (PIN), followed by progression to invasive adenocarcinoma. The Pten conditional knockout mice [(Pten-loxp/loxp:PB-Cre4(+)) (Pten-KO)] provide a unique preclinical model to evaluate agents for efficacy for both the prevention and treatment of prostate cancer. We present here for the first time that dietary plumbagin, a medicinal plant-derived naphthoquinone (200 or 500 ppm) inhibits tumor development in intact as well as castrated Pten-KO mice. Plumbagin has shown no signs of toxicity at either of these doses. Plumbagin treatment resulted in a decrease expression of PKCε, AKT, Stat3, and COX2 compared with the control mice. Plumbagin treatment also inhibited the expression of vimentin and slug, the markers of epithelial-to-mesenchymal transition (EMT) in prostate tumors. In summary, the results indicate that dietary plumbagin inhibits growth of both primary and castration-resistant prostate cancer (CRPC) in Pten-KO mice, possibly via inhibition of PKCε, Stat3, AKT, and EMT markers (vimentin and slug), which are linked to the induction and progression of prostate cancer.


Sujet(s)
Adénocarcinome/prévention et contrôle , Antinéoplasiques d'origine végétale/pharmacologie , Carcinogenèse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Naphtoquinones/pharmacologie , Tumeurs de la prostate/prévention et contrôle , Adénocarcinome/anatomopathologie , Animaux , Antinéoplasiques d'origine végétale/usage thérapeutique , Marqueurs biologiques/métabolisme , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Transition épithélio-mésenchymateuse/génétique , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Naphtoquinones/usage thérapeutique , Orchidectomie , Phosphohydrolase PTEN/génétique , Tumeur intraépithéliale prostate/traitement médicamenteux , Tumeur intraépithéliale prostate/anatomopathologie , Tumeurs de la prostate/anatomopathologie , Protein kinase C-epsilon/métabolisme , Facteur de transcription STAT-3/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique
15.
J Invest Dermatol ; 135(4): 1098-1107, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25337691

RÉSUMÉ

We present here that heat-shock protein 90 (Hsp90) inhibitor 17-(allylamino)-17-demethoxygeldanamycin (17AAG), when topically applied to mouse skin, inhibits UVR-induced development of cutaneous squamous cell carcinoma (SCC). In these experiments, DMSO:acetone (1:40 v/v) solution of 17AAG (500 nmol) was applied topically to mouse skin in conjunction with each UVR exposure (1.8 kJ m(-2)). The UVR source was Kodacel-filtered FS-40 sun lamps (approximately 60% UVB and 40% UVA). In independent experiments with three separate mouse lines (SKH-1 hairless mice, wild-type FVB, and protein kinase C epsilon (PKCɛ)-overexpressing transgenic FVB mice), 17AAG treatment increased the latency and decreased both the incidence and multiplicity of UVR-induced SCC. Topical 17AAG alone or in conjunction with UVR treatments elicited neither skin nor systemic toxicity. 17AAG-caused inhibition of SCC induction was accompanied by a decrease in UVR-induced (1) hyperplasia, (2) Hsp90ß-PKCɛ interaction, and (3) expression levels of Hsp90ß, Stat3, pStat3Ser727, pStat3Tyr705, pAktSer473, and matrix metalloproteinase (MMP). The results presented here indicate that topical Hsp90 inhibitor 17AAG is effective in prevention of UVR-induced epidermal hyperplasia and SCC. One may conclude from the preclinical data presented here that topical 17AAG may be useful for prevention of UVR-induced inflammation and cutaneous SCC either developed in UVR-exposed or organ transplant population.


Sujet(s)
Benzoquinones/pharmacologie , Protéines du choc thermique HSP90/métabolisme , Lactames macrocycliques/pharmacologie , Mélanome/métabolisme , Tumeurs radio-induites/métabolisme , Tumeurs cutanées/métabolisme , Acétone/composition chimique , Animaux , Diméthylsulfoxyde/composition chimique , Épiderme/métabolisme , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Humains , Inflammation , Matrix metalloproteinases/métabolisme , Mélanome/prévention et contrôle , Souris , Tumeurs radio-induites/prévention et contrôle , Protein kinase C-epsilon/métabolisme , Peau/effets des médicaments et des substances chimiques , Tumeurs cutanées/traitement médicamenteux , Rayons ultraviolets ,
16.
Antioxid Redox Signal ; 21(5): 682-99, 2014 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-24295217

RÉSUMÉ

AIMS: Pancreatic cancer (PC) is the most aggressive malignant disease, ranking as the fourth most leading cause of cancer-related death among men and women in the United States. In this study, we provide evidence of chemotherapeutic effects of α-mangostin, a dietary antioxidant isolated from the pericarp of Garcinia mangostana L. against human PC. RESULTS: The chemotherapeutic effect of α-mangostin was determined using four human PC cells (PL-45, PANC1, BxPC3, and ASPC1). α-Mangostin resulted in a significant inhibition of PC cells viability without having any effects on normal human pancreatic duct epithelial cells. α-Mangostin showed a dose-dependent increase of apoptosis in PC cells. Also, α-mangostin inhibited the expression levels of pNF-κB/p65Ser552, pStat3Ser727, and pStat3Tyr705. α-Mangostin inhibited DNA binding activity of nuclear factor kappa B (NF-κB) and signal transducer and activator 3 (Stat3). α-Mangostin inhibited the expression levels of matrix metallopeptidase 9 (MMP9), cyclin D1, and gp130; however, increased expression of tissue inhibitor of metalloproteinase 1 (TIMP1) was observed in PC cells. In addition, i.p. administration of α-mangostin (6 mg/kg body weight, 5 days a week) resulted in a significant inhibition of both primary (PL-45) and secondary (ASPC1) human PC cell-derived orthotopic and ectopic xenograft tumors in athymic nude mice. No sign of toxicity was observed in any of the mice administered with α-mangostin. α-Mangostin treatment inhibited the biomarkers of cell proliferation (Ki-67 and proliferating cell nuclear antigen [PCNA]) in the xenograft tumor tissues. INNOVATION: We present, for the first time, that dietary antioxidant α-mangostin inhibits the growth of PC cells in vitro and in vivo. CONCLUSION: These results suggest the potential therapeutic efficacy of α-mangostin against human PC.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Antioxydants/pharmacologie , Modèles animaux de maladie humaine , Garcinia mangostana/composition chimique , Tumeurs du pancréas/traitement médicamenteux , Xanthones/pharmacologie , Animaux , Antinéoplasiques d'origine végétale/administration et posologie , Antinéoplasiques d'origine végétale/isolement et purification , Antioxydants/administration et posologie , Antioxydants/isolement et purification , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Humains , Injections péritoneales , Souris , Souris nude , Tumeurs du pancréas/anatomopathologie , Relation structure-activité , Xanthones/administration et posologie , Xanthones/isolement et purification , Tests d'activité antitumorale sur modèle de xénogreffe
17.
J Skin Cancer ; 2013: 452425, 2013.
Article de Anglais | MEDLINE | ID: mdl-23738074

RÉSUMÉ

To find clues about the mechanism by which kinase C epsilon (PKC ε ) may impart susceptibility to ultraviolet radiation (UVR)-induced development of cutaneous squamous cell carcinomas (SCC), we compared PKC ε transgenic (TG) mice and their wild-type (WT) littermates for (1) the effects of UVR exposures on percent of putative hair follicle stem cells (HSCs) and (2) HSCs proliferation. The percent of double HSCs (CD34+ and α 6-integrin or CD34+/CD49f+) in the isolated keratinocytes were determined by flow cytometric analysis. Both single and chronic UVR treatments (1.8 kJ/m(2)) resulted in an increase in the frequency of double positive HSCs in PKC ε TG mice as compared to their WT littermates. To determine the rate of proliferation of bulge region stem cells, a 5-bromo-2'-deoxyuridine labeling (BrdU) experiment was performed. In the WT mice, the percent of double positive HSCs retaining BrdU label was 28.4 ± 0.6% compared to 4.0 ± 0.06% for the TG mice, an approximately 7-fold decrease. A comparison of gene expression profiles of FACS sorted double positive HSCs showed increased expression of Pes1, Rad21, Tfdp1 and Cks1b genes in TG mice compared to WT mice. Also, PKC ε over expression in mice increased the clonogenicity of isolated keratinocytes, a property commonly ascribed to stem cells.

18.
Mol Oncol ; 7(3): 428-39, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23273564

RÉSUMÉ

We present here first time that Plumbagin (PL), a medicinal plant-derived 1,4-naphthoquinone, inhibits the growth and metastasis of human prostate cancer (PCa) cells in an orthotopic xenograft mouse model. In this study, human PCa PC-3M-luciferase cells (2 × 10(6)) were injected into the prostate of athymic nude mice. Three days post cell implantation, mice were treated with PL (2 mg/kg body wt. i.p. five days in a week) for 8 weeks. Growth and metastasis of PC-3M-luciferase cells was examined weekly by bioluminescence imaging of live mice. PL-treatment significantly (p = 0.0008) inhibited the growth of orthotopic xenograft tumors. Results demonstrated a significant inhibition of metastasis into liver (p = 0.037), but inhibition of metastasis into the lungs (p = 0.60) and lymph nodes (p = 0.27) was not observed to be significant. These results were further confirmed by histopathology of these organs. Results of histopathology demonstrated a significant inhibition of metastasis into lymph nodes (p = 0.034) and lungs (p = 0.028), and a trend to significance in liver (p = 0.075). None of the mice in the PL-treatment group showed PCa metastasis into the liver, but these mice had small metastasis foci into the lymph nodes and lungs. However, control mice had large metastatic foci into the lymph nodes, lungs, and liver. PL-caused inhibition of the growth and metastasis of PC-3M cells accompanies inhibition of the expression of: 1) PKCε, pStat3Tyr705, and pStat3Ser727, 2) Stat3 downstream target genes (survivin and Bcl(xL)), 3) proliferative markers Ki-67 and PCNA, 4) metastatic marker MMP9, MMP2, and uPA, and 5) angiogenesis markers CD31 and VEGF. Taken together, these results suggest that PL inhibits tumor growth and metastasis of human PCa PC3-M-luciferase cells, which could be used as a therapeutic agent for the prevention and treatment of human PCa.


Sujet(s)
Antinéoplasiques d'origine végétale/usage thérapeutique , Naphtoquinones/usage thérapeutique , Métastase tumorale/prévention et contrôle , Plumbaginaceae/composition chimique , Prostate/anatomopathologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Animaux , Antinéoplasiques d'origine végétale/composition chimique , Antinéoplasiques d'origine végétale/isolement et purification , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mâle , Matrix metalloproteinase 2/génétique , Matrix metalloproteinase 9/génétique , Souris , Souris nude , Naphtoquinones/composition chimique , Naphtoquinones/isolement et purification , Métastase tumorale/anatomopathologie , Nitric oxide synthase type II/génétique , Prostate/effets des médicaments et des substances chimiques , Prostate/métabolisme , Tumeurs de la prostate/génétique , Protéine kinase C/génétique , Facteur de transcription STAT-3/génétique
19.
Carcinogenesis ; 33(12): 2586-92, 2012 Dec.
Article de Anglais | MEDLINE | ID: mdl-22976928

RÉSUMÉ

Plumbagin (PL), 5-hydroxy-2-methyl-1,4-naphthoquinone, is a quinoid constituent isolated from the roots of the medicinal plant Plumbago zeylanica L. (also known as chitrak). PL has also been found in Juglans regia (English Walnut), Juglans cinerea (whitenut) and Juglans nigra (blacknut). The roots of P. zeylanica have been used in Indian and Chinese systems of medicine for more than 2500 years for the treatment of various types of ailments. We were the first to report that PL inhibits the growth and invasion of hormone refractory prostate cancer (PCa) cells [Aziz,M.H. et al. (2008) Plumbagin, a medicinal plant-derived naphthoquinone, is a novel inhibitor of the growth and invasion of hormone-refractory prostate cancer. Cancer Res., 68, 9024-9032.]. Now, we present that PL inhibits in vivo PCa development in the transgenic adenocarcinoma of mouse prostate (TRAMP). PL treatment (2 mg/kg body weight i.p. in 0.2 ml phosphate-buffered saline, 5 days a week) to FVB-TRAMP resulted in a significant (P < 0.01) decrease in prostate tumor size and urogenital apparatus weights at 13 and 20 weeks. Histopathological analysis revealed that PL treatment inhibited progression of prostatic intraepithelial neoplasia (PIN) to poorly differentiated carcinoma (PDC). No animal exhibited diffuse tumor formation in PL-treated group at 13 weeks, whereas 75% of the vehicle-treated mice elicited diffuse PIN and large PDC at this stage. At 20 weeks, 25% of the PL-treated animals demonstrated diffuse PIN and 75% developed small PDC, whereas 100% of the vehicle-treated mice showed large PDC. PL treatment inhibited expression of protein kinase C epsilon (PKCε), signal transducers and activators of transcription 3 phosphorylation, proliferating cell nuclear antigen and neuroendocrine markers (synaptophysin and chromogranin-A) in excised prostate tumor tissues. Taken together, these results further suggest PL could be a novel chemopreventive agent against PCa.


Sujet(s)
Adénocarcinome/prévention et contrôle , Anticarcinogènes/usage thérapeutique , Chromogranine A/antagonistes et inhibiteurs , Naphtoquinones/usage thérapeutique , Tumeurs de la prostate/prévention et contrôle , Protein kinase C-epsilon/antagonistes et inhibiteurs , Facteur de transcription STAT-3/antagonistes et inhibiteurs , Synaptophysine/antagonistes et inhibiteurs , Adénocarcinome/anatomopathologie , Animaux , Antigènes transformants de polyomavirus/analyse , Modèles animaux de maladie humaine , Mâle , Souris , Souris transgéniques , Phosphorylation , Antigène nucléaire de prolifération cellulaire/analyse , Tumeurs de la prostate/anatomopathologie
20.
Int J Cancer ; 131(9): 2175-86, 2012 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-22322442

RÉSUMÉ

Pancreatic cancer (PC) is the most aggressive malignant disease, ranks as the fourth most leading cause of cancer-related death among men and women in the United States. We present here that plumbagin (PL), a quinoid constituent isolated from the roots of the medicinal plant Plumbago zeylanica L, inhibits the growth of PC cells both in vitro and in vivo model systems. PL treatment induces apoptosis and inhibits cell viability of PC cells (PANC1, BxPC3 and ASPC1). In addition, i.p. administration of PL (2 mg/kg body weight, 5 days a week) in severe combined immunodeficiency (SCID) mice beginning 3 days after ectopic implantation of PANC1 cells resulted in a significant (P < 0.01) inhibition of both tumor weight and volume. PL treatment inhibited (1) constitutive expression of epidermal growth factor receptor (EGFR), pStat3Tyr705 and pStat3Ser727, (2) DNA binding of Stat3 and (3) physical interaction of EGFR with Stat3, in both cultured PANC1 cells and their xenograft tumors. PL treatment also inhibited phosphorylation and DNA-binding activity of NF-κB in both cultured PC cells (PANC1 and ASPC1) and in PANC1 cells xenograft tumors. Downstream target genes (cyclin D1, MMP9 and Survivin) of Stat3 and NF-κB were similarly inhibited. These results suggest that PL may be used as a novel therapeutic agent against human PC. Published 2012 Wiley-Liss, Inc. This article is a US Government work, and, as such, is in the public domain in the United States of America.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Récepteurs ErbB/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Naphtoquinones/pharmacologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Facteur de transcription STAT-3/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cycline D1/antagonistes et inhibiteurs , Humains , Protéines IAP/antagonistes et inhibiteurs , Mâle , Inhibiteurs de métalloprotéinases matricielles , Souris , Souris SCID , Phosphorylation , Extraits de plantes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Survivine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE