Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 5 de 5
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Mol Med Rep ; 27(4)2023 04.
Article de Anglais | MEDLINE | ID: mdl-36825563

RÉSUMÉ

Signal transducer and activator of transcription 3 (STAT3) signalling serves an important role in carcinogenesis and cellular senescence, and its inhibition in tumour cells represents an attractive therapeutic target. Premature cellular senescence, a process of permanent proliferative arrest of cells in response to various inducers, such as cytostatic drugs or ionizing radiation, is accompanied by morphological and secretory changes, and by altered susceptibility to chemotherapeutic agents, which can thereby complicate their eradication by cancer therapies. In the present study, the responsiveness of proliferating and docetaxel (DTX)­induced senescent cancer cells to small molecule STAT3 inhibitor Stattic and its analogues was evaluated using tumour cell lines. These agents displayed cytotoxic effects in cell viability assays on both proliferating and senescent murine TRAMP­C2 and TC­1 cells; however, senescent cells were markedly more resistant. Western blot analysis revealed that Stattic and its analogues effectively inhibited constitutive STAT3 phosphorylation in both proliferating and senescent cells. Furthermore, whether the Stattic­derived inhibitor K1836 could affect senescence induction or modulate the phenotype of senescent cells was evaluated. K1836 treatment demonstrated no effect on senescence induction by DTX. However, the K1836 compound significantly modulated secretion of certain cytokines (interleukin­6, growth­regulated oncogene α and monocyte chemoattractant protein­1). In summary, the present study demonstrated differences between proliferating and senescent tumour cells in terms of their susceptibility to STAT3 inhibitors and demonstrated the ability of the new STAT3 inhibitor K1836 to affect the secretion of essential components of the senescence­associated secretory phenotype. The present study may be useful for further development of STAT3 inhibitor­based therapy of cancer or age­related diseases.


Sujet(s)
Cytokines , Facteur de transcription STAT-3 , Animaux , Souris , Phosphorylation , Facteur de transcription STAT-3/métabolisme , Expression des gènes , Docetaxel/pharmacologie , Cytokines/métabolisme , Vieillissement de la cellule
2.
J Enzyme Inhib Med Chem ; 36(1): 410-424, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-33440995

RÉSUMÉ

Twelve novel analogs of STAT3 inhibitor BP-1-102 were designed and synthesised with the aim to modify hydrophobic fragments of the molecules that are important for interaction with the STAT3 SH2 domain. The cytotoxic activity of the reference and novel compounds was evaluated using several human and two mouse cancer cell lines. BP-1-102 and its two analogs emerged as effective cytotoxic agents and were further tested in additional six human and two murine cancer cell lines, in all of which they manifested the cytotoxic effect in a micromolar range. Reference compound S3I-201.1066 was found ineffective in all tested cell lines, in contrast to formerly published data. The ability of selected BP-1-102 analogs to induce apoptosis and inhibition of STAT3 receptor-mediated phosphorylation was confirmed. The structure-activity relationship confirmed a demand for two hydrophobic substituents, i.e. the pentafluorophenyl moiety and another spatially bulky moiety, for effective cytotoxic activity and STAT3 inhibition.


Sujet(s)
Acides amino-salicyliques/pharmacologie , Antinéoplasiques/pharmacologie , Conception de médicament , Facteur de transcription STAT-3/antagonistes et inhibiteurs , Sulfonamides/pharmacologie , Acides amino-salicyliques/synthèse chimique , Acides amino-salicyliques/composition chimique , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Relation dose-effet des médicaments , Humains , Interactions hydrophobes et hydrophiles , Souris , Souris de lignée C57BL , Structure moléculaire , Phosphorylation/effets des médicaments et des substances chimiques , Facteur de transcription STAT-3/métabolisme , Relation structure-activité , Sulfonamides/synthèse chimique , Sulfonamides/composition chimique
3.
Int J Oncol ; 53(5): 1997-2009, 2018 Nov.
Article de Anglais | MEDLINE | ID: mdl-30226595

RÉSUMÉ

Cellular senescence is the process of the permanent proliferative arrest of cells in response to various inducers. It is accompanied by typical morphological changes, in addition to the secretion of bioactive molecules, including proinflammatory cytokines and chemokines [known as the senescence-associated secretory phenotype (SASP)]. Thus, senescent cells may affect their local environment and induce a so-called 'bystander' senescence through the state of SASP. The phenotypes of senescent cells are determined by the type of agent inducing cellular stress and the cell lineages. To characterise the phenotypes of senescent cancer cells, two murine cell lines were employed in the present study: TC-1 and B16F10 (B16) cells. Two distinct senescence inductors were used: Chemotherapeutic agent docetaxel (DTX) and a combination of immunomodulatory cytokines, including interferon Î³ (IFNγ) and tumour necrosis factor α (TNFα). It was demonstrated that DTX induced senescence in TC-1 and B16 tumour cell lines, which was demonstrated by growth arrest, positive ß-galactosidase staining, increased p21Waf1 (p21) expression and the typical SASP capable of inducing a 'bystander' senescence. By contrast, treatment with a combination of T helper cell 1 cytokines, IFNγ and TNFα, induced proliferation arrest only in B16 cells. Despite the presence of certain characteristic features resembling senescent cells (proliferation arrest, morphological changes and increased p21 expression), these cells were able to form tumours in vivo and started to proliferate upon cytokine withdrawal. In addition, B16 cells were not able to induce a 'bystander' senescence. In summary, the present study described cell line- and treatment-associated differences in the phenotypes of senescent cells that may be relevant in optimization of cancer chemo- and immunotherapy.


Sujet(s)
Antinéoplasiques/pharmacologie , Effet bystander/immunologie , Vieillissement de la cellule/immunologie , Docetaxel/pharmacologie , Interféron gamma/métabolisme , Tumeurs/immunologie , Facteur de nécrose tumorale alpha/métabolisme , Animaux , Antinéoplasiques/usage thérapeutique , Effet bystander/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Docetaxel/usage thérapeutique , Humains , Interféron gamma/immunologie , Mâle , Souris , Souris de lignée C57BL , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Phénotype , Facteur de nécrose tumorale alpha/immunologie
4.
Aging (Albany NY) ; 10(3): 434-462, 2018 03 28.
Article de Anglais | MEDLINE | ID: mdl-29615539

RÉSUMÉ

Aging involves tissue accumulation of senescent cells (SC) whose elimination through senolytic approaches may evoke organismal rejuvenation. SC also contribute to aging-associated pathologies including cancer, hence it is imperative to better identify and target SC. Here, we aimed to identify new cell-surface proteins differentially expressed on human SC. Besides previously reported proteins enriched on SC, we identified 78 proteins enriched and 73 proteins underrepresented in replicatively senescent BJ fibroblasts, including L1CAM, whose expression is normally restricted to the neural system and kidneys. L1CAM was: 1) induced in premature forms of cellular senescence triggered chemically and by gamma-radiation, but not in Ras-induced senescence; 2) induced upon inhibition of cyclin-dependent kinases by p16INK4a; 3) induced by TGFbeta and suppressed by RAS/MAPK(Erk) signaling (the latter explaining the lack of L1CAM induction in RAS-induced senescence); and 4) induced upon downregulation of growth-associated gene ANT2, growth in low-glucose medium or inhibition of the mevalonate pathway. These data indicate that L1CAM is controlled by a number of cell growth- and metabolism-related pathways during SC development. Functionally, SC with enhanced surface L1CAM showed increased adhesion to extracellular matrix and migrated faster. Our results provide mechanistic insights into senescence of human cells, with implications for future senolytic strategies.


Sujet(s)
Molécule d'adhérence cellulaire neurale L-1/métabolisme , Adhérence cellulaire/physiologie , Cycle cellulaire , Lignée cellulaire tumorale , Mouvement cellulaire/physiologie , Vieillissement de la cellule , Régulation négative , Fibroblastes , Rayons gamma , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des radiations , Humains , Molécule d'adhérence cellulaire neurale L-1/génétique , Interférence par ARN , Réaction de polymérisation en chaine en temps réel , RT-PCR , Transduction du signal , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/pharmacologie
5.
Oncotarget ; 7(34): 54952-54964, 2016 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-27448982

RÉSUMÉ

Standard-of-care chemo- or radio-therapy can induce, besides tumor cell death, also tumor cell senescence. While senescence is considered to be a principal barrier against tumorigenesis, senescent cells can survive in the organism for protracted periods of time and they can promote tumor development. Based on this emerging concept, we hypothesized that elimination of such potentially cancer-promoting senescent cells could offer a therapeutic benefit. To assess this possibility, here we first show that tumor growth of proliferating mouse TC-1 HPV-16-associated cancer cells in syngeneic mice becomes accelerated by co-administration of TC-1 or TRAMP-C2 prostate cancer cells made senescent by pre-treatment with the anti-cancer drug docetaxel, or lethally irradiated. Phenotypic analyses of tumor-explanted cells indicated that the observed acceleration of tumor growth was attributable to a protumorigenic environment created by the co-injected senescent and proliferating cancer cells rather than to escape of the docetaxel-treated cells from senescence. Notably, accelerated tumor growth was effectively inhibited by cell immunotherapy using irradiated TC-1 cells engineered to produce interleukin IL-12. Collectively, our data document that immunotherapy, such as the IL-12 treatment, can provide an effective strategy for elimination of the detrimental effects caused by bystander senescent tumor cells in vivo.


Sujet(s)
Vieillissement de la cellule/effets des médicaments et des substances chimiques , Immunothérapie adoptive/méthodes , Interleukine-12/pharmacologie , Tumeurs expérimentales/thérapie , Taxoïdes/pharmacologie , Charge tumorale/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques/pharmacologie , Effet bystander/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Association thérapeutique , Cytokines/génétique , Cytokines/métabolisme , Docetaxel , Interleukine-12/biosynthèse , Mâle , Souris de lignée C57BL , Tumeurs expérimentales/génétique , Tumeurs expérimentales/métabolisme , Facteurs temps
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...