Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 43(17): 1263-1273, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38433256

RESUMO

DNA double-strand breaks (DSBs) contribute to genome instability, a key feature of cancer. DSBs are mainly repaired by homologous recombination (HR) and non-homologous end-joining (NHEJ). We investigated the role of an isoform of the multifunctional cyclin-dependent kinase 9, CDK9-55, in DNA repair, by generating CDK9-55-knockout HeLa clones (through CRISPR-Cas9), which showed potential HR dysfunction. A phosphoproteomic screening in these clones treated with camptothecin revealed that CDC23 (cell division cycle 23), a component of the E3-ubiquitin ligase APC/C (anaphase-promoting complex/cyclosome), is a new substrate of CDK9-55, with S588 being its putative phosphorylation site. Mutated non-phosphorylatable CDC23(S588A) affected the repair pathway choice by impairing HR and favouring error-prone NHEJ. This CDK9 role should be considered when designing CDK-inhibitor-based cancer therapies.

2.
Viruses ; 13(12)2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34960727

RESUMO

Malignant mesothelioma (MM) is an aggressive asbestos-related cancer, against which no curative modalities exist. Oncolytic virotherapy is a promising therapeutic approach, for which MM is an ideal candidate; indeed, the pleural location provides direct access for the intra-tumoral injection of oncolytic viruses (OVs). Some non-human OVs offer advantages over human OVs, including the non-pathogenicity in humans and the absence of pre-existing immunity. We previously showed that caprine herpesvirus 1 (CpHV-1), a non-pathogenic virus for humans, can kill different human cancer cell lines. Here, we assessed CpHV-1 effects on MM (NCI-H28, MSTO, NCI-H2052) and non-tumor mesothelial (MET-5A) cells. We found that CpHV-1 reduced cell viability and clonogenic potential in all MM cell lines without affecting non-tumor cells, in which, indeed, we did not detect intracellular viral DNA after treatment. In particular, CpHV-1 induced MM cell apoptosis and accumulation in G0/G1 or S cell cycle phases. Moreover, CpHV-1 strongly synergized with cisplatin, the drug currently used in MM chemotherapy, and this agent combination did not affect normal mesothelial cells. Although further studies are required to elucidate the mechanisms underlying the selective CpHV-1 action on MM cells, our data suggest that the CpHV-1-cisplatin combination could be a feasible strategy against MM.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Cisplatino/farmacologia , Mesotelioma Maligno/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Varicellovirus/fisiologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Humanos , Mesotelioma Maligno/tratamento farmacológico , Mesotelioma Maligno/fisiopatologia , Mesotelioma Maligno/virologia , Vírus Oncolíticos/genética , Varicellovirus/genética
3.
Int J Mol Sci ; 21(19)2020 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-33020398

RESUMO

Malignant mesothelioma (MM) is a very aggressive asbestos-related cancer, for which no therapy proves to be effective. We have recently shown that the oncolytic adenovirus dl922-947 had antitumor effects in MM cell lines and murine xenografts. Previous studies demonstrated that dl922-947-induced host cell cycle checkpoint deregulation and consequent DNA lesions associated with the virus efficacy. However, the cellular DNA damage response (DDR) can counteract this virus action. Therefore, we assessed whether AZD1775, an inhibitor of the G2/M DNA damage checkpoint kinase WEE1, could enhance MM cell sensitivity to dl922-947. Through cell viability assays, we found that AZD1775 synergized with dl922-947 selectively in MM cell lines and increased dl922-947-induced cell death, which showed hallmarks of apoptosis (annexinV-positivity, caspase-dependency, BCL-XL decrease, chromatin condensation). Predictably, dl922-947 and/or AZD1775 activated the DDR, as indicated by increased levels of three main DDR players: phosphorylated histone H2AX (γ-H2AX), phospho-replication protein A (RPA)32, phospho-checkpoint kinase 1 (CHK1). Dl922-947 also increased inactive Tyr-15-phosphorylated cyclin-dependent kinase 1 (CDK1), a key WEE1 substrate, which is indicative of G2/M checkpoint activation. This increase in phospho-CDK1 was effectively suppressed by AZD1775, thus suggesting that this compound could, indeed, abrogate the dl922-947-induced DNA damage checkpoint in MM cells. Overall, our data suggest that the dl922-947-AZD1775 combination could be a feasible strategy against MM.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Sobrevivência Celular/efeitos dos fármacos , Mesotelioma Maligno/tratamento farmacológico , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirazóis/farmacologia , Pirimidinonas/farmacologia , Adenoviridae/genética , Apoptose/efeitos dos fármacos , Amianto/toxicidade , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/farmacologia , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Humanos , Mesotelioma Maligno/induzido quimicamente , Mesotelioma Maligno/genética , Mesotelioma Maligno/virologia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases , Proteínas Tirosina Quinases/genética
4.
Cell Death Dis ; 11(10): 867, 2020 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-33067416

RESUMO

Author Francesca Pentimalli was incorrectly associated with Histopathological Unit, IRCCS-Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy. The author's actual affiliation is Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, I-80131 Napoli, Italy.

5.
Cell Death Dis ; 11(9): 748, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32929059

RESUMO

Malignant pleural mesothelioma (MPM) is an aggressive cancer, related to asbestos exposure, which has a dismal prognosis. MPM diagnosis is late and often challenging, suggesting the need to identify more reliable molecular biomarkers. Here, we set out to identify differentially expressed miRNAs in epithelioid, biphasic, and sarcomatoid MPMs versus normal mesothelium and explored specific miRNA contribution to mesothelial tumorigenesis. We screened an LNA™-based miRNA-microrray with 14 formalin-fixed paraffin-embedded (FFPE) MPMs and 6 normal controls. Through real-time qRT-PCR we extended the analysis of a miRNA subset and further investigated miR-320a role through state-of-the-art techniques. We identified 16 upregulated and 32 downregulated miRNAs in MPMs versus normal tissue, including the previously identified potential biomarkers miR-21, miR-126, miR-143, miR-145. We showed in an extended series that miR-145, miR-10b, and miR-320a levels can discriminate tumor versus controls with high specificity and sensitivity. We focused on miR-320a because other family members were found downregulated in MPMs. However, stable miR-320a ectopic expression induced higher proliferation and migration ability, whereas miR-320a silencing reduced these processes, not supporting a classic tumor-suppressor role in MPM cell lines. Among putative targets, we found that miR-320a binds the 3'-UTR of the immune inhibitory receptor ligand PDL1 and, consistently, miR-320a modulation affects PDL1 levels in MPM cells. Finally, we showed that p53 over-expression induces the upregulation of miR-320a, along with miR-200a and miR-34a, both known to target PDL1, and reduces PDL1 levels in MPM cells. Our data suggest that PDL1 expression might be due to a defective p53-regulated miRNA response, which could contribute to MPM immune evasion or tumorigenesis through tumor-intrinsic roles.


Assuntos
Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Mesotelioma Maligno/metabolismo , MicroRNAs/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Regulação para Baixo , Células HEK293 , Humanos , Mesotelioma Maligno/genética , Mesotelioma Maligno/patologia , MicroRNAs/genética , Transfecção , Proteína Supressora de Tumor p53/genética
6.
Cancers (Basel) ; 12(7)2020 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-32664483

RESUMO

Abstract: Malignant mesothelioma (MM) is a deadly tumor mainly caused by exposure to asbestos. Unfortunately, no current treatment is able to change significantly the natural history of the disease, which has a poor prognosis in the majority of patients. The non-receptor tyrosine kinase SRC and other SRC family kinase (SFK) members are frequently hyperactivated in many cancer types, including MM. Several works have indeed suggested that SFKs underlie MM cell proliferation, survival, motility, and invasion, overall affecting multiple oncogenic pathways. Consistently, SFK inhibitors effectively counteracted MM cancerous features at the preclinical level. Dasatinib, a multi-kinase inhibitor targeting SFKs, was also assessed in clinical trials either as second-line treatment for patients with unresectable MM or, more recently, as a neoadjuvant agent in patients with resectable MM. Here, we provide an overview of the molecular mechanisms implicating SFKs in MM progression and discuss possible strategies for a more successful clinical application of SFK inhibitors. Our aim is to stimulate discussion and further consideration of these agents in better designed preclinical and clinical studies to make the most of another class of powerful antitumoral drugs, which too often are lost in translation when applied to MM.

7.
J Cell Mol Med ; 24(10): 5565-5577, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32301278

RESUMO

Malignant mesothelioma (MM) is an aggressive asbestos-related cancer of the serous membranes. Despite intensive treatment regimens, MM is still a fatal disease, mainly due to the intrinsic resistance to current therapies and the lack of predictive markers and new valuable molecular targets. Protein arginine methyltransferase 5 (PRMT5) inhibition has recently emerged as a potential therapy against methylthioadenosine phosphorylase (MTAP)-deficient cancers, in which the accumulation of the substrate 5'-methylthioadenosine (MTA) inhibits PRMT5 activity, thus sensitizing the cells to further PRMT5 inhibition. Considering that the MTAP gene is frequently codeleted with the adjacent cyclin-dependent kinase inhibitor 2A (CDKN2A) locus in MM, we assessed whether PRMT5 could represent a therapeutic target also for this cancer type. We evaluated PRMT5 expression, the MTAP status and MTA content in normal mesothelial and MM cell lines. We found that both administration of exogenous MTA and stable PRMT5 knock-down, by short hairpin RNAs (shRNAs), selectively reduced the growth of MTAP-deleted MM cells. We also observed that PRMT5 knock-down in MTAP-deficient MM cells reduced the expression of E2F1 target genes involved in cell cycle progression and of factors implicated in epithelial-to-mesenchymal transition. Therefore, PRMT5 targeting could represent a promising new therapeutic strategy against MTAP-deleted MMs.


Assuntos
Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Mesotelioma/genética , Proteína-Arginina N-Metiltransferases/genética , Purina-Núcleosídeo Fosforilase/genética , Linhagem Celular Tumoral , Cromatografia Líquida , Transição Epitelial-Mesenquimal/genética , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Mesotelioma/metabolismo , Mesotelioma/patologia , Espectrometria de Massas em Tandem
9.
J Cell Physiol ; 235(6): 5353-5362, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31957873

RESUMO

Psychological distress is a common consequence of breast cancer diagnosis and treatment and could further exacerbate therapy side effects. Interventions increasing treatment tolerance are crucial to improve both patients' quality of life and adherence to therapies. Virtual reality (VR) has emerged as an effective distraction tool for different medical procedures. Here, we assessed the efficacy of immersive and interactive VR in alleviating chemotherapy-related psychological distress in a cohort of Italian breast cancer patients, also comparing its effects with those of music therapy (MT). Thirty patients were included in the VR group, 30 in the MT group, and 34 in the control group, consisting of patients receiving standard care during chemotherapy. Our data suggest that both VR and MT are useful interventions for alleviating anxiety and for improving mood states in breast cancer patients during chemotherapy. Moreover, VR seems more effective than MT in relieving anxiety, depression, and fatigue.


Assuntos
Ansiedade/terapia , Neoplasias da Mama/tratamento farmacológico , Transtornos do Humor/terapia , Musicoterapia , Adolescente , Adulto , Idoso , Ansiedade/patologia , Neoplasias da Mama/patologia , Neoplasias da Mama/psicologia , Feminino , Humanos , Pessoa de Meia-Idade , Transtornos do Humor/patologia , Qualidade de Vida , Realidade Virtual , Adulto Jovem
10.
J Cell Physiol ; 235(6): 5213-5222, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31838757

RESUMO

In Italy, in the eastern area of the Campania region, the illegal dumping and burning of waste have been documented, which could potentially affect the local population's health. In particular, toxic waste exposure has been suggested to associate with increased cancer development/mortality in these areas, although a causal link has not yet been established. In this pilot study, we evaluated blood levels of toxic heavy metals and persistent organic pollutants (POPs) in 95 patients with different cancer types residing in this area and in 27 healthy individuals. While we did not find any significant correlation between the blood levels of POPs and the provenance of the patients, we did observe high blood concentrations of heavy metals in some municipalities, including Giugliano, where many illegal waste disposal sites have previously been documented. Our results showed that patients with different cancer types from Giugliano had higher blood levels of heavy metals than healthy controls. Despite the obvious limitations of this exploratory study, our preliminary observations encourage further research assessing the possible association between exposure to hazardous waste, increased blood metals, and increased risk of cancer.


Assuntos
Detecção Precoce de Câncer , Metais Pesados/sangue , Neoplasias/sangue , Poluentes Orgânicos Persistentes/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Feminino , Humanos , Itália/epidemiologia , Masculino , Metais Pesados/toxicidade , Pessoa de Meia-Idade , Neoplasias/induzido quimicamente , Neoplasias/patologia , Poluentes Orgânicos Persistentes/toxicidade , Adulto Jovem
11.
Front Oncol ; 9: 564, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31355131

RESUMO

Background: Malignant pleural mesothelioma (MPM) is an aggressive cancer associated with asbestos exposure that urgently requires effective therapeutic strategies. Current treatments are unable to increase significantly patient survival, which is often limited to <1 year from diagnosis. Virotherapy, based on the use of oncolytic viruses that exert anti-cancer effects by direct cell lysis and through the induction of anti-tumor immune response, represents an alternative therapeutic option for rare tumors with limited life expectancy. In this study, we propose the use of the adenovirus dl922-947, engineered to allow selective replication in cancer cells, to counteract MPM. Methods: We performed a thorough preclinical assessment of dl922-947 effects in a set of MPM cell lines and xenografts. Cytotoxicity of dl922-947 alone and in combination assays was evaluated by sulforhodamine B assay. Cell cycle, calreticulin expression, and high mobility group box protein 1 (HMGB1) secretion were determined by flow cytometry, whereas ATP content was determined by a luminescence-based bioassay. The modulation of angiogenic factors in MPM-infected cells was evaluated through ELISA. Results: We found that dl922-947 infection exhibits cytotoxic effects in MPM cell lines, affecting cell viability, cell cycle progression, and regulating main hallmarks of immunogenic cell death inducing calreticulin surface exposure, HMGB1 and ATP release. Our results also suggest that dl922-947 may affect angiogenic signals by regulation of VEGF-A and IL-8 secretion. Furthermore, dl922-947 shows anti-tumor efficacy in murine xenograft models reducing tumor growth and enhancing survival. Finally, the combination with cisplatin potentiated the cytotoxic effect of dl922-947. Conclusions: Overall our data identify virotherapy, based on the use of dl922-947, as a new possible therapeutic strategy against MPM, which could be used alone, in combination with standard chemotherapy drugs, as shown here, or other approaches also aimed at enhancing the antitumoral immune response elicited by the virus.

12.
Biochem Pharmacol ; 166: 323-334, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31176618

RESUMO

The retinoblastoma RB1 gene has been identified in the 80s as the first tumor suppressor. RB1 loss of function, as well alterations in its pathway, occur in most human cancers and often have prognostic value. RB1 has a key role in restraining cell cycle entry and, along with its family members, regulates a myriad of cellular processes and affects cell response to a variety of stimuli, ultimately determining cell fate. Consistently, RB1 status is a crucial determinant of the cell response to antitumoral therapies, impacting on the outcome of both traditional and modern anti-cancer strategies, including precision medicine approaches, such as kinase inhibitors, and immunotherapy. Despite many efforts however, the predictive value of RB1 status in the clinical practice is still underused, mainly owing to the complexity of RB1 function, to differences depending on the cellular context and on the therapeutic strategies, and, not-lastly, to technical issues. Here, we provide an overview of studies analyzing the role of RB1 in response to conventional cytotoxic and cytostatic therapeutic agents in different cancer types, including hormone dependent ones. We also review RB1 predictive value in the response to the last generation CDK4/6 inhibitors, other kinase inhibitors, and immunotherapy and discuss new emerging non-canonical roles of RB1 that could impact on the response to antitumoral treatments.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Retina/metabolismo , Proteínas de Ligação a Retinoblastoma/metabolismo , Retinoblastoma/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antineoplásicos/farmacologia , Humanos , Valor Preditivo dos Testes , Neoplasias da Retina/tratamento farmacológico , Neoplasias da Retina/genética , Retinoblastoma/tratamento farmacológico , Retinoblastoma/genética , Proteínas de Ligação a Retinoblastoma/genética , Ubiquitina-Proteína Ligases/genética
13.
Int J Oncol ; 54(6): 2189-2199, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31081046

RESUMO

Glioblastoma (GB) is the most common and aggressive malignant tumor of the central nervous system. Despite current intensive treatment regimens, consisting of surgical resection followed by radiotherapy with concomitant and adjuvant temozolomide (TMZ) chemotherapy, the prognosis of patients with GB remains extremely poor. Considering that alterations of the p53 tumor suppressor pathway have a key role in both GB development and resistance to TMZ treatment, the re­activation of p53 could be an effective therapeutic approach against GB. In this study, we challenged p53 wild­type and mutant GB cell lines with RITA, a molecule originally identified for its ability to restore p53 functions, although it was subsequently shown to act also through p53­independent mechanisms. We examined the effects of RITA on GB cell viability, through MTS and clonogenic assays, and analyzed cell death through cytoflourimetric analyses. In all the tested GB cell lines, RITA significantly reduced the cell proliferative and clonogenic potential and induced cell accumulation in the S and/or G2/M cell cycle phases and massive p53­dependent apoptosis. Moreover, RITA was more effective than the well­known p53 re­activating molecule, nutlin­3, and did not affect the viability of normal astrocytes. In addition, RITA decreased survivin expression and induced DNA damage, two mechanisms that likely contribute to its anti­tumor effects. Furthermore, RITA synergized with TMZ and was able to decrease the expression of MGMT, which is a crucial player in TMZ resistance. Thus, although further studies are warranted to clarify the exact mechanisms of action of RITA, the data of this study suggest the potential of such an approach for GB therapy, which may also help to overcome resistance to TMZ.


Assuntos
Neoplasias Encefálicas/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Furanos/farmacologia , Glioblastoma/metabolismo , Temozolomida/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Humanos , Imidazóis/farmacologia , Mutação , Piperazinas/farmacologia , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/genética
14.
J Cell Physiol ; 233(9): 7391-7401, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29659015

RESUMO

Malignant mesothelioma (MM) is a very aggressive asbestos-related cancer, whose incidence is increasing worldwide. Unfortunately, no effective therapies are currently available and the prognosis is extremely poor. Recently, the anti-helminthic drug pyrvinium pamoate has attracted a strong interest for its anti-cancer activity, which has been demonstrated in many cancer models. Considering the previously established inhibitory effect of pyrvinium pamoate on the Wnt/ß-catenin pathway and given the important role of this pathway in MM, we investigated the potential anti-tumor activity of this drug in MM cell lines. We observed that pyrvinium pamoate significantly impairs MM cell proliferation, cloning efficiency, migration, and tumor spheroid formation. At the molecular level, our data show that pyrvinium pamoate down-regulates the expression of ß-catenin and Wnt-regulates genes. Overall, our study suggests that the repurposing of pyrvinium pamoate for MM treatment could represent a new promising therapeutic approach.


Assuntos
Reposicionamento de Medicamentos , Mesotelioma/tratamento farmacológico , Compostos de Pirvínio/uso terapêutico , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Mesotelioma/genética , Mesotelioma/patologia , Compostos de Pirvínio/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Fatores de Tempo , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética , beta Catenina/metabolismo
15.
Oncogene ; 37(27): 3657-3671, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29606701

RESUMO

The retinoblastoma (RB) protein family includes RB1/p105, RBL1/p107, and RBL2/p130, which are key factors in cell-cycle regulation and stand at the crossroads of multiple pathways dictating cell fate decisions. The role of RB proteins in apoptosis is controversial because they can inhibit or promote apoptosis depending on the context, on the apoptotic stimuli and on their intrinsic status, impacting on the response to antitumoral treatments. Here we identified RBL2/p130 as a direct substrate of the AKT kinase, a key antiapoptotic factor hyperactive in multiple cancer types. We showed that RBL2/p130 and AKT1 physically interact and AKT phosphorylates RBL2/p130 Ser941, located in the pocket domain, but not when this residue is mutated into Ala. We found that pharmacological inhibition of AKT, through the highly selective AKT inhibitor VIII (AKTiVIII), impairs RBL2/p130 Ser941 phosphorylation and increases RBL2/p130 stability, mRNA expression and nuclear levels in both lung cancer and mesothelioma cell lines, mirroring the more extensively studied effects on the p27 cell-cycle inhibitor. Consistently, AKT inhibition reduced cell viability, induced cell accumulation in G0/G1, and triggered apoptosis, which proved to be largely dependent on RBL2/p130 itself, as shown upon RBL2/p130 silencing. AKT inhibition induced RBL2/p130-dependent apoptosis also in HEK-293 cells, in which re-expression of a short hairpin-resistant RBL2/p130 was able to rescue AKTiVIII-induced apoptosis upon RBL2/p130 silencing. Our data also showed that the combination of AKT and cyclin-dependent kinases (CDK) inhibitors, which converge on the re-activation of RBL2/p130 antitumoral potential, could be a promising anticancer strategy.


Assuntos
Apoptose/fisiologia , Neoplasias Pulmonares/patologia , Mesotelioma/patologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína p130 Retinoblastoma-Like/metabolismo , Células A549 , Benzimidazóis/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Mesotelioma/genética , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Quinoxalinas/farmacologia , RNA Mensageiro/biossíntese , Proteína p130 Retinoblastoma-Like/genética
16.
Clin J Pain ; 34(9): 858-877, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29485536

RESUMO

OBJECTIVES: This review aims to provide a framework for evaluating the utility of virtual reality (VR) as a distraction intervention to alleviate pain and distress during medical procedures. We first describe the theoretical bases underlying the VR analgesic and anxiolytic effects and define the main factors contributing to its efficacy, which largely emerged from studies on healthy volunteers. Then, we provide a comprehensive overview of the clinical trials using VR distraction during different medical procedures, such as burn injury treatments, chemotherapy, surgery, dental treatment, and other diagnostic and therapeutic procedures. METHODS: A broad literature search was performed using as main terms "virtual reality," "distraction," and "pain." No date limit was applied and all the retrieved studies on immersive VR distraction during medical procedures were selected. RESULTS: VR has proven to be effective in reducing procedural pain, as almost invariably observed even in patients subjected to extremely painful procedures, such as patients with burn injuries undergoing wound care, and physical therapy. Moreover, VR seemed to decrease cancer-related symptoms in different settings, including during chemotherapy. Only mild and infrequent side effects were observed. DISCUSSION: Despite these promising results, future long-term randomized controlled trials with larger sample sizes and evaluating not only self-report measures but also physiological variables are needed. Further studies are also required both to establish predictive factors to select patients who can benefit from VR distraction and to design hardware/software systems tailored to the specific needs of different patients and able to provide the greatest distraction at the lowest cost.


Assuntos
Dor Processual/terapia , Estresse Psicológico/etiologia , Estresse Psicológico/terapia , Realidade Virtual , Atenção , Humanos
17.
J Cell Physiol ; 233(2): 1266-1277, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28488765

RESUMO

Gastric cancer represents a diffuse and aggressive neoplasm, whose mortality index is among the highest in the world. Predisposing factors are E-cadherin mutations, Helicobacter pylori infection, and a diet rich in salted and smoked food, with a low intake of fresh fruits and vegetables. Here, we analyzed the effect of total lipophilic extracts of two Southern Italy tomato varieties, San Marzano and Corbarino, on an in vitro model of gastric cancer, YCC-1, YCC-2 and YCC-3 cell lines, characterized by different aggressiveness. Our results showed a possible role of these two varieties of tomatoes against typical neoplastic features. The treatment with tomato extracts affected cancer cell ability to grow both in adherence and in semisolid medium, reducing also cell migration ability. No toxic effects were observed on non-tumoral cells. We found, on gastric cancer cell lines, effects on both cell cycle progression and apoptosis modulation. The extent of antineoplastic effects, however, did not seem to correlate with the carotenoid content and antioxidant activity of the two tomato varieties. Our data indicate that San Marzano and Corbarino intake might be further considered as nutritional support not only in cancer prevention, but also for cancer patient diet.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Antioxidantes/farmacologia , Carotenoides/farmacologia , Solanum lycopersicum/química , Neoplasias Gástricas/tratamento farmacológico , Antineoplásicos Fitogênicos/isolamento & purificação , Antioxidantes/isolamento & purificação , Apoptose/efeitos dos fármacos , Carotenoides/isolamento & purificação , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Frutas/química , Humanos , Itália , Invasividade Neoplásica , Fitoterapia , Plantas Medicinais , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Fatores de Tempo
18.
J Cell Physiol ; 232(1): 129-35, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27037775

RESUMO

Ewing sarcoma (ES) is a highly aggressive bone and soft tissue cancer, representing the second most common primary malignant bone tumor in children and adolescents. Although the development of a multimodal therapy, including both local control (surgery and/or radiation) and systemic multidrug chemotherapy, has determined a significant improvement in survival, patients with metastatic and recurrent disease still face a poor prognosis. Moreover, considering that ES primarily affects young patients, there are concerns about long-term adverse effects of the therapy. Therefore, more rational strategies, targeting specific molecular alterations underlying ES, are required. Recent studies suggest that SRC family kinases (SFKs), which are aberrantly activated in most cancer types, could represent key therapeutic targets also for ES. Here, we challenged ES cell lines with a recently developed selective SFK inhibitor (a pyrazolo[3,4-d]pyrimidine derivative, called SI221), which was previously shown to be a valuable proapoptotic agent in other tumor types while not affecting normal cells. We observed that SI221 significantly reduced ES cell viability and proved to be more effective than the well-known SFK inhibitor PP2. SI221 was able to induce apoptosis in ES cells and also reduced ES cell clonogenic potential. Furthermore, SI221 was also able to reduce ES cell migration. At the molecular level, our data suggest that SFK inhibition through SI221 could reduce ES cell viability at least in part by hindering an SFK-NOTCH1 receptor-p38 mitogen-activated protein kinase (MAPK) axis. Overall, our study suggests a potential application of specific SFK inhibition in ES therapy. J. Cell. Physiol. 232: 129-135, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Neoplasias Ósseas/metabolismo , Movimento Celular/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Sarcoma de Ewing/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases da Família src/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Pirazóis/química , Pirimidinas/química , Quinases da Família src/metabolismo
19.
Oncotarget ; 6(20): 17873-90, 2015 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-26160835

RESUMO

Inactivation of the retinoblastoma (RB1) tumor suppressor is one of the most frequent and early recognized molecular hallmarks of cancer. RB1, although mainly studied for its role in the regulation of cell cycle, emerged as a key regulator of many biological processes. Among these, RB1 has been implicated in the regulation of apoptosis, the alteration of which underlies both cancer development and resistance to therapy. RB1 role in apoptosis, however, is still controversial because, depending on the context, the apoptotic cues, and its own status, RB1 can act either by inhibiting or promoting apoptosis. Moreover, the mechanisms whereby RB1 controls both proliferation and apoptosis in a coordinated manner are only now beginning to be unraveled. Here, by reviewing the main studies assessing the effect of RB1 status and modulation on these processes, we provide an overview of the possible underlying molecular mechanisms whereby RB1, and its family members, dictate cell fate in various contexts. We also describe the current antitumoral strategies aimed at the use of RB1 as predictive, prognostic and therapeutic target in cancer. A thorough understanding of RB1 function in controlling cell fate determination is crucial for a successful translation of RB1 status assessment in the clinical setting.


Assuntos
Apoptose , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Neoplasias/metabolismo , Proteína do Retinoblastoma/metabolismo , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Fosforilação , Proteína do Retinoblastoma/genética , Transdução de Sinais , Resultado do Tratamento
20.
Oncotarget ; 6(14): 12421-35, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25762618

RESUMO

Recent data suggest that SRC family kinases (SFKs) could represent potential therapeutic targets for rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma in children. Here, we assessed the effect of a recently developed selective SFK inhibitor (a pyrazolo[3,4-d]pyrimidine derivative, called SI221) on RMS cell lines. SI221, which showed to be mainly effective against the SFK member YES, significantly reduced cell viability and induced apoptosis, without affecting non-tumor cells, such as primary human skin fibroblasts and differentiated C2C12 cells. Moreover, SI221 decreased in vitro cell migration and invasion and reduced tumor growth in a RMS xenograft model. SFK inhibition also induced muscle differentiation in RMS cells by affecting the NOTCH3 receptor-p38 mitogen-activated protein kinase (MAPK) axis, which regulates the balance between proliferation and differentiation. Overall, our findings suggest that SFK inhibition, besides reducing RMS cell growth and invasive potential, could also represent a differentiation therapeutic strategy for RMS.


Assuntos
Antineoplásicos/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Rabdomiossarcoma/patologia , Quinases da Família src/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Diferenciação Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Imunofluorescência , Humanos , Camundongos , Reação em Cadeia da Polimerase , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...