Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Toxicol Appl Pharmacol ; 467: 116497, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37003365

RESUMO

Novel psychoactive substances (NPS) consumption has increased in recent years, thus NPS-induced cognitive decline is a current source of concern. Alpha-pyrrolidinovalerophenone (α-PVP), as a member of NPS, is consumed throughout regions like Washington, D.C., Eastern Europe, and Central Asia. Mitochondrial dysfunction plays an essential role in NPS-induced cognitive impairment. Meanwhile, no investigations have been conducted regarding the α-PVP impact on spatial learning/memory and associated mechanisms. Consequently, our study investigated the α-PVP effect on spatial learning/memory and brain mitochondrial function. Wistar rats received different α-PVP doses (5, 10, and 20 mg/kg) intraperitoneally for 10 sequential days; 24 h after the last dose, spatial learning/memory was evaluated by the Morris Water Maze (MWM). Furthermore, brain mitochondrial protein yield and mitochondrial function variables (Mitochondrial swelling, succinate dehydrogenase (SDH) activity, lipid peroxidation, Mitochondrial Membrane Potential (MMP), Reactive oxygen species (ROS) level, brain ADP/ATP proportion, cytochrome c release, Mitochondrial Outer Membrane (MOM) damage) were examined. α-PVP higher dose (20 mg/kg) significantly impaired spatial learning/memory, mitochondrial protein yield, and brain mitochondrial function (caused reduced SDH activity, increased mitochondrial swelling, elevated ROS generation, increased lipid peroxidation, collapsed MMP, increased cytochrome c release, elevated brain ADP/ATP proportion, and MOM damage). Moreover, the lower dose of α-PVP (5 mg/kg) did not alter spatial learning/memory and brain mitochondrial function. These findings provide the first evidence regarding impaired spatial learning/memory following repeated administration of α-PVP and the possible role of brain mitochondrial dysfunction in these cognitive impairments.


Assuntos
Encefalopatias , Aprendizagem Espacial , Ratos , Animais , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Citocromos c/metabolismo , Aprendizagem em Labirinto , Mitocôndrias , Encéfalo , Trifosfato de Adenosina/metabolismo , Hipocampo , Estresse Oxidativo
2.
Mol Biol Rep ; 50(9): 7639-7647, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37528313

RESUMO

BACKGROUND: Colorectal Cancer (CC) is among the most prevalent cancers in elderly persons. Radiotherapy is usually prescribed as CC develops, however, radiation beams indiscriminately affect normal cells. Previous studies nominated that probiotics and their metabolites can be used to minimize the side effects of radiotherapy. Hereby, the aim of this study was to investigate the probable correlation between cell-free supernatant of Bacillus subtilis and radiation response in normal and cancerous cell lines. METHODS AND RESULTS: IEC-18 and SW-48 cells were treated with different concentrations of B. subtilis supernatant. To evaluate the effect of probiotic treatments under radiation and the normal situation, the cytotoxicity of the treatments was measured using the MTT method. The cell cycle status was analyzed by flow cytometry. The expression levels of Bax, Bcl-2, and Caspase 3 genes were also determined by real-time (RT) PCR. B. subtilis supernatant increased the viability of normal cells under radiation treatment, although this effect was not significant. 40% v/v of this mixture could amplify the lethal effect of radiation and decreased the viability of cancer cells. SW-48 cells that received 40% v/v of the supernatant had a significantly higher rate of apoptosis. Probiotic supernatant effectively induced the expression of proapoptotic Bax and Caspase 3 genes. CONCLUSION: Presented results confirmed that the supernatant of B. subtilis can be supposed as a clue to improve the efficacy of radiation therapy in CC patients as it increased the sensitivity of cancerous cells and protected normal epithelial cells from detrimental effects of radiation.


Assuntos
Bacillus subtilis , Probióticos , Ratos , Animais , Bacillus subtilis/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo , Regulação para Cima , Células Epiteliais/metabolismo , Apoptose , Probióticos/farmacologia
3.
Mikrochim Acta ; 190(2): 77, 2023 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-36715890

RESUMO

Recently, electrochemiluminescent (ECL) immunosensors have received much attention in the field of biomarker detection. Here, a highly enhanced ECL immunosensing platform was designed for ultrasensitive detection of carcinoembryonic antigen (CEA). The surface of the glassy carbon electrode was enhanced by applying functional nanostructures such as thiolated graphene oxide (S-GO) and streptavidin-coated gold nanoparticles (SA-AuNPs). The selectivity and sensitivity of the designed immunosensor were improved by entrapping CEA biomolecules using a sandwich approach. Luminol/silver nanoparticles (Lu-SNPs) were applied as the main core of the signaling probe, which were then coated with streptavidin to provide overloading of the secondary antibody. The highly ECL signal enhancement was obtained due to the presence of horseradish peroxidase (HRP) in the signaling probe, in which the presence of H2O2 further amplified the intensity of the signals. The engineered immunosensor presented excellent sensitivity for CEA detection, with limit of detection (LOD) and linear detection range (LDR) values of 58 fg mL-1 and 0.1 pg mL-1 to 5 pg mL-1 (R2 = 0.9944), respectively. Besides its sensitivity, the fabricated ECL immunosensor presented outstanding selectivity for the detection of CEA in the presence of various similar agents. Additionally, the developed immunosensor showed an appropriate repeatability (RSD 3.8%) and proper stability (2 weeks). Having indicated a robust performance in the real human serum with stated LOD and LDR, the engineered immunosensor can be considered for the detection and monitoring of CEA in the clinic.


Assuntos
Técnicas Biossensoriais , Nanopartículas Metálicas , Nanocompostos , Humanos , Luminol/química , Antígeno Carcinoembrionário , Ouro/química , Prata/química , Nanopartículas Metálicas/química , Peróxido de Hidrogênio , Estreptavidina , Medições Luminescentes , Imunoensaio , Nanocompostos/química
4.
Analyst ; 146(22): 6902-6916, 2021 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-34636832

RESUMO

The contribution of oxidative stress in several chronic and degenerative diseases suggests that antioxidant therapy can be a promising therapeutic strategy. However, in the case of many antioxidants, their biodistribution and bioactivity are restricted due to low water solubility. Curcumin is a powerful free radical scavenger that upon conjugation to gold nanoparticles results in the formation of stable gold nanoparticles that act as highly water-soluble carriers for the curcumin molecules. In the present study, the effect of curcumin-coated gold nanoparticles (Cur-GNPs) on the H2O2-treated human neuroblastoma (SK-N-SH) cell line was evaluated by using Fourier transform infrared (FTIR) microspectroscopy. Biochemical changes in cells resulting from exposure to reactive oxygen species (ROS) and antioxidant treatment on cells were investigated. Analyzing changes in PO2- bands and amide bands in the fingerprint region and also changes in the ratio of CH2(asym) to CH3(asym) bands in the lipid region revealed that post-treatment with Cur-GNPs could effectively decrease the damage on DNA caused by H2O2 treatment, whereas pre-treatment of cells with Cur-GNPs was found to be more effective at preventing lipid peroxidation than post-treatment. Further analysis of the CH2(asym) to CH3(asym) ratio provided information on not only the lipid peroxidation level in cells, but also the interaction of nanoparticles with the plasma membrane, as confirmed by lactate dehydrogenase assay.


Assuntos
Curcumina , Nanopartículas Metálicas , Nanopartículas , Neuroblastoma , Curcumina/farmacologia , Ouro , Humanos , Peróxido de Hidrogênio/toxicidade , Nanopartículas Metálicas/toxicidade , Estresse Oxidativo , Distribuição Tecidual
5.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-34502144

RESUMO

Impaired wound healing in people with diabetes has multifactorial causes, with insufficient neovascularization being one of the most important. Hypoxia-inducible factor-1 (HIF-1) plays a central role in the hypoxia-induced response by activating angiogenesis factors. As its activity is under precise regulatory control of prolyl-hydroxylase domain 2 (PHD-2), downregulation of PHD-2 by small interfering RNA (siRNA) could stabilize HIF-1α and, therefore, upregulate the expression of pro-angiogenic factors as well. Intracellular delivery of siRNA can be achieved with nanocarriers that must fulfill several requirements, including high stability, low toxicity, and high transfection efficiency. Here, we designed and compared the performance of layer-by-layer self-assembled siRNA-loaded gold nanoparticles with two different outer layers-Chitosan (AuNP@CS) and Poly L-arginine (AuNP@PLA). Although both formulations have exactly the same core, we find that a PLA outer layer improves the endosomal escape of siRNA, and therefore, transfection efficiency, after endocytic uptake in NIH-3T3 cells. Furthermore, we found that endosomal escape of AuNP@PLA could be improved further when cells were additionally treated with desloratadine, thus outperforming commercial reagents such as Lipofectamine® and jetPRIME®. AuNP@PLA in combination with desloratadine was proven to induce PHD-2 silencing in fibroblasts, allowing upregulation of pro-angiogenic pathways. This finding in an in vitro context constitutes a first step towards improving diabetic wound healing with siRNA therapy.


Assuntos
Indutores da Angiogênese/metabolismo , Angiopatias Diabéticas/metabolismo , Ouro , Hipóxia/metabolismo , Lisossomos , Nanopartículas , RNA Interferente Pequeno/genética , Animais , Sobrevivência Celular , Fenômenos Químicos , Angiopatias Diabéticas/etiologia , Angiopatias Diabéticas/patologia , Composição de Medicamentos , Endossomos/metabolismo , Técnicas de Transferência de Genes , Hipóxia/genética , Loratadina/análogos & derivados , Loratadina/química , Loratadina/farmacologia , Camundongos , Células NIH 3T3 , Nanopartículas/química , RNA Interferente Pequeno/administração & dosagem
6.
Nanomedicine ; 29: 102243, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32623018

RESUMO

Many tumors develop resistance to most of the apoptosis-based cancer therapies. In this sense targeting non-apoptotic forms of cell death including necroptosis, autophagy and ferroptosis may have therapeutic benefits in apoptosis-defective cancer cells. Nanomaterials have shown great advantages in cancer treatment owing to their unique characteristics. Besides, the capability of nanomaterials to induce different forms of cell death has gained widespread attention in cancer treatment. Reports in this field reflect the therapeutic potential of necroptotic cell death induced by nanomaterials in cancer. Also, autophagic cell death induced by nanomaterials alone and as a part of chemo-, radio- and photothermal therapy holds great promise as anticancer therapeutic option. Besides, ferroptosis induction by iron-based nanomaterials in drug delivery, immunotherapy, hyperthermia and imaging systems shows promising results in malignancies. Hence, this review is devoted to the latest efforts and the challenges in this field of research and its clinical merits.


Assuntos
Morte Celular/efeitos dos fármacos , Nanoestruturas/uso terapêutico , Necroptose/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Apoptose/genética , Autofagia/efeitos dos fármacos , Autofagia/genética , Morte Celular/genética , Ferroptose/efeitos dos fármacos , Ferroptose/genética , Humanos , Necroptose/genética , Neoplasias/genética , Neoplasias/patologia
7.
J Cell Biochem ; 120(6): 9125-9137, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30614034

RESUMO

Fragile histidine trail (FHIT) is a tumor suppressor in response to DNA damage which has been deleted in various tumors. However, the signaling mechanisms and interactions of FHIT with regard to apoptotic proteins including p53 and p38 in the DNA damage-induced apoptosis are not well described. In the present study, we used etoposide-induced DNA damage in MCF-7 as a model to address these crosstalks. The time course study showed that the expression of FHIT, p53, and p38MAPK started after 1 hour following etoposide treatment. FHIT overexpression led to increase p53 expression, p38 activation, and augmented apoptosis following etoposide-induced DNA damage compared to wild-type cells. However, FHIT knockdown blocked p53 expression, delayed p38 activation, and completely inhibited etoposide-induced apoptosis. Inhibition of p38 activity prevented induction of p53, FHIT, and apoptosis in this model. Thus, activation of p38 upon etoposide treatment leads to increase in FHIT and p53 expression. In p53 knockdown MCF-7, the FHIT induction was hampered but p38 activation was induced in lower doses of etoposide. In p53 knockdown cells, inhibition of p38 induced FHIT expression and apoptosis. Our data demonstrated that the exposure of MCF-7 cells to etoposide increases apoptosis through a mechanism involving the activation of the p38-FHIT-p53 pathway. Moreover, our findings suggest signaling interaction for these pathways may represent a promising therapy for breast cancer.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Apoptose/efeitos dos fármacos , Etoposídeo/farmacologia , Proteínas de Neoplasias/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Humanos , Células MCF-7 , Transdução de Sinais/efeitos dos fármacos
8.
Cancer Immunol Immunother ; 68(7): 1039-1058, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31165204

RESUMO

The emergence of immunotherapy has revolutionized medical oncology with unprecedented advances in cancer treatment over the past two decades. However, a major obstacle in cancer immunotherapy is identifying appropriate tumor-specific antigens to make targeted therapy achievable with fewer normal cells being impaired. The similarity between placentation and tumor development and growth has inspired many investigators to discover antigens for effective immunotherapy of cancers. Placenta-specific 1 (PLAC1) is one of the recently discovered placental antigens with limited normal tissue expression and fundamental roles in placental function and development. There is a growing body of evidence showing that PLAC1 is frequently activated in a wide variety of cancer types and promotes cancer progression. Based on the restricted expression of PLAC1 in testis, placenta and a wide variety of cancers, we have designated this molecule with new terminology, cancer-testis-placenta (CTP) antigen, a feature that PLAC1 shares with many other cancer testis antigens. Recent reports from our lab provide compelling evidence on the preferential expression of PLAC1 in prostate cancer and its potential utility in prostate cancer immunotherapy. PLAC1 may be regarded as a potential CTP antigen for targeted cancer immunotherapy based on the available data on its promoting function in cancer development and also its expression in cancers of different histological origin. In this review, we will summarize current data on PLAC1 with emphasis on its association with cancer development and immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Antineoplásicos Imunológicos/uso terapêutico , Biomarcadores Tumorais/antagonistas & inibidores , Neoplasias/terapia , Proteínas da Gravidez/antagonistas & inibidores , Antígenos de Neoplasias/metabolismo , Antineoplásicos Imunológicos/farmacologia , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Progressão da Doença , Feminino , Humanos , Imunoterapia/métodos , Masculino , Terapia de Alvo Molecular/métodos , Neoplasias/imunologia , Neoplasias/patologia , Placenta/patologia , Gravidez , Proteínas da Gravidez/imunologia , Proteínas da Gravidez/metabolismo , Testículo/patologia
9.
Strahlenther Onkol ; 195(10): 934-939, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31363801

RESUMO

PURPOSE: The urinary bladder is one major organ at risk in radiotherapy of pelvic malignancies. The radiation response manifests in early and chronic changes in bladder function. These are based on inflammatory effects and changes in urothelial cell function and proliferation. This study evaluates the effect of bortezomib as an anti-proliferative and anti-inflammatory compound in an established mouse bladder model. The early radiation-induced bladder dysfunction in the mouse occurs in two phases during the first month after irradiation (phase I: day 0-15, phase II: days 16-30). MATERIALS AND METHODS: Daily bortezomib injections (0.02 mg/ml, subcutaneously) were administered between days 0-15 or 15-30 in separate groups. Single graded radiation doses were administered in five dose groups. Cystometry was carried out before (individual control) and during the first month after irradiation. When bladder capacity was decreased by ≥50%, mice were considered as responders. Statistical analysis was performed by the SPSS software version 24. RESULTS: Daily bortezomib injections between days 0-15 resulted in a significant decrease in responders for phase I. There was no significant effect with daily bortezomib injections between days 16-30. CONCLUSION: Two separate waves of acute radiation-induced urinary bladder dysfunction have distinct mechanisms that need further biological studies.


Assuntos
Bortezomib/farmacologia , Lesões Experimentais por Radiação/tratamento farmacológico , Bexiga Urinária/efeitos da radiação , Animais , Relação Dose-Resposta à Radiação , Esquema de Medicação , Feminino , Humanos , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C3H , Tamanho do Órgão/efeitos dos fármacos , Tamanho do Órgão/efeitos da radiação , Órgãos em Risco/efeitos da radiação , Bexiga Urinária/efeitos dos fármacos , Urodinâmica/efeitos dos fármacos , Urodinâmica/efeitos da radiação
10.
Cell Biochem Funct ; 37(5): 320-330, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31172564

RESUMO

GDF15 plays a paradoxical role during carcinogenesis; it inhibits tumour growth in the early stages and promotes tumour cell proliferation in the late stages of cancer. Besides, GDF15 can induce apoptosis in some cancer cells including A549 but not in some others. Moreover, as a potential receptor for GDF15, TGFBR2 is inactivated during carcinogenesis in many types of cancers, and it is not present in cells with no GDF15 induced apoptosis. Thus, we tested whether GDF15 overexpression and/or TGFBR2 silencing can affect the GDF15 induced apoptosis in A549 cells. The full and mature forms of GDF15 were cloned and overexpressed in A549 cells. The TGFBR2 was silenced using specific siRNA and confirmed by real-time PCR. Results indicated that overexpression of full and mature forms of GDF15 as well as TGFBR2 knocked down reduced A549 cell viability in 24 and 48 hours. Flow cytometric analysis of annexin V/PI indicated induction of apoptosis in A549 cells by overexpression of GDF15 or silencing TGFBR2. Interestingly, the silencing of TGFBR2 inhibited the GDF15 induced cytotoxicity and apoptosis in A549 cells. Overexpression of GDF15 activated caspase-9 and caspase-3 and inhibited ERK1/2 and p38 phosphorylation in A549 cells. TGFBR2 knocked down inhibited GDF15 effects on caspases, ERK1/2, and p38MAPK activation. Our results indicated that the effect of GDF15 on apoptosis and activation of MAPK in A549 cells depends on TGFBR2 expression. These findings may point to mechanisms in which GDF15 exerts dual effect during carcinogenesis with regard to TGFBR2 expression. SIGNIFICANCE OF THE STUDY: GDF15 plays a tumour suppressor or promotor roles during carcinogenesis. The expression of GDF15 induced cytotoxicity, apoptosis, and inhibition of MAPK in A549 cells. All these effects were blocked by silencing TGFBR2 expression. These findings may point to mechanisms in which GDF15 exerts dual effect during carcinogenesis with regard to TGFBR2 expression.


Assuntos
Apoptose/efeitos dos fármacos , Fator 15 de Diferenciação de Crescimento/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Células A549 , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Perfilação da Expressão Gênica , Fator 15 de Diferenciação de Crescimento/genética , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptor do Fator de Crescimento Transformador beta Tipo II/deficiência , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Células Tumorais Cultivadas
11.
J Appl Toxicol ; 39(2): 375-384, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30294794

RESUMO

Benzo[α]pyrene (BaP) can have significant role in the development of breast cancer via aryl hydrocarbon receptor (AhR) activation. AhR activation has been studied in several functions such as survival, migration and invasion of cancer cells. In cancer, integrins contribute to the migration/invasion process and are regulated by nuclear factor of activated T cells (NFAT) and transforming growth factor (TGF) beta pathways. The aim of the present study was to examine the effect of BaP, an activator of AhR and cyclosporine A (CsA), as inhibitor of NFAT on migration and invasion of MDA-MB-231 cells. Furthermore, the effects of BaP and CsA were evaluated regarding the crosstalk of AhR, NFAT1 and TGF-ß receptor 1 signaling. Treatment of MDA-MB-231 with BaP resulted in significantly more live cells in low doses; however, blocking NFAT with CsA decreased the viability of the cells. Activation of AhR by BaP induced invasion as well as migration in MDA-MB-231 cells, which was blocked by AhR antagonist. Unlike BaP, block of NFAT with CsA inhibited cell migration and cell invasion. In these cells, BaP significantly reduced AhR expression while this reduction was reversed by CH-223191; however, CsA treatment lowered the AhR expression only at low dose. The level of ß4 integrin was significantly reduced by CsA at 1 and 2.5 µm. Protein levels of Snail and TGF-ß receptor 1 were not significantly altered by BaP and CsA treatments. Considering these findings, the low AhR expression and high ß4 integrin level following BaP and/or CsA treatments may contribute to the higher invasion/migration in MDA-MB-231 cells.


Assuntos
Benzo(a)pireno/toxicidade , Movimento Celular/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Integrina beta4/metabolismo , Fatores de Transcrição NFATC/antagonistas & inibidores , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclosporina/farmacologia , Relação Dose-Resposta a Droga , Humanos , Invasividade Neoplásica , Transdução de Sinais
12.
Inflammopharmacology ; 27(5): 1071-1080, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30929154

RESUMO

Ischemic heart disease is a leading cause of death on a global scale, placing major socio-economic burdens on health systems worldwide. Myocardial ischaemia and reperfusion (I/R)-induced tissue injury is associated with alteration in activity of inflammatory system and nitric oxide pathway. Sumatriptan, which is mainly used to relieve migraine headache, has recently been shown to exert anti-inflammatory properties. In this study, we aimed to assess the possible cardioprotective effect of sumatriptan in a rat model of I/R injury. Male Wistar rats were subjected to 30-min ligation of left anterior descending coronary artery and 120-min reperfusion. Animals were randomly divided into five groups: (1) Sham (2) I/R (3) I/R treated with sumatriptan (0.3 mg/kg i.p.) 20 min after induction of I/R rats, (4) GR127935 (a selective antagonist of 5-HT1B/D serotonin receptors; 0.3 mg/kg) 20 min after induction of I/R, and (5) GR127935 (0.3 mg/kg) 15 min before administration of sumatriptan. Post-infarct treatment with sumatriptan increased left ventricular function, which was damaged in I/R animal's heart. Sumatriptan (0.3 mg/kg) decreased lipid peroxidation, CK-MB and lactate dehydrogenase levels; tumor necrosis factor concentration; and Nf-Ò¡B' protein production. Treatment with sumatriptan significantly increased the endothelial nitric oxide synthase (eNOS) expression consequences nitric oxide metabolites' level in I/R rats. Also, injection of sumatriptan remarkably decreased myocardial tissue injury assessed by histopathological study. These findings suggest that sumatriptan may attenuate I/R injury via modulating the inflammatory responses and endothelial NOS activity. But therapeutic index of sumatriptan is narrow according to the result of this study.


Assuntos
Inflamação/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Substâncias Protetoras/farmacologia , Sumatriptana/farmacologia , Animais , Cardiotônicos/farmacologia , Inflamação/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos
13.
Microb Pathog ; 123: 28-35, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29908306

RESUMO

The Lactococcus lactis is known as a probiotic bacterium and also as a producer of nisin. Nisin has been approved by related legal agencies to be used as an antimicrobial peptide in food preservation. In fact, the L. lactis is present in different food products along with other micro-organisms especially pathogenic bacteria. So, it is important to predict the behavior of nisin-producer strain in contact with other pathogens. In this regard, nisin gene expression and the level of secreted biologically active form of nisin by L. lactis subsp. lactis in modified MRS broth and whey solution in co-culture with Listeria monocytogenes or Salmonella enterica were studied. The nisin concentration was determined by microbiological assay method and the transcription level of nisin gene was assayed through quantitative reverse transcription PCR (RT-qPCR). According to our results, the highest concentration of nisin and its gene transcription level were detected in mono- and co-cultures after 16 h of incubation, concurrent with the end of L. lactis exponential phase of growth. The nisin mRNA copies in co-cultures were higher than mono-cultures only at 16 h of incubation. But, differences between nisin concentrations in mono- and co-cultures were significant at 16, 24 h and at 12, 16, 24 h of incubation in the modified MRS medium and whey solution, respectively. This incompatibility could be related to the low availability of components required for nisin precursor modification, transportation and processing in mono-cultures. Overall, the L. lactis produced more mature and active nisin when it was in contact with pathogenic bacteria.


Assuntos
Antibacterianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Lactococcus lactis/metabolismo , Listeria monocytogenes/crescimento & desenvolvimento , Interações Microbianas/fisiologia , Nisina/metabolismo , Salmonella enterica/crescimento & desenvolvimento , Técnicas de Cocultura , Microbiologia de Alimentos , Conservação de Alimentos , Lactococcus lactis/genética , Lactococcus lactis/crescimento & desenvolvimento , Nisina/genética
14.
Tumour Biol ; 39(3): 1010428317695021, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28349824

RESUMO

Dendritic cells are important in initiating immune responses; therefore, a range of dendritic cell-based approaches have been established to induce immune response against cancer cells. However, the presence of immunosuppressive mediators such as adenosine in the tumor microenvironment reduces the efficacy of dendritic cell-based cancer immunotherapy. In this study, we investigated whether blockade of the A2A adenosine receptor with a selective antagonist and a CD73 inhibitor may increase the efficacy of a dendritic cell-based cancer vaccine. According to the findings, this therapeutic combination reduced tumor growth, prolonged survival of tumor-bearing mice, and enhanced specific antitumor immune responses. Thus, we suggest that targeting cancer-derived adenosine improves the outcomes of dendritic cell-based cancer immunotherapy.


Assuntos
5'-Nucleotidase/antagonistas & inibidores , Neoplasias da Mama/tratamento farmacológico , Vacinas Anticâncer/imunologia , Neoplasias Experimentais/tratamento farmacológico , Antagonistas de Receptores Purinérgicos P1/administração & dosagem , Receptor A2A de Adenosina/imunologia , 5'-Nucleotidase/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Terapia Baseada em Transplante de Células e Tecidos , Células Dendríticas/imunologia , Feminino , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Camundongos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Receptor A2A de Adenosina/genética , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
15.
Mol Cell Biochem ; 425(1-2): 95-102, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27826746

RESUMO

Protein tyrosine phosphatase 1B (PTP1B) has been shown to regulate multiple cellular events such as differentiation, cell growth, and proliferation; however, the role of PTP1B in differentiation of embryonic stem (ES) cells into cardiomyocytes remains unexplored. In the present study, we investigated the effects of PTP1B inhibition on differentiation of ES cells into cardiomyocytes. PTP1B mRNA and protein levels were increased during the differentiation of ES cells into cardiomyocytes. Accordingly, a stable ES cell line expressing PTP1B shRNA was established. In vitro, the number and size of spontaneously beating embryoid bodies were significantly decreased in PTP1B-knockdown cells, compared with the control cells. Decreased expression of cardiac-specific markers Nkx2-5, MHC-α, cTnT, and CX43, as assessed by real-time PCR analysis, was further confirmed by immunocytochemistry of the markers. The results also showed that PTP1B inhibition induced apoptosis in both differentiated and undifferentiated ES cells, as presented by increasing the level of cleaved caspase-3, cytochrome C, and cleaved PARP. Further analyses revealed that PTP1B inhibition did not change proliferation and pluripotency of undifferentiated ES cells. Taken together, the data presented here suggest that PTP1B is essential for proper differentiation of ES cells into cardiomyocytes.


Assuntos
Diferenciação Celular , Regulação Enzimológica da Expressão Gênica , Células-Tronco Embrionárias Murinas/enzimologia , Miócitos Cardíacos/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/biossíntese , Animais , Antígenos de Diferenciação/biossíntese , Linhagem Celular , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Miócitos Cardíacos/citologia
16.
Cell Biochem Funct ; 35(7): 407-413, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28975647

RESUMO

Mesenchymal stem cells (MSCs) have therapeutic potential for treatment of diabetes. However, in vitro behavior of MSCs in high glucose condition as well as presence of glucose lowering agents is not fully understood. Because MSCs have an important role in tissue repair, we examined the effects of metformin and celecoxib on viability of MSCs in different glucose conditions. MSCs, from umbilical cord blood, were cultured in normoglycemic (glucose 5.5 mM), midglycemic (glucose 10 mM), and hyperglycemic (glucose 25 mM) conditions, and the cell viability was evaluated by MTT assay. The cytotoxicity and secretion of GDF-15 were further tested in MSCs treated with metformin and celecoxib in various glucose concentrations. Our results showed that high glucose condition lowered viability of MSCs. Metformin treatment also inhibited proliferation of MSCs, but its toxicity was not changed in high glucose condition. Celecoxib induced cytotoxicity in MSCs, and the toxicity was increased in high glucose condition. Metformin and celecoxib induced release from MSCs; however, high glucose inhibited the metformin-induced GDF-15 release. These findings suggested that metformin did not increase the cytotoxicity of high glucose condition in MSCs. Moreover, celecoxib treatment in diabetic condition can reduce the viability of MSCs to proliferate and regenerate perhaps via change in release of GDF-15.


Assuntos
Celecoxib/toxicidade , Proliferação de Células/efeitos dos fármacos , Glucose/farmacologia , Fator 15 de Diferenciação de Crescimento/metabolismo , Metformina/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sangue Fetal/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo
17.
Pharm Dev Technol ; 22(3): 426-435, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27575893

RESUMO

Multiple sclerosis (MS) is a chronic central nervous system (CNS) inflammation. Efficient drug delivery to brain is however hampered by blood-brain barrier (BBB). In order to have highly efficient and safe delivery of drugs to brain, solid lipid nanoparticles (SLNs) have indicated promising potentials as smart carriers that can pass the blood-brain barrier and deliver therapeutic biomolecules to the brain. In this study, PEGylated SLNs surface modified using anti-Contactin-2 or anti-Neurofascin, two axo-glial-glycoprotein antigens located in node of Ranvier, were prepared. These targeting moieties are considered as the main targets of autoimmune reaction in MS. The targeted SLNs were then characterized and their in vitro release profile together with their cell viability and uptake were studied. Their brain uptakes were also probed following injections in MS-induced mice. It was found that the targeted PEGylated SLNs had no significant cytotoxicity on U87MG cells although their cellular uptake was increased 4- and 8-fold when surface modified with anti-Contactin-2 or anti-Neurofascin, respectively, compared to control. Brain uptake results demonstrated higher uptake of surface-modified SLNs in the brain tissue compared with the PEGylated SLNs. The results of this report will help scientist to design more efficient nanocarriers for treatment of MS.


Assuntos
Anti-Inflamatórios/administração & dosagem , Encéfalo/metabolismo , Moléculas de Adesão Celular/antagonistas & inibidores , Contactina 2/antagonistas & inibidores , Portadores de Fármacos/química , Metilprednisolona/administração & dosagem , Nanopartículas/química , Fatores de Crescimento Neural/antagonistas & inibidores , Animais , Anti-Inflamatórios/farmacocinética , Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/química , Encéfalo/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Endocitose/efeitos dos fármacos , Humanos , Metilprednisolona/farmacocinética , Metilprednisolona/uso terapêutico , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microscopia Eletrônica de Varredura , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Propriedades de Superfície
18.
Toxicol Ind Health ; 32(7): 1246-53, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27353300

RESUMO

OBJECTIVE: Polycyclic aromatic hydrocarbons (PAHs) are potent environmental pollutants. Benzo[α]pyrene (B[α]P) is the major compound of PAHs that acts by activating aryl hydrocarbon receptor (AhR) in cells. B[α]P is a known carcinogen and an immunotoxicant; however, its role with regard to nuclear factor of activated T cell (NFAT) pathway is unclear. AhR and NFAT signaling pathways have common roles in pathological functions in immunotoxicity and lung cancer. In this study, the effect of AhR activation on expression and signaling cross talk of AhR and NFATc1 pathways in mouse lung tissue has been investigated. METHODS: Swiss albino mice were randomly allocated to five groups and administered with cyclosporin A (CsA) and B[α]P for seven constitutive days. Animals were then killed, and lung tissues were obtained after washing the whole blood. Paraffin-embedded blocks were prepared, and 5 µm sections were cut for histopathological and immunohistochemical assessments. The results were scored by observer and digitally analyzed using ImageJ software. RESULTS: Our data showed that CsA administration resulted in a significant reduction of AhR expression. This effect was partly blocked in mice coadministrated with B[α]P and CsA. NFATc1 expression was also reduced in CsA-treated animals. Furthermore, CsA inhibited the pathological effects of B[α]P in mouse lung tissue. CONCLUSION: AhR expression is dependent on NFATc1 activation, and NFATc1 inhibition remarkably decreases AhR expression. However, it seems that total expression of NFATc1 is not dependent on AhR expression or activation. Moreover, CsA can prevent B[α]P-induced lung tissue damage, and it remarkably decreases NFATc1 expression. The results from this study point toward the molecular interactions of AhR and NFATc1 activation in lung tissue and the benefit of CsA treatment in B[α]P-induced lung damage.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Benzo(a)pireno/toxicidade , Pulmão/efeitos dos fármacos , Fatores de Transcrição NFATC/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Carcinógenos/toxicidade , Ciclosporina/farmacologia , Regulação da Expressão Gênica , Pulmão/metabolismo , Camundongos , Fatores de Transcrição NFATC/genética , Receptores de Hidrocarboneto Arílico/genética
19.
Phytother Res ; 29(7): 1046-53, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25857366

RESUMO

Despite several introduced preventive modalities, cisplatin nephrotoxicity remains a clinical problem. Some in vitro and in vivo studies have addressed the protective effects of silymarin against cisplatin nephrotoxicity. This study evaluated the effects of silymarin administration on cisplatin nephrotoxicity as the first human study. During this pilot, randomized, double-blinded, placebo-controlled clinical trial, the effect of oral silymarin 420 mg daily in three divided doses starting 24-48 h before the initiation of cisplatin infusion and continuing to the end of three 21-day cisplatin-containing chemotherapy courses on cisplatin-induced renal electrolytes wasting and kidney function were assessed. Cisplatin-associated acute kidney injury (AKI) occurred in 8% of the patients. Urine neutrophil gelatinase-associated lipocalin to urine creatinine ratio (NGAL/Cr) and urinary magnesium and potassium wasting increased significantly after cisplatin infusion in both groups. Significant positive correlation was found between cumulative dose of cisplatin and urine NGAL/Cr after three courses of cisplatin infusion. Incidence of AKI and the magnitude of urinary magnesium and potassium wasting did not differ between silymarin and placebo groups. No adverse reaction was reported by silymarin administration. Prophylactic administration of conventional form of silymarin tablets could not prevent cisplatin-induced urine electrolyte wasting or renal function impairment.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Cisplatino/efeitos adversos , Rim/efeitos dos fármacos , Silimarina/farmacologia , Proteínas de Fase Aguda/urina , Adulto , Creatinina/sangue , Creatinina/urina , Método Duplo-Cego , Feminino , Humanos , Testes de Função Renal , Lipocalina-2 , Lipocalinas/urina , Magnésio/urina , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Potássio/urina , Proteínas Proto-Oncogênicas/urina
20.
Toxicol Mech Methods ; 25(5): 388-95, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25980655

RESUMO

Previous report of the vast effectiveness of opium derivatives in cancer therapy is leading us to see possible effects of these derivatives on cancer stem cells in order to find new agent for cancer therapy. In this study, cells were stained for CSC markers and sorted by magnetic beads. CSCs exhibit the characteristic CD44(+)/CD24(-/low)/ESA(+) phenotype. Noscapine and papaverine (alkaloids) showed anti-proliferative activity on MCF-7 and MDA-MB-231 cell lines. It was observed that noscapine has more cytotoxic effect on CSC derived from both cell lines compared with their parental cells. Papaverine has more cytotoxic effect on MCF-7 CSCs in comparison with parental cells, while CSCs population of MDA-MB-231 is more resistant to papaverine compared with MDA-MB-231 cells. Noscapine enhances apoptosis in MDA-MB-231 CSCs more than parent cells, while in MCF-7 CSCs the apoptosis is less than parent cells. Our results show that papverine is less active in terms of apoptotic effect on CSCs in both cell lines. Moreover, noscapine arrests MCF-7 and MDA-MB-231 CSCs cell cycle at G2/M phase, while papverine arrests cell cycle at G0/G1 phase. It was suggested different mechanism for apoptotic cytotoxicity. The results of this study show possible specific effects of noscapine on these breast cell lines CSCs.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Noscapina/farmacologia , Papaverina/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Humanos , Concentração Inibidora 50 , Células MCF-7 , Células-Tronco Neoplásicas/patologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa