Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Biol Rep ; 51(1): 599, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689181

RESUMEN

BACKGROUND: CPUK02 (15-Oxosteviol benzyl ester) is a semi-synthetic derivative of stevioside known for its anticancer effects. It has been reported that the natural compound of stevioside and its associated derivatives enhances the sensitivity of cancer cells to conventional anti-cancer agents by inducing endoplasmic reticulum (ER) stress. In response to ER stress, autophagy and unfolded protein responses (UPR) are activated to restore cellular homeostasis. Consequently, the primary aim of this study is to investigate the impact of CPUK02 treatment on UPR and autophagy markers in two colorectal cancer cell lines. METHODS: HCT116 and SW480 cell lines were treated with various concentrations of CPUK02 for 72 h. The expression levels of several proteins and enzymes were evaluated to investigate the influence of CPUK02 on autophagy and UPR pathways. These include glucose-regulated protein 78 (GRP78), Inositol-requiring enzyme 1-α (IRE1-α), spliced X-box binding protein 1 (XBP-1 s), protein kinase R-like ER kinase (PERK), C/EBP homologous protein (CHOP), Beclin-1, P62 and Microtubule-associated protein 1 light chain 3 alpha (LC3ßII). The evaluation was conducted using western blotting and quantitative real-time PCR techniques. RESULTS: The results obtained indicate that the treatment with CPUK02 reduced the expression of UPR markers, including GRP78 and IRE1-α at protein levels and XBP-1 s, PERK, and CHOP at mRNA levels in both HCT116 and SW480 cell lines. Furthermore, CPUK02 also influenced autophagy by decreasing Beclin-1 and increasing P62 and LC3ßII at mRNA levels in both HCT116 and SW480 treated cells. CONCLUSIONS: The study findings suggest CPUK02 may exert its cytotoxic effects by inhibiting UPR and autophagy flux in colorectal cancer cells.


Asunto(s)
Autofagia , Neoplasias Colorrectales , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Respuesta de Proteína Desplegada , Humanos , Autofagia/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células HCT116 , Línea Celular Tumoral , Diterpenos de Tipo Kaurano/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Proteína 1 de Unión a la X-Box/metabolismo , Proteína 1 de Unión a la X-Box/genética
2.
Ecotoxicol Environ Saf ; 276: 116316, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38615640

RESUMEN

Aflatoxins B1 (AFB1) a dangerous type of aflatoxin, poses a serious threat to human health. Meanwhile, Taraxasterol, a bioactive compound in dandelion, exhibits strong anti-inflammatory and antioxidant activity. Therefore, the aim of this study was to investigate the impact of AFB1 on the intrinsic and extrinsic pathways of apoptosis, as well as evaluate the protective role of taraxasterol in the TM3 Leydig cell line. Cell viability was evaluated using an MTT assay, measuring the effects of 3.6 µM AFB1 and varying concentrations of taraxasterol. Expression levels of Caspase 3,8, and 9 were analyzed with RT-qPCR, and flow cytometry was used to assess cell cycle progression and apoptotic alterations. The findings of this study demonstrated that exposure to 3.6 µM of AFB1 resulted in an upregulation of Caspase 3 and Caspase 9 expression, indicating an activation of apoptotic pathways in TM3 cells. Additionally, the analysis of apoptosis revealed a significant increase in cellular apoptosis at this AFB1 concentration. However, when TM3 cells were exposed to 5 µM of taraxasterol, a downregulation of Caspase 3 and Caspase 9 expression was observed, suggesting a protective effect against apoptosis. Moreover, the apoptotic rate in TM3 cells was reduced in the presence of 5 µM of taraxasterol. Consequently, this study highlights the potential of taraxasterol as a protective agent against AFB1-induced apoptosis and suggest its potential application in regulating cell survival and apoptosis-related processes. Further investigations are necessary to elucidate the underlying mechanisms and evaluate the clinical implications of taraxasterol in the context of fertility disorders and other conditions associated with AFB1 exposure.


Asunto(s)
Aflatoxina B1 , Apoptosis , Supervivencia Celular , Células Intersticiales del Testículo , Triterpenos , Aflatoxina B1/toxicidad , Apoptosis/efectos de los fármacos , Células Intersticiales del Testículo/efectos de los fármacos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ratones , Masculino , Triterpenos/farmacología , Esteroles/farmacología , Caspasa 3/metabolismo , Sustancias Protectoras/farmacología , Caspasa 9/metabolismo
3.
Front Immunol ; 14: 1280601, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38022534

RESUMEN

Malignancies contain a relatively small number of Mesenchymal stem/stromal cells (MSCs), constituting a crucial tumor microenvironment (TME) component. These cells comprise approximately 0.01-5% of the total TME cell population. MSC differentiation potential and their interaction with the tumor environment enable these cells to affect tumor cells' growth, immune evasion, metastasis, drug resistance, and angiogenesis. This type of MSC, known as cancer-associated mesenchymal stem/stromal cells (CA-MSCs (interacts with tumor/non-tumor cells in the TME and affects their function by producing cytokines, chemokines, and various growth factors to facilitate tumor cell migration, survival, proliferation, and tumor progression. Considering that the effect of different cells on each other in the TME is a multi-faceted relationship, it is essential to discover the role of these relationships for targeting in tumor therapy. Due to the immunomodulatory role and the tissue repair characteristic of MSCs, these cells can help tumor growth from different aspects. CA-MSCs indirectly suppress antitumor immune response through several mechanisms, including decreasing dendritic cells (DCs) antigen presentation potential, disrupting natural killer (NK) cell differentiation, inducing immunoinhibitory subsets like tumor-associated macrophages (TAMs) and Treg cells, and immune checkpoint expression to reduce effector T cell antitumor responses. Therefore, if these cells can be targeted for treatment so that their population decreases, we can hope for the treatment and improvement of the tumor conditions. Also, various studies show that CA-MSCs in the TME can affect other vital aspects of a tumor, including cell proliferation, drug resistance, angiogenesis, and tumor cell invasion and metastasis. In this review article, we will discuss in detail some of the mechanisms by which CA-MSCs suppress the innate and adaptive immune systems and other mechanisms related to tumor progression.


Asunto(s)
Células Madre Mesenquimatosas , Neoplasias , Humanos , Citocinas/metabolismo , Diferenciación Celular , Inmunidad , Células Madre Mesenquimatosas/metabolismo , Microambiente Tumoral
4.
Cell J ; 25(7): 470-482, 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37543860

RESUMEN

OBJECTIVE: Endoplasmic reticulum-metallopeptidase 1 (ERMP1) is involved in cellular response to oxidative stress. However, its functional role in proliferation and progression of cancer cells remains unknown. The focus of this study was to investigate the molecular-mechanisms in which ERMP1 modulates the proliferation and progression of colorectal cancer (CRC) cells under normal and environment stress conditions. MATERIALS AND METHODS: In this experimental study, ERMP1 expression was evaluated using reverse transcriptionquantitative polymerase chain reaction (RT-qPCR) in CRC cells. ERMP1 was knocked down using lentiviral transduction of ERMP1-specific shRNA into HCT116 cells. ERMP1 was also upregulated using lipofectamine transfection of ERMP1-overexpressing vector into SW48 cells. To evaluate the role of ERMP1 in the cellular and environmental stress conditions, ERMP1-downregulated cells were exposed to stressful conditions including starvation, serum free medium, and treatment with redox or chemotherapy agents for 72 hours. The expression of AKT, p-AKT, phospho-mammalian target of rapamycin (p-mTOR), ß-catenin, p-ß-catenin, E-cadherin, and Glucose-regulating protein 78 (GRP78) proteins was evaluated by western blotting. The expression of ERMP1, CYCLIN D, and c-MYC was evaluated by RT-qPCR. The cell surface localization of GRP78, cell cycle distribution, and apoptosis were determined by Flow cytometry. RESULTS: ERMP1 knock-down reduced the cellular proliferation, inactivated the PI3K/AKT pathway, prompted the G1 arrest, and attenuated the free ß-catenin and CYCLIN D expression. Opposite results were obtained in ERMP1- overexpressed cells. Knock-down of ERMP1 also reduced the GRP78 localization at the cell surface. Various environmental stress conditions differently affected the ERMP1-downregulated cells. CONCLUSION: ERMP1 functioned as an oncogene in CRC cells by promoting malignant characteristics. The phosphoinositide 3-kinases (PI3K)/AKT/ß-catenin pathway and localization of GRP78 were closely related to the effects of ERMP1. Consequently, ERMP1 might be regarded as a promising target in therapeutic strategies related to CRC.

5.
Exp Neurol ; 366: 114434, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37201745

RESUMEN

Long non-coding RNAs (lncRNAs) cannot be coded to proteins; however, they can display important functions in several aspects of cell biology. Their abnormal expression is verified in various disorders, including neurodegenerative diseases, especially Alzheimer's disease (AD). By acting as a cell cycle suppressor or promotor, lncRNAs mediate some signaling pathways, which in turn lead to exacerbation or improvement of AD. Wnt/ß-catenin signaling pathway, as an important pathway in the pathogenesis of AD, can extremely be affected by lncRNAs. This pathway participates in various biological processes, such as embryogenesis and tissue homeostasis, and is involved in expanding the central nervous system, such as synaptogenesis, plasticity, and hippocampal neurogenesis. lncRNAs can regulate the expression of Wnt pathway target genes by interacting with various components of this pathway. This article discusses lncRNAs and their associated mechanisms in the alteration of Wnt/ß-catenin signaling, which can be regarded as a new aspect of diagnosing and treating AD.


Asunto(s)
Enfermedad de Alzheimer , ARN Largo no Codificante , Humanos , Enfermedad de Alzheimer/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Ciclo Celular , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Vía de Señalización Wnt/genética
6.
Biomed Pharmacother ; 162: 114615, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37011484

RESUMEN

Cell therapy is one of the methods that have shown promising results in treating diseases in recent decades. However, the use of different types of cells comes with limitations. The application of immune cells in cell therapy can lead to cytokine storms and inappropriate responses to self-antigens. Also, the use of stem cells has the potential to create tumors. Also, cells may not migrate to the injury site after intravenous injection. Therefore, using exosomes from different cells as therapeutic candidates were proposed. Due to their small size and favorable characteristics, such as biocompatibility and immunocompatibility, the easy storage and isolation, exosomes have attracted much attention. They are used in treating many diseases, including cardiovascular diseases, orthopedic diseases, autoimmune diseases, and cancer. However, the results of various studies have shown that the therapeutic efficiency of exosomes (Exo) can be increased by loading different drugs and microRNAs inside them (encapsulated exosomes). Therefore, analyzing studies investigating encapsulated exosomes' therapeutic ability is critical. In this study, we have examined the studies related to the use of encapsulated exosomes in treating diseases such as cancer and infectious diseases and their use in regenerative medicine. Compared to intact exosomes, the results show that the application of encapsulated exosomes has a higher therapeutic ability. Therefore it is suggested to use this method depending on the treatment type to increase the treatment's efficiency.


Asunto(s)
Exosomas , MicroARNs , Exosomas/metabolismo , MicroARNs/metabolismo , Células Madre , Medicina Regenerativa
7.
Mol Ther Nucleic Acids ; 29: 705-717, 2022 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-35992045

RESUMEN

The occurrence of viral infections and approaches to handling them are very challenging and require prompt diagnosis and timely treatment. Recently, genomic medicine approaches have come up with the discovery of the competing endogenous RNA (ceRNA) network, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) on the basis of gene silencing. CircRNAs, as a group of non-encoded RNAs, make a loop-like structure by back-splicing through 3' and 5' ends. They are stable, abundant, specific, and highly conserved and can be quickly generated at large scales in vitro. CircRNAs have the potential to contribute in several cellular processes in a way that some serve as microRNA sponges, cellular transporters, protein-binding RNAs, transcriptional regulators, and immune system modulators. CircRNAs can even play an important role in modulating antiviral immune responses. In the present review, circRNAs' biogenesis, function, and biomarker and therapeutic potential as well as their prospective applications as vaccines against viral infections such as SARS-CoV-2 are explained. By considering their unique properties, their potential to be used as novel vaccines, biomarkers, and a therapeutic approach appears possible.

8.
Mini Rev Med Chem ; 22(12): 1619-1630, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34645371

RESUMEN

Sarcoidosis is a worldwide inflammatory disorder of unknown etiology that is characterized by the formation of non-caseating immune granulomas in involved organs,most commonly in the lungs and eyes. Although clinical manifestations of sarcoidosis depend on the organs involved, the most common symptoms include fatigue, fever, weight loss, eye pain, dyspnea, and chest pain. Sarcoidosis usually undergoes spontaneous regression, yet its chronic form progressively threatens the involved organs through the induction of fibrotic damage. Despite decades of medical research, the etiology of sarcoidosis still remains unclear. Nevertheless, a combination of contributors, including genetic factors, environmental exposures, and microbial agents, is believed to trigger the inflammatory state observed in this disease. Furthermore, a highly polarized Th1 and Th17 response with diminished immunomodulatory mechanisms constitute the most significant immunological event associated with this disorder. Indeed, sarcoid granulomas, which consist of highly activated antigen-presenting cells (APCs) and lymphocytes, maintain a robust specialized niche to facilitate antigen presentation and exaggerated immune responses. Both the unknown etiology and multisystem nature of the disease have hampered the development of specific therapeutics and definitive diagnostic assays for sarcoidosis. Consequently, its diagnosis and treatment still represent a challenging task for clinicians. In this article, we aim to summarize contemporary findings of sarcoidosis and its etiology, pathogenesis, and treatment.


Asunto(s)
Sarcoidosis , Granuloma/diagnóstico , Granuloma/tratamiento farmacológico , Humanos , Sarcoidosis/diagnóstico , Sarcoidosis/tratamiento farmacológico
9.
Int J Mol Sci ; 22(11)2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-34206057

RESUMEN

The COVID-19 pandemic is caused by the 2019-nCoV/SARS-CoV-2 virus. This severe acute respiratory syndrome is currently a global health emergency and needs much effort to generate an urgent practical treatment to reduce COVID-19 complications and mortality in humans. Viral infection activates various cellular responses in infected cells, including cellular stress responses such as unfolded protein response (UPR) and autophagy, following the inhibition of mTOR. Both UPR and autophagy mechanisms are involved in cellular and tissue homeostasis, apoptosis, innate immunity modulation, and clearance of pathogens such as viral particles. However, during an evolutionary arms race, viruses gain the ability to subvert autophagy and UPR for their benefit. SARS-CoV-2 can enter host cells through binding to cell surface receptors, including angiotensin-converting enzyme 2 (ACE2) and neuropilin-1 (NRP1). ACE2 blockage increases autophagy through mTOR inhibition, leading to gastrointestinal complications during SARS-CoV-2 virus infection. NRP1 is also regulated by the mTOR pathway. An increased NRP1 can enhance the susceptibility of immune system dendritic cells (DCs) to SARS-CoV-2 and induce cytokine storm, which is related to high COVID-19 mortality. Therefore, signaling pathways such as mTOR, UPR, and autophagy may be potential therapeutic targets for COVID-19. Hence, extensive investigations are required to confirm these potentials. Since there is currently no specific treatment for COVID-19 infection, we sought to review and discuss the important roles of autophagy, UPR, and mTOR mechanisms in the regulation of cellular responses to coronavirus infection to help identify new antiviral modalities against SARS-CoV-2 virus.


Asunto(s)
Autofagia , COVID-19/patología , Neuropilina-1/metabolismo , Respuesta de Proteína Desplegada , Antivirales/farmacología , Autofagia/efectos de los fármacos , COVID-19/virología , Humanos , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo
10.
Front Endocrinol (Lausanne) ; 12: 576412, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746897

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has recently emerged, which was then spread rapidly in more than 190 countries worldwide so far. According to the World Health Organization, 3,232,062 global cases of COVID-19 were confirmed on April 30th with a mortality rate of 3.4%. Notably, the symptoms are almost similar to those of flu such as fever, cough, and fatigue. Unfortunately, the global rates of morbidity and mortality caused by this disease are more and still increasing on a daily basis. The rates for patients suffering from inflammatory diseases like diabetes, is even further, due to their susceptibility to the pathogenesis of COVID-19. In this review, we attempted to focus on diabetes to clarify the physiological and immunological characteristics of diabetics before and after the infection with COVID-19. We hope these conceptions could provide a better understanding of the mechanisms involved in COVID-19 susceptibility and increase the awareness of risk to motivate behavior changes in vulnerable people for enhancing the prevention. Up to now, the important role of immune responses, especially the innate ones, in the development of the worst signs in COVID-19 infection have been confirmed. Therefore, to better control patients with COVID-19, it is recommended to consider a history of chronic inflammatory diseases as well as the way of controlling immune response in these patients.


Asunto(s)
COVID-19/epidemiología , Complicaciones de la Diabetes/epidemiología , Inflamación/complicaciones , COVID-19/complicaciones , Enfermedad Crónica , Humanos , Inflamación/epidemiología
12.
Bioorg Chem ; 105: 104429, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33161256

RESUMEN

Human serum albumin (HSA) as the most abundant protein in human blood plasma, can be a good indicator for evaluating severity of some diseases in the clinic. HSA can be find in two forms: reduced albumin (human mercaptalbumin (HMA)) and oxidized albumin (human non-mercaptalbumin (HNA)). The rate of oxidized albumin to total albumin can be enhanced in multiple diseases. Increase in HNA level have been demonstrated in liver, diabetes plus fatigue and coronary artery diseases. In liver patients, this enhancement can reach to 50-200 percent which can then lead to bacterial/viral infections and eventually death in severe conditions. Due to the induction of cytokine storm, we can say that the level of HNA in serum of coronavirus disease 2019 (COVID-19) patients may be a positive predictor of mortality, especially in patients with underlying diseases such as cardiovascular disease (CVD), diabetes, aging and other inflammatory diseases. We suggest that checking oxidized albumin in COVID-19 patients may provide new therapeutic and diagnostic opportunities to better combat COVID-19.


Asunto(s)
COVID-19/diagnóstico , Albúmina Sérica Humana/análisis , COVID-19/terapia , COVID-19/virología , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Hígado/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/química , Especies Reactivas de Oxígeno/metabolismo , SARS-CoV-2/aislamiento & purificación , Albúmina Sérica/análisis , Albúmina Sérica/química , Albúmina Sérica Humana/química
13.
Int Immunopharmacol ; 84: 106602, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32417655

RESUMEN

OBJECTIVES: Mesenchymal stem cells (MSCs) can influence immune effector cells. It is proved that MSCs respond to various Toll-like receptor (TLR) ligands, which could ultimately result in changes in their immunomodulatory effects. Neutrophils play an essential role in the first line defense system and their function can be regulated by MSCs. Estrogen is a female hormone that contributes to sex differences in several immune-related diseases. With regard to the stated facts, this research aims to elucidate the effects of estrogen treatment on the ability of TLR4-primed MSCs to regulate neutrophil functions. METHODS: Following isolation and characterization, MSCs were stimulated with LPS as a TLR4 ligand and subsequently incubated with different concentrations (0, 10, 20 and 40 nM) of estrogen for 48 hrs. Then, MSCs were co-cultured with neutrophils to investigate the vitality and function of the co-cultured neutrophils. RESULTS: Our results indicated that TLR4-primed MSCs could decrease the viability and neutral red uptake potential of co-cultured neutrophils. Furthermore, neutrophils co-cultured with TLR4-primed MSCs exhibited a decrease in the respiratory burst intensity after being challenged with opsonized yeast. Interestingly, treating TLR4-primed MSCs with estrogen reversed the observed alterations in neutrophil functions. CONCLUSION: It appears that estrogen can alter the interaction between MSCs and neutrophils.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Receptor Toll-Like 4/inmunología , Adipogénesis , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Lipopolisacáridos/farmacología , Masculino , Osteogénesis , Ratas Wistar
14.
Asian Pac J Cancer Prev ; 19(10): 2821-2829, 2018 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-30361551

RESUMEN

Background: Due to the possible biomedical potential of nanoparticles, titanium dioxide nanoparticles (TiO2 NPs) have received great attention in cancer research. Although selectivity of cytotoxicity with TiO2 NPs in various cells is clinically significant comparisons of cancer and non-cancer cells have been limited. Therefore, we here studied exposure to TiO2 NPs in colorectal cancer cells (CRCs) and human umbilical vein endothelial cells (HUVECs). Methods: After characterization of TiO2 NPs, culture and treatment of cells (HCT116, HT29 and HUVEC), viability was assessed by MTT assay and in terms of morphological features. Acridine orange (AO) and propidium iodide (PI) assays were carried out to estimate the incidence of apoptosis. The RT-PCR method was also employed to evaluate the expression of P53, Bax, Bcl-2 and Caspase 3. Results: Exposure to increasing concentrations of TiO2 NPs enhanced overall cell survival of HCT116 cells and reduced the Bcl-2 and Caspase 3 expression while the ratio of Bax/Bcl-2 was down-regulated. TiO2 NPs at 400 and 50 µg/ml concentrations suppressed cell proliferation and induced apoptosis of HT29 cells and also up-regulated P53 and Bax at the mRNA level, enhanced the Bax/Bcl-2 ratio and eventually up-regulated Caspase 3 mRNA. Although, inhibition of cell proliferation in HUVECs was seen at 200 and 400 µg/ml TiO2 NPs, it was not marked. Conclusion: TiO2 NPs have selective bio-effects on exposed cells with dose- and cell-dependent influence on viability. Cell proliferation in HCT116 as a metastatic colorectal cancer cell line appeared to be stimulated via multiple signaling pathways, with promotion of apoptosis in less metastatic cells at 50 and 400 µg/ml concentrations. This was associated with elevated P53, Bax and Caspase 3 mRNA and reduced Bcl-2 expression. However, TiO2 NPs did not exert any apparent significant effects on HUVECs as hyperproliferative angiogenic cells.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Nanopartículas del Metal/administración & dosificación , Titanio/farmacología , Venas Umbilicales/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Células Endoteliales/metabolismo , Células HCT116 , Células HT29 , Células Endoteliales de la Vena Umbilical Humana , Humanos , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Venas Umbilicales/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
15.
Asian Pac J Cancer Prev ; 19(7): 1895-1902, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-30049202

RESUMEN

Background: Immunotherapy is one promising therapeutic strategy against glioma, an aggressive form of brain cancer. Previous studies have demonstrated that multiple tumor antigens exist and can be used to induce tumor specific T cell responses. Furthermore, recently it was shown that TLR4-primed mesenchymal stem cells (MSCs), also known as MSC1, mostly elaborate pro-inflammatory mediators. Compared to MSCs, MSC-derived microvesicles (MVs) have advantageous properties that present them as stable, long lasting effectors with no risk of immune rejection. Therefore, peripheral blood monocyte derived dendritic cells (MoDCs) have been used to load tumor antigens and stimulate T cell mediated responses in the presence of MSC1-derived MVs in vitro. Methods: The B92 tumor cell line was heated to 43°C for 90 min prior to preparation of tumor cell lysates. MVs were purified by differential ultracentrifugation after isolation, stimulation of proliferation and treatment of MSCs. Autologous T cells isolated from non-adherent cells were harvested during the procedure to generate MoDCs and then incubated with heat stressed tumor cell lysate pulsed DCs in the presence of MSC1-derived MVs. T cells were then co-cultured with tumor cells in 96-well plates at a final volume of 200 µl CM at an effector: target ratio of 100:1 to determine their specific cytotoxic activity. Results: Flow cytometric analysis, T cell mediated cytotoxicity showed that heat stressed tumor antigen pulsed MoDCs and MSC1-derived MVs primed T cells elicited non-significantly enhanced cytotoxic activity toward B92 tumor cells (P≥0.05). Conclusion: These findings may offer new insights into tumor antigen presenting technology involving dendritic cells and MSC1-derived MVs. Further exploration of the potential of such nanoscale particles in immunotherapy and in novel cancer vaccine settings appears warranted.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/inmunología , Glioma/inmunología , Células Madre Mesenquimatosas/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Técnicas de Cocultivo , Glioma/terapia , Inmunoterapia , Activación de Linfocitos , Ratas , Ratas Wistar , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...