Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 22(1): 530, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38831317

RESUMEN

BACKGROUND: Cancer stem-like cells (CSCs) have been extensively researched as the primary drivers of therapy resistance and tumor relapse in patients with breast cancer. However, due to lack of specific molecular markers, increased phenotypic plasticity and no clear clinicopathological features, the assessment of CSCs presence and functionality in solid tumors is challenging. While several potential markers, such as CD24/CD44, have been proposed, the extent to which they truly represent the stem cell potential of tumors or merely provide static snapshots is still a subject of controversy. Recent studies have highlighted the crucial role of the tumor microenvironment (TME) in influencing the CSC phenotype in breast cancer. The interplay between the tumor and TME induces significant changes in the cancer cell phenotype, leading to the acquisition of CSC characteristics, therapeutic resistance, and metastatic spread. Simultaneously, CSCs actively shape their microenvironment by evading immune surveillance and attracting stromal cells that support tumor progression. METHODS: In this study, we associated in vitro mammosphere formation assays with bulk tumor microarray profiling and deconvolution algorithms to map CSC functionality and the microenvironmental landscape in a large cohort of 125 breast tumors. RESULTS: We found that the TME score was a significant factor associated with CSC functionality. CSC-rich tumors were characterized by an immune-suppressed TME, while tumors devoid of CSC potential exhibited high immune infiltration and activation of pathways involved in the immune response. Gene expression analysis revealed IFNG, CXCR5, CD40LG, TBX21 and IL2RG to be associated with the CSC phenotype and also displayed prognostic value for patients with breast cancer. CONCLUSION: These results suggest that the characterization of CSCs content and functionality in tumors can be used as an attractive strategy to fine-tune treatments and guide clinical decisions to improve patients therapy response.


Asunto(s)
Neoplasias de la Mama , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas , Microambiente Tumoral , Humanos , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Femenino , Transcripción Genética , Perfilación de la Expresión Génica , Línea Celular Tumoral , Esferoides Celulares/patología , Esferoides Celulares/metabolismo , Fenotipo
2.
Molecules ; 28(11)2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37298962

RESUMEN

In women, breast cancer is the most commonly diagnosed cancer (11.7% of total cases) and the leading cause of cancer death (6.9%) worldwide. Bioactive dietary components such as Sea buckthorn berries are known for their high carotenoid content, which has been shown to possess anti-cancer properties. Considering the limited number of studies investigating the bioactive properties of carotenoids in breast cancer, the aim of this study was to investigate the antiproliferative, antioxidant, and proapoptotic properties of saponified lipophilic Sea buckthorn berries extract (LSBE) in two breast cancer cell lines with different phenotypes: T47D (ER+, PR+, HER2-) and BT-549 (ER-, PR-, HER2-). The antiproliferative effects of LSBE were evaluated by an Alamar Blue assay, the extracellular antioxidant capacity was evaluated through DPPH, ABTS, and FRAP assays, the intracellular antioxidant capacity was evaluated through a DCFDA assay, and the apoptosis rate was assessed by flow cytometry. LSBE inhibited the proliferation of breast cancer cells in a concentration-dependent manner, with a mean IC50 of 16 µM. LSBE has proven to be a good antioxidant both at the intracellular level, due to its ability to significantly decrease the ROS levels in both cell lines (p = 0.0279 for T47D, and p = 0.0188 for BT-549), and at the extracellular level, where the ABTS and DPPH inhibition vried between 3.38-56.8%, respectively 5.68-68.65%, and 35.6 mg/L equivalent ascorbic acid/g LSBE were recorded. Based on the results from the antioxidant assays, LSBE was found to have good antioxidant activity due to its rich carotenoid content. The flow cytometry results revealed that LSBE treatment induced significant alterations in late-stage apoptotic cells represented by 80.29% of T47D cells (p = 0.0119), and 40.6% of BT-549 cells (p = 0.0137). Considering the antiproliferative, antioxidant, and proapoptotic properties of the carotenoids from LSBE on breast cancer cells, further studies should investigate whether these bioactive dietary compounds could be used as nutraceuticals in breast cancer therapy.


Asunto(s)
Hippophae , Neoplasias , Humanos , Antioxidantes/química , Carotenoides/química , Hippophae/química , Células MCF-7 , Frutas/química , Extractos Vegetales/química
3.
Medicina (Kaunas) ; 58(2)2022 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-35208642

RESUMEN

Glioblastoma multiforme (GBM) is one of the deadliest brain tumors. Current standard therapy includes tumor resection surgery followed by radiotherapy and chemotherapy. Due to the tumors invasive nature, recurrences are almost a certainty, giving the patients after diagnosis only a 12-15 months average survival time. Therefore, there is a dire need of finding new therapies that could potentially improve patient outcomes. Ferroptosis is a newly described form of cell death with several implications in cancer, among which GBM. Agents that target different molecules involved in ferroptosis and that stimulate this process have been described as potentially adjuvant anti-cancer treatment options. In GBM, ferroptosis stimulation inhibits tumor growth, improves patient survival, and increases the efficacy of radiation and chemotherapy. This review provides an overview of the current knowledge regarding ferroptosis modulation in GBM.


Asunto(s)
Neoplasias Encefálicas , Ferroptosis , Glioblastoma , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Humanos
4.
Nanotechnology ; 31(31): 315102, 2020 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-32315999

RESUMEN

Nowadays, extensive research is being carried out to find innovative solutions for the development of stable, reproductible, and highly efficient fluorescent contrast agents with the ability of targeting specific cells, which can be further implemented for fluorescent-guided surgery in a real clinical setting. The present study is focused on the development of fluorescent dye-loaded protein nanoparticles (NPs) to overcome the drawbacks of the standard administration of free organic fluorophores, such as cytotoxicity, aqueousinstability, and rapid photo-degradation. Precisely, human serum albumin (HSA) NPs loaded with two different FDA approved dyes, namely indocyanine green (ICG) and fluorescein isothiocyanate (FITC), with a fluorescence response in the near-infrared and visible spectral domains, respectively, have been successfully designed. Even though the diameter of fluorescent HSA NPs is around 30 nm as proven by dynamic light scattering and transmission electron microscopy investigations, they present good loading efficiencies of almost 50% for ICG, and over 30% for FITC and a high particle yield of over 75%. Molecular docking simulations of ICG and FITC within the structure of HSA confirmed that the dyes were loaded inside the NPs, and docked in Site I (subdomain IIA) of the HSA molecule. After the confirmation of their high fluorescence photostability, the NPs were covalently conjugated with folic acid (HSA-FA NPs) in order to bind specifically to the folate receptor alpha (FRα) protein overexpressed on NIH:OVCAR3 ovarian cancer cells. Finally, fluorescence microscopy imaging investigations validate the improved internalization of folate targeted HSA&FITC NPs compared to cells treated with untargeted ones. Furthermore, TEM examinations of the distribution of HSA NPs into the NIH:OVCAR3 cells revealed anincreased number of NP-containing vesicles for the cells treated with HSA-FA NPs, compared to the cells exposed to untargeted HAS NPs, upholding the enhanced cellular uptake through FRα-mediated potocytosis.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Colorantes Fluorescentes/química , Ácido Fólico/farmacología , Neoplasias Ováricas/metabolismo , Albúmina Sérica Humana/química , Línea Celular Tumoral , Supervivencia Celular , Femenino , Fluoresceína-5-Isotiocianato/química , Ácido Fólico/química , Humanos , Verde de Indocianina/química , Simulación del Acoplamiento Molecular , Nanopartículas , Regulación hacia Arriba
5.
Cell Mol Biol (Noisy-le-grand) ; 65(8): 18-22, 2019 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-32133974

RESUMEN

The aim of the present study was to examine characteristics of tuberculosis (TB) patients with different clinical forms and to study the frequency of Regulatory T cells (Treg cells) and Activated T cells in patients with new active and relapse TB. Forty-five pulmonary TB patients and a control group of 15 healthy individuals were enrolled in this study. Of the 45 TB patients, 15 were new cases with drug-susceptible active TB and 30 were relapsed cases (15 drug-susceptible and 15 multidrug resistant-TB). The age of study participants ranged from 21 to 68 years old. According to sex presentation, males were appreciably highly affected than females with a sex ratio of 2. The patients reported a mean recent weight loss of 8.9 kg. The Erythrocyte Sedimentation Rate was high in TB group, far exceeding the normal value. The results revealed that the number of CD3+ CD4+ T-cells significantly decreased whereas the level of blood Treg cells and expression of activation markers CD38 and HLA-DR on CD4+ T-cells significantly increased in TB group compared with the control group (p<0.05). The frequency of Treg cells was significantly higher in the TB group than the control group. Both the patients with new active TB and relapse TB demonstrated significantly higher levels of CD4+FoxP3+ Treg compared to healthy subjects (p<0.05). A high and significant percentage of Treg cells were found in patients with DS active TB than patients with MDR relapse TB. Interestingly, the frequency of CD4+FoxP3+ cells also differs according to the sputum smear microscopy status. The presence of high numbers of Treg cells and corresponding high immune activation may be an unfavourable factor that can predispose individuals to different clinical forms of TB, including relapse TB.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T Reguladores/metabolismo , Tuberculosis/patología , ADP-Ribosil Ciclasa 1/metabolismo , Adulto , Anciano , Sedimentación Sanguínea , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Estudios de Casos y Controles , Susceptibilidad a Enfermedades , Femenino , Factores de Transcripción Forkhead/metabolismo , Antígenos HLA-DR/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Recurrencia , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Tuberculosis/inmunología , Tuberculosis Resistente a Múltiples Medicamentos/inmunología , Tuberculosis Resistente a Múltiples Medicamentos/patología , Adulto Joven
6.
J BUON ; 23(3): 800-813, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30003755

RESUMEN

PURPOSE: Glioblastoma is a rapidly evolving lethal disease mainly due to its highly chemo- and radioresistant glioblastoma stem cells (GSCs). Herein, we tested if chitosan-capped gold nanoparticles (Chit-GNPs) may overcome the limitations of drug concentrations by increased cell internalization in GSCs and if such GNPs could enhance the response to irradiation. METHODS: Chitosan was used for Chit-GNP synthesis as a reducing and stabilizing agent. Chit-GNPs were characterized by spectroscopy, dark field, transmission electron microscopy and zeta potential measurements. Patient-derived GSCs and human osteoblasts were treated with increasing concentrations of nanoparticles and irradiated. The uptake and cytotoxicity of Chit-GNPs were compared to that of uncoated GNPs. RESULTS: The positively-charged, 26 nm-sized, spherical Chit-GNPs, showed a huge intracellular accumulation into the cytosol, lysosomes and near the nucleus, whereas no uncoated GNPs were internalized within GSCs. Surprisingly, Chit-GNPs were highly cytotoxic for GSCs irrespective of cell irradiation, that failed to add an additional benefit when combined with Chit-GNPs/GNPs. Moreover, Chit-GNPs were selectively cytotoxic for GSCs and did not affect the normal cells, despite an increased nanoparticle internalization. CONCLUSIONS: The important Chit-GNP internalization and their selective cytotoxicity for GSCs make this compound a potential novel anticancer agent and a promising backbone for drug delivery in glioblastoma.


Asunto(s)
Quitosano/administración & dosificación , Glioblastoma/tratamiento farmacológico , Oro/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Células Madre/efectos de los fármacos , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Humanos
7.
Mol Cell Biochem ; 419(1-2): 75-82, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27370646

RESUMEN

Oral squamous cell carcinoma (OSCC) is a malignancy with elevated prevalence and somber prognosis due to the fact that most of the patients are diagnosed at an advanced stage. p53 has a crucial role in proliferation and apoptosis during the occurrence and development of numerous malignant tumors. The impact of mutated p53 on the development and progression of OSCC is unclear and might have therapeutic implications. Using an in vitro RNA interference experiment, we have evaluated the impact of p53 knockdown on cell viability, apoptosis, migration, and gene expression for key genes involved in apoptosis and angiogenesis. We observed that inhibiting the expression of p53 decreased the proliferation ability and induced apoptosis/autophagy in SSC-4 cells. Moreover, we observed that this has decreased migration and has blocked the expression of VEGF. In conclusion, our research provides a proof that a direct connection between p53 knockdown and OSCC cell death can be established, therefore opening new potential directions in OSCC molecular therapeutics and management.


Asunto(s)
Apoptosis/genética , Autofagia/genética , Carcinoma de Células Escamosas , Movimiento Celular/genética , Neoplasias de la Boca , Proteína p53 Supresora de Tumor/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología
8.
Pharm Res ; 33(9): 2059-77, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27230936

RESUMEN

Extensive hypoxic regions are the daunting hallmark of glioblastoma, as they host aggressive stem-like cells, hinder drug delivery and shield cancer cells from the effects of radiotherapy. Nanotechnology could address most of these issues, as it employs nanoparticles (NPs) carrying drugs that selectively accumulate and achieve controlled drug release in tumor tissues. Methods overcoming the stiff interstitium and scarce vascularity within hypoxic zones include the incorporation of collagenases to degrade the collagen-rich tumor extracellular matrix, the use of multistage systems that progressively reduce NP size or of NP-loaded cells that display inherent hypoxia-targeting abilities. The unfavorable hypoxia-induced low pH could be converted into a therapeutical advantage by pH-responsive NPs or multilayer NPs, while overexpressed markers of hypoxic cells could be specifically targeted for an enhanced preferential drug delivery. Finally, promising new gene therapeutics could also be incorporated into nanovehicles, which could lead to silencing of hypoxia-specific genes that are overexpressed in cancer cells. In this review, we highlight NPs which have shown promising results in targeting cancer hypoxia and we discuss their applicability in glioblastoma, as well as possible limitations. Novel research directions in this field are also considered.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Nanopartículas/administración & dosificación , Animales , Preparaciones de Acción Retardada/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Humanos
9.
J Appl Toxicol ; 36(3): 373-84, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26397720

RESUMEN

Two polyoxometalates (POMs) with W were synthesized by a two-step, self-assembling method. They were used for stimulation of mesenchymal stem cell differentiation into insulin-producing cells. The nanocompounds (tris(vanadyl)-substituted tungsto-antimonate(III) anions [POM1] and tris-butyltin-21-tungsto-9-antimonate(III) anions [POM2]) were characterized by analytical techniques, including ultraviolet-visible, Fourier transform infrared, nuclear magnetic resonance spectroscopy, and transmission electron microscopy. We found that these polyoxotungstates, with 2-4 nm diameters, did not present toxic effects at the tested concentrations. In vitro, POM1 stimulated differentiation of a greater number of dithizone-positive cells (also organized in clusters) than the second nanocompound (POM2). Based on our in vitro studies, we have concluded that both the POMs tested had significant biological activity acting as active stimuli for differentiation of stem cells into insulin-producing cells.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Nanopartículas del Metal , Compuestos de Tungsteno/farmacología , Células Madre Adultas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Espectroscopía de Resonancia Magnética , Células Madre Mesenquimatosas/metabolismo , Nanopartículas del Metal/química , Nanopartículas del Metal/toxicidad , Microscopía Electrónica de Transmisión , Fenotipo , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja por Transformada de Fourier , Compuestos de Tungsteno/síntesis química , Compuestos de Tungsteno/toxicidad
10.
Microsc Microanal ; 22(5): 1018-1033, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27608930

RESUMEN

Resin composite materials that are used to restore tooth cervical lesions associated with gingival recessions can hamper healing after root coverage surgeries. This study evaluates the in vitro cytotoxic effect of five resin composites (two commercial and three experimental) on oral mesenchymal stem cells (MSCs) and the persistence of stemness properties in high passage MSCs. Sorption and solubility tests were made for all materials. MSCs were isolated from re-entry palatal and periodontal granulation tissues and were characterized and cultured on composite discs. Cytotoxicity of the materials was evaluated by the Alamar Blue viability test, by Paul Karl Horan (PKH) labeling, and by immunocytochemical staining for actin. Water and saliva sorption and solubility data revealed that two of the experimental materials behaved comparable with the marketed resin composites. The Alamar Blue viability test shows that both cell lines grew well on composite discs that seemed to induce no apparent toxic effects. No signs of disruption of cytoskeleton organization was seen. Experimental resin composites can be recommended for further investigation for obtaining approval for use. The standard minimal criteria were fulfilled for high passage MSCs. Palatal tissue regains its regenerative properties in terms of MSC presence in the re-entry area after 6 months of healing.


Asunto(s)
Resinas Compuestas/toxicidad , Materiales Dentales/toxicidad , Células Madre Mesenquimatosas/efectos de los fármacos , Línea Celular , Humanos , Ensayo de Materiales , Solubilidad
11.
J BUON ; 21(1): 199-207, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27061549

RESUMEN

PURPOSE: Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor. Despite maximal cytoreductive surgery followed by chemoradiotherapy the prognosis is still poor. Although surgery and radiotherapy may have reached maximal effectiveness, chemotherapy has the potential to improve survival. The aim of this study was to evaluate in vitro the antitumor efficacy of tamoxifen (TAM), raloxifen (RAL), pyrimethamine (PYR) and alphanizomenon flos-aquae (AFA) in combination with temozolomide (TMZ) plus ionizing radiation against cultured glioblastoma stem-like cells and primary glioblastoma cells , as possible way to increase the treatment options in patients with therapy-refractory GBM. METHODS: Stem-like tumor cells and glioblastoma cells isolated from two GBM biopsies were established by cell proliferation assays. TAM, RAL, PYR and AFA were added prior to TMZ and ionizing irradiation. RESULTS: All tested drugs enhanced the cytotoxic effect of TMZ and sensitized cancer cells to radiotherapy as demonstrated by MTT assay and different staining reagents (TMRE, Hoechst dye and Annexin V) used for monitoring of several events in apoptosis. CONCLUSION: The administration of certain selective estrogen receptor modulators (SERMs) (TAM and RAL), PYR and AFA before conventional postoperative chemo radiotherapy for glioblastoma might increase therapy efficiency compared to standard oncological treatment. These results need to be extended in vivo on laboratory animals in order to test the encouraging results obtained in vitro.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Glioblastoma/terapia , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Quimioradioterapia , Dacarbazina/uso terapéutico , Resistencia a Antineoplásicos , Glioblastoma/patología , Humanos , Temozolomida
12.
J Cell Mol Med ; 19(2): 371-82, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25311137

RESUMEN

The platelet-derived growth factor (PDGF) signalling pathway has been reported to play an important role in human cancers by modulating autocrine and paracrine processes such as tumour growth, metastasis and angiogenesis. Several clinical trials document the benefits of targeting this pathway; however, in cervical cancer the role of PDGF signalling in still unclear. In this study, we used siRNA against PDGF beta (PDGFBB) to investigate the cellular and molecular mechanisms of PDGFBB signalling in Ca Ski and HeLa cervical cancer cells. Our results show that PDGFBB inhibition in Ca Ski cells led to rapid alterations of the transcriptional pattern of 579 genes, genes that are known to have antagonistic roles in regulating tumour progression. Concomitantly, with the lack of significant effects on cervical cancer cells proliferation, apoptosis, migration or invasion, these findings suggests that cervical cancer cells shift between compensatory signalling pathways to maintain their behaviour. The observed autocrine effects were limited to cervical cancer cells ability to adhere to an endothelial cell (EC) monolayer. However, by inhibiting PDGFBB on cervical cells, we achieved reduced proliferation of ECs in co-culture settings and cellular aggregation in conditioned media. Because of lack of PDGF receptor expression on ECs, we believe that these effects are a result of indirect PDGFBB paracrine signalling mechanisms. Our results shed some light into the understanding of PDGFBB signalling mechanism in cervical cancer cells, which could be further exploited for the development of synergistic anti-tumour and anti-angiogenic therapeutic strategies.


Asunto(s)
Carcinogénesis/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Transducción de Señal/fisiología , Neoplasias del Cuello Uterino/metabolismo , Becaplermina , Línea Celular Tumoral , Femenino , Células HeLa , Humanos
13.
Drug Metab Rev ; 47(4): 455-69, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26689239

RESUMEN

Craniofacial bone structures are frequently and extensively affected by trauma, tumors, bone infections and diseases, age-related degeneration and atrophy, as well as congenital malformations and developmental anomalies. Consequently, severe encumbrances are imposed on both patients and healthcare systems due to the complex and lengthy treatment duration. The search for alternative methods to bone transplantation, grafting and the use of homologous or heterologous bone thus responds to one of the most significant problems in human medicine. This review focuses on the current consensus of bone-tissue engineering in the craniofacial area with emphasis on drug-induced stem cell differentiation and induced bone regeneration.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Cráneo/efectos de los fármacos , Ingeniería de Tejidos/métodos , Animales , Trasplante Óseo/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Humanos , Cráneo/patología , Cráneo/trasplante , Células Madre/citología , Células Madre/efectos de los fármacos , Andamios del Tejido
14.
BMC Biotechnol ; 15: 114, 2015 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-26718927

RESUMEN

BACKGROUND: We aimed to demonstrate that DF stem cells from impacted molars and canines can be used to improve bone regeneration on titanium implants surfaces. This study highlights the presence of stem cells in DF, their potential to adhere and differentiate into osteoblasts on different types of titanium surfaces. RESULTS: Isolated cells from the harvested DF tissue from impacted canine/molars, expressed stem cells markers. Differentiation into bone cells was induced in presence or absence of BMP-2 and TGFß1. The presence of growth factors until 28 days in medium maintained the cells in an earlier stage of differentiation with a lower level of specific bone proteins and a higher expression of alkaline phosphatase (ALP). Influence of titanium implants with different bioactive coatings, hydroxyapatite (TiHA) and with silicatitanate (TiSiO2), and porous Ti6Al7Nb implants as control (TiCtrl), was studied in terms of cell adhesion and viability. Ti HA implants proved to be more favorable for adhesion and proliferation of DF stem cells in first days of cultivation. The influence of titanium coatings and osteogenic differentiation mediums with or without growth factors were evaluated. Additional BMP-2 in the medium did not allow DF stem cells to develop a more mature phenotype, leaving them in a pre-osteogenic stage. The best sustained mineralization process evaluated by immuno-cytochemical staining, scanning electron microscopy and Ca(2+) quantification was observed for TiHA implants with a higher expression of ALP, collagen and Ca(2+) deposition. Long term culturing (70 days) on titanium surfaces of DF stem cells in standard medium without soluble osteogenic inducers, indicated that HA coating is more favorable, with the acquisition of a more mature osteoblastic phenotype as shown by immunocytochemical staining. These findings demonstrated that even in absence of exogenous osteogenic factors, TiHA implants and in a lesser extent TiCtrl and TiSiO2 implants can induce and sustain osteogenic differentiation of DF stem cells, by their chemical and topographical properties. CONCLUSIONS: Our research demonstrated that DF stem cells have a spontaneous tendency for osteogenic differentiation and can be used for improving bone regeneration on titanium implants surfaces.


Asunto(s)
Regeneración Ósea/fisiología , Implantes Dentales , Saco Dental/citología , Células Madre/citología , Titanio , Adolescente , Adulto , Fosfatasa Alcalina/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/metabolismo , Diente Canino/citología , Durapatita/química , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Diente Molar/citología , Osteoblastos/citología , Osteoblastos/fisiología , Osteogénesis , Adulto Joven
15.
Mol Cell Biochem ; 409(1-2): 163-76, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26187676

RESUMEN

Different molecular changes have been previously associated with therapeutic response and recurrent disease, however, the detailed mechanism of action in triple-negative breast cancer subtype remains elusive. In this study, we investigated the cellular and molecular signaling of two claudin-low triple-negative breast cancer cells to doxorubicin and docetaxel treatment. Whole human transcriptomic evaluation was used to identify the subsequent changes in gene expression, while biological effects were measured by means of proliferation and anchorage-independent growth assays. Microarray analysis revealed changes in stem cell-related signaling pathways, suggesting that doxorubicin treatment affects the balance between self-renewal and differentiation. While the treatment reduced the proliferation, aggregation and mammosphere forming ability of stem-like cells derived from Hs578T cell line, stem-like cells derived from MDA-MB-231 cells were not significantly affected. Our results suggest that claudin-low triple-negative breast cancer cells might predominantly alter stem cell-related signaling pathways to promote stem-like cells activity as an innate resistance mechanism to doxorubicin treatment.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/genética , Células Madre Neoplásicas/patología , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Docetaxel , Femenino , Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Esferoides Celulares/efectos de los fármacos , Taxoides/farmacología , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas
16.
J Appl Toxicol ; 35(10): 1200-10, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25900356

RESUMEN

The purpose of this study was to evaluate the biocompatibility of some composites consisting of different proportions of graphene in combination with gold nanoparticles (AuNPs) and nanostructured hydroxyapatite (HA) on osteoblast viability, proliferation and differentiation. Au/HA@graphene composites synthesized by the catalytic chemical vapor deposition induction heating method with acetylene as the carbon source and over an Au/HA catalyst, were characterized by transmission electron microscopy, thermogravimetric analysis and Raman spectroscopy and showed that the few-layer graphene was grown over the Au/HA catalyst. The cytocompatibility study was performed using the fluorescein diacetate assay for assessment of the viability and proliferation of osteoblasts cultivated in the presence of HA, Au/HA and Au/HA@graphene composites as colloidal suspensions or as substrates. The most favorable composites for cell adhesion and proliferation were HA, Au/HA and Au/HA composites with 1.6% and 3.15% concentration of graphenes. Immunocytochemical staining performed after 19 days of osteoblasts cultivation on substrates showed that the graphene composites induced low expression of alkaline phosphatase compared to the control group and HA and Au/HA substrates. The presence of graphene in the substrate composition also induced an increased level of intracellular osteopontin and cytoskeleton reorganization (actin-F) depending on graphene concentration, suggesting cell activation, increased cellular adhesion and acquisition of a mechanosensorial osteocyte phenotype.


Asunto(s)
Materiales Biocompatibles/toxicidad , Resinas Compuestas/toxicidad , Durapatita/toxicidad , Oro/toxicidad , Grafito/toxicidad , Nanopartículas del Metal/toxicidad , Osteoblastos/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Ensayo de Materiales
17.
Microsc Microanal ; 21(5): 1249-63, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26315895

RESUMEN

The aim of the present study was to isolate human mesenchymal stem cells (MSCs) from palatal connective and periodontal granulation tissues and to comparatively evaluate their properties. MSCs were isolated using the explant culture method. Adherence to plastic, specific antigen makeup, multipotent differentiation potential, functionality, and ultrastructural characteristics were investigated. The frequency of colony-forming unit fibroblasts for palatal-derived mesenchymal stem cells (pMSCs) was significantly higher than that of granulation tissue-derived mesenchymal stem cells (gtMSCs). A significantly higher population doubling time and lower migration potential were recorded for gtMSCs than for pMSCs. Both cell lines were positive for CD105, CD73, CD90, CD44, and CD49f, and negative for CD34, CD45, and HLA-DR, but the level of expression was different. MSCs from both sources were relatively uniform in their ultrastructure. Generally, both cell lines possessed a large, irregular-shaped euchromatic nucleus, and cytoplasm rich in mitochondria, lysosomes, and endoplasmic reticulum. The periphery of the plasma membrane displayed many small filopodia. MSCs from both cell lines were successfully differentiated into osteogenic, adiopogenic, and chondrogenic lineages. Both healthy and diseased tissues may be considered as valuable sources of MSCs for regenerative medicine owing to the high acceptance and fewer complications during harvesting.


Asunto(s)
Tejido Conectivo , Células Madre Mesenquimatosas/fisiología , Mucosa Bucal/citología , Adulto , Antígenos CD/análisis , Diferenciación Celular , Membrana Celular/ultraestructura , Movimiento Celular , Proliferación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Perfilación de la Expresión Génica , Antígenos HLA-DR/análisis , Voluntarios Sanos , Humanos , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/ultraestructura , Microscopía , Orgánulos/ultraestructura , Enfermedades Periodontales , Adulto Joven
18.
J BUON ; 20(4): 1107-14, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26416064

RESUMEN

PURPOSE: Oral squamous cell carcinoma (OSCC) is a disease with increased prevalence and unfavorable prognosis calling for development of novel therapeutic strategies. Tumor necrosis factor-α (TNF-α) is a pro-inflammatory cytokine implicated in the development and progression of cancer. The present study was designed to assess the impact of TNF-α specific inhibition using small interference RNA (siRNA) in SSC-4 cells, a representative model for OSCC. METHODS: The present study evaluated the effect of TNF-α inhibition using siRNA as inhibitory mechanism on SCC-4 cells. The study focused on the effect of TNF-α inhibition on apoptosis, autophagy and invasion in parallel with a panel of 20 genes involved in apoptosis and angiogenesis. RESULTS: TNF-α inhibition was related with reduction of cell viability, activation of apoptosis and autophagy in parallel with the inhibition of migration in SCC-4 cells. Evaluating the impact on gene expression levels, inhibition of FASL-FADD, NFκB, SEMA 3C, TNF-α, TGFB1, VEGFA, along with activation of PDGFB and SEMA 3D was observed. Our study confirms the important role of TNF-α and sustains that it might be a therapeutic target in OSCC. CONCLUSIONS: TNF-α is a key mediator of the immune system, with important role in OSCC tumorigenesis, and might be considered as a therapeutic target using siRNA technology, particularly for those risk cases having FASL/FADD overexpressed.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Neoplasias de la Boca/terapia , Interferencia de ARN , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Apoptosis , Autofagia , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Epistasis Genética , Humanos , Neoplasias de la Boca/patología , Factor de Crecimiento Transformador beta1/fisiología
19.
Nanotechnology ; 25(6): 065102, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24434767

RESUMEN

Multidimensional scaffolds are considered to be ideal candidates for regenerative medicine and tissue engineering based on their potential to provide an excellent microenvironment and direct the fate of the cultured cells. More recently, the use of stem cells in medicine has opened a new technological opportunity for controlled tissue formation. However, the mechanism through which the substrate directs the differentiation of stem cells is still rather unclear. Data concerning its specific surface chemistry, topology, and its signaling ability need to be further understood and analyzed. In our study, atomic force microscopy was used to study the stiffness, roughness, and topology of the collagen (Coll) and metallized collagen (MC) substrates, proposed as an excellent substrate for regenerative medicine. The importance of signaling molecules was studied by constructing a new hybrid signaling substrate that contains both collagen and laminin extracellular matrix (ECM) proteins. The cellular response-such as attachment capability, proliferation and cardiac and neuronal phenotype expression on the metallized and non-metallized hybrid substrates (collagen + laminin)-was studied using MTT viability assay and immunohistochemistry studies. Our findings indicate that such hybrid materials could play an important role in the regeneration of complex tissues.


Asunto(s)
Biomimética , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Materiales Biocompatibles/química , Supervivencia Celular , Colágeno/química , Matriz Extracelular/metabolismo , Humanos , Inmunohistoquímica , Laminina/química , Nanopartículas del Metal/química , Microscopía de Fuerza Atómica , Miocitos Cardíacos/citología , Neuronas/efectos de los fármacos , Fenotipo , Espectroscopía de Fotoelectrones , Regeneración , Transducción de Señal , Células Madre/citología , Propiedades de Superficie , Andamios del Tejido
20.
J BUON ; 19(2): 502-11, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24965413

RESUMEN

PURPOSE: Glioblastoma stem cells (GSCs), responsible for the dismal disease prognosis after conventional treatments, are driven by overactive signaling pathways, such as PI3K/ AKT/mTOR and RAS/RAF/MAPK. The objective of our study was to target in vitro-GSCs by combining metformin (Met) as a mTOR inhibitor, with sorafenib (Soraf) as a RAF inhibitor. METHODS: GSCs cultured under basal conditions were treated with Met, temozolomide (TMZ), Soraf, Met+TMZ and Met+Soraf; as untreated arm served as control. At 4 hrs of drug exposure, we measured the level of reactive oxygen species (ROS) by 2',7'-dichlorofluorescein diacetate (DCFDA) assay, apoptosis by prodium iodide (PI)-V Annexin staining and efflux pump activity by using the fluorescent dye rhodamine 123. At 24 hrs, we measured cell proliferation by 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) assay, apoptosis and malondialdehyde (MDA) levels. MTT results were compared with corresponding measurements on cultures of non-stem glioblastoma cells and osteoblasts. RESULTS: Met+Soraf exerted the highest antiproliferative effects in GSCs and non-stem glioblastoma cells (p<0.001). Both Met and Soraf monotherapy exhibited a selective cytotoxic effect on GSCs (p<0.001), while no effect was detected on non-stem glioblastoma cells (p>0.05). Soraf, but not Met, impacted the proliferation of normal cells. Soraf displayed synergism with Met in producing high levels of ROS, decreasing efflux pump activity and generating the highest apoptotic rates when compared to either drug alone (p<0.001). CONCLUSION: GSCs were highly sensitive to the combination of Met and Soraf which reduced cell proliferation, increased oxidative stress, inhibited efflux pump activity and ultimately killed GSCs. We strongly believe that these results warrant further in vivo exploration.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Metformina/administración & dosificación , Células Madre Neoplásicas/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Quinasas raf/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Dacarbazina/uso terapéutico , Resistencia a Antineoplásicos , Glioblastoma/patología , Humanos , Peroxidación de Lípido , Células Madre Neoplásicas/metabolismo , Niacinamida/administración & dosificación , Estrés Oxidativo , Rodamina 123/metabolismo , Sorafenib , Temozolomida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA