Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Genomics ; 116(3): 110853, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38701988

RESUMEN

Atg8 family proteins play crucial roles in autophagy to maintain cellular homeostasis. However, the physiological roles of Atg8 family proteins have not been systematically determined. In this study, we generated Atg8a and Atg8b (homologs of Atg8 in Drosophila melanogaster) knockout flies. We found that the loss of Atg8a affected autophagy and resulted in partial lethality, abnormal wings, decreased lifespan, and decreased climbing ability in flies. Furthermore, the loss of Atg8a resulted in reduced muscle integrity and the progressive degeneration of the neuron system. We also found that the phosphorylation at Ser88 of Atg8a is important for autophagy and neuronal integrity. The loss of Atg8b did not affect autophagy but induced male sterility in flies. Here, we take full advantage of the fly system to elucidate the physiological function of Atg8a and Atg8b in Drosophila.


Asunto(s)
Familia de las Proteínas 8 Relacionadas con la Autofagia , Autofagia , Proteínas de Drosophila , Drosophila melanogaster , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Masculino , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Drosophila melanogaster/metabolismo , Familia de las Proteínas 8 Relacionadas con la Autofagia/metabolismo , Familia de las Proteínas 8 Relacionadas con la Autofagia/genética , Fosforilación , Longevidad , Neuronas/metabolismo , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo
2.
Nanoscale Adv ; 6(1): 72-78, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38125595

RESUMEN

Nanovaccines have emerged as promising agents for cancer therapy because of their ability to induce specific immune responses without off-target effects. However, inadequate cytotoxic T lymphocyte response and low antigen/adjuvant encapsulation remain major obstacles to vaccinating against cancer. Herein, we designed a stimulator of interferon genes (STING) pathway-activating nanovaccine based on hollow metal-organic frameworks (MOFs) for tumor treatment. The nanovaccine (OVA@HZIF-Mn) was constructed by encapsulating a model antigen ovalbumin (OVA) into zeolitic imidazolate framework-8, followed by etching with tannic acid and functionalizing with manganese ions. Studies have shown that the nanovaccine can effectively enhance antigen uptake, STING pathway activation and dendritic cell maturation, triggering a robust immune response to inhibit tumor growth. In addition, no infection or pathological signs were observed in mice organs after multiple administrations. This study combines a simple assembly approach and superior therapeutic effect, providing a promising strategy for engineering effective nanovaccines.

3.
Adv Healthc Mater ; 12(22): e2300249, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37016572

RESUMEN

Nanovaccine-based immunotherapy can initiate strong immune responses and establish a long-term immune memory to prevent tumor invasion and recurrence. Herein, the assembly of redox-responsive antigen nanoparticles (NPs) conjugated with imidazoquinoline-based TLR7/8 agonists for lymph node-targeted immune activation is reported, which can potentiate tumor therapy and prevention. Antigen NPs are assembled via the templating of zeolitic imidazolate framework-8 NPs to cross-link ovalbumin with disulfide bonds, which enables the NPs with redox-responsiveness for improved antigen cross-presentation and dendritic cell activation. The formulated nanovaccines promote the lymphatic co-delivery of antigens and agonists, which can trigger immune responses of cytotoxic T lymphocytes and strong immunological memory. Notably, nanovaccines demonstrate their superiority for tumor prevention owing to the elicited robust antitumor immunity. The reported strategy provides a rational design of nanovaccines for enhanced cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Nanopartículas , Neoplasias , Humanos , Animales , Ratones , Receptor Toll-Like 7 , Antígenos/química , Neoplasias/terapia , Adyuvantes Inmunológicos , Inmunoterapia , Nanopartículas/química , Vacunación , Células Dendríticas , Ratones Endogámicos C57BL
4.
ACS Appl Mater Interfaces ; 14(41): 46176-46187, 2022 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-36206454

RESUMEN

Hepatitis B virus (HBV) infection remains a major challenge to global health due to unsatisfactory treatment efficacy, side effects of current therapies, and immune tolerance. Toll-like receptors 7/8 (TLR7/8) agonists have shown great potential in chronic hepatitis B (CHB) cure, but systemic administration often induces severe side effects due to rapid dispersion into the microvasculature. Herein, we encapsulate an imidazoquinoline-based TLR7/8 agonist (IMDQ) into zeolitic imidazolate framework 8 nanoparticles (IMDQ@ZIF-8 NPs) for HBV immunotherapy. Compared with free IMDQ, IMDQ@ZIF-8 NPs efficiently accumulate in the liver and are selectively taken up by antigen-presenting cells (APCs), leading to enhanced APC activation and efficient viral elimination in HBV-infected models. Strikingly, MDQ@ZIF-8 NP treatment results in the obvious production of anti-HBs antibody and seroconversion in HBV-infected mice. Overall, this study on the convergence of a facile assembly approach and efficient therapeutic effects represents a promising strategy for HBV treatment.


Asunto(s)
Hepatitis B Crónica , Hepatitis B , Estructuras Metalorgánicas , Animales , Ratones , Adyuvantes Inmunológicos/farmacología , Hepatitis B/tratamiento farmacológico , Virus de la Hepatitis B , Hepatitis B Crónica/tratamiento farmacológico , Estructuras Metalorgánicas/farmacología , Estructuras Metalorgánicas/uso terapéutico , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8
5.
Chem Commun (Camb) ; 58(14): 2315-2318, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35076033

RESUMEN

Herein, we report the design of therapeutic nanoparticles by encapsulating photosensitizers and aluminum ions into metal-organic frameworks. The nanoparticles could significantly inhibit the growth of primary and rechallenged tumors by a combination of photothermal therapy and immunotherapy. This work offers a promising strategy to design an immunologic nanoplatform for "cold" tumor therapy.


Asunto(s)
Antineoplásicos/farmacología , Inmunoterapia , Estructuras Metalorgánicas/farmacología , Terapia Fototérmica , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Estructuras Metalorgánicas/síntesis química , Estructuras Metalorgánicas/química , Ratones , Estructura Molecular , Tamaño de la Partícula
6.
Biomater Sci ; 9(20): 6940-6949, 2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34528658

RESUMEN

Combination cancer immunotherapy that synergizes the advantages of multiple therapeutic agents has shown great potential in tumor treatment. Herein, we report the one-step assembly of therapeutic nanoparticles (NPs) to co-deliver photosensitizers and adjuvants for combination photodynamic therapy (PDT) and immunotherapy. The NPs are obtained via self-assembly of chlorin e6 (Ce6) and imidazoquinoline-based TLR7 agonists (IMDQ), which results in a high loading efficacy of 72.2% and 27.8% for Ce6 and IMDQ, respectively. Upon laser irradiation, the resulting NPs could not only effectively induce photodynamic immunogenic cancer cell death, but also elicit robust antitumor immunity, leading to significant inhibition of both primary and distant tumors in a bilateral tumor model. This study demonstrates the potential of self-assembled NPs in co-delivering multiple therapeutics for potential immunotherapy to enhance the antitumor efficacy.


Asunto(s)
Nanopartículas , Fotoquimioterapia , Porfirinas , Línea Celular Tumoral , Inmunoterapia , Fármacos Fotosensibilizantes
7.
ACS Appl Mater Interfaces ; 13(12): 13978-13989, 2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33749241

RESUMEN

We report the one-step assembly of vaccine particles by encapsulating ovalbumin (OVA) and cytosine-phosphate-guanine oligodeoxynucleotides (CpG) into poly(ethylene glycol) (PEG)-mediated zeolitic imidazolate framework-8 nanoparticles (OVA-CpG@ZIF-8 NPs), where PEG improves the stability and dispersity of ZIF-8 NPs and the NPs protect the encapsulated OVA and CpG to circumvent the cold chain issue. Compared with free OVA and OVA-encapsulated ZIF-8 (OVA@ZIF-8) NPs, OVA-CpG@ZIF-8 NPs can enhance antigen uptake, cross-presentation, dendritic cell (DC) maturation, production of specific antibody and cytokines, and CD4+ T and CD8+ T cell activation. More importantly, the vaccine particles retain their bioactivity against enzymatic degradation, elevated temperatures, and long-term storage at ambient temperature. The study highlights the importance of PEG-mediated ZIF-8 NPs as a vaccine delivery system for the promising application of effective and cold chain-independent vaccination against diseases.


Asunto(s)
Inmunogenicidad Vacunal , Nanopartículas/química , Oligodesoxirribonucleótidos/administración & dosificación , Ovalbúmina/administración & dosificación , Polietilenglicoles/química , Vacunas/administración & dosificación , Animales , Citocinas/inmunología , Sistemas de Liberación de Medicamentos , Femenino , Imidazoles/química , Activación de Linfocitos , Estructuras Metalorgánicas/química , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/inmunología , Ovalbúmina/inmunología , Células RAW 264.7 , Vacunas/inmunología , Zeolitas/química
8.
Chem Commun (Camb) ; 56(75): 11078-11081, 2020 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-32812555

RESUMEN

We report a one-pot approach for the scalable synthesis of zeolitic imidazolate framework-8 nanoparticles (ZIF-8 NPs) using poly(ethylene glycol) as the mineralizer, where drugs and proteins can be encapsulated in the ZIF-8 NPs for intracellular delivery. The ZIF-8 NPs exhibit high colloidal dispersity and stability (above two weeks) in cell medium.

9.
Biomed Pharmacother ; 107: 606-614, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30114644

RESUMEN

Previous studies have shown that BMS-345541 (BMS, a specific IκB kinase ß inhibitor) sensitized various tumor cells including MCF-7 breast cancer cells to ionizing radiation (IR). However, the mechanisms of BMS action are unknown. Since the expression of E1A-like inhibitor of differentiation 3 (EID3) was highly upregulated in MCF-7 cells after BMS treatment, we investigated the role of EID3 in the response of MCF-7 cells to IR. We found that BMS induced EID3 expression in MCF-7 cells in a time- and dose-dependent manner. Knockdown of EID3 by specific shRNA attenuated BMS-induced radiosensitization in MCF-7 cells. In contrast, induction of EID3 expression in an inducible EID3 expressing MCF-7 cell line with doxycycline sensitized the cells to IR. EID3-mediated sensitization of MCF-7 cells to IR was not attributed to an increase in apoptosis. Instead, EID3-expressing MCF-7 cells exhibited significantly higher levels of senescence associated ß-galactosidase (SA-ß-gal) activity and higher levels of p21 and p57 than EID3-MCF-7 cells without induction of EID3 after exposure to IR. Similar findings were observed when EID3-expressing MCF-7 cells were treated with etoposide, a topoisomerase II inhibitor. Taken together, our findings reveal a novel function of EID3 and suggest that the induction of EID3 by BMS may be exploited as a new strategy to sensitize breast cancer cells to IR and chemotherapy by inducing cancer cell senescence.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas Portadoras/genética , Senescencia Celular/efectos de la radiación , Radiación Ionizante , Regulación hacia Arriba/genética , Proteínas Portadoras/metabolismo , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Etopósido/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Imidazoles/farmacología , Células MCF-7 , Quinoxalinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/efectos de la radiación
10.
Immunobiology ; 223(11): 663-670, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30005969

RESUMEN

Porcine circovirus type 2 (PCV2)-associated diseases have led to great economic losses to the pig industry. Our lab previously found that conjugation of chitosan oligosaccharides (COS) or via a carrier protein enhanced the immunogenicity of PCV2 vaccine against infectious pathogens. However, precise mechanisms and signal transduction pathways underlying the efficacy of COS conjugation remains poorly defined. In this study, to better understand the effects and mechanism of COS conjugates maintain the adjuvant potential in vivo, we investigated its augmentation of macrophage function, including cell activation, NO production, cytokine production and phagocytosis. Additionally, the role of Toll-like receptors (TLR) proteins in this process was also assessed. The results indicate that, as compared to the PCV and PCV/COS, conjugation of COS effectively enhanced the NO production, cytokines generation and phagocytosis activity of macrophages. Noticeably, the generation of NO and proinflammatory cytokines was closely related to the TLR2/4 signaling pathways, strongly suggesting that conjugation of COS regulates innate and adaptive immunity by activation of macrophages, resulting in immune enhancement. In summary, the present study provides a potential mechanism of COS conjugation as a novel adjuvant to improve immune responses against various diseases.


Asunto(s)
Quitosano/inmunología , Infecciones por Circoviridae/inmunología , Circovirus/fisiología , Macrófagos/inmunología , Vacunas Virales/inmunología , Adyuvantes Inmunológicos , Animales , Anticuerpos Antivirales/sangre , Quitosano/química , Activación de Macrófagos , Óxido Nítrico/metabolismo , Fagocitosis , Transducción de Señal , Porcinos , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
11.
Glycoconj J ; 35(5): 451-459, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30051156

RESUMEN

Porcine circovirus type 2 (PCV2)-associated diseases have led to huge economic losses in pig industry. Our laboratory previously found that conjugation of chitosan oligosaccharides (COS) enhanced the immunogenicity of PCV2 vaccine against infectious pathogens. In this study, an effective adjuvant system was developed by covalent conjugation of COS via a carrier protein (Ovalbumin, OVA) to further increase the immunogenicity of vaccine. Its effect on dendritic cells maturation was assessed in vitro and its immunogenicity was investigated in mice. The results indicated that, as compared to the PCV2 and COS-PCV2, COS-OVA-PCV2 stimulated dendritic cells to express higher maturation markers (CD80, CD86, CD40 and MHC class II) and remarkably promoted both humoral and cellular immunity against PCV2 by enhancing the lymphocyte proliferation and inducing a mixed Th1/Th2 response, including the increased production of PCV2-specific antibodies and raised levels of inflammatory cytokines. Furthermore, it displayed better immune-stimulating effects than the physical mixture of vaccine and ISA206 (a commercialized adjuvant). In conclusion, conjugation of COS via a carrier protein might be a promising strategy to enhance the immunogenicity of vaccines.


Asunto(s)
Quitosano/metabolismo , Circovirus/fisiología , Inmunoconjugados/metabolismo , Oligosacáridos/metabolismo , Ovalbúmina/metabolismo , Vacunas Virales/inmunología , Animales , Formación de Anticuerpos , Proteínas Portadoras , Proliferación Celular , Quitosano/química , Citocinas/metabolismo , Inmunoconjugados/química , Inmunoconjugados/aislamiento & purificación , Linfocitos/citología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Oligosacáridos/química , Porcinos , Vacunas Virales/química
12.
Mar Drugs ; 16(5)2018 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-29734657

RESUMEN

Bacteria biofilm helps bacteria prevent phagocytosis during infection and increase resistance to antibiotics. Staphylococcus aureus is a Gram-positive pathogenic bacterium and is tightly associated with biofilm-related infections, which have led to great threat to human health. Chitosan, the only cationic polysaccharide in nature, has been demonstrated to have antimicrobial and anti-biofilm activities, which, however, require a relative high dosage of chitosan. Moreover, poor water solubility further restricts its applications on anti-infection therapy. Inulins are a group of polysaccharides produced by many types of plants, and are widely used in processed foods. Compared to chitosan, inulin is very soluble in water and possesses a mild antibacterial activity against certain pathogenic bacteria. In order to develop an effective strategy to treat biofilm-related infections, we introduce a method by covalent conjugation of inulin to chitosan. The physicochemical characterization of the inulin⁻chitosan conjugate was assayed, and the anti-biofilm activity was evaluated against S. aureus biofilm. The results indicated that, as compared to chitosan, this novel polysaccharide⁻polysaccharide conjugate significantly enhanced activities against S. aureus either in a biofilm or planktonic state. Of note, the conjugate also showed a broad spectrum anti-biofilm activity on different bacteria strains and low cellular toxicity to mammalian cells. These results suggested that chitosan conjugation of inulin was a viable strategy for treatment against biofilm-related infections. This finding may further spread the application of natural polysaccharides on treatments of infectious disease.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Quitosano/química , Inulina/química , Animales , Línea Celular , Línea Celular Tumoral , Células Hep G2 , Humanos , Ratones , Pruebas de Sensibilidad Microbiana/métodos , Células RAW 264.7 , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos
14.
Mar Drugs ; 15(8)2017 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-28933754

RESUMEN

Chitosan oligosaccharides (COS), the degraded products of chitosan, have been demonstrated to have versatile biological functions. In primary studies, it has displayed significant adjuvant effects when mixed with other vaccines. In this study, chitosan oligosaccharides with different deacetylation degrees were prepared and conjugated to porcine circovirus type 2 (PCV2) subunit vaccine to enhance its immunogenicity. The vaccine conjugates were designed by the covalent linkage of COSs to PCV2 molecules and administered to BALB/c mice three times at two-week intervals. The results indicate that, as compared to the PCV2 group, COS-PCV2 conjugates remarkably enhanced both humoral and cellular immunity against PCV2 by promoting lymphocyte proliferation and initiating a mixed T-helper 1 (Th1)/T-helper 2 (Th2) response, including raised levels of PCV2-specific antibodies and an increased production of inflammatory cytokines. Noticeably, with the increasing deacetylation degree, the stronger immune responses to PCV2 were observed in the groups with COS-PCV2 vaccination. In comparison with NACOS (chitin oligosaccharides)-PCV2 and LCOS (chitosan oligosaccharides with low deacetylation degree)-PCV2, HCOS (chitosan oligosaccharides with high deacetylation degree)-PCV2 showed the highest adjuvant effect, even comparable to that of PCV2/ISA206 (a commercialized adjuvant) group. In summary, COS conjugation might be a viable strategy to enhance the immune response to PCV2 subunit vaccine, and the adjuvant effect was positively correlated with the deacetylation degree of COS.


Asunto(s)
Quitosano/inmunología , Circovirus/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Inmunidad Celular/efectos de los fármacos , Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Vacunas Conjugadas/inmunología , Vacunas de Subunidad/inmunología , Vacunas Virales/farmacología
15.
Carbohydr Polym ; 166: 64-72, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28385249

RESUMEN

This study aimed to investigate the effect of chitosan oligosaccharide (COS) conjugation on the immunogenicity of porcine circovirus type-2 (PCV2) vaccine. Two conjugates (PCV2-COS-1 and PCV2-COS-2) were designed by covalent conjugation of an inactivated PCV2 vaccine with COS, and administered to C57BL/6 mice three times at two-week intervals. The results indicate that, as compared to PCV2 alone group, the PCV2-COS conjugates remarkably enhanced both humoral and cellular immunity against PCV2 by promoting T lymphocyte proliferation and initiating a mixed Th1/Th2 response, including the elevated production of PCV-2 specific antibodies and up-regulated secretion of inflammatory cytokines. Noticeably, the immunization with PCV2-COS-1 conjugate displayed similar or even better immune-stimulating effects than that by PCV2/ISA206 (a commercialized adjuvant) and showed no infection or pathological signs at injection sites of the mice. Presumably, the covalent linkage of PCV2 vaccine to COS might be a viable strategy to increase the efficacy against PCV2-associated diseases.


Asunto(s)
Quitosano , Infecciones por Circoviridae/prevención & control , Inmunidad Celular , Inmunidad Humoral , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Infecciones por Circoviridae/inmunología , Circovirus , Ratones , Ratones Endogámicos C57BL , Porcinos
16.
Mol Cell Biochem ; 430(1-2): 47-56, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28190168

RESUMEN

The knowledge regarding the importance of long non-coding RNAs (lncRNAs), a new class of genes, is very sparse in osteosarcoma. In the present study, we describe the expression profile of lncRNAs in osteosarcomas compared with paired adjacent non-cancerous tissue (n = 7) using microarray analysis. A total of 25,733 lncRNAs were identified in osteosarcoma; 1995 lncRNAs were consistently upregulated and 2226 lncRNAs were consistently under-regulated in all samples analyzed (≥2.0-fold, p < 0.05). We have validated three over-regulated and three under-regulated lncRNAs in patient samples (n = 7). The antisense transcript of SATB2 protein (SATB2-AS1) was identified as one of the upregulated lncRNAs. The SATB2-AS1 is a 3197-bp lncRNA on chromosome 2. This is the first report, where we have documented the increased expression of SATB2-AS1 in osteosarcoma patients and in human osteosarcoma cancer cell lines (U2OS, HOS, MG63). SATB2-AS1 expression was significantly higher in the metastatic tumors compared to non-metastatic tumors. In vitro gain and loss of function approaches demonstrated that SATB2-AS1 regulates cell cycle, cell proliferation, and cell growth. In addition, SATB2-AS1 affects the translational expression of SATB2 gene. Our data demonstrate that an antisense non-coding RNA regulates the expression of its sense gene, and increases the cell growth, therefore pointing the pivotal functions of SATB2-AS1 in osteosarcoma.


Asunto(s)
Neoplasias Óseas/metabolismo , Ciclo Celular , Regulación Neoplásica de la Expresión Génica , Osteosarcoma/metabolismo , ARN Largo no Codificante/biosíntesis , ARN Neoplásico/biosíntesis , Neoplasias Óseas/genética , Línea Celular Tumoral , Femenino , Humanos , Masculino , Proteínas de Unión a la Región de Fijación a la Matriz/biosíntesis , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Osteosarcoma/genética , ARN Largo no Codificante/genética , ARN Neoplásico/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
17.
Oncol Rep ; 36(6): 3545-3551, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27748909

RESUMEN

Osteosarcoma is a high­grade malignant tumor frequently found in children and adolescents. Thalidomide has been reported for treatment of various malignancies. Thalidomide was added to osteosarcoma cells and studied by cytotoxicity assay, evaluating apoptosis, cell cycle arrest, mitochondrial membrane potential (ΔΨm), and reactive oxygen species (ROS) levels and the expression of Bcl­2, Bax, caspase­3 and NF­κB. The results showed that thalidomide could inhibit the proliferation of MG­63 and U2OS cells in a concentration­ and time­dependent manner. Morphological changes of apoptosis were also observed. Thalidomide increased the apoptosis rate of MG­63 cells and induced cell cycle arrest by increasing the number of cells in the G0/G1 phase and decreasing the percentage of S phase in MG­63 cells. Further investigation showed that a disruption of ΔΨm and upregulation of ROS were induced by thalidomide in high concentration. By western blot analysis, thalidomide resulted in the decreasing expression of Bcl­2 and NF­κB, and the increasing expression of Bcl­2/Bax and caspase­3. Here, we provide evidence that thalidomide could cause apoptosis in osteosarcoma cells. Taken together, these results indicate that thalidomide could be an antitumor drug in the therapy of osteosarcoma.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias Óseas/tratamiento farmacológico , Osteosarcoma/tratamiento farmacológico , Talidomida/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Forma de la Célula/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , FN-kappa B/metabolismo , Osteosarcoma/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo
18.
J Membr Biol ; 249(4): 483-92, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27007877

RESUMEN

A new Ru(II) complex [Ru(dmp)2(NMIP)](ClO4)2 (dmp = 2,9-dimethyl-1,10-phenanthroline, NMIP = 2'-(2″-nitro-3″,4″-methylenedioxyphenyl)imidazo[4',5'-f][1,10]-phenanthroline) was synthesized and characterized by elemental analysis, ESI-MS and (1)H NMR. The cytotoxic activity of the complex against MG-63, U2OS, HOS, and MC3T3-e1 cell lines was investigated by MTT method. The complex shows moderate cytotoxicity toward HOS (IC50 = 35.6 ± 2.6 µM) and MC3T3-e1 (IC50 = 41.6 ± 2.8 µM) cell lines. The morphological studies show that the complex can induce apoptosis in HOS cells and cause an increase of reactive oxygen species levels and a decrease in the mitochondrial membrane potential. The cell cycle distribution demonstrates that the complex inhibits the cell growth at S phase. Additionally, the antitumor activity in vivo reveals that the complex can induce a decrease in tumor weight.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Organometálicos/farmacología , Rutenio , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Estructura Molecular , Compuestos Organometálicos/química , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/metabolismo , Osteosarcoma/patología , Especies Reactivas de Oxígeno/metabolismo , Rutenio/química , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Artículo en Inglés | MEDLINE | ID: mdl-26956530

RESUMEN

The cytotoxic activity of two Ru(II) complexes against A549, BEL-7402, HeLa, PC-12, SGC-7901 and SiHa cell lines was investigated by MTT method. Complexes 1 and 2 show moderate cytotoxicity toward BEL-7402 cells with an IC50 value of 53.9 ± 3.4 and 39.3 ± 2.1 µM. The effects of the complexes inducing apoptosis, cellular uptake, reactive oxygen species and mitochondrial membrane potential in BEL-7402 cells have been studied by fluorescence microscopy. The percentages of apoptotic and necrotic cells and cell cycle arrest were studied by flow cytometry. The BSA-binding behaviors were investigated by UV/visible and fluorescent spectra.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Rutenio/química , Rutenio/farmacología , 2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Células HeLa , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...