Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Biol Rep ; 51(1): 358, 2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38400849

RESUMEN

BACKGROUND: Cardiomyocytes form, transport, and metabolize the omnipresent metabolite adenosine. Depending upon the adenosine concentrations and the pharmacological properties of receptor subtypes, adenosine exerts (patho)physiological responses in the cardiovascular system. The objective of this review is to present different protective mechanisms of A1-adenosine receptor inhibitors in cardiovascular diseases. METHODS AND RESULTS: Literature references were collected and sorted using relevant keywords and key phrases as search terms in scientific databases such as Web of Science, PubMed and Google Scholar. A1 adenosine receptor regulates free fatty acid metabolism, lipolysis, heart rate, blood pressure, and cardiovascular toxicity. The evidence clearly supporting the therapeutic potency of pharmacological A1 adenosine receptors agonists and antagonists in modulating cardiovascular risk factor parameters and treatment of cardiovascular diseases. CONCLUSION: This review summarizes the protective role of pharmacological A1-adenosine receptor regulators in the pathogenesis of cardiovascular diseases for a better management of cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Antagonistas de Receptores Purinérgicos P1 , Humanos , Antagonistas de Receptores Purinérgicos P1/farmacología , Enfermedades Cardiovasculares/tratamiento farmacológico , Presión Sanguínea , Adenosina , Receptores Purinérgicos P1
2.
BMC Med ; 21(1): 193, 2023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37226233

RESUMEN

BACKGROUND: Immunotherapy has emerged as an efficient therapeutic approach for cancer management. However, stimulation of host immune system against cancer cells often fails to achieve promising clinical outcomes mainly owing to the immunosuppressive characteristics of the tumor microenvironment (TME). Combination therapeutics that can trigger sustained immunogenic cell death (ICD) have provided new opportunities for cancer treatment. METHODS: In this study, we designed and applied an ICD inducer regimen, including a genetically engineered oncolytic virus (miRNA-modified coxsackieviruses B3, miR-CVB3), a pore-forming lytic peptide (melittin, found in bee venom), and a synthetic toll-like receptor 9 ligand (CpG oligodeoxynucleotides), for breast cancer and melanoma treatment. We compared the anti-tumor efficacy of miR-CVB3 and CpG-melittin (CpGMel) alone and in combination (miR-CVB3 + CpGMel) and investigated possible mechanisms involved. RESULTS: We demonstrated that miR-CVB3 + CpGMel had no major impact on viral growth, while enhancing the cellular uptake of CpGMel in vitro. We further showed that combination therapy led to significant increases in tumor cell death and release of damage-associated molecular patterns compared with individual treatment. In vivo studies in 4T1 tumor-bearing Balb/c mice revealed that both primary and distant tumors were significantly suppressed, and the survival rate was significantly prolonged after administration of miR-CVB3 + CpGMel compared with single treatment. This anti-tumor effect was accompanied by increased ICD and immune cell infiltration into the TME. Safety analysis showed no significant pathological abnormalities in Balb/c mice. Furthermore, the developed therapeutic regimen also demonstrated a great anti-tumor activity in B16F10 melanoma tumor-bearing C57BL/6 J mice. CONCLUSIONS: Overall, our findings indicate that although single treatment using miR-CVB3 or CpGMel can efficiently delay tumor growth, combining oncolytic virus-based therapy can generate even stronger anti-tumor immunity, leading to a greater reduction in tumor size.


Asunto(s)
Melanoma , Virus Oncolíticos , Ratones , Animales , Ratones Endogámicos C57BL , Meliteno , Virus Oncolíticos/genética , Inmunoterapia , Melanoma/terapia , Microambiente Tumoral
3.
J Neuroinflammation ; 19(1): 16, 2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-35022041

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of the motor neuron system associated with both genetic and environmental risk factors. Infection with enteroviruses, including poliovirus and coxsackievirus, such as coxsackievirus B3 (CVB3), has been proposed as a possible causal/risk factor for ALS due to the evidence that enteroviruses can target motor neurons and establish a persistent infection in the central nervous system (CNS), and recent findings that enteroviral infection-induced molecular and pathological phenotypes closely resemble ALS. However, a causal relationship has not yet been affirmed. METHODS: Wild-type C57BL/6J and G85R mutant superoxide dismutase 1 (SOD1G85R) ALS mice were intracerebroventricularly infected with a sublethal dose of CVB3 or sham-infected. For a subset of mice, ribavirin (a broad-spectrum anti-RNA viral drug) was given subcutaneously during the acute or chronic stage of infection. Following viral infection, general activity and survival were monitored daily for up to week 60. Starting at week 20 post-infection (PI), motor functions were measured weekly. Mouse brains and/or spinal cords were harvested at day 10, week 20 and week 60 PI for histopathological evaluation of neurotoxicity, immunohistochemical staining of viral protein, neuroinflammatory/immune and ALS pathology markers, and NanoString and RT-qPCR analysis of inflammatory gene expression. RESULTS: We found that sublethal infection (mimicking chronic infection) of SOD1G85R ALS mice with CVB3 resulted in early onset and progressive motor dysfunction, and shortened lifespan, while similar viral infection in C57BL/6J, the background strain of SOD1G85R mice, did not significantly affect motor function and mortality as compared to mock infection within the timeframe of the current study (60 weeks PI). Furthermore, we showed that CVB3 infection led to a significant increase in proinflammatory gene expression and immune cell infiltration and induced ALS-related pathologies (i.e., TAR DNA-binding protein 43 (TDP-43) pathology and neuronal damage) in the CNS of both SOD1G85R and C57BL/6J mice. Finally, we discovered that early (day 1) but not late (day 15) administration of ribavirin could rescue ALS-like neuropathology and symptoms induced by CVB3 infection. CONCLUSIONS: Our study identifies a new risk factor that contributes to early onset and accelerated progression of ALS and offers opportunities for the development of novel targeted therapies.


Asunto(s)
Esclerosis Amiotrófica Lateral , Enfermedades Neurodegenerativas , Esclerosis Amiotrófica Lateral/patología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Médula Espinal/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
4.
J Virol ; 95(12)2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33827951

RESUMEN

During viral infection, the dynamic virus-host relationship is constantly in play. Many cellular proteins, such as RNA-binding proteins (RBPs), have been shown to mediate antiviral responses during viral infection. Here, we report that the RBP FUS/TLS (fused in sarcoma/translocated in liposarcoma) acts as a host-restricting factor against infection with coxsackievirus B3 (CVB3). Mechanistically, we found that deletion of FUS leads to increased viral RNA transcription and enhanced internal ribosome entry site (IRES)-driven translation, with no apparent impact on viral RNA stability. We further demonstrated that FUS physically interacts with the viral genome, which may contribute to direct inhibition of viral RNA transcription/translation. Moreover, we identified a novel function for FUS in regulating host innate immune response. We show that in the absence of FUS, gene expression of type I interferons and proinflammatory cytokines elicited by viral or bacterial infection is significantly impaired. Emerging evidence suggests a role for stress granules (SGs) in antiviral innate immunity. We further reveal that knockout of FUS abolishes the ability to form SGs upon CVB3 infection or poly(I·C) treatment. Finally, we show that, to avoid FUS-mediated antiviral response and innate immunity, CVB3 infection results in cytoplasmic mislocalization and cleavage of FUS through the enzymatic activity of viral proteases. Together, our findings in this study identify FUS as a novel host antiviral factor which restricts CVB3 replication through direct inhibition of viral RNA transcription and protein translation and through regulation of host antiviral innate immunity.IMPORTANCE Enteroviruses are common human pathogens, including those that cause myocarditis (coxsackievirus B3 [CVB3]), poliomyelitis (poliovirus), and hand, foot, and mouth disease (enterovirus 71). Understanding the virus-host interaction is crucial for developing means of treating and preventing diseases caused by these pathogens. In this study, we explored the interplay between the host RNA-binding protein FUS/TLS and CVB3 and found that FUS/TLS restricts CVB3 replication through direct inhibition of viral RNA transcription/translation and through regulation of cellular antiviral innate immunity. To impede the antiviral role of FUS, CVB3 targets FUS for mislocalization and cleavage. Findings from this study provide novel insights into interactions between CVB3 and FUS, which may lead to novel therapeutic interventions against enterovirus-induced diseases.


Asunto(s)
Enterovirus Humano B/inmunología , Enterovirus Humano B/fisiología , Inmunidad Innata , Proteína FUS de Unión a ARN/metabolismo , Proteasas Virales 3C/metabolismo , Animales , Antivirales/farmacología , Autofagia , Línea Celular , Cisteína Endopeptidasas/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Citoplasma/metabolismo , Gránulos Citoplasmáticos/metabolismo , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Genoma Viral , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Sitios Internos de Entrada al Ribosoma , Ratones , Neuronas Motoras/virología , Poli I-C/farmacología , Biosíntesis de Proteínas , ARN Viral/genética , ARN Viral/metabolismo , Proteína FUS de Unión a ARN/genética , Estrés Fisiológico , Transcripción Genética , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
5.
Biochem Biophys Res Commun ; 540: 75-82, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33450483

RESUMEN

The ongoing pandemic of COVID-19 alongside the outbreaks of SARS in 2003 and MERS in 2012 underscore the significance to understand betacoronaviruses as a global health challenge. SARS-CoV-2, the etiological agent for COVID-19, has infected over 50 million individuals' worldwide with more than ∼1 million fatalities. Autophagy modulators have emerged as potential therapeutic candidates against SARS-CoV-2 but recent clinical setbacks urge for better understanding of viral subversion of autophagy. Using MHV-A59 as a model betacoronavirus, time-course infections revealed significant loss in the protein level of ULK1, a canonical autophagy-regulating kinase, and the concomitant appearance of a possible cleavage fragment. To investigate whether virus-encoded proteases target ULK1, we conducted in-vitro and cellular cleavage assays and identified ULK1 as a novel bona fide substrate of SARS-CoV-2 papain-like protease (PLpro). Mutagenesis studies discovered that ULK1 is cleaved at a conserved PLpro recognition sequence (LGGG) after G499, separating its N-terminal kinase domain from a C-terminal substrate recognition region. Over-expression of SARS-CoV-2 PLpro is sufficient to impair starvation-induced autophagy and disrupt formation of ULK1-ATG13 complex. Finally, we demonstrated a dual role for ULK1 in MHV-A59 replication, serving a pro-viral functions during early replication that is inactivated at late stages of infection. In conclusion, our study identified a new mechanism by which PLpro of betacoronaviruses induces viral pathogenesis by targeting cellular autophagy.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Autofagia , Proteasas Similares a la Papaína de Coronavirus/metabolismo , SARS-CoV-2/enzimología , Animales , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Células Cultivadas , Ratones
6.
J Nanobiotechnology ; 18(1): 180, 2020 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-33298099

RESUMEN

Breast cancer continues to be the most frequently diagnosed malignancy among women, putting their life in jeopardy. Cancer immunotherapy is a novel approach with the ability to boost the host immune system to recognize and eradicate cancer cells with high selectivity. As a promising treatment, immunotherapy can not only eliminate the primary tumors, but also be proven to be effective in impeding metastasis and recurrence. However, the clinical application of cancer immunotherapy has faced some limitations including generating weak immune responses due to inadequate delivery of immunostimulants to the immune cells as well as uncontrolled modulation of immune system, which can give rise to autoimmunity and nonspecific inflammation. Growing evidence has suggested that nanotechnology may meet the needs of current cancer immunotherapy. Advanced biomaterials such as nanoparticles afford a unique opportunity to maximize the efficiency of immunotherapy and significantly diminish their toxic side-effects. Here we discuss recent advancements that have been made in nanoparticle-involving breast cancer immunotherapy, varying from direct activation of immune systems through the delivery of tumor antigens and adjuvants to immune cells to altering immunosuppression of tumor environment and combination with other conventional therapies.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Inmunoterapia , Nanomedicina , Adyuvantes Inmunológicos , Animales , Materiales Biocompatibles , Neoplasias de la Mama/genética , Vacunas contra el Cáncer , Células Dendríticas , Técnicas de Inactivación de Genes , Humanos , Nanopartículas , Metástasis de la Neoplasia , Microambiente Tumoral
7.
Biochem Genet ; 58(4): 518-532, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32519277

RESUMEN

Endothelial Nitric Oxide Synthase (eNOS) is an indispensable regulator of blood pressure through producing Nitric Oxide (NO). There is some evidence to suggest that eNOS gene polymorphisms are associated with Essential Hypertension (EHT). In this study, the potential association between eNOS 4a/4b, A922G, G894T, T786C gene polymorphisms and EHT as individual risk factors and as haplotypes are examined in the southern population of Iran (Bandar-Abbas). In this study, 200 EHT patients and 200 normotensive subjects which were matched for age and sex were included. Genotyping was performed by either utilizing Polymerase Chain Reaction (PCR) or PCR followed by Restriction Fragment length Polymorphism (RFLP) method. Our results demonstrated statistically significant associations between T786C, G894T, and 4a/4a and EHT (p < 0.05); however, A922G had no significant association with EHT (p > 0.05). Haplotype analysis also suggested that - 786C/- 922A/4a, - 786C/- 922A/4b and - 786C/- 922G/4a haplotypes were more frequent in EHT group than control group, hypothesizing a positive association with EHT. The present study has identified that the eNOS genetic variations are associated with EHT in southern population of Iran (Bandar-Abbas). These findings also suggested that a number of haplotypes of eNOS gene may be a driving factor for EHT susceptibility in respected population.


Asunto(s)
Hipertensión Esencial/enzimología , Hipertensión Esencial/genética , Haplotipos , Intrones , Óxido Nítrico Sintasa de Tipo III/genética , Polimorfismo de Nucleótido Simple , Adulto , Estudios de Casos y Controles , Hipertensión Esencial/sangre , Hipertensión Esencial/epidemiología , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Irán/epidemiología , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
8.
Drug Chem Toxicol ; 43(5): 514-521, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30714419

RESUMEN

Ulcerative colitis is a chronic inflammatory bowel disease with high incidence and prevalence worldwide. To investigate the therapeutic potency of crocin, as a pharmacologically active component of saffron, in dextran sodium sulfate (DSS)-induced colitis mice model. Experimental colitis was induced by 7-day administration of DSS dissolved in water at a concentration of 1.5% (w/v). The animals were randomly divided into four groups (n»6 for each group). (1) Control group received regular drinking water for four weeks, (2) the second group of mice received regular drinking water for three weeks and then received DSS for one week, (3) and (4) the other two groups received 50-ppm or 200-ppm crocin for three weeks, respectively, and then treated with DSS for one week. Our results showed that Crocin attenuates colitis disease activity index including body weight loss, diarrhea, rectal bleeding, and colon shortening in crocin pre-tread mice. Comparison of histology of colon tissues between groups showed that crocin significantly decreases colon histopathological score, at least partially, by eliciting anti-inflammatory responses in DSS-induced colitis mice. These results clearly showed that crocin is a novel therapeutic agent with low toxicity as well as great clinical significance in treatment of colitis.


Asunto(s)
Carotenoides/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Animales , Carotenoides/efectos adversos , Carotenoides/farmacología , Colitis Ulcerosa/inducido químicamente , Colon/efectos de los fármacos , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL
9.
J Cell Physiol ; 234(9): 14601-14611, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30673132

RESUMEN

Crocin is the major component of saffron, which is used in phytomedicine for the treatment of several diseases including diabetes, fatty liver, depression, menstruation disorders, and, of special interest in this review, inflammatory diseases. Promising selective anti-inflammatory properties of this pharmacological active component have been observed in several studies. Saffron has been shown to exert anti-inflammatory properties against several inflammatory diseases and can be used as a novel therapeutic agent for the treatment of inflammatory diseases either alone or in combination with other standard anti-inflammatory agents. This review summarizes the protective role of saffron and its pharmacologically active constituents in the pathogenesis of inflammatory diseases including digestive diseases, dermatitis, asthma, atherosclerosis, and neurodegenerative diseases for a better understanding and hence a better management of these diseases.

10.
J Cell Physiol ; 234(2): 1295-1299, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30146778

RESUMEN

Adenosine and its analogs are of particular interest as potential therapeutic agents for treatment of cardiovascular diseases (CVDs). A2 adenosine receptor subtypes (A2a and A2b) are extensively expressed in cardiovascular system, and modulation of these receptors using A2 adenosine receptor agonists or antagonists regulates heart rate, blood pressure, heart rate variability, and cardiovascular toxicity during both normoxia and hypoxia conditions. Regulation of A2 adenosine receptor signaling via specific and novel pharmacological regulators is a potentially novel therapeutic approach for a better understanding and hence a better management of CVDs. This review summarizes the role of pharmacological A2 adenosine receptor regulators in the pathogenesis of CVDs.


Asunto(s)
Agonistas del Receptor de Adenosina A2/uso terapéutico , Antagonistas del Receptor de Adenosina A2/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Sistema Cardiovascular/efectos de los fármacos , Receptores de Adenosina A2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Agonistas del Receptor de Adenosina A2/efectos adversos , Antagonistas del Receptor de Adenosina A2/efectos adversos , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatología , Humanos , Terapia Molecular Dirigida , Receptores de Adenosina A2/metabolismo , Resultado del Tratamiento
11.
J Cell Physiol ; 234(6): 8075-8081, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30317621

RESUMEN

Aberrant microRNA (miR) expression is implicated in multiple human malignancies. miR-21, acting as a proto-oncogene, is involved in a variety of cellular processes and tumorigenesis and is frequently overexpressed in some cancer types. Several tumor suppressors, metastatic, and apoptotic genes have been identified as miR-21 targets, including Ras homolog gene family member B, PTEN, Sprouty2, programmed cell death 4, Integrin-ß4, and E-cadherin thereby regulating tumor growth, invasion, and metastasis. There is a growing evidence that miR-21 expression is associated with clinical outcomes in patients with colorectal cancer (CRC). In this review, we summarize the potential diagnostic, prognostic, and therapeutic values of miR-21 in CRC progression for a better understanding and hence a better management of this disease.


Asunto(s)
Proliferación Celular/genética , Neoplasias del Colon/genética , MicroARNs/genética , Pronóstico , Apoptosis/genética , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Fosfohidrolasa PTEN/genética , Proto-Oncogenes Mas , Proteína de Unión al GTP rhoB/genética
12.
J Cell Physiol ; 234(3): 2329-2336, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30191994

RESUMEN

The hypoxic niche of tumor leads to a tremendous increase in the extracellular adenosine concentration through alteration of adenosine metabolism in the tumor microenvironment (TME). This consequently affects cancer progression, local immune responses, and apoptosis of tumor cells. Regulatory effect of adenosine on apoptosis in TME depends on the cancer cell type, pharmacological characteristics of adenosine receptor subtypes, and the adenosine concentration in the tumor niche. Exploiting specific pharmacological adenosine receptor agonist and antagonist inducing apoptosis in cancer cells can be considered as a proper procedure to control cancer progression. This review summarizes the regulatory role of adenosine in cancer cell apoptosis for a better understanding, and hence better management of the disease.


Asunto(s)
Neoplasias/tratamiento farmacológico , Agonistas del Receptor Purinérgico P1/uso terapéutico , Antagonistas de Receptores Purinérgicos P1/uso terapéutico , Receptores Purinérgicos P1/genética , Adenosina Trifosfato/genética , Apoptosis/efectos de los fármacos , Neoplasias/genética , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
13.
J Cell Biochem ; 120(3): 2766-2773, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30321455

RESUMEN

Brain tumors are the most common form of solid tumors in children and is presently a serious therapeutic challenge worldwide. Traditional treatment with chemotherapy and radiotherapy was shown to be unsuccessful in targeting brain tumor cancer stem cells (CSCs), leading to recurrent, treatment-resistant secondary malignancies. Oncolytic virotherapy (OV) is an effective antitumor therapeutic strategy which offers a novel, targeted approach for eradicating pediatric brain tumor CSCs by utilizing mechanisms of cell killing that differ from conventional therapies. A number of studies and some clinical trials have therefore investigated the effects of combined therapy of radiations or chemotherapies with oncolytic viruses which provide new insights regarding the effectiveness and improvement of treatment responses for brain cancer patients. This review summarizes the current knowledge of the therapeutic potency of OVs-induced CSCs targeting in the treatment of brain tumors for a better understanding and hence a better management of this disease.


Asunto(s)
Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Terapia Molecular Dirigida , Células Madre Neoplásicas/patología , Viroterapia Oncolítica/tendencias , Humanos , Virus Oncolíticos
14.
Anal Biochem ; 575: 1-9, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30926271

RESUMEN

AS1411 is a 26-mer G-rich DNA aptamer which has been broadly employed in the field of targeted drug delivery, due to its capability to bind to nucleolin protein on the surface of cancer cells. In this work, it has been shown for the first time that in addition to nucleolin, AS1411 aptamer could bind to copper ions (Cu2+) with high affinity and selectivity, affecting AS1411 usage in drug delivery systems as a targeting agent. In this study, besides the evaluations of the affinity of AS1411 to different ions and the impact of Cu2+ on targeted drug delivery employed AS1411 aptamer as a targeting agent, a simple and ultra-sensitive fluorescent aptasensor was fabricated for Cu2+ detection through applying AS1411, its complementary strand and magnetic beads coated with streptavidin. Gel Red (GR) was also used as a fluorescent dye. The fabricated aptasensor offered the possibility of condensing samples with different volumes. The detection limit of the sensor was 0.01 µM towards Cu2+ in serum samples. The efficacy of this sensor was further confirmed by comparing Cu2+ levels in serums of healthy people with patients suffering from Wilson's disease, Alzheimer's disease and Diabetes Mellitus using the proposed sensing platform.


Asunto(s)
Aptámeros de Nucleótidos/química , Cobre/análisis , Oligodesoxirribonucleótidos/química , Cobre/química , Epirrubicina/química , Colorantes Fluorescentes/química , Humanos , Límite de Detección , Células MCF-7 , Prueba de Estudio Conceptual
15.
Drug Dev Ind Pharm ; 45(4): 603-610, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30633594

RESUMEN

To explore the effect of combination therapy of epirubicin (Epi) and melittin (Mel) to cancer cells, calcium carbonate nanoparticles (CCN), as carriers, were developed which were modified with MUC1-Dimer aptamers as targeting agents. Both Epi and Mel were delivered at the same time to cancer cells overexpressing the target of MUC1 aptamer, mucin 1 glycoproteins (MCF7 and C26 cells). CCN were prepared with a water-in-oil emulsion method. Epi and Mel were separately encapsulated in CCN and the nanoparticles were modified with MUC1-Dimer aptamers. In vitro studies, including MTT assay, flow cytometry analysis and fluorescence imaging were applied to investigate the targeting and cell proliferation inhibition capabilities of MUC1-Dimer aptamer-CCN-Mel complex and MUC1-Dimer aptamer-CCN-Epi complex in the target (MCF-7 and C26 cells) and nontarget (HepG2) cells. Also, the function of the developed complexes was analyzed using in vivo tumor growth inhibition. The release of Epi from MUC1-Dimer aptamer-CCN-Epi complex was pH-sensitive. Cellular uptake studies showed more internalization of the MUC1-Dimer aptamer-CCN-Epi complex into MCF-7 and C26 cells (target) compared to HepG2 cells (nontarget). Interestingly, the MUC1-Dimer aptamer-CCN-Mel complex and MUC1-Dimer aptamer-CCN-Epi complex indicated very low toxicity as compared to target cells. Moreover, co-delivery of Epi and Mel using the mixture of MUC1-Dimer aptamer-CCN-Mel complex and MUC1-Dimer aptamer-CCN-Epi complex exhibited strong synergistic cytotoxicity in MCF-7 and C26 cells. Furthermore, the presented complexes had a better function to control tumor growth in vivo compared to free Epi.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Portadores de Fármacos/química , Epirrubicina/farmacología , Meliteno/farmacología , Neoplasias/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Aptámeros de Péptidos/química , Carbonato de Calcio/química , Línea Celular Tumoral/trasplante , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacocinética , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Epirrubicina/uso terapéutico , Femenino , Humanos , Meliteno/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Mucina-1/química , Nanopartículas/química , Neoplasias/patología , Resultado del Tratamiento
16.
J Cell Physiol ; 233(4): 2733-2740, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28661026

RESUMEN

Increasing evidence suggests that adenosine is dysregulated in ulcerative colitis (UC), potentially affecting UC pathogenesis, diagnosis, and therapy. Dysregulation of the activity of adenosine generating enzymes including adenosine deaminase in serum of patients with acute colitis supports the role of this omnipresent metabolite in the pathogenesis of colitis. Adenosine regulates inflammatory responses including epithelial barrier hyper-permeability, myeloperoxidase activity, and neuromuscular motility in colitis, supporting the therapeutic potency of adenosine receptors agonists and antagonists in this disease. Depending upon the adenosine receptor subtype, activation or suppression of the receptor with pharmacological agonists or antagonists attenuates colitis pathological symptoms in colitis model. This review summarizes the role of adenosine receptors agonists and antagonists in the pathogenesis of colitis for a better understanding and hence a better management of this disease.


Asunto(s)
Colitis/tratamiento farmacológico , Agonistas del Receptor Purinérgico P1/uso terapéutico , Antagonistas de Receptores Purinérgicos P1/uso terapéutico , Adenosina/metabolismo , Animales , Colitis/patología , Humanos , Inflamación/patología , Transducción de Señal
17.
J Cell Physiol ; 233(6): 4783-4790, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29165795

RESUMEN

Mammalian target of rapamycin (mTOR) signaling pathway controls cell energy metabolism. There is an interplay between mTOR and proinflammatory signaling pathways, supporting the role of the pathway in the pathogenesis of inflammatory diseases. Inhibition of mTOR signaling using specific pharmacological inhibitors could offer therapeutic promise in several inflammatory-associated diseases. In this review, we summarize recent findings on the regulatory effects of mTOR signaling on inflammation and the therapeutic potency of mTOR pharmacological inhibitors in the treatment of inflammatory diseases including cancer, neurodegenerative diseases, atherosclerosis, sepsis, and rheumatoid arthritis for a better understanding and hence a better management of these diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Mediadores de Inflamación/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/enzimología , Artritis Reumatoide/inmunología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/enzimología , Aterosclerosis/inmunología , Humanos , Inflamación/enzimología , Inflamación/inmunología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/inmunología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/inmunología , Sepsis/tratamiento farmacológico , Sepsis/enzimología , Sepsis/inmunología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo
18.
J Cell Physiol ; 233(3): 1836-1843, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28383816

RESUMEN

The plasma level of adenosine increases under ischemic and inflamed conditions in tumor microenvironment. Adenosine elicits a range of signaling pathways in tumors, resulting in either inhibition or enhancement of tumor growth depending upon different subtypes of adenosine receptors activation and type of cancer. Metabolism of adenosine-5'-triphosphate (ATP) and its derivatives including adenosine is dysregulated in the breast tumor microenvironment, supporting the role of this metabolite in the pathogenesis of breast cancer. Adenosine regulates inflammation, apoptosis, cell proliferation, and metastasis in breast cancer cells. This review summarizes the role of adenosine in the pathogenesis of breast cancer for a better understanding and hence a better management of this disease.


Asunto(s)
Adenosina/metabolismo , Apoptosis/fisiología , Neoplasias de la Mama/patología , Proliferación Celular/fisiología , Inflamación/patología , Metástasis de la Neoplasia/patología , 5'-Nucleotidasa/metabolismo , Adenosina/sangre , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Receptores de Adenosina A2/metabolismo , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología
19.
J Cell Physiol ; 233(4): 2715-2722, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28617999

RESUMEN

Extracellular concentration of adenosine increases in the hypoxic tumor microenvironment. Adenosine signaling regulates apoptosis, angiogenesis, metastasis, and immune suppression in cancer cells. Adenosine-induced cell responses depend upon different subtypes of adenosine receptors activation and type of cancer. Suppression of adenosine signaling via inhibition of adenosine receptors or adenosine generating enzymes including CD39 and CD73 on ovarian or cervical cancer cells is a potentially novel therapeutic approach for gynecological cancer patients. This review summarizes the role of adenosine in the pathogenesis of gynecological cancer for a better understanding and hence a better management of this disease.


Asunto(s)
Adenosina/metabolismo , Neoplasias de los Genitales Femeninos/etiología , Neoplasias de los Genitales Femeninos/metabolismo , Animales , Femenino , Neoplasias de los Genitales Femeninos/patología , Humanos , Modelos Biológicos , Transducción de Señal
20.
J Cell Biochem ; 119(10): 7905-7912, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30011093

RESUMEN

The concentrations of adenosine may increase under ischemic conditions in the tumor microenvironment, and then it enters the systemic circulation. Adenosine controls cancer progression and responses to therapy by regulating angiogenesis, cell survival, apoptosis, cell proliferation, and metastases in tumors. Hence, adenosine metabolism, adenosine-generating enzymes, and adenosine signaling are potentially novel therapeutic targets in a wide range of pathological conditions, including cerebral and cardiac ischemic diseases, inflammatory disorders, immunomodulatory disorders, and, of special interest in this review, cancer. This review summarizes the role of adenosine in the pathogenesis of head and neck cancer for a better understanding of how this may be applied to treating this type of cancer.


Asunto(s)
Adenosina/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Animales , Humanos , Neovascularización Patológica/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA