Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Immunol Invest ; 50(6): 671-684, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32597289

RESUMEN

Antigen-specific immune responses are crucially involved in both multiple sclerosis (MS) and myasthenia gravis (MG). Teriflunomide is an immunomodulatory agent approved for treatment of MS through inhibition of lymphocyte proliferation. MG associated with muscle-specific tyrosine kinase (MuSK) antibodies often manifests with a severe disease course, prompting development of effective treatment methods. To evaluate whether teriflunomide treatment may ameliorate MuSK-autoimmunity, experimental autoimmune MG (EAMG) was induced by immunizing C57BL/6 (B6) mice three times with MuSK in complete Freund's adjuvant (CFA) (n = 17). MuSK-immunized mice were treated daily with teriflunomide (n = 8) or PBS (n = 9) starting from the third immunization (week 8) to termination (week 14). Clinical severity of EAMG was monitored. Immunological alterations were evaluated by measurement of anti-MuSK IgG, neuromuscular junction deposits, and flow cytometric analysis of lymph node cells. In MS patients under teriflunomide treatment, the peripheral blood B cell subset profile was analyzed. B6 mice treated with teriflunomide displayed relatively preserved body weight, lower EAMG prevalence, reduced average clinical grades, higher inverted screen scores, diminished anti-MuSK antibody and NMJ deposit levels. Amelioration of EAMG findings was associated with reduced memory B cell ratios in the lymph nodes. Similarly, MS patients under teriflunomide treatment showed reduced memory B cell, plasma cell, and plasmablast ratios. Teriflunomide treatment has effectively ameliorated MuSK-autoimmunity and thus may putatively be used in long-term management of MuSK-MG as an auxiliary treatment method. Teriflunomide appears to exert beneficial effects through inhibition of effector B cells.


Asunto(s)
Subgrupos de Linfocitos B/efectos de los fármacos , Crotonatos/administración & dosificación , Hidroxibutiratos/administración & dosificación , Esclerosis Múltiple/tratamiento farmacológico , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Nitrilos/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/inmunología , Toluidinas/administración & dosificación , Adulto , Animales , Subgrupos de Linfocitos B/inmunología , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Esclerosis Múltiple/sangre , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/inmunología , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/diagnóstico , Miastenia Gravis Autoinmune Experimental/inmunología , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Receptores Colinérgicos/administración & dosificación , Resultado del Tratamiento
2.
Cancer Treat Rev ; 89: 102062, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32659623

RESUMEN

Cabozantinib is an oral tyrosine kinase inhibitor (TKI) approved for the treatment of patients with advanced renal cell carcinoma (RCC) at a dose of 60 mg/day. As with other TKIs, cabozantinib is associated with high interpatient variability in drug clearance and exposure that can significantly impact safety and tolerability across a patient population. To optimize cabozantinib exposure (maintaining efficacy and tolerability) for the individual, patients may require treatment interruption with dose reduction (40 mg/day and then 20 mg/day). In the pivotal Phase 3 METEOR trial, cabozantinib significantly improved overall survival, progression-free survival and the objective response rate compared with everolimus in patients with advanced RCC who had received previous treatment with a VEGFR TKI. Dose reductions were common for patients receiving cabozantinib (60%) but effective as only 9% discontinued treatment due to adverse events (AEs). In this review, we discuss pharmacometric analyses that evaluated the impact of cabozantinib dose on efficacy and safety outcomes during the METEOR study. Exposure-response models demonstrate that the risk of experiencing adverse events and dose reduction is increased in patients with low cabozantinib clearance versus typical clearance and decreased in patients with high clearance. Dose reduction of cabozantinib to manage AEs is predicted to have minimal impact on efficacy as AEs are more likely to occur in patients with low clearance and higher exposure to cabozantinib. These analyses further support a dose modification strategy to optimize cabozantinib exposure for individual patients.


Asunto(s)
Anilidas/administración & dosificación , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Piridinas/administración & dosificación , Anilidas/efectos adversos , Anilidas/farmacocinética , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Carcinoma de Células Renales/metabolismo , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Renales/metabolismo , Modelos Estadísticos , Supervivencia sin Progresión , Piridinas/efectos adversos , Piridinas/farmacocinética , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/efectos adversos , Proteínas Tirosina Quinasas Receptoras/farmacocinética
3.
Front Immunol ; 11: 403, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32256489

RESUMEN

Myasthenia gravis (MG) with antibodies to the muscle-specific receptor tyrosine kinase (MuSK) is a distinct sub-group of MG, affecting 5-8% of all MG patients. MuSK, a receptor tyrosine kinase, is expressed at the neuromuscular junctions (NMJs) from the earliest stages of synaptogenesis and plays a crucial role in the development and maintenance of the NMJ. MuSK-MG patients are more severely affected and more refractory to treatments currently used for MG. Most patients require long-term immunosuppression, stressing the need for improved treatments. Ideally, preferred treatments should specifically delete the antigen-specific autoimmune response, without affecting the entire immune system. Mucosal tolerance, induced by oral or nasal administration of an auto-antigen through the mucosal system, resulting in an antigen-specific immunological systemic hyporesponsiveness, might be considered as a treatment of choice for MuSK-MG. In the present study we have characterized several immunological parameters of murine MuSK-EAMG and have employed induction of oral tolerance in mouse MuSK-EAMG, by feeding with a recombinant MuSK protein one week before disease induction. Such a treatment has been shown to attenuate MuSK-EAMG. Both induction and progression of disease were ameliorated following oral treatment with the recombinant MuSK fragment, as indicated by lower clinical scores and lower anti-MuSK antibody titers.


Asunto(s)
Tolerancia Inmunológica/inmunología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Administración Oral , Animales , Femenino , Ratones , Proteínas Tirosina Quinasas Receptoras/administración & dosificación
4.
J Neuroimmunol ; 339: 577136, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31855721

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease affecting the neuromuscular junction. Approximately 9% of MG patients have autoantibodies targeting the muscle specific kinase (MuSK), and are challenging therapeutically, since they often present with more severe symptoms. A useful therapy is plasmapheresis, but it is highly non-specific. Antigen-specific immunoadsorption would only remove the pathogenic autoantibodies, minimizing the possible side effects and maximizing the benefit. We used rats with human MuSK-induced experimental autoimmune MG to perform antigen-specific immunoadsorptions, and found it very effective, resulting in a dramatic autoantibody titer decrease, while immunoadsorbed, but not mock-treated, animals showed an significant improvement of their clinical symptoms. Overall, the procedure was efficient, supporting its application for MG treatment.


Asunto(s)
Autoanticuerpos/administración & dosificación , Antígenos de Histocompatibilidad Clase II/administración & dosificación , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Plasmaféresis/métodos , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Receptores Colinérgicos/administración & dosificación , Animales , Autoanticuerpos/inmunología , Femenino , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Inmunización/métodos , Miastenia Gravis Autoinmune Experimental/inmunología , Ratas , Ratas Endogámicas Lew , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/inmunología , Resultado del Tratamiento
6.
Mol Cancer Ther ; 16(10): 2144-2156, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28536313

RESUMEN

NADPH oxidase-derived reactive oxygen species (ROS) potentiate receptor tyrosine kinase (RTK) signaling, resulting in enhanced angiogenesis and tumor growth. In this study, we report that BJ-1301, a hybrid of pyridinol and alpha-tocopherol, exerts anticancer effects by dual inhibition of NADPH oxidase and RTK activities in endothelial and lung cancer cells. BJ-1301 suppresses ROS production by blocking translocation of NADPH oxidase cytosolic subunits to the cell membrane, thereby inhibiting activation. The potency of RTK inhibition by BJ-1301 was lower than that of sunitinib (a multi-RTK inhibitor), but the inhibition of downstream signaling pathways (e.g., ROS generation) and subsequent biological changes (e.g., NOX2 induction) by BJ-1301 was superior. Consistently, BJ-1301 inhibited cisplatin-resistant lung cancer cell proliferation more than sunitinib did. In xenograft chick or mouse tumor models, BJ-1301 inhibited lung tumor growth, to an extent greater than that of sunitinib or cisplatin. Treatments with BJ-1301 induced regression of tumor growth, potentially due to downregulation of autocrine-stimulatory ligands for RTKs, such as TGFα and stem cell factor, in tumor tissues. Taken together, the current study demonstrates that BJ-1301 is a promising anticancer drug for the treatment of lung cancer. Mol Cancer Ther; 16(10); 2144-56. ©2017 AACR.


Asunto(s)
Comunicación Autocrina/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , NADPH Oxidasa 2/genética , Neovascularización Patológica/tratamiento farmacológico , alfa-Tocoferol/análogos & derivados , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Pollos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , NADPH Oxidasa 2/antagonistas & inhibidores , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Oxidación-Reducción/efectos de los fármacos , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , alfa-Tocoferol/farmacología , alfa-Tocoferol/uso terapéutico
7.
Clin Cancer Res ; 23(15): 4251-4258, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28183714

RESUMEN

Purpose: ALK rearrangement detection using FISH is the standard test to identify patients with non-small cell lung carcinoma (NSCLC) eligible for treatment with ALK inhibitors. Recently, ALK protein expression in resectable NSCLC showed predictive value. We evaluated tumor response rate and survival after crizotinib treatment of patients with advanced NSCLC with ALK activation using both dichotomous immunohistochemical (IHC) staining and FISH.Experimental Design: Patients with stage IV NSCLC treated with crizotinib were selected. Tumor response was assessed. ALK rearrangements were detected by FISH (Vysis ALK-break-apart FISH-Probe KIT) and IHC [Ventana ALK (D5F3) CDx assay]. Cohorts of patients with ALK-FISH-positive advanced NSCLC from four other hospitals were used for validation.Results: Twenty-nine consecutive patients with ALK-positive advanced NSCLC diagnosed by FISH and/or IHC on small biopsies or fine-needle aspirations (FNA) were treated with ALK inhibitors. All ALK-IHC-positive patients responded to crizotinib except three with primary resistance. No tumor response was observed in 13 ALK-FISH-positive but ALK-IHC-negative patients. This was confirmed in an external cohort of 16 patients. Receiver operator characteristic (ROC) curves for ALK-IHC and ALK-FISH compared with treatment outcome showed that dichotomous ALK-IHC outperforms ALK-FISH [tumor response area under the curve: (AUC), 0.86 vs. 0.64, P = 0.03; progression-free survival (PFS): AUC 0.86 vs. 0.36, P = 0.005; overall survival (OS): AUC, 0.78 vs. 0.41, P = 0.01, respectively].Conclusions: Dichotomous ALK-IHC is superior to ALK-FISH on small biopsies and FNA to predict tumor response and survival to crizotinib for patients with advanced NSCLC. Our data strongly suggest adapting the guidelines and using dichotomous ALK-IHC as standard companion diagnostic test to select patients with NSCLC who benefit from ALK-targeting therapy. Clin Cancer Res; 23(15); 4251-8. ©2017 AACR.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Pronóstico , Proteínas Tirosina Quinasas Receptoras/genética , Adulto , Anciano , Quinasa de Linfoma Anaplásico , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/aislamiento & purificación , Biopsia con Aguja Fina , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Crizotinib , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Reordenamiento Génico/genética , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Pirazoles/administración & dosificación , Piridinas/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/aislamiento & purificación
8.
J Neuroinflammation ; 12: 231, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26646841

RESUMEN

BACKGROUND: Myasthenia gravis (MG) is an antibody-mediated autoimmune disease of the neuromuscular junction (NMJ), mostly associated with acetylcholine receptor (AChR) antibodies. Around 5-10 % of MG patients show antibodies to muscle-specific tyrosine kinase (MuSK). Mesenchymal stem cell (MSC) administration has been shown to ameliorate muscle weakness in the experimental autoimmune myasthenia gravis (EAMG) model induced by AChR immunization. METHODS: To investigate the efficacy of stem cell treatment in MuSK-related EAMG, clinical and immunological features of MuSK-immunized mice with or without dental follicle MSC (DFMSC) treatment were compared. RESULTS: MuSK-immunized mice intravenously treated with DFMSC after second and third immunizations showed significantly lower EAMG incidence and severity and reduced serum anti-MuSK antibody, NMJ IgG, and C3 deposit levels and CD11b+ lymph node cell ratios. Moreover, lymph node cells of DFMSC-administered mice showed reduced proliferation and IL-6 and IL-12 production responses to MuSK stimulation. By contrast, proportions of B and T cell populations and production of a wide variety of cytokines were not affected from DFMSC treatment. CONCLUSIONS: Our results suggest that DFMSC treatment shows its beneficial effects mostly through suppression of innate immune system, whereas other immune functions appear to be preserved. Stem cell treatment might thus constitute a specific and effective treatment method in MuSK-associated MG.


Asunto(s)
Saco Dental/trasplante , Inmunización/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Debilidad Muscular/terapia , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Receptores Colinérgicos/administración & dosificación , Animales , Células Cultivadas , Saco Dental/citología , Saco Dental/inmunología , Femenino , Humanos , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Endogámicos C57BL , Debilidad Muscular/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/inmunología
9.
Invest Ophthalmol Vis Sci ; 53(4): 1895-904, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22408006

RESUMEN

PURPOSE: The absence of Mertk in RCS rats results in defective RPE phagocytosis, accumulation of outer segment (OS) debris in the subretinal space, and subsequent death of photoreceptors. Previous research utilizing Mertk gene replacement therapy in RCS rats provided proof of concept for treatment of this form of recessive retinitis pigmentosa (RP); however, the beneficial effects on retinal function were transient. In the present study, we evaluated whether delivery of a MERTK transgene using a tyrosine-mutant AAV8 capsid could lead to more robust and longer-term therapeutic outcomes than previously reported. METHODS: An AAV8 Y733F vector expressing a human MERTK cDNA driven by a RPE-selective promoter was administrated subretinally at postnatal day 2. Functional and morphological analyses were performed at 4 months and 8 months post-treatment. Retinal vasculature and Müller cell activation were analyzed by quantifying acellular capillaries and glial fibrillary acidic protein immunostaining, respectively. RESULTS: Electroretinographic responses from treated eyes were more than one-third of wild-type levels and OS were well preserved in the injection area even at 8 months. Rescue of RPE phagocytosis, prevention of retinal vasculature degeneration, and inhibition of Müller cell activation were demonstrated in the treated eyes for at least 8 months. CONCLUSIONS: This research describes a longer and much more robust functional and morphological rescue than previous studies. We also demonstrate for the first time that an AAV8 mutant capsid serotype vector has a substantial therapeutic potential for RPE-specific gene delivery. These results suggest that tyrosine-mutant AAV8 vectors hold promise for the treatment of individuals with MERTK-associated RP.


Asunto(s)
Terapia Genética/métodos , Proteínas Proto-Oncogénicas/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Retinitis Pigmentosa/terapia , Animales , Animales Recién Nacidos , Western Blotting , Modelos Animales de Enfermedad , Electrorretinografía , Estudios de Seguimiento , Vectores Genéticos , Humanos , Inmunohistoquímica , Inyecciones , Microscopía Electrónica de Transmisión , Mutación , Plásmidos , Proteínas Proto-Oncogénicas/uso terapéutico , ARN/genética , Ratas , Ratas Mutantes , Proteínas Tirosina Quinasas Receptoras/uso terapéutico , Retina , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/ultraestructura , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Tomografía de Coherencia Óptica , Transgenes , Tirosina/genética , Tirosina Quinasa c-Mer
10.
J Neurol Sci ; 316(1-2): 150-7, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22251934

RESUMEN

Myasthenia Gravis (MG) patients with antibodies against the muscle specific tyrosine kinase (MuSK+) typically present with focal fatigue and atrophy of the facial and bulbar muscles, along with unbeneficial reactions upon administration of acetylcholinesterase inhibitors (AChEIs). This study addresses the neurophysiological characteristics in facial versus limb muscles, before and after intraperitoneal injection of AChEIs, in mice immunized with MuSK. We performed in-vivo neurophysiological examinations in the masseter and gastrocnemius muscles of mice with MuSK+experimental autoimmune MG (EAMG) and in healthy control mice before and after administration of AChEIs. Abnormal spontaneous activity (fibrillations) was observed in the masseter muscle of MuSK+mice. Furthermore, 94% of MuSK-immunized mice displayed so called extra discharges (EDs) upon administration of a therapeutic AChEI dose, in contrast to 22% of the control mice, indicating neuromuscular hyperactivity. These findings support functional denervation in the masseter muscle and neuromuscular hypersensitivity already at a standard dose of AChEIs in MuSK+EAMG.


Asunto(s)
Autoanticuerpos/biosíntesis , Inhibidores de la Colinesterasa/farmacología , Miastenia Gravis/inmunología , Miastenia Gravis/fisiopatología , Proteínas Tirosina Quinasas Receptoras/inmunología , Animales , Desnervación/métodos , Femenino , Músculo Masetero/efectos de los fármacos , Músculo Masetero/inervación , Músculo Masetero/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/inmunología , Músculo Esquelético/inervación , Miastenia Gravis/genética , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/inmunología , Distribución Aleatoria , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/genética
11.
J Clin Neuromuscul Dis ; 12(2): 76-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21386774

RESUMEN

Familial occurrence of myasthenia gravis is uncommon and reports of maternal transmission of muscle-specific tyrosine kinase (MuSK) antibody myasthenia are rarer still. We report two families with maternal transmission of MuSK antibody myasthenia gravis to the offspring by different mechanisms. The first family demonstrates transmission genetic susceptibility of inheriting myasthenia gravis from MuSK antibodies, whereas the second one demonstrates transplacental transmission of MuSK antibodies at birth.


Asunto(s)
Inmunidad Materno-Adquirida , Intercambio Materno-Fetal , Miastenia Gravis/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Colinérgicos/genética , Adulto , Autoanticuerpos/administración & dosificación , Autoanticuerpos/biosíntesis , Niño , Femenino , Humanos , Inmunidad Materno-Adquirida/genética , Recién Nacido , Masculino , Intercambio Materno-Fetal/genética , Miastenia Gravis/inmunología , Miastenia Gravis/metabolismo , Embarazo , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Receptores Colinérgicos/administración & dosificación , Adulto Joven
12.
Rio de Janeiro; s.n; 2009. 102 p. ilus.
Tesis en Portugués | LILACS | ID: lil-578261

RESUMEN

Células de todos os organismos vivos restauram lesões em DNA preservando a integridade da informação genética, através de mecanismos de reparo. A não eliminação, ou o acúmulo destas lesões ou a deficiência em uma via de reparo, leva ao acúmulo de mutações em células, podendo contribuir para o câncer. Lesões no DNA podem estar associadas com Diabetes Mellitus (DM) e pouco se sabe a respeito de possíveis distúrbios no sistema de reparo, associado a esta síndrome. A metfomina (MET), um sensibilizador de insulina, pode estar associado a incidência reduzida e melhora no prognóstico de certos tipos de cânceres em DM. Neste estudo, estamos sugerindo em eucariotos e procariotos, deficientes em enzimas de reparo de DNA, que a MET, age via enzimas de reparo, para proteger a célula contra os danos em DNA. Isto porque células XPD, deficientes na enzima de reparo XPD, pré-tratadas com MET (1mmM) e irradiadas com UVC, tiveram sobrevivência menores, avaliadas através da viabilidade celular pela exclusão do Azul de Trypan, em comparação com controle, MRC5. O mesmo ocorrendo com as cepas bacterianas AB1885, BH20 e AB2463 deficientes nas enzimas de reparo, UvrB e Fpg e do sistema SOS, respectivamente, quando foram pré-tratadas com MET (20mg=168mM) e irradiadas com UVC comparadas com suas sobrevivências sem UVC. Cepas AB1885 e BH20 transformadas em proficientes de todos os sistemas de reparo e submetidas ao mesmo tratamento com MET, mas com a dose de UVC igual à administrada para a cepa selvagem (AB1157), observou-se um aumento de sobrevivência. E ao se utilizar a cepa GY4765 (SOS+), proficiente da atividade autocatalítica do sistema SOS, nas mesmas condições de tratamento, houve também aumento da sobrevivência em relação ao controle, sendo também o sistema testado através do SOS Cromoteste onde a MET expressou este sistema. Através do ensaio da eletroforese alcalina em gel de agarose, onde se avaliou a atividade de reparo da cepa bacteriana AB1157, após o pré-tratamento...


Cells prevent the formation of injuries in DNA preserving integrity of the genetic information, through repair mechanisms. The accumulation of these injuries or the deficiency in one repair mechanism can produce mutations in cells, leading to neoplasic transformation. Injuries in the DNA can be associated with Diabetes Mellitus (DM) and little is known regarding possible disturbance in the repair system, associated with this syndrome. The use of metformin (MET), a insulin sensitizer, can be associated with reduced incidence and improvement in the prognostic of certain types of cancer in DM. In this study, we are suggesting in prokaryotes and eukaryotes deficient in repair enzymes that MET acts through these enzymes, to protect against the damages in DNA. Cells XPD, deficient in the enzyme of repair XPD, pre-treated with MET (1mM) and radiated with UVC, showed lower survival, evaluated through Trypan Blue test of cell viability in comparison with MRC5control cells. The same occurring with bacterial strains AB1885, BH20 and AB2463 deficient in repair enzymes UvrB and Fpg and the SOS system, respectively, when pre-treated with MET (20mg/ml=168mM) and radiated with UVC compared with its survival without UVC. Strains AB1885 and BH20 transformed into proficient of all the repair systems through, and submitted to the same treatment with MET and equal dose of UVC the used in the wild strain (AB1157), we observed an increase of its survival. Studying the strains GY4765 (SOS+) proficient on the autocatalytic activity of the SOS system, using the same conditions of treatment, also had increase of its survival in relation to the control, being also this system tested through Chromotest SOS where the MET expressed the SOS genes. Through alkaline electroforese in agarose gel to evaluate the repair activity on the bacterial strain AB1157 (proficient of all repair systems) the pre-treatment with MET and irradiation with UVC, confirmed the action of MET in activating repair...


Asunto(s)
Humanos , Animales , Quimioterapia Combinada , Daño del ADN/genética , Diabetes Mellitus/tratamiento farmacológico , Insulina/uso terapéutico , Metformina/farmacología , Metformina/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Reparación del ADN , Neoplasias/etiología , Neoplasias/terapia
13.
Expert Rev Anticancer Ther ; 7(12 Suppl): S29-36, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18076315

RESUMEN

The current standard of care for malignant gliomas consists of surgery, radiotherapy and conventional (DNA-damaging) chemotherapies. These treatments are relatively nonspecific and may be applied to all glioma subtypes. Developments in cancer medicine, however, now offer the opportunity to direct therapies to specific molecular pathways involved in tumorigenesis. This offers the potential to tailor treatments to tumor subtypes--perhaps with greater efficacy and less toxicity. Many of the so-called targeted therapies are under investigation in the treatment of malignant glioma. In this review, we will focus on the use of agents that affect signal transduction. In particular, we will review the potential role for inhibitors of: tyrosine kinases, targets of rapamycin, farnesyl transferase and histone deacetylase. Inhibitors of angiogenesis will also be discussed. Some 'targeted' therapies are less specific than others, working on more than one pathway or receptor, thus complex interactions are possible.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Glioma/patología , Glioma/terapia , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Encefálicas/mortalidad , Terapia Combinada , Sistemas de Liberación de Medicamentos , Femenino , Glioma/mortalidad , Humanos , Masculino , Estadificación de Neoplasias , Procedimientos Neuroquirúrgicos/métodos , Pronóstico , Radioterapia Adyuvante , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Medición de Riesgo , Análisis de Supervivencia , Resultado del Tratamiento
14.
J Neuroimmunol ; 175(1-2): 107-17, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16697051

RESUMEN

UNLABELLED: Myasthenia gravis (MG) is mostly caused by anti-acetylcholine receptor (AChR) auto-antibodies (Abs). Such Abs are undetectable in 10-15% of MG patients, but many have anti-muscle-specific kinase (MuSK) Abs. We injected recombinant rat-MuSK extracellular domain in H-2(a), H-2(b), H-2(bm12) and H-2(d) mice. Certain strains exhibited exercise-induced fatigue, tremors, weight loss, and some died after 2-3 injections. Compound muscle action potentials showed decrement with low-frequency repetitive nerve stimulation. Miniature endplate potentials decreased, suggesting lower numbers of endplates functional AChRs. Myasthenic sera inhibited agrin-induced AChR aggregation in C2C12 myotubes. CONCLUSION: Anti-MuSK Abs induce MG, which might also result from blocking the agrin-signaling pathway.


Asunto(s)
Líquido Extracelular/enzimología , Miastenia Gravis/enzimología , Miastenia Gravis/inmunología , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/administración & dosificación , Receptores Colinérgicos/inmunología , Potenciales de Acción/inmunología , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/sangre , Células Cultivadas , Cricetinae , Líquido Extracelular/inmunología , Femenino , Inmunización , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología
15.
J Clin Invest ; 115(2): 237-46, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15650770

RESUMEN

Mechanisms regulating thrombus stabilization remain largely unknown. Here, we report that loss of any 1 of the Gas6 receptors (Gas6-Rs), i.e., Tyro3, Axl, or Mer, or delivery of a soluble extracellular domain of Axl that traps Gas6 protects mice against life-threatening thrombosis. Loss of a Gas6-R does not prevent initial platelet aggregation but impairs subsequent stabilization of platelet aggregates, at least in part by reducing "outside-in" signaling and platelet granule secretion. Gas6, through its receptors, activates PI3K and Akt and stimulates tyrosine phosphorylation of the beta3 integrin, thereby amplifying outside-in signaling via alphaIIbbeta3. Blocking the Gas6-R-alphaIIbbeta3 integrin cross-talk might be a novel approach to the reduction of thrombosis.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Agregación Plaquetaria/fisiología , Transducción de Señal/fisiología , Trombosis/metabolismo , Animales , Integrina beta3/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Agregación Plaquetaria/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/genética , Trombosis/tratamiento farmacológico , Trombosis/genética , Trombosis/patología
16.
J Immunol ; 170(2): 861-9, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12517951

RESUMEN

Thyroid epithelial cells frequently express one or more members of the rearranged during transfection/papillary thyroid carcinoma (RET/PTC) fusion oncogene family during early stages of cancer, and fusion gene transcripts have been found in inflammatory conditions of the thyroid such as the autoimmune disease, Hashimoto's thyroiditis. Because these oncogenes encode chimeric proteins, novel RET/PTC epitopes may be targets of antitumor immune responses. We have been interested in the RET/PTC3 (RP3) fusion protein because this family member is more frequently expressed in radiation-induced and childhood papillary carcinomas than other members of the fusion oncogene family. We hypothesized that the activated kinase of c-RET, in the form of RP3, when expressed in patients with thyroid disease, presents an unusual altered self target for T cell recognition. Interestingly, we find that immunization with mouse RP3 protein can induce a strongly immunogenic response to RP3, although this response is not directed against the peptide comprising the unique fusion region. Rather, the responses are specific for the carboxyl-terminal portion of RP3 that is derived from the self protein c-RET. Furthermore, transplantation of RP3-expressing thyroid tumors into naive mice resulted in leukocytic infiltration, tumor rejection, and induction of RP3-specific T cells. Thus, the somatic fusion of two unrelated self proteins results in the development of a uniquely immunogenic response directed against self epitopes within RP3. These studies may better define the mechanisms controlling the initiation of thyroid-specific immune responses and provide insight into the design of novel molecules for invoking tumor-specific immunity.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Autoantígenos/metabolismo , Proteínas de Drosophila , Proteínas del Ojo , Proteínas de Fusión Oncogénica/biosíntesis , Neoplasias de la Tiroides/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Autoantígenos/administración & dosificación , Autoantígenos/genética , Subgrupos de Linfocitos B/inmunología , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Femenino , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Tolerancia Inmunológica/genética , Inmunización , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Biosíntesis de Proteínas , Estructura Terciaria de Proteína/genética , Proteínas/administración & dosificación , Proteínas/genética , Proteínas/fisiología , Proteínas Proto-Oncogénicas/administración & dosificación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas c-ret , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/inmunología , Subgrupos de Linfocitos T/inmunología , Glándula Tiroides/metabolismo , Glándula Tiroides/trasplante , Neoplasias de la Tiroides/genética , Tiroiditis Autoinmune/genética , Tiroiditis Autoinmune/inmunología , Vaccinia/genética
17.
Circulation ; 105(9): 1110-5, 2002 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-11877364

RESUMEN

BACKGROUND: It remains unclear whether vascular endothelial growth factor (VEGF) is a proarteriosclerotic or an antiarteriosclerotic factor. We recently reported that long-term inhibition of nitric oxide by administering Nomega-nitro-L-arginine methyl ester (L-NAME) induces coronary vascular inflammation and arteriosclerosis. METHODS AND RESULTS: We used this animal model to investigate the role of VEGF in arteriosclerosis. We blocked VEGF activity in vivo by transfecting with plasmid DNA encoding the murine soluble FLT-1 (sFLT-1) gene into thigh muscle. Soluble FLT-1 can suppress VEGF activity both by sequestering VEGF and by functioning as a dominant-negative inhibitor of VEGF receptors. We observed vascular inflammation associated with increased VEGF expression within 3 days of L-NAME administration, which was prevented by pretreatment with ACE inhibitor, angiotensin II receptor antagonist, or neutralizing monocyte chemoattractant protein-1 antibody. The sFLT-1 gene transfer attenuated the early vascular inflammation and prevented late arteriosclerosis. The sFLT-1 gene transfer also inhibited increased expression of monocyte chemoattractant protein-1 and transforming growth factor-beta, indicating creation of a positive feedback loop to cause arteriosclerosis. CONCLUSIONS: VEGF is necessary in the development of arteriosclerosis by mediating monocyte recruitment and activation in this model.


Asunto(s)
Arteriosclerosis/metabolismo , Factores de Crecimiento Endotelial/metabolismo , Linfocinas/metabolismo , Monocitos/metabolismo , Óxido Nítrico/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Animales , Arteriosclerosis/etiología , Arteriosclerosis/inmunología , Arteriosclerosis/patología , Bioensayo , Presión Sanguínea/efectos de los fármacos , División Celular/efectos de los fármacos , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/inmunología , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Factores de Crecimiento Endotelial/antagonistas & inhibidores , Inhibidores Enzimáticos/administración & dosificación , Expresión Génica/efectos de los fármacos , Transferencia de Gen Horizontal , Vectores Genéticos/administración & dosificación , Vectores Genéticos/biosíntesis , Vectores Genéticos/genética , Inflamación/inmunología , Inflamación/patología , Inyecciones Intramusculares , Linfocinas/antagonistas & inhibidores , Masculino , Ratones , Monocitos/inmunología , Monocitos/patología , NG-Nitroarginina Metil Éster/administración & dosificación , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico/sangre , Peptidil-Dipeptidasa A/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas WKY , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Tiempo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...