Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
1.
Pak J Pharm Sci ; 34(3): 925-932, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34602415

RESUMEN

MicroRNA (miRNA)-26a is one of the tumor suppressor genes that has been down regulated during the development of hepatocellular carcinoma (HCC). This work was conducted to evaluate the possible preventive effect of exogenous miRNA-26a administration on diethylnitrosamine (DEN)-mediated HCC. Balb/C mice were intraperitoneally injected with saline (Normal group), DEN (HCC group) or miRNA-26a (HCC+miRNA-26a group). On week 8, 12, 16 and 20, the concentrations of alpha-fetoprotein (AFP), des-gamma carboxyprothrombin (DCP), the levels of helper T cells-associated cytokines, and the vascular endothelial growth factor (VEGF), were measured. Flow cytometry determined the frequencies of regulatory T (Treg) cells. The concentrations of AFP, DCP and VEGF, as well as the frequency of Treg cells showed significantly lower values following miRNA-26a administration than in HCC group. miRNA-26a administration has reduced the levels of IL (interleukin)-2 and TNF (tumor necrosis factor)-α, in contrast, IL-10 level was markedly elevated in comparison to HCC model at all experimental time points. The restore of miRNA-26a function significantly (P<0.001) down regulated the expression levels of survivin & caspase-3 compared to HCC group. The obtained data introduce an evidence for the suppressive impact of miRNA-26a on liver tumor formation and its possible manipulation as a therapeutic design for HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas/metabolismo , Hígado/efectos de los fármacos , MicroARNs/farmacología , Alquilantes/toxicidad , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Dietilnitrosamina/toxicidad , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/patología , Ratones , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/metabolismo , Protrombina/efectos de los fármacos , Protrombina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Survivin/efectos de los fármacos , Survivin/metabolismo , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , alfa-Fetoproteínas/efectos de los fármacos , alfa-Fetoproteínas/metabolismo
2.
Clin Nutr ; 40(10): 5288-5297, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34534897

RESUMEN

BACKGROUND &AIM: The MaPLE study was a randomized, controlled, crossover trial involving adults ≥60 y.o. (n = 51) living in a residential care facility during an 8-week polyphenol-rich (PR)-diet. Results from the MaPLE trial showed that the PR-diet reduced the intestinal permeability (IP) in older adults by inducing changes to gut microbiota (GM). The present work aimed at studying the changes in serum metabolome in the MaPLE trial, as a further necessary step to depict the complex crosstalk between dietary polyphenols, GM, and intestinal barrier. METHODS: Serum metabolome was monitored using a semi-targeted UHPLC-MS/MS analysis. Metataxonomic analysis (16S rRNA gene profiling) of GM was performed on faecal samples. Clinical characteristics and serum levels of the IP marker zonulin were linked to GM and metabolomics data in a multi-omics network. RESULTS: Compared to the control diet, the PR-diet increased serum metabolites related to polyphenols and methylxanthine intake. Theobromine and methylxanthines, derived from cocoa and/or green tea, were positively correlated with butyrate-producing bacteria (the order Clostridiales and the genera Roseburia, Butyricicoccus and Faecalibacterium) and inversely with zonulin. A direct correlation between polyphenol metabolites hydroxyphenylpropionic acid-sulfate, 2-methylpyrogallol-sulfate and catechol-sulfate with Butyricicoccus was also observed, while hydroxyphenylpropionic acid-sulfate and 2-methylpyrogallol-sulfate negatively correlated with Methanobrevibacter. The multi-omics network indicated that participant's age, baseline zonulin levels, and changes in Porphyromonadaceae abundance were the main factors driving the effects of a PR-diet on zonulin. CONCLUSION: Overall, these results reveal the complex relationships among polyphenols consumption, intestinal permeability, and GM composition in older adults, and they may be important when setting personalized dietary interventions for older adults. TRIAL REGISTRATION NUMBER: ISRCTN10214981.


Asunto(s)
Dieta/métodos , Microbioma Gastrointestinal/efectos de los fármacos , Haptoglobinas/efectos de los fármacos , Mucosa Intestinal/metabolismo , Metaboloma/efectos de los fármacos , Polifenoles/farmacología , Precursores de Proteínas/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Estudios Cruzados , Femenino , Haptoglobinas/metabolismo , Humanos , Masculino , Permeabilidad , Precursores de Proteínas/metabolismo
3.
Am J Physiol Renal Physiol ; 321(3): F305-F321, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34282956

RESUMEN

Although vasopressin V1B receptor (V1BR) mRNA has been detected in the kidney, the precise renal localization as well as pharmacological and physiological properties of this receptor remain unknown. Using the selective V1B agonist d[Leu4, Lys8]VP, either fluorescent or radioactive, we showed that V1BR is mainly present in principal cells of the inner medullary collecting duct (IMCD) in the male rat kidney. Protein and mRNA expression of V1BR were very low compared with the V2 receptor (V2R). On the microdissected IMCD, d[Leu4, Lys8]VP had no effect on cAMP production but induced a dose-dependent and saturable intracellular Ca2+ concentration increase mobilization with an EC50 value in the nanomolar range. This effect involved both intracellular Ca2+ mobilization and extracellular Ca2+ influx. The selective V1B antagonist SSR149415 strongly reduced the ability of vasopressin to increase intracellular Ca2+ concentration but also cAMP, suggesting a cooperation between V1BR and V2R in IMCD cells expressing both receptors. This cooperation arises from a cross talk between second messenger cascade involving PKC rather than receptor heterodimerization, as supported by potentiation of arginine vasopressin-stimulated cAMP production in human embryonic kidney-293 cells coexpressing the two receptor isoforms and negative results obtained by bioluminescence resonance energy transfer experiments. In vivo, only acute administration of high doses of V1B agonist triggered significant diuretic effects, in contrast with injection of selective V2 agonist. This study brings new data on the localization and signaling pathways of V1BR in the kidney, highlights a cross talk between V1BR and V2R in the IMCD, and suggests that V1BR may counterbalance in some pathophysiological conditions the antidiuretic effect triggered by V2R activation.NEW & NOTEWORTHY Although V1BR mRNA has been detected in the kidney, the precise renal localization as well as pharmacological and physiological properties of this receptor remain unknown. Using original pharmaceutical tools, this study brings new data on the localization and signaling pathways of V1BR, highlights a cross talk between V1BR and V2 receptor (V2R) in the inner medullary collecting duct, and suggests that V1BR may counterbalance in some pathophysiological conditions the antidiuretic effect triggered by V2R activation.


Asunto(s)
Receptores de Vasopresinas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Vasopresinas/farmacología , Animales , Arginina Vasopresina/farmacología , Masculino , Neurofisinas/efectos de los fármacos , Precursores de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Vasopresinas/metabolismo , Vasopresinas/efectos de los fármacos
4.
Microbiol Immunol ; 64(12): 792-809, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32970362

RESUMEN

Hepatitis B virus (HBV), a major global health problem, can cause chronic hepatitis, liver cirrhosis, and hepatocellular carcinomas in chronically infected patients. However, before HBV infection can be adequately controlled, many mysteries about the HBV life cycle must be solved. In this study, TIMM29, an inner mitochondrial membrane protein, was identified as an interaction partner of the preS1 region of the HBV large S protein. The interaction was verified by both an immunoprecipitation with preS1 peptides and a GST-pulldown assay. Immunofluorescence studies also showed colocalization of preS1 and TIMM29. Moreover, it was determined that the preS1 bound with amino acids 92-189 of the TIMM29 protein. Infection of HBV in TIMM29-overexpressing NTCP/G2 cells resulted in a significant decrease of HBeAg and both extracellular particle-associated and core particle-associated HBV DNA without affecting cccDNA formation. Comparable results were obtained with TIMM29-overexpressing HB611 cells, which constitutively produce HBV. In contrast, knockout of TIMM29 in NTCP/G2 cells led to a higher production of HBV including HBeAg expression, as did knockout of TIMM29 in HB611. Collectively, these results suggested that TIMM29 interacts with the preS1 region of the HBV large S protein and modulates HBV amplification.


Asunto(s)
Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Precursores de Proteínas/metabolismo , Carcinoma Hepatocelular , Línea Celular , Proliferación Celular , Escherichia coli , Expresión Génica , Técnicas de Inactivación de Genes , Células Hep G2 , Hepatitis B/virología , Antígenos de Superficie de la Hepatitis B/efectos de los fármacos , Antígenos de Superficie de la Hepatitis B/genética , Antígenos e de la Hepatitis B , Virus de la Hepatitis B/efectos de los fármacos , Humanos , Estadios del Ciclo de Vida , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/farmacología , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Transportadores de Anión Orgánico Sodio-Dependiente , Dominios y Motivos de Interacción de Proteínas , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/genética , Simportadores , Transcriptoma
5.
Am J Physiol Endocrinol Metab ; 319(1): E81-E90, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32396496

RESUMEN

We have previously shown that systemic injection of erythropoietin-producing hepatocellular receptor A7 (EPHA7)-Fc raises serum luteinizing hormone (LH) levels before ovulation in female rats, indicating the induction of EPHA7 in ovulation. In this study, we aimed to identify the mechanism and hypothalamus-pituitary-ovary (HPO) axis level underlying the promotion of LH secretion by EPHA7. Using an ovariectomized (OVX) rat model, in conjunction with low-dose 17ß-estradiol (E2) treatment, we investigated the association between EPHA7-ephrin (EFN)A5 signaling and E2 negative feedback. Various rat models (OVX, E2-treated OVX, and abarelix treated) were injected with the recombinant EPHA7-Fc protein through the caudal vein to investigate the molecular mechanism underlying the promotion of LH secretion by EPHA7. Efna5 was observed strongly expressed in the arcuate nucleus of the female rat by using RNAscope in situ hybridization. Our results indicated that E2, combined with estrogen receptor (ER)α, but not ERß, inhibited Efna5 and gonadotropin-releasing hormone 1 (Gnrh1) expressions in the hypothalamus. In addition, the systemic administration of EPHA7-Fc restrained the inhibition of Efna5 and Gnrh1 by E2, resulting in increased Efna5 and Gnrh1 expressions in the hypothalamus as well as increased serum LH levels. Collectively, our findings demonstrated the involvement of EPHA7-EFNA5 signaling in the regulation of LH and the E2 negative feedback pathway in the hypothalamus, highlighting the functional role of EPHA7 in female reproduction.


Asunto(s)
Efrina-A5/metabolismo , Receptor alfa de Estrógeno/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Precursores de Proteínas/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Efrina-A5/efectos de los fármacos , Efrina-A5/genética , Estradiol/farmacología , Receptor beta de Estrógeno/metabolismo , Estrógenos/farmacología , Retroalimentación Fisiológica/efectos de los fármacos , Retroalimentación Fisiológica/fisiología , Femenino , Hormona Liberadora de Gonadotropina/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/efectos de los fármacos , Hormona Luteinizante/efectos de los fármacos , Oligopéptidos/farmacología , Ovariectomía , Ovario/efectos de los fármacos , Ovario/metabolismo , Precursores de Proteínas/efectos de los fármacos , Ratas , Receptor EphA7/genética , Receptor EphA7/metabolismo , Receptor EphA7/farmacología , Proteínas Recombinantes
6.
Neurotox Res ; 38(2): 370-384, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32378057

RESUMEN

Major depression disorder is one of the most common psychiatric disorders that greatly threaten the mental health of a large population worldwide. Previous studies have shown that endoplasmic reticulum (ER) stress plays an important role in the pathophysiology of depression, and current research suggests that brain-derived neurotrophic factor precursor (proBDNF) is involved in the development of depression. However, the relationship between ER and proBDNF in the pathophysiology of depression is not well elucidated. Here, we treated primary hippocampal neurons of mice with corticosterone (CORT) and evaluated the relationship between proBDNF and ERS. Our results showed that CORT induced ERS and upregulated the expression of proBDNF and its receptor, Follistatin-like protein 4 (FSTL4), which contributed to significantly decreased neuronal viability and expression of synaptic-related proteins including NR2A, PSD95, and SYN. Anti-proBDNF neutralization and ISRIB (an inhibitor of the ERS) treatment, respective ly, protected neuronal viabilities and increased the expression of synaptic-related proteins in corticosterone-exposed neurons. ISRIB treatment reduced the expression of proBDNF and FSTL4, whereas anti-proBDNF treatment did not affect ERS markers (Grp78, p-PERK, ATF4) expression. Our study presented evidence that CORT-induced ERS negatively regulated the neuronal viability and the level of synaptic-related protein of primary neurons via the proBDNF/FSTL4 pathway.


Asunto(s)
Antiinflamatorios/farmacología , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Corticosterona/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Hipocampo/citología , Neuronas/efectos de los fármacos , Precursores de Proteínas/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastorno Depresivo Mayor , Homólogo 4 de la Proteína Discs Large/efectos de los fármacos , Homólogo 4 de la Proteína Discs Large/metabolismo , Chaperón BiP del Retículo Endoplásmico , Proteínas Relacionadas con la Folistatina/efectos de los fármacos , Proteínas Relacionadas con la Folistatina/metabolismo , Ratones , Neuronas/metabolismo , Cultivo Primario de Células , Precursores de Proteínas/metabolismo , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Sinaptofisina/metabolismo
7.
J Virol ; 92(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212943

RESUMEN

Productive picornavirus infection requires the hijacking of host cell pathways to aid with the different stages of virus entry, synthesis of the viral polyprotein, and viral genome replication. Many picornaviruses, including foot-and-mouth disease virus (FMDV), assemble capsids via the multimerization of several copies of a single capsid precursor protein into a pentameric subunit which further encapsidates the RNA. Pentamer formation is preceded by co- and posttranslational modification of the capsid precursor (P1-2A) by viral and cellular enzymes and the subsequent rearrangement of P1-2A into a structure amenable to pentamer formation. We have developed a cell-free system to study FMDV pentamer assembly using recombinantly expressed FMDV capsid precursor and 3C protease. Using this assay, we have shown that two structurally different inhibitors of the cellular chaperone heat shock protein 90 (hsp90) impeded FMDV capsid precursor processing and subsequent pentamer formation. Treatment of FMDV permissive cells with the hsp90 inhibitor prior to infection reduced the endpoint titer by more than 10-fold while not affecting the activity of a subgenomic replicon, indicating that translation and replication of viral RNA were unaffected by the drug.IMPORTANCE FMDV of the Picornaviridae family is a pathogen of huge economic importance to the livestock industry due to its effect on the restriction of livestock movement and necessary control measures required following an outbreak. The study of FMDV capsid assembly, and picornavirus capsid assembly more generally, has tended to be focused upon the formation of capsids from pentameric intermediates or the immediate cotranslational modification of the capsid precursor protein. Here, we describe a system to analyze the early stages of FMDV pentameric capsid intermediate assembly and demonstrate a novel requirement for the cellular chaperone hsp90 in the formation of these pentameric intermediates. We show the added complexity involved for this process to occur, which could be the basis for a novel antiviral control mechanism for FMDV.


Asunto(s)
Proteínas de la Cápside/metabolismo , Virus de la Fiebre Aftosa/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Ensamble de Virus , Proteasas Virales 3C , Animales , Benzoquinonas/farmacología , Proteínas de la Cápside/efectos de los fármacos , Línea Celular , Supervivencia Celular , Sistema Libre de Células , Cricetinae , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Fiebre Aftosa/metabolismo , Virus de la Fiebre Aftosa/genética , Virus de la Fiebre Aftosa/crecimiento & desarrollo , Proteínas HSP90 de Choque Térmico/efectos de los fármacos , Isoxazoles/farmacología , Lactamas Macrocíclicas/farmacología , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , ARN Viral/genética , ARN Viral/metabolismo , Resorcinoles/farmacología , Proteínas Virales/efectos de los fármacos , Proteínas Virales/genética , Proteínas Virales/metabolismo , Ensamble de Virus/genética , Ensamble de Virus/fisiología , Replicación Viral
8.
Diabetes Obes Metab ; 19(6): 901-905, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28105731

RESUMEN

We assessed the effects of liraglutide treatment on five cardiovascular risk biomarkers, reflecting different pathophysiology: tumour necrosis factor (TNF)-α; soluble urokinase plasminogen activator receptor (suPAR); mid-regional pro-adrenomedullin (MR-proADM); mid-regional pro-atrial natriuretic peptide (MR-proANP); and copeptin, in people with type 2 diabetes with albuminuria. In a randomized, double-blind, placebo-controlled, crossover trial we enrolled people with type 2 diabetes and persistent albuminuria (urinary albumin-to-creatinine ratio [UACR] >30 mg/g) and estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73 m2 . Participants received liraglutide (1.8 mg/d) and matched placebo for 12 weeks, in random order. The primary endpoint was change in albuminuria; this was a prespecified sub-study. A total of 32 participants were randomized, of whom 27 completed the study. TNF-α level was 12% (95% confidence interval [CI] 3; 20) lower after liraglutide treatment compared with placebo (P = .012); MR-proADM level was 4% (95% CI 0; 8) lower after liraglutide treatment compared with placebo (P = .038), and MR-proANP level was 13% (95% CI 4; 21) lower after liraglutide treatment compared with placebo (P = .006). In the present study, we showed anti-inflammatory effects of liraglutide treatment, reflected in reductions in levels of TNF-α and MR-proADM, while the reduction in MR-proANP levels may represent a clinically relevant benefit with regard to heart failure.


Asunto(s)
Albuminuria/sangre , Enfermedades Cardiovasculares/etiología , Diabetes Mellitus Tipo 2/sangre , Hipoglucemiantes/uso terapéutico , Liraglutida/uso terapéutico , Adrenomedulina/sangre , Adrenomedulina/efectos de los fármacos , Anciano , Albuminuria/tratamiento farmacológico , Albuminuria/etiología , Factor Natriurético Atrial/sangre , Factor Natriurético Atrial/efectos de los fármacos , Biomarcadores/sangre , Estudios Cruzados , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Método Doble Ciego , Femenino , Glicopéptidos/sangre , Glicopéptidos/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/efectos de los fármacos , Precursores de Proteínas/sangre , Precursores de Proteínas/efectos de los fármacos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/sangre , Receptores del Activador de Plasminógeno Tipo Uroquinasa/efectos de los fármacos , Factores de Riesgo , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/efectos de los fármacos
9.
Am J Physiol Endocrinol Metab ; 311(2): E380-95, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27329801

RESUMEN

Type 1 diabetes (T1D) originates from autoimmune ß-cell destruction. IMT504 is an immunomodulatory oligonucleotide that increases mesenchymal stem cell cloning capacity and reverts toxic diabetes in rats. Here, we evaluated long-term (20 doses) and short-term (2-6 doses) effects of IMT504 (20 mg·kg(-1)·day(-1) sc) in an immunodependent diabetes model: multiple low-dose streptozotocin-injected BALB/c mice (40 mg·kg(-1)·day(-1) ip for 5 consecutive days). We determined blood glucose, glucose tolerance, serum insulin, islet morphology, islet infiltration, serum cytokines, progenitor cell markers, immunomodulatory proteins, proliferation, apoptosis, and islet gene expression. IMT504 reduced glycemia, induced ß-cell recovery, and impaired islet infiltration. IMT504 induced early blood glucose decrease and infiltration inhibition, increased ß-cell proliferation and decreased apoptosis, increased islet indoleamine 2,3-dioxygenase (IDO) expression, and increased serum tumor necrosis factor and interleukin-6 (IL-6). IMT504 affected islet gene expression; preproinsulin-2, proglucagon, somatostatin, nestin, regenerating gene-1, and C-X-C motif ligand-1 cytokine (Cxcl1) increased in islets from diabetic mice and were decreased by IMT504. IMT504 downregulated platelet endothelial cell adhesion molecule-1 (Pecam1) in islets from control and diabetic mice, whereas it increased regenerating gene-2 (Reg2) in islets of diabetic mice. The IMT504-induced increase in IL-6 and islet IDO expression and decreased islet Pecam1 and Cxcl1 mRNA expression could participate in keeping leukocyte infiltration at bay, whereas upregulation of Reg2 may mediate ß-cell regeneration. We conclude that IMT504 effectively reversed immunodependent diabetes in mice. Corroboration of these effects in a model of autoimmune diabetes more similar to human T1D could provide promising results for the treatment of this disease.


Asunto(s)
Glucemia/efectos de los fármacos , Citocinas/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Oligodesoxirribonucleótidos/farmacología , ARN Mensajero/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Glucemia/metabolismo , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL1/efectos de los fármacos , Quimiocina CXCL1/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Prueba de Tolerancia a la Glucosa , Indolamina-Pirrol 2,3,-Dioxigenasa/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-6/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Litostatina/efectos de los fármacos , Litostatina/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Nestina/efectos de los fármacos , Nestina/genética , Proteínas Asociadas a Pancreatitis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Proglucagón/efectos de los fármacos , Proglucagón/genética , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/genética , Proteínas/efectos de los fármacos , Proteínas/genética , ARN Mensajero/metabolismo , Somatostatina/efectos de los fármacos , Somatostatina/genética , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Transcriptoma/efectos de los fármacos , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
10.
Oncotarget ; 7(20): 29492-506, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27121052

RESUMEN

The binding of hepatocyte growth factor (HGF) to its receptor MET activates a signaling cascade that promotes cell survival, proliferation, cell scattering, migration and invasion of malignant cells. HGF is secreted by cancer cells or by tumor-associated fibroblasts as pro-HGF, an inactive precursor. A key step in the regulation of HGF/MET signaling is proteolytic processing of pro-HGF to its active form by one of the three serine proteases, matriptase, hepsin or HGF activator (HGFA).We developed SRI 31215, a small molecule that acts as a triplex inhibitor of matriptase, hepsin and HGFA and mimics the activity of HAI-1/2, endogenous inhibitors of HGF activation. We demonstrated that SRI 31215 inhibits fibroblast-induced MET activation, epithelial-mesenchymal transition and migration of cancer cells. SRI 31215 overcomes primary resistance to cetuximab and gefitinib in HGF-producing colon cancer cells and prevents fibroblast-mediated resistance to EGFR inhibitors. Thus, SRI 31215 blocks signaling between cancer cells and fibroblasts and inhibits the tumor-promoting activity of cancer-associated fibroblasts.Aberrant HGF/MET signaling supports cell survival, proliferation, angiogenesis, invasion and metastatic spread of cancer cells, establishing HGF and MET as valid therapeutic targets. Our data demonstrate that inhibitors of HGF activation, such as SRI 31215, merit investigation as potential therapeutics in tumors that are addicted to HGF/MET signaling. The findings reported here also indicate that inhibitors of HGF activation overcome primary and acquired resistance to anti-EGFR therapy, providing a rationale for concurrent inhibition of EGFR and HGF to prevent therapeutic resistance and to improve the outcome of cancer patients.


Asunto(s)
Antineoplásicos/farmacología , Benzamidinas/farmacología , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Precursores de Proteínas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Pirimidinonas/farmacología , Transducción de Señal/efectos de los fármacos , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Precursores de Proteínas/efectos de los fármacos
11.
Am J Physiol Heart Circ Physiol ; 310(6): H667-80, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26747502

RESUMEN

Mitochondrial dysfunction has been implicated as a cause of energy deprivation in heart failure (HF). Herein, we tested individual and combined effects of two pathogenic factors of nonischemic HF, inhibition of nitric oxide synthesis [with l-N(G)-nitroarginine methyl ester (l-NAME)] and hypertension [with angiotensin II (AngII)], on myocardial mitochondrial function, oxidative stress, and metabolic gene expression. l-NAME and AngII were administered individually and in combination to mice for 5 wk. Although all treatments increased blood pressure and reduced cardiac contractile function, the l-NAME + AngII group was associated with the most severe HF, as characterized by edema, hypertrophy, oxidative stress, increased expression of Nppa and Nppb, and decreased expression of Atp2a2 and Camk2b. l-NAME + AngII-treated mice exhibited robust deterioration of cardiac mitochondrial function, as observed by reduced respiratory control ratios in subsarcolemmal mitochondria and reduced state 3 levels in interfibrillar mitochondria for complex I but not for complex II substrates. Cardiac myofibrils showed reduced ADP-supported and oligomycin-inhibited oxygen consumption. Mitochondrial functional impairment was accompanied by reduced mitochondrial DNA content and activities of pyruvate dehydrogenase and complex I but increased H2O2 production and tissue protein carbonyls in hearts from AngII and l-NAME + AngII groups. Microarray analyses revealed the majority of the gene changes attributed to the l-NAME + AngII group. Pathway analyses indicated significant changes in metabolic pathways, such as oxidative phosphorylation, mitochondrial function, cardiac hypertrophy, and fatty acid metabolism in l-NAME + AngII hearts. We conclude that l-NAME + AngII is associated with impaired mitochondrial respiratory function and increased oxidative stress compared with either l-NAME or AngII alone, resulting in nonischemic HF.


Asunto(s)
Angiotensina II/farmacología , Inhibidores Enzimáticos/farmacología , Insuficiencia Cardíaca/etiología , Mitocondrias Cardíacas/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Vasoconstrictores/farmacología , Animales , Factor Natriurético Atrial , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Cardiomegalia , ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/metabolismo , Complejo I de Transporte de Electrón/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/efectos de los fármacos , Complejo II de Transporte de Electrones/metabolismo , Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Ratones , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Miocardio/patología , Péptido Natriurético Encefálico/efectos de los fármacos , Péptido Natriurético Encefálico/genética , Péptido Natriurético Tipo-C/efectos de los fármacos , Péptido Natriurético Tipo-C/genética , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/genética , Complejo Piruvato Deshidrogenasa/efectos de los fármacos , Complejo Piruvato Deshidrogenasa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética
12.
Peptides ; 64: 1-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25541043

RESUMEN

OBJECTIVE: Hemokinin-1, the newest tachykinin encoded by the preprotachykinin C (Tac4) gene, is predominatly produced by immune cells. Similarly to substance P, it has the greatest affinity to the tachykinin NK1 receptor, but has different binding site and signaling mechanisms. Furthermore, several recent data indicate the existence of a not yet identified own receptor and divergent non-NK1-mediated actions. Since there is no information on its functions in the airways, we investigated its role in endotoxin-induced pulmonary inflammation. METHODS: Acute pneumonitis was induced in Tac4 gene-deleted (Tac4(-/-)) mice compared to C57Bl/6 wildtypes by intranasal E. coli lipopolysaccharide (LPS). Airway responsiveness to inhaled carbachol was measured with unrestrained whole body plethysmography 24h later. Semiquantitative histopathological scoring was performed; reactive oxygen species (ROS) production was measured with luminol bioluminescence, myeloperoxidase activity with spectrophotometry, and inflammatory cytokines with Luminex. RESULTS: All inflammatory parameters, such as histopathological alterations (perivascular edema, neutrophil/macrophage accumulation, goblet cell hyperplasia), myeloperoxidase activity, ROS production, as well as interleukin-1beta, interleukin-6, tumor necrosis factor alpha, monocyte chemoattractant protein-1 and keratinocyte chemoattractant concentrations were significantly diminished in the lung of Tac4(-/-) mice. However, bronchial hyperreactivity similarly developed in both groups. Interestingly, in LPS-treated Tac4(-/-) mouse lungs, bronchus-associated, large, follicle-like lymphoid structures developed. CONCLUSIONS: We provide the first evidence that hemokinin-1 plays a crucial pro-inflammatory role in the lung by increasing inflammatory cell activities, and might also be a specific regulator of lymphocyte functions.


Asunto(s)
Neumonía/fisiopatología , Precursores de Proteínas/fisiología , Taquicininas/fisiología , Enfermedad Aguda , Animales , Citocinas/metabolismo , Femenino , Lipopolisacáridos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Neumonía/inducido químicamente , Neumonía/inmunología , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/inmunología , Taquicininas/efectos de los fármacos , Taquicininas/inmunología
13.
Wound Repair Regen ; 22(5): 640-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25039304

RESUMEN

The development of new methods to improve skin wound healing may affect the outcomes of a number of medical conditions. Here, we evaluate the molecular and clinical effects of topical 5-azacytidine on wound healing in rats. 5-Azacytidine decreases the expression of follistatin-1, which negatively regulates activins. Activins, in turn, promote cell growth in different tissues, including the skin. Eight-week-old male Wistar rats were submitted to 8.0-mm punch-wounding in the dorsal region. After 3 days, rats were randomly assigned to receive either a control treatment or the topical application of a solution containing 5-azacytidine (10 mM) once per day. Photo documentation and sample collection were performed on days 5, 9, and 15. Overall, 5-azacytidine promoted a significant acceleration of complete wound healing (99.7% ± 0.7.0 vs. 71.2% ± 2.8 on day 15; n = 10; p < 0.01), accompanied by up to threefold reduction in follistatin expression. Histological examination of the skin revealed efficient reepithelization and cell proliferation, as evaluated by the BrdU incorporation method. 5-Azacytidine treatment also resulted in increased gene expression of transforming growth factor-beta and the keratinocyte markers involucrin and cytokeratin, as well as decreased expression of cytokines such as tumor necrosis factor-alpha and interleukin-10. Lastly, when recombinant follistatin was applied to the skin in parallel with topical 5-azacytidine, most of the beneficial effects of the drug were lost. Thus, 5-azacytidine acts, at least in part through the follistatin/activin pathway, to improve skin wound healing in rodents.


Asunto(s)
Azacitidina/farmacología , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Folistatina/efectos de los fármacos , Piel/lesiones , Cicatrización de Heridas/efectos de los fármacos , Activinas/efectos de los fármacos , Administración Cutánea , Animales , Expresión Génica/efectos de los fármacos , Interleucina-10/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinas/efectos de los fármacos , Queratinas/metabolismo , Masculino , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
14.
Artículo en Inglés | MEDLINE | ID: mdl-23643583

RESUMEN

OBJECTIVE: Surgical approaches for the mobilization of impacted teeth involve the use of gold chains to connect the impacted tooth with the orthodontic appliance. In this study we have compared the local effects gold plated stainless steel with stainless steel specimen using an in vitro model of the gingival mucosa and monolayer cultures of cells of the alveolus. STUDY DESIGN: Local effects on differentiation, proliferation, and apoptosis and inflammatory response were tested using organotypic cultures of gingival cells. Cytotoxicity was measured by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assays with monolayer cultures of human periodontal cells. RESULTS: The data obtained in this study could not reveal any differences in favor of using gold plated chains during the mobilization of impacted teeth. CONCLUSION: For patients not suffering from allergies against nickel there might be no rationale to favor gold plated chains, as there are no functional aspects to favor gold plated chains over stainless steel chains.


Asunto(s)
Materiales Biocompatibles Revestidos/toxicidad , Aleaciones de Oro/toxicidad , Aparatos Ortodóncicos , Extrusión Ortodóncica/instrumentación , Acero Inoxidable/química , Diente Impactado/terapia , Apoptosis/efectos de los fármacos , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Galvanoplastia , Fibroblastos/efectos de los fármacos , Proteínas Filagrina , Encía/citología , Encía/efectos de los fármacos , Humanos , Interleucina-1beta/efectos de los fármacos , Interleucina-6/análisis , Proteínas de Filamentos Intermediarios/efectos de los fármacos , Queratina-14/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Ensayo de Materiales , Osteoblastos/efectos de los fármacos , Ligamento Periodontal/citología , Ligamento Periodontal/efectos de los fármacos , Precursores de Proteínas/efectos de los fármacos
15.
ACS Chem Neurosci ; 4(5): 747-60, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23496922

RESUMEN

Accumulating evidence supports the value of 5-HT1A receptor (5-HT1AR) agonists for dyskinesias that arise with long-term L-DOPA therapy in Parkinson's disease (PD). Yet, how 5-HT1AR stimulation directly influences the dyskinetogenic D1 receptor (D1R)-expressing striatonigral pathway remains largely unknown. To directly examine this, one cohort of hemiparkinsonian rats received systemic injections of Vehicle + Vehicle, Vehicle + the D1R agonist SKF81297 (0.8 mg/kg), or the 5-HT1AR agonist ±8-OH-DPAT (1.0 mg/kg) + SKF81297. Rats were examined for changes in abnormal involuntary movements (AIMs), rotations, striatal preprodynorphin (PPD), and glutamic acid decarboxylase (GAD; 65 and 67) mRNA via RT-PCR. In the second experiment, hemiparkinsonian rats received intrastriatal pretreatments of Vehicle (aCSF), ±8-OH-DPAT (7.5 mM), or ±8-OH-DPAT + the 5-HT1AR antagonist WAY100635 (4.6 mM), followed by systemic Vehicle or SKF81297 after which AIMs, rotations, and extracellular striatal glutamate and nigral GABA efflux were measured by in vivo microdialysis. Results revealed D1R agonist-induced AIMs were reduced by systemic and intrastriatal 5-HT1AR stimulation while rotations were enhanced. Although ±8-OH-DPAT did not modify D1R agonist-induced increases in striatal PPD mRNA, the D1R/5-HT1AR agonist combination enhanced GAD65 and GAD67 mRNA. When applied locally, ±8-OH-DPAT alone diminished striatal glutamate levels while the agonist combination increased nigral GABA efflux. Thus, presynaptic 5-HT1AR stimulation may attenuate striatal glutamate levels, resulting in diminished D1R-mediated dyskinetic behaviors, but maintain or enhance striatal postsynaptic factors ultimately increasing nigral GABA levels and rotational activity. The current findings offer a novel mechanistic explanation for previous results concerning 5-HT1AR agonists for the treatment of dyskinesia.


Asunto(s)
Agonistas de Dopamina/farmacología , Actividad Motora/efectos de los fármacos , Neostriado/efectos de los fármacos , Receptor de Serotonina 5-HT1A , Receptores de Dopamina D1/agonistas , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Sustancia Negra/efectos de los fármacos , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Conducta Animal/efectos de los fármacos , Benzazepinas/farmacología , Dinorfinas/efectos de los fármacos , Dinorfinas/metabolismo , Discinesia Inducida por Medicamentos , Glutamato Descarboxilasa/efectos de los fármacos , Glutamato Descarboxilasa/metabolismo , Trastornos Parkinsonianos , Piperazinas/farmacología , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/metabolismo , Piridinas/farmacología , Ratas , Antagonistas del Receptor de Serotonina 5-HT1/farmacología
17.
Acta Med Indones ; 44(2): 140-4, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22745145

RESUMEN

AIM: to determine the effect of oral N-acetylcysteine (NAC) on plasma levels of inflammatory markers in Continuous Ambulatory Peritoneal Dialysis (CAPD) patients. METHODS: we performed a placebo-controlled study over 8 weeks in 32 patients on regular CAPD. The patients were divided into 2 groups of 16 patients matched for age and gender. The first group was given NAC 2x600 mg/day for 8 weeks and inflammatory parameter was compared with control group. The immune system is determined from the average levels of Procalcitonin, IL-6, IL-1, C3, SICAM, hsCRP, and TNF- before and after treatment with NAC. Student t-test was performed to compare the means between NAC receiving and control groups. All statistics were done using SPSS software (SPSS Ver 16.0). RESULTS: administration of NAC, significantly diminished PCT (-0.38±0.57 vs 0.09±0.14; p=0.004), IL-6 (-1.94±3.03 vs 1.19±1.99; p=0.002), IL-1 (-0.14±0.21 vs 0.01±0.04; p=0.010), C3 (-7.40±12.04 vs 4.60±8.12; p=0.002), sICAM (-80.59±29.18 vs -35.02±46.99; p=0.007), hsCRP (-1.50±1.32 vs 0.81±1.17; p<0.001) and TNF- (-0.73±0.47 vs 0.14±0.74; p<0.001) levels compared control to group. CONCLUSION: short-term oral NAC treatment resulted in reduction of circulating PCT, IL-6, IL-1, C3, sICAM, hsCRP, and TNF- in CAPD patients.


Asunto(s)
Acetilcisteína/farmacología , Antioxidantes/farmacología , Inflamación/sangre , Inflamación/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Diálisis Peritoneal Ambulatoria Continua , Adulto , Biomarcadores/sangre , Proteína C-Reactiva/efectos de los fármacos , Proteína C-Reactiva/metabolismo , Calcitonina/sangre , Calcitonina/efectos de los fármacos , Péptido Relacionado con Gen de Calcitonina , Complemento C3/efectos de los fármacos , Complemento C3/metabolismo , Femenino , Humanos , Interleucina-1/sangre , Interleucina-6/sangre , Fallo Renal Crónico/inmunología , Fallo Renal Crónico/terapia , Masculino , Persona de Mediana Edad , Precursores de Proteínas/sangre , Precursores de Proteínas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/efectos de los fármacos
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 27(3): 337-8, 2011 Mar.
Artículo en Chino | MEDLINE | ID: mdl-21638936

RESUMEN

AIM: To investigate hCG on the expression of epidermal growth factor (EGF) in renal tissues. METHODS: 20 male SD rats weighed 300 - 350 g were injected hCG 200 IU/d for 7 days, and 20 male rats were injected sa-line at equal volume as a control. Kidney tissues were per-formed with immunohistochemical staining to evaluate the distribution of EGF and quantitative PCR (Q-PCR) was used to evaluate the EGF mRNA. RESULTS: Immunohistochemtry showed that the positive rate of EGF in experimental group was significantly higher than that in control group(P<0.01), but Q-PCR results showed no significant difference between two groups ( P > 0. 05). CONCLUSION: hCG increased the concentration of EGF by maturing pre-pro-EGF not expressing EGF from de novo synthesis.


Asunto(s)
Gonadotropina Coriónica/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Riñón/metabolismo , Precursores de Proteínas/efectos de los fármacos , ARN Mensajero/metabolismo , Animales , Factor de Crecimiento Epidérmico/efectos de los fármacos , Inmunohistoquímica , Riñón/citología , Masculino , Ratas , Ratas Sprague-Dawley
19.
Acta Pharmacol Sin ; 31(7): 765-74, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20581854

RESUMEN

AIM: To investigate the protective effects of octacosanol in 6-hydroxydopamine-induced Parkinsonian rats and find whether octacosanol has effects on pro nerve growth factor (pro-NGF), NGF and the downstream effector proteins. METHODS: Behavioral tests, enzymatic assay, tyrosine hydroxylase immunohistochemistry, TUNEL and Western blot were used to investigate the effects of octacosanol in this rat model of PD. RESULTS: Oral administration of octacosanol (35-70 mg/kg, po for 14 d) significantly improved the behavioral impairments in rats induced by 6-OHDA and dose-dependently preserved the free radical scavenging capability of the striatum. Octacosanol treatment also effectively ameliorated morphological appearances of TH-positive neuronal cells in nigrostriatal systems and decreased the apoptotic cells induced by 6-OHDA in striatum. In addition, octacosanol strikingly blocked the 6-OHDA-induced increased expression of proNGF-p75NTR-sortilin death signaling complex and its downstream effector proteins. Meantime, octacosanol prevented the decreased levels of NGF, its receptors TrkA and p-Akt which together mediated the cell survival pathway. CONCLUSION: The findings implicated that the anti-parkinsonism effects afforded by octacosanol might be mediated by its neuro-microenvironment improving potency through retrieving the ratios of proNGF:NGF and the respective receptors p75NTR:TrkA in vivo. Due to its excellent tolerability and non-toxicity, octacosanol may be a promising agent for PD treatment.


Asunto(s)
Antiparkinsonianos/farmacología , Alcoholes Grasos/farmacología , Trastornos Parkinsonianos/tratamiento farmacológico , Animales , Antiparkinsonianos/administración & dosificación , Antiparkinsonianos/toxicidad , Western Blotting , Relación Dosis-Respuesta a Droga , Alcoholes Grasos/administración & dosificación , Alcoholes Grasos/toxicidad , Masculino , Factor de Crecimiento Nervioso/efectos de los fármacos , Factor de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/efectos de los fármacos , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso , Oxidopamina , Trastornos Parkinsonianos/fisiopatología , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor trkA/metabolismo , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/metabolismo
20.
Artículo en Inglés | MEDLINE | ID: mdl-20383340

RESUMEN

The purpose of this study was to investigate the effects of capsaicin (CAP) and resiniferatoxin (RTX) on substance P (SP) release and transient receptor potential vanilloid type 1 (TRPV1) expression of cultured rat dorsal root ganglion (DRG) neurons. Dissociated DRG cells of embryonic 15-day-old Wistar rat were cultured for 3 days and then exposed to CAP (1 micromol/L, 10 micromol/L) or RTX (10 nmol/L, 100 nmol/L) for 10 min. At 3 days of culture growth, SP release increased significantly after 10 min of stimulation with CAP or RTX as compared with controls. Six days after acute exposure to CAP or RTX, SP release and SP expression of cultured rat DRG neurons decreased significantly in both CAP- and RTX-treated cultures as compared with controls. Preprotachykinin (PPT) mRNA, TRPV1 mRNA and TRPV1 protein expression decreased in CAP-treated cultures and RTX cultures treated with the higher concentration, whereas RTX cultures treated with the lower concentration were not affected. The results indicate that CAP and high concentrations of RTX are more neurotoxic to cultured rat DRG neurons while the inability of the neurons to express SP or TRPV1 after acute exposure to the lower concentration of RTX could be partially reversed after a period of incubation. The efficacy and therapeutic potential of the reversible action of RTX are more applicable as strategies for pain or neurogenic inflammation therapy.


Asunto(s)
Capsaicina/farmacología , Diterpenos/farmacología , Sustancia P/efectos de los fármacos , Canales Catiónicos TRPV/efectos de los fármacos , Animales , Capsaicina/administración & dosificación , Capsaicina/toxicidad , Células Cultivadas , Diterpenos/administración & dosificación , Diterpenos/toxicidad , Relación Dosis-Respuesta a Droga , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Fármacos del Sistema Sensorial/administración & dosificación , Fármacos del Sistema Sensorial/farmacología , Fármacos del Sistema Sensorial/toxicidad , Sustancia P/metabolismo , Canales Catiónicos TRPV/genética , Taquicininas/efectos de los fármacos , Taquicininas/genética , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA