Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Bull Exp Biol Med ; 172(1): 18-21, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34796426

RESUMEN

Proinflammatory S100A9 protein is a promoter of inflammation-linked neurodegeneration and the Tnfrsf1A gene encodes the TNF receptor 1A that binds TNFα to function as a regulator of inflammation. We studied the effects of chronic intranasal administration of in vitro prepared S100A9 fibrils alone or in combination with anti-glutamate antibodies on the expression of the Tnfrsf1A gene in the hippocampus, prefrontal cortex, and cerebellum of aging C57BL/6 mice under conditions of impaired spatial memory. A differential cerebral pattern of Tnfrsf1A gene activity and its modification by S100A9 fibrillar structures were observed: inhibition of Tnfrsf1A gene expression in the hippocampus and cerebellum and its activation in the prefrontal cortex. Anti-glutamate antibodies normalized the expression of the Tnfrsf1A gene in the prefrontal cortex by affecting the TNF signaling pathway and preventing the development of inflammation.


Asunto(s)
Envejecimiento/fisiología , Amnesia/patología , Calgranulina B/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Memoria Espacial/fisiología , Animales , Anticuerpos/inmunología , Cerebelo/metabolismo , Ácido Glutámico/inmunología , Hipocampo/metabolismo , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Corteza Prefrontal/metabolismo , Navegación Espacial/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
2.
Neuromolecular Med ; 22(4): 517-533, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32813234

RESUMEN

Impact of reactive oxygen species (ROS) in development of hyperalgesia has recently motivated scientists to focus on ROS as novel target of anti-hyperalgesic interventions. Studies have indicated the usefulness of ROS scavengers and exogenous antioxidants as anti-nociceptive agents in animal models of neuropathic and inflammatory hyperalgesia. In present study, we suggest the anti-hyperalgesic potential of the dietary antioxidant quercetin on chronic inflammatory hyperalgesia induced by Complete Freund's Adjuvant (CFA). Three doses of quercetin (25, 50 and 75 mg/kg body weight) for consecutive 7 days were used for the study. Thermal hyperalgesia was assessed by paw withdrawal latency (PWL) test and inflammation was checked in terms of changes in paw edema. The insight of molecular signaling during chronic hyperalgesia was analyzed by TNF-α-TNFR1-ERK1/2 pathway in relation to change in ROS level in DRG and spinal cord. CFA-induced hyperalgesia was confirmed by decreased PWL and increased c-Fos activity in dorsal horn of spinal cord, determined by immunohistochemical analysis. It was characterized with elevated level of ROS and TNF-α estimated by ELISA. The activation of ERK1/2 and NF-κB in DRG and spinal cord and over-expression of TNFR1 in DRG were analyzed by Western blotting. Up-regulation of Iba1 and GFAP indicates glial activation in spinal cord. Expression of GFAP and its co-localization with NF-κB were examined by immunofluorescence. All the molecular modulators of hyperalgesia were brought towards normal after quercetin treatment showing its anti-hyperalgesic activity, indicating that repeated quercetin treatment is able to alleviate chronic inflammatory hyperalgesia by attenuating TNF-α-TNFR1-ERK1/2 signaling pathway via modulation of ROS and by suppression of central sensitization via inhibition of spinal glial activation.


Asunto(s)
Astrocitos/efectos de los fármacos , Adyuvante de Freund/toxicidad , Hiperalgesia/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microglía/efectos de los fármacos , Quercetina/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Animales , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/inducido químicamente , Hiperalgesia/etiología , Hiperalgesia/patología , Inflamación , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Quercetina/farmacología , Distribución Aleatoria , Ratas , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Factor de Necrosis Tumoral alfa/metabolismo
3.
J Clin Invest ; 129(8): 3103-3120, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31112137

RESUMEN

Mechanisms underlying motor neuron degeneration in amyotrophic lateral sclerosis (ALS) are yet unclear. Specific deletion of the ER-component membralin in astrocytes manifested postnatal motor defects and lethality in mice, causing the accumulation of extracellular glutamate through reducing the glutamate transporter EAAT2. Restoring EAAT2 levels in membralin KO astrocytes limited astrocyte-dependent excitotoxicity in motor neurons. Transcriptomic profiles from mouse astrocytic membralin KO motor cortex indicated significant perturbation in KEGG pathway components related to ALS, including downregulation of Eaat2 and upregulation of Tnfrsf1a. Changes in gene expression with membralin deletion also overlapped with mouse ALS models and reactive astrocytes. Our results shown that activation of TNF receptor (TNFR1)-NFκB pathway known to suppress Eaat2 transcription was upregulated with membralin deletion. Further, reduced membralin and EAAT2 levels correlated with disease progression in spinal cord from SOD1-mutant mouse models, and reductions in membralin/EAAT2 were observed in human ALS spinal cord. Importantly, overexpression of membralin in SOD1G93A astrocytes decreased TNFR1 levels and increased EAAT2 expression, and improved motor neuron survival. Importantly, upregulation of membralin in SOD1G93A mice significantly prolonged mouse survival. Together, our study provided a mechanism for ALS pathogenesis where membralin limited glutamatergic neurotoxicity, suggesting that modulating membralin had potentials in ALS therapy.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Corteza Motora/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/patología , Regulación hacia Abajo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Ácido Glutámico/genética , Humanos , Ratones , Ratones Noqueados , Corteza Motora/patología , Proteínas del Tejido Nervioso/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Transcripción Genética , Regulación hacia Arriba
4.
Environ Pollut ; 244: 486-494, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30366296

RESUMEN

Environmental estrogens are capable of interfering with the spermatogenesis and fertility of fish. However in natural waters, these chemicals are more likely to occur as a combination rather than a single stressor. Whether and how the mixture of xenoestrogens with environmental relevant concentrations may affect fish spermatogenesis remains largely unknown. In this study, male zebrafish adults were administered to 17alpha-ethinylestradiol (EE2) and a mixture of xenoestrogens (Mix (E2, EE2, DES, 4-t-OP, 4-NP and BPA)), with the estrogenic potency equivalent to EE2. After a 60-day exposures, elevated mRNA levels of vitellogenin 1 (vtg1) and estrogen receptor 1 (esr1) in the liver of fish in both treated groups were observed. Moreover, the plasma level of E2 declined significantly in the Mix group and the ratio of 11-KT/E2 was significantly elevated in both treated groups. Consistently, the mRNA level of P450 side-chain cleavage (scc) in the EE2 group and ovarian type aromatase (cyp19a1a) in the Mix group was significantly suppressed. In addition, decreased gonadosomatic index and sperm count in the fish of Mix group were present. Furthermore, increased number of the proliferating germ cells (such as spermatogonia and spermatocytes) was observed in the fish of both groups, suggesting a stimulated germ cell proliferation and meiosis. Accordingly, both exposures significantly up-regulated the mRNA levels of genes in mitosis (cyclinb1) and meiosis (cyp26a1 in EE2 group, aldh1a2, cyp26a1, sycp3 and spo11 in Mix). In addition, decreased number of spermatozoa and increased number of TUNEL-positive signals were present in the testis of fish in the Mix group, indicating an enhanced apoptosis. Further analyses demonstrated the significant elevated expressions of tnfrsf1a and the ratio of tnfrsf1a/tnfrsf1b in the Mix group, suggesting an elevated apoptosis in the testis of fish in the Mix group via extrinsic pathway. The present study greatly extends our understanding of the underlying mechanisms of the reproductive toxicity of xenoestrogens on fish.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Estrógenos/sangre , Etinilestradiol/toxicidad , Meiosis/efectos de los fármacos , Espermatogénesis/fisiología , Espermatozoides/citología , Animales , Exposición a Riesgos Ambientales , Receptor alfa de Estrógeno/metabolismo , Femenino , Hormonas Esteroides Gonadales , Masculino , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Reproducción/fisiología , Ácido Retinoico 4-Hidroxilasa/biosíntesis , Recuento de Espermatozoides , Espermatogonias/citología , Testículo/metabolismo , Vitelogeninas/metabolismo , Pez Cebra
5.
Clin Exp Med ; 18(4): 547-554, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29802567

RESUMEN

The aims of the study were to compare the levels of tumor necrosis factor alpha (TNF-α) and its soluble type I (sTNF-R1) and type II (sTNF-R2) receptors detected in intracystic liquid and serum from benign and malignant ovarian neoplasms and to relate them to prognostic factors in epithelial ovarian cancer. The patients were divided into benign ovarian neoplasms (n = 46) and malignant ovarian neoplasms (n = 17). The serum and intracystic samples were collected before and during surgery for ovarian cyst, respectively. The levels of TNF-α, sTNF-R1, and sTNF-R2 were measured using ELISA. Results were compared with the Mann-Whitney test. Concentration of sTNF-R2 in the intracystic samples collected from the malignant neoplasia was significantly higher than that of the benign neoplasias (p = 0.02). Higher intracystic levels of sTNF-R2 exhibited a significant association with tumor differentiation grades 2 and 3 (p = 0.0087). There was no statistical significance in relation to serum levels. Tumor microenvironment levels of sTNF-R2 may represent a factor of poor prognosis in epithelial ovarian cancer.


Asunto(s)
Carcinoma Epitelial de Ovario/metabolismo , Neoplasias Ováricas/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/biosíntesis , Carcinoma Epitelial de Ovario/diagnóstico , Femenino , Humanos , Neoplasias Ováricas/diagnóstico , Pronóstico
6.
Exp Neurol ; 306: 190-198, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29772244

RESUMEN

This study investigated the effects of progesterone (PROG) on neonatal hypoxic/ischemic (NHI) brain injury, the differences in effects between genders, and the underlying mechanisms. NHI brain injury was established in both male and female neonatal mice induced by occlusion of the left common carotid artery followed by hypoxia. The mice were treated with PROG or vehicle. Fluoro-Jade B staining (F-JB), long term behavior testing, and brain magnetic resonance image (MRI) were applied to evaluate neuronal death, neurological function, and brain damage. The underlying molecular mechanisms were also investigated by Western blots. The results showed that, in the male mice, administration of PROG significantly reduced neuronal death, improved the learning and memory function impaired by cerebral HI, decreased infarct size, and maintained the thickness of the cortex after cerebral HI. PROG treatment, however, did not show significant neuroprotective effects on female mice subjected to HI. In addition, the data demonstrated a gender difference in the expression of tumor necrosis factor receptor 1 (TNFR1), TNF receptor associated factor 6 (TRAF6), Fas associated protein with death domain (FADD), and TIR-domain-containing adapter-inducing interferon-ß (TRIF) between males and females. Our results indicated that treatment with PROG had beneficial effects on NHI injured brain in acute stage and improved the long term cognitive function impaired by cerebral HI in male mice. In addition, the activation of TNF and TRIF mediated signaling in response to cerebral HI and the treatment of PROG varied between genders, which highly suggested that gender differences should be emphasized in evaluating neonatal HI brain injury and PROG effects, as well as the underlying mechanisms.


Asunto(s)
Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Progesterona/uso terapéutico , Animales , Animales Recién Nacidos , Encéfalo/patología , Estenosis Carotídea , Cognición/efectos de los fármacos , Femenino , Hipoxia-Isquemia Encefálica/patología , Imagen por Resonancia Magnética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Caracteres Sexuales , Factor 6 Asociado a Receptor de TNF/biosíntesis , Factor 6 Asociado a Receptor de TNF/genética
7.
J Musculoskelet Neuronal Interact ; 17(3): 226-236, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28860425

RESUMEN

BACKGROUND: The peritendinous connective tissues can have importance in chronic tendon pain. Recently cytokine TNF-α has been suggested to be involved in tendinopathic processes. It is not known how TNF-α and its receptors TNFR1 and TNFR2 are expressed in peritendinous tissues. METHODS: The objective for this study was to immunohistochemically evaluate the expression patterns of these in the peritendinous tissue located between the plantaris and Achilles tendons and the one located superficially to the extensor origin at the elbow region for patients with tendinopathy/tennis elbow. RESULTS: The nerve fascicles were of two types, one type being homogenously stained for the nerve markers ßIII-tubulin and neurofilament and the other showing deficits for these suggesting features of axonal damage. Much more distinct TNFR1/TNFR2 immunoreactions were seen for the latter nerve fascicles. TNFR1 was seen in axons, TNFR2 mainly in Schwann cells. TNFR1 and particularly TNFR2 were seen in walls of parts of blood vessels. The dispersed cells showed frequently TNFR1 and TNFR2 immunoreactivity. DISCUSSION: These findings suggest that TNF-α can be related to degenerative events but also attempts for healing concerning the nerve structures. The marked expression of the TNF-α system in the peritendinous tissue suggests an impact of TNF-α in tendinopathy/tennis elbow.


Asunto(s)
Tejido Conectivo/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Codo de Tenista/metabolismo , Adulto , Axones/patología , Tejido Conectivo/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nervios Periféricos , Codo de Tenista/patología
8.
Am J Physiol Heart Circ Physiol ; 313(4): H744-H756, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28710070

RESUMEN

In systolic heart failure (HF), circulating proinflammatory cytokines upregulate inflammation and renin-angiotensin system (RAS) activity in cardiovascular regions of the brain, contributing to sympathetic excitation and cardiac dysfunction. Important among these is the subfornical organ (SFO), a forebrain circumventricular organ that lacks an effective blood-brain barrier and senses circulating humors. We hypothesized that the tumor necrosis factor-α (TNF-α) receptor 1 (TNFR1) in the SFO contributes to sympathetic excitation and cardiac dysfunction in HF rats. Rats received SFO microinjections of a TNFR1 shRNA or a scrambled shRNA lentiviral vector carrying green fluorescent protein, or vehicle. One week later, some rats were euthanized to confirm the accuracy of the SFO microinjections and the transfection potential of the lentiviral vector. Other rats underwent coronary artery ligation (CL) to induce HF or a sham operation. Four weeks after CL, vehicle- and scrambled shRNA-treated HF rats had significant increases in TNFR1 mRNA and protein, NF-κB activity, and mRNA for inflammatory mediators, RAS components and c-Fos protein in the SFO and downstream in the hypothalamic paraventricular nucleus, along with increased plasma norepinephrine levels and impaired cardiac function, compared with vehicle-treated sham-operated rats. In HF rats treated with TNFR1 shRNA, TNFR1 was reduced in the SFO but not paraventricular nucleus, and the central and peripheral manifestations of HF were ameliorated. In sham-operated rats treated with TNFR1 shRNA, TNFR1 expression was also reduced in the SFO but there were no other effects. These results suggest a key role for TNFR1 in the SFO in the pathophysiology of systolic HF.NEW & NOTEWORTHY Activation of TNF-α receptor 1 in the subfornical organ (SFO) contributes to sympathetic excitation in heart failure rats by increasing inflammation and renin-angiotensin system activity in the SFO and downstream in the hypothalamic paraventricular nucleus. Cytokine receptors in the SFO may be a target for central intervention in cardiovascular conditions characterized by peripheral inflammation.


Asunto(s)
Circulación Coronaria/genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Órgano Subfornical/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Animales , Electrocardiografía , Técnicas de Silenciamiento del Gen , Hemodinámica/efectos de los fármacos , Masculino , FN-kappa B/metabolismo , Norepinefrina/sangre , Proteínas Proto-Oncogénicas c-fos/biosíntesis , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Sistema Renina-Angiotensina , Factor de Necrosis Tumoral alfa/farmacología
9.
Cancer Chemother Pharmacol ; 79(4): 747-758, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28315953

RESUMEN

PURPOSE: Hepatocellular carcinoma (HCC) is a common digestive system malignancy that is associated with a poor prognosis. This study researched the interaction of tumor necrosis factor-α (TNF-α) and angiotensin II (Ang II) in HCC cells proliferation, migration and invasion and examined their influence on the expression of G protein-coupled receptor kinase 2 (GRK2) and relevant receptors. METHODS: Cell Counting Kit-8 and Transwell assays were performed to evaluate the effects of TNF-α and Ang II on HepG2 cells proliferation, migration and invasion. Flow cytometry was used to investigate the expression of tumor necrosis factor receptor 1 (TNFR1), angiotensin II type 1 (AT1R) and type 2 receptors (AT2R) on the surface of HepG2 cells. Additionally, Western blot was performed to assess the modulation of GRK2 expression by TNF-α and Ang II in HepG2 cells. Meanwhile, GRK2 siRNA-transfected HepG2 cells were used to confirm the effects of GRK2, TNF-α and Ang II on the proliferation, migration and invasion of GRK2-knockdown HCC cells. Finally, the expression of TNF-α, Ang II, TNFR1, AT1R, AT2R and GRK2 proteins in HCC, tumor-adjacent and normal liver tissues were tested by immunohistochemistry. RESULTS: The data demonstrated that TNF-α and Ang II can enhance the proliferation, migration and invasion of HepG2 cells through suppressing GRK2 expression but that the two reagents combined did not have synergistic effects. Moreover,overexpression of TNFR1 and AT1R perhaps promoted the formation and progression of HCC, while high AT2R expression had the opposite effect. CONCLUSIONS: This study provides new ideas for the prevention and treatment of HCC by researching the interaction and probable mechanism of different bioactive factors associated with HCC.


Asunto(s)
Angiotensina II/farmacología , Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasa 2 del Receptor Acoplado a Proteína-G/biosíntesis , Factor de Necrosis Tumoral alfa/farmacología , Angiotensina II/uso terapéutico , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Citometría de Flujo , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica/patología , ARN Interferente Pequeño/genética , Receptor de Angiotensina Tipo 1/biosíntesis , Receptor de Angiotensina Tipo 2/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Factor de Necrosis Tumoral alfa/uso terapéutico
10.
Perit Dial Int ; 37(3): 250-258, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28096440

RESUMEN

♦ BACKGROUND: Peritoneal dialysis (PD) coincides with high concentrations of proinflammatory cytokines, such as tumor necrosis factor (TNF), in the peritoneal cavity. During treatment, chronic inflammatory processes lead to damage of the peritoneal membrane and a subsequent ultrafiltration failure. Human peritoneal mesothelial cells (HPMCs) play a central role as mediators and targets of PD-related inflammatory changes. Although TNF Receptor 1 (TNFR1) is expressed in high numbers on the cells, TNF-induced apoptosis is inhibited. Here, the underlying molecular mechanisms of TNFR1 signaling in HPMCs are investigated. ♦ METHODS: Human peritoneal mesothelial cells were isolated from the omentum of healthy donors and the dialysis solution of PD patients. Flow cytometry was applied to determine cell surface expression of TNFR1 on HPMCS from healthy donors in absence or presence of TNF or PD fluid (PDF) and were compared to TNFR1 expression on cells from PD patients. To investigate TNFR1-mediated signaling, HPMCs were treated with PDF or TNF, and expression patterns of proteins involved in the TNFR1 signaling pathway were assessed by western blot. ♦ RESULTS: Incubation with PDF led to a significant up-regulation of TNFR1 on the cell surface correlating with elevated TNFR1 numbers on HPMCs from PD patients. Investigations of underlying molecular mechanisms of TNFR1 signaling showed that PDF affects TNFR1 signaling at the proapoptotic signaling pathway by upregulation of IκBα and downregulation of cFLIPL. In contrast, TNF exclusively induces the activation of NFκB by an increase of phosphorylated IκBα. ♦ CONCLUSIONS: Novel and relevant insights into the mechanisms of TNFR1-mediated signaling in HPMCs with an impact on our understanding of PD-associated damage of the peritoneal membrane are shown.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Inflamación/genética , Epiplón/metabolismo , Diálisis Peritoneal/efectos adversos , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Apoptosis , Western Blotting , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/biosíntesis , Supervivencia Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/patología , Citometría de Flujo , Humanos , Inflamación/metabolismo , Inflamación/patología , Microscopía Fluorescente , Epiplón/patología , ARN Mensajero/genética , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Transducción de Señal
11.
Oncotarget ; 8(11): 17726-17737, 2017 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-28099148

RESUMEN

Suberoylanilide hydroxamic acid (SAHA) as a histone deacetylase (HDAC) inhibitor has anti-cancer effect. Here, we evaluated the effect of SAHA on HDAC activity and cell growth in many normal lung and cancer cells. We observed that the HDAC activities of lung cancer cells were higher than that of normal lung cells. SAHA inhibited the growth of lung cancer cells regardless of the inhibitory effect on HDAC. This agent induced a G2/M phase arrest and apoptosis, which was accompanied by mitochondrial membrane potential (MMP: ΔΨm) loss in lung cancer cells. However, SAHA did not induce cell death in normal lung cells. All tested caspase inhibitors prevented apoptotic cell death in SAHA-treated A549 and Calu-6 lung cancer cells. Treatment with tumor necrosis factor-alpha (TNF-α) enhanced apoptosis in SAHA-treated lung cancer cells through caspase-8 and caspase-9 activations. Especially, SAHA increased the expression level of TNF-α receptor 1 (TNFR1), especially acetylation of the region of TNFR1 promoter -223/-29 in lung cancer cells. The down-regulation of TNFR1 suppressed apoptosis in TNF-α and SAHA-treated lung cancer cells. In conclusion, SAHA inhibited the growth of lung cancer cells via a G2/M phase arrest and caspase-dependent apoptosis. SAHA also enhanced apoptotic effect of TNF-α in human lung cancer cells through up-regulation of TNFR1. TNF-α may be a key to improve anti-cancer effect of HDAC inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Neoplasias Pulmonares/patología , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo , Células A549 , Acetilación , Apoptosis/efectos de los fármacos , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Histona Desacetilasas/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba , Vorinostat
12.
J Biol Chem ; 291(38): 20247-59, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27502281

RESUMEN

Recent studies have shown that myocardial ischemia/reperfusion (I/R)-induced necrosis can be controlled by multiple genes. In this study, we observed that both strands (5p and 3p) of miR-223 were remarkably dysregulated in mouse hearts upon I/R. Precursor miR-223 (pre-miR-223) transgenic mouse hearts exhibited better recovery of contractile performance over reperfusion period and lesser degree of myocardial necrosis than wild type hearts upon ex vivo and in vivo myocardial ischemia. Conversely, pre-miR-223 knock-out (KO) mouse hearts displayed opposite effects. Furthermore, we found that the RIP1/RIP3/MLKL necroptotic pathway and inflammatory response were suppressed in transgenic hearts, whereas they were activated in pre-miR-223 KO hearts upon I/R compared with wild type controls. Accordingly, treatment of pre-miR-223 KO mice with necrostatin-1s, a potent necroptosis inhibitor, significantly decreased I/R-triggered cardiac necroptosis, infarction size, and dysfunction. Mechanistically, we identified two critical cell death receptors, TNFR1 and DR6, as direct targets of miR-223-5p, whereas miR-223-3p directly suppressed the expression of NLRP3 and IκB kinase α, two important mediators known to be involved in I/R-induced inflammation and cell necroptosis. Our findings indicate that miR-223-5p/-3p duplex works together and cooperatively inhibits I/R-induced cardiac necroptosis at multiple layers. Thus, pre-miR-223 may constitute a new therapeutic agent for the treatment of ischemic heart disease.


Asunto(s)
MicroARNs/biosíntesis , Daño por Reperfusión Miocárdica/metabolismo , Animales , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Imidazoles/farmacología , Indoles/farmacología , Ratones , Ratones Noqueados , MicroARNs/genética , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Necrosis , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética
13.
BMC Cancer ; 16: 279, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27098354

RESUMEN

BACKGROUND: Acquired resistance towards apoptosis is a hallmark of cancer. Elimination of cells bearing activated oncogenes or stimulation of tumor suppressor mediators may provide a selection pressure to overcome resistance. KC-53 is a novel biyouyanagin analogue known to elicit strong anti-inflammatory and anti-viral activity. The current study was designed to evaluate the anticancer efficacy and molecular mechanisms of KC-53 against human cancer cells. METHODS: Using the MTT assay we examined initially how KC-53 affects the proliferation rates of thirteen representative human cancer cell lines in comparison to normal peripheral blood mononuclear cells (PBMCs) and immortalized cell lines. To decipher the key molecular events underlying its mode of action we selected the human promyelocytic leukemia HL-60 and the acute lymphocytic leukemia CCRF/CEM cell lines that were found to be the most sensitive to the antiproliferative effects of KC-53. RESULTS: KC-53 promoted rapidly and irreversibly apoptosis in both leukemia cell lines at relatively low concentrations. Apoptosis was characterized by an increase in membrane-associated TNFR1, activation of Caspase-8 and proteolytic inactivation of the death domain kinase RIP1 indicating that KC-53 induced mainly the extrinsic/death receptor apoptotic pathway. Regardless, induction of the intrinsic/mitochondrial pathway was also achieved by Caspase-8 processing of Bid, activation of Caspase-9 and increased translocation of AIF to the nucleus. FADD protein knockdown restored HL-60 and CCRF/CEM cell viability and completely blocked KC-53-induced apoptosis. Furthermore, KC-53 administration dramatically inhibited TNFα-induced serine phosphorylation on TRAF2 and on IκBα hindering therefore p65/NF-κΒ translocation to nucleus. Reduced transcriptional expression of pro-inflammatory and pro-survival p65 target genes, confirmed that the agent functionally inhibited the transcriptional activity of p65. CONCLUSIONS: Our findings demonstrate, for the first time, the selective anticancer properties of KC-53 towards leukemic cell lines and provide a detailed understanding of the molecular events underlying its dual anti-proliferative and pro-apoptotic properties. These results provide new insights into the development of innovative and targeted therapies for the treatment of some forms of leukemia.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Leucemia/tratamiento farmacológico , Proteínas de Neoplasias/biosíntesis , Sesquiterpenos/administración & dosificación , Compuestos de Espiro/administración & dosificación , Apoptosis/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HL-60 , Humanos , Leucemia/genética , Leucemia/patología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , FN-kappa B/biosíntesis , FN-kappa B/genética , Proteínas de Neoplasias/genética , Fosforilación , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Sesquiterpenos/química , Transducción de Señal/efectos de los fármacos , Compuestos de Espiro/química
14.
Stem Cells Transl Med ; 5(3): 301-13, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26819253

RESUMEN

T helper 17 (Th17) cells play an important role in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Th17 cell differentiation from naïve T cells can be induced in vitro by the cytokines transforming growth factor ß1 and interleukin-6. However, it remains unclear whether other regulatory factors control the differentiation of Th17 cells. Mesenchymal stem cells (MSCs) have emerged as a promising candidate for inhibiting Th17 cell differentiation and autoimmune diseases. Despite the fact that several molecules have been linked to the immunomodulatory function of MSCs, many other key MSC-secreted regulators that are involved in inhibiting Th17 cell polarization are ill-defined. In this study, we demonstrated that the intraperitoneal administration of skin-derived MSCs (S-MSCs) substantially ameliorated the development of EAE in mice. We found that the proinflammatory cytokine tumor necrosis factor (TNF)-α, a key mediator in the pathophysiology of MS and EAE, was capable of promoting Th17 cell differentiation. Moreover, under inflammatory conditions, we demonstrated that S-MSCs produced high amounts of soluble TNF receptor 1 (sTNFR1), which binds TNF-α and antagonizes its function. Knockdown of sTNFR1 in S-MSCs decreased their inhibitory effect on Th17 cell differentiation ex vivo and in vivo. Thus, our data identified sTNFR1 and its target TNF-α as critical regulators for Th17 cell differentiation, suggesting a previously unrecognized mechanism for MSC therapy in Th17-mediated autoimmune diseases.


Asunto(s)
Diferenciación Celular/genética , Encefalomielitis Autoinmune Experimental/genética , Esclerosis Múltiple/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-6/biosíntesis , Interleucina-6/genética , Activación de Linfocitos , Células Madre Mesenquimatosas/metabolismo , Ratones , Esclerosis Múltiple/patología , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Células Th17/inmunología , Células Th17/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Factor de Necrosis Tumoral alfa/biosíntesis
15.
J Hepatol ; 64(5): 1108-1117, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26708145

RESUMEN

BACKGROUND & AIMS: The liver exhibits a unique capacity for regeneration in response to injury. Lymphotoxin-ß receptor (LTßR), a core member of the tumor necrosis factor (TNF)/tumor necrosis factor receptor (TNFR) superfamily is known to play an important role in this process. However, the function of LTßR during pathophysiological alterations and its molecular mechanisms during liver regeneration are so far ill-characterized. METHODS: LTßR(-/-) mice were subjected to 70% hepatectomy and liver regeneration capacity, bile acid profiles, and transcriptome analysis were performed. RESULTS: LTßR(-/-) deficient mice suffered from increased and prolonged liver tissue damage after 70% hepatectomy, accompanied by deregulated bile acid homeostasis. Pronounced differences in the expression patterns of genes relevant for bile acid synthesis and recirculation were observed. LTßR and TNFRp55 share downstream signalling elements. Therefore, LTßR(-/-) mice were treated with etanercept to create mice functionally deficient in both signalling pathways. Strikingly, the combined blockade of TNFRp55 and LTßR signalling leads to complete failure of liver regeneration resulting in death within 24 to 48h after PHx. Transcriptome analysis revealed a marked disparity in gene expression programs in livers of LTßR(-/-) and etanercept-treated LTßR(-/-) vs. wild-type animals after PHx. Murinoglobulin 2 was identified as a significantly differentially regulated gene. CONCLUSIONS: LTßR is essential for efficient liver regeneration and cooperates with TNFRp55 in this process. Differences in survival kinetics strongly suggest distinct functions for these two cytokine receptors in liver regeneration. Failure of TNFR and LTßR signalling renders liver regeneration impossible.


Asunto(s)
ADN/genética , Regulación de la Expresión Génica , Hepatopatías/genética , Regeneración Hepática/genética , Receptor beta de Linfotoxina/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Señuelo del Factor de Necrosis Tumoral/genética , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Hepatopatías/metabolismo , Hepatopatías/patología , Receptor beta de Linfotoxina/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Señuelo del Factor de Necrosis Tumoral/biosíntesis
16.
Eur J Pharmacol ; 771: 48-55, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26688568

RESUMEN

Ginsenoside metabolite compound K (CK), metabolite of the ginsenoside, is considered to exert numerous pharmacological efficacies of ginsenoside, including anti-inflammation and immunoregulatory effects. Rheumatoid arthritis (RA) is a multi-systemic autoimmune disease characterized by hyperplastic synovial membrane and systemic inflammation, which ultimately lead to progressive destructive inflammatory arthropathy. To evaluate the potential joint-protective effects of CK and the underlying mechanism, adjuvant arthritis (AA) was induced by complete Freund's adjuvant in rats. After the onset of arthritis, The effect of CK on AA rats was evaluated by histopathology of the joint. The proliferation of fibroblast-like synoviocyte(FLS) was assayed by the Cell Counting Kit-8.The migration of FLS was assayed by transwell migration assay. Cytokines in the supernatant from FLS were measured by ELISA kit. Expression of Tumor Necrosis Factor Receptor Type 1(TNFR1) and Tumor Necrosis Factor Receptor Type 2(TNFR2) were detected by immunostaining analysis and western blot analysis. CK (80mg/kg) significantly ameliorated the histopathological change of joint in AA rats, balanced the RANKL/OPG ratio and attenuated the proliferation and migration of AA-FLS. CK suppressed the secretion of proinflammatory cytokines TNF-α and downregulated the expression of TNFR2 on AA-FLS. In vitro CK also significantly suppressed proliferation, migration and secretion of AA-FLS mediated by TNF-α. Further studies showed that the effects of CK on AA-FLS were reversed by using glucocorticoid receptor (GR) antagonist (mifepristone). Our data suggest that CK exerts joint-protective effect by interfering with synoviocyte function mediated by TNF-α and TNFR2, and this effect may be mediated by GR.


Asunto(s)
Antiinflamatorios/farmacología , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Sapogeninas/farmacología , Membrana Sinovial/citología , Membrana Sinovial/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Masculino , Mifepristona/farmacología , Ligando RANK/biosíntesis , Ligando RANK/genética , Ratas , Ratas Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Membrana Sinovial/patología
17.
Cell Metab ; 22(5): 799-810, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26456334

RESUMEN

Primary hyperparathyroidism (PHPT) is a common cause of bone loss that is modeled by continuous PTH (cPTH) infusion. Here we show that the inflammatory cytokine IL-17A is upregulated by PHPT in humans and cPTH in mice. In humans, IL-17A is normalized by parathyroidectomy. In mice, treatment with anti-IL-17A antibody and silencing of IL-17A receptor IL-17RA prevent cPTH-induced osteocytic and osteoblastic RANKL production and bone loss. Mechanistically, cPTH stimulates conventional T cell production of TNFα (TNF), which increases the differentiation of IL-17A-producing Th17 cells via TNF receptor 1 (TNFR1) signaling in CD4(+) cells. Moreover, cPTH enhances the sensitivity of naive CD4(+) cells to TNF via GαS/cAMP/Ca(2+) signaling. Accordingly, conditional deletion of GαS in CD4(+) cells and treatment with the calcium channel blocker diltiazem prevents Th17 cell expansion and blocks cPTH-induced bone loss. Neutralization of IL-17A and calcium channel blockers may thus represent novel therapeutic strategies for hyperparathyroidism.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Hiperparatiroidismo Primario/metabolismo , Interleucina-17/metabolismo , Animales , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Enfermedades Óseas Metabólicas/etiología , Enfermedades Óseas Metabólicas/patología , Bloqueadores de los Canales de Calcio/uso terapéutico , Humanos , Hiperparatiroidismo Primario/complicaciones , Hiperparatiroidismo Primario/tratamiento farmacológico , Hiperparatiroidismo Primario/patología , Interleucina-17/biosíntesis , Ratones , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/patología , Factor de Necrosis Tumoral alfa/biosíntesis
18.
Tumour Biol ; 36(12): 9421-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26113409

RESUMEN

Although the expression of tumor necrosis factor receptors (TNFRs) has been associated with clinicopathologic features of some other cancers, their roles in hypopharyngeal squamous cell carcinoma (HPSCC) have not been documented. Forty-five HPSCC specimens were analyzed for the expression of TNFR1 and TNFR2 and its relationship with clinicopathologic factors. Interaction between the two receptors and its effects on TNF-α was investigated by neutralizing TNFR1 and upregulation of TNFR2. The results indicated that, in HPSCC specimens, the expression of TNFR1 but not TNFR2 is associated with clinical staging, T stage, cervical lymph node metastasis, and histologic grade in HPSCC. In Fadu cells, when conjugating with its receptors, TNF-α mediates proliferation effects, and neutralizing TNFR1 and/or upregulating TNFR2 evokes proliferation-inhibiting and apoptosis-inducing effects and potentiates cisplatin (DDP)-induced growth inhibition and apoptosis induction. In conclusion, interaction of TNFR1 with TNFR2 determines the biological characters of HPSCC, and TNFR1 may dominate this process. Moreover, interaction between the two receptors plays important roles in determining the fates of HPSCC cells and thus may serve as a therapeutic target for developing new therapeutic strategies for HPSCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias Hipofaríngeas/genética , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Adulto , Anciano , Apoptosis/genética , Carcinoma de Células Escamosas/patología , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hipofaríngeas/patología , Masculino , Persona de Mediana Edad , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética
19.
EMBO Mol Med ; 7(8): 989-91, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26071594

RESUMEN

Tumor necrosis factor (TNF) is a fascinating anti­tumoral cytokine, which plays a key role in orchestrating the fight of innate immunity against infection. Concomitantly, TNF is a major player of the inflammatory response, as illustrated by successful therapeutic strategies targeting TNF in patho logies such as Crohn's diseases, rheumatoid arthritis or psoriasis. In mice, TNF is able to induce tissue injuries and lethal shock. In this issue of EMBO Molecular Medicine, Puimège et al (2015) elegantly demonstrated that the lethal shock induced by TNF reflects high levels of its receptor TNFRI as seen in sensitive (Mus musculus), vs. resistant (Mus spretus) mice, where TNFR1 expression is low. They reported that this expression is under the control of a microRNA (miR­511), which itself is induced by glucocorticoids.


Asunto(s)
Glucocorticoides/metabolismo , MicroARNs/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo , Animales
20.
Tumour Biol ; 36(11): 8913-24, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26076811

RESUMEN

Cancer cells are characterized by increased production of reactive oxygen species (ROS) and an altered redox environment as compared to normal cells. Continuous accumulation of ROS triggers oxidative stress leading to hyper-activation of signaling pathways that promote cell proliferation, survival, and metabolic adaptation to the tumor microenvironment. Therefore, antioxidants are proposed to contribute to cancer prevention. Protein kinase C (PKC) is a crucial regulator of diverse cellular processes and contributes to cancer progression. The activation of PKC is partially dependent on ROS signaling. In the present study, cancer preventive activity of natural flavonoid quercetin is analyzed in ascite cells of Dalton's lymphoma-bearing mice. The total ROS level and activity of PKC were downregulated after quercetin treatment in lymphoma-bearing mice. Quercetin modulates the expression of almost all isozymes of classical, novel, and atypical PKC as well as downregulates the level and expression of PKCα. Further, quercetin improves apoptotic potential, as observed by the levels of caspase 3, caspase 9, PARP, PKCδ, and nuclear condensation. Additionally, quercetin reduces cell survival and promotes death receptor-mediated apoptosis via differential localization of the TNFR1 level in ascite cells. The overall result suggests the cancer preventive activity of quercetin via the induction of apoptosis and modulates PKC signaling with the reduction of oxidative stress in ascite cells of lymphoma-bearing mice.


Asunto(s)
Linfoma/tratamiento farmacológico , Proteína Quinasa C-alfa/biosíntesis , Quercetina/administración & dosificación , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Isoenzimas/biosíntesis , Linfoma/genética , Linfoma/patología , Ratones , Estrés Oxidativo/efectos de los fármacos , Proteína Quinasa C-alfa/genética , Especies Reactivas de Oxígeno/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...