Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
1.
Nature ; 618(7967): 1072-1077, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37196676

RESUMEN

Plasma membrane rupture (PMR) in dying cells undergoing pyroptosis or apoptosis requires the cell-surface protein NINJ11. PMR releases pro-inflammatory cytoplasmic molecules, collectively called damage-associated molecular patterns (DAMPs), that activate immune cells. Therefore, inhibiting NINJ1 and PMR may limit the inflammation that is associated with excessive cell death. Here we describe an anti-NINJ1 monoclonal antibody that specifically targets mouse NINJ1 and blocks oligomerization of NINJ1, preventing PMR. Electron microscopy studies showed that this antibody prevents NINJ1 from forming oligomeric filaments. In mice, inhibition of NINJ1 or Ninj1 deficiency ameliorated hepatocellular PMR induced with TNF plus D-galactosamine, concanavalin A, Jo2 anti-Fas agonist antibody or ischaemia-reperfusion injury. Accordingly, serum levels of lactate dehydrogenase, the liver enzymes alanine aminotransaminase and aspartate aminotransferase, and the DAMPs interleukin 18 and HMGB1 were reduced. Moreover, in the liver ischaemia-reperfusion injury model, there was an attendant reduction in neutrophil infiltration. These data indicate that NINJ1 mediates PMR and inflammation in diseases driven by aberrant hepatocellular death.


Asunto(s)
Anticuerpos Monoclonales , Membrana Celular , Inflamación , Hígado , Factores de Crecimiento Nervioso , Daño por Reperfusión , Animales , Ratones , Alanina Transaminasa , Alarminas , Anticuerpos Monoclonales/inmunología , Aspartato Aminotransferasas , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/inmunología , Moléculas de Adhesión Celular Neuronal/ultraestructura , Muerte Celular , Membrana Celular/patología , Membrana Celular/ultraestructura , Concanavalina A , Galactosamina , Hepatocitos/patología , Hepatocitos/ultraestructura , Inflamación/patología , Lactato Deshidrogenasas , Hígado/patología , Microscopía Electrónica , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/inmunología , Factores de Crecimiento Nervioso/ultraestructura , Infiltración Neutrófila , Daño por Reperfusión/patología
2.
Science ; 374(6571): 1076-1080, 2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34822265

RESUMEN

Inflammatory processes that recruit leukocytes to injured or infected tissues are crucial for tissue repair and the elimination of pathogens. However, excessive or chronic inflammation promotes tissue damage and disease, as in arthritis, atherosclerosis, inflammatory bowel disease, and COVID-19. Intracellular constituents released from dying cells are among the stimuli that trigger proinflammatory gene expression programs in innate immune cells. We explore how programmed cell death mechanisms­apoptosis, necroptosis, and pyroptosis­may contribute to inflammatory disease. We discuss inhibition of cell death as a potential therapeutic strategy, focusing on the targets RIPK1 (receptor interacting serine/threonine kinase 1), NLRP3 (NLR family pyrin domain containing 3), and GSDMD (gasdermin D) as important mediators of lytic cell death. We also consider the potential benefits of limiting membrane rupture rather than cell death by targeting NINJ1.


Asunto(s)
Apoptosis , Inflamación/fisiopatología , Necroptosis , Piroptosis , Animales , Caspasa 8/metabolismo , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Humanos , Inflamasomas/metabolismo , Inflamación/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
3.
Bioorg Chem ; 116: 105323, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34482170

RESUMEN

Diabetic retinopathy (DR) remains high incidence and accounts for severe impact on vision in diabetics, but its mechanism is still poorly understood. Abnormal migration and proliferation of endothelial cells (ECs) drive neovascular retinopathies, which has an important role in promoting the occurrence and development of DR. In this study, we designed and synthesized a series of PEDF-derived peptides as angiogenesis inhibitors. Especially, compound G24 significantly inhibited the cell proliferation in VEGF-activated human umbilical vein endothelial cells (HUVECs) with IC50 values of 2.88 ± 0.19 µM. Further biological evaluation demonstrated that compound G24 exhibited strong inducing-effects on cell apoptosis and internalization of 67LR, and advanced inhibitory potency in cell migration and angiogenesis formed by HUVECs in vitro. In summary, the optimal compound G24 as a novel angiogenesis inhibitor showed the potentiality in the further research for the treatment for DR.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Proteínas del Ojo/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Péptidos/farmacología , Receptores de Laminina/antagonistas & inhibidores , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Proteínas del Ojo/metabolismo , Humanos , Estructura Molecular , Factores de Crecimiento Nervioso/metabolismo , Péptidos/síntesis química , Péptidos/química , Receptores de Laminina/metabolismo , Serpinas/metabolismo , Relación Estructura-Actividad
4.
Biochem Biophys Res Commun ; 524(4): 869-875, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32051089

RESUMEN

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a neuroprotective factor produced in response to endoplasmic reticulum (ER) stress induced by various stressors, but its involvement in the radioresistance of tumor cells is unknown. Here, we found that MANF is released after γ-irradiation (2 Gy and 4 Gy) of B16 melanoma cells, and its release was suppressed by 4-phenylbutyric acid, an ER stress inhibitor. MANF was not released after low-dose (1 Gy) γ-irradiation, but pretreatment of 1 Gy-irradiated cells with recombinant MANF enhanced the cellular DNA damage response and attenuated reproductive cell death. In MANF-knockdown cells, the DNA damage response and p53 activation by γ-irradiation (2 Gy) were suppressed, and reproductive cell death was increased. MANF also activated the ERK signaling pathway. Our findings raise the possibility that MANF could be a new target for overcoming radioresistance.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de la radiación , Retículo Endoplásmico/efectos de la radiación , Regulación Neoplásica de la Expresión Génica , Factores de Crecimiento Nervioso/genética , Tolerancia a Radiación/genética , Animales , Línea Celular Tumoral , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Rayos gamma , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/radioterapia , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/metabolismo , Fenilbutiratos/farmacología , Fosforilación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
5.
J Clin Pathol ; 72(7): 460-467, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31072837

RESUMEN

The neurotrophic tyrosine receptor kinase (NTRK) gene family encodes three tropomyosin receptor kinases (TRKA, TRKB, TRKC) that contribute to central and peripheral nervous system development and function. NTRK gene fusions are oncogenic drivers of various adult and paediatric tumours. Several methods have been used to detect NTRK gene fusions including immunohistochemistry, fluorescence in situ hybridisation, reverse transcriptase polymerase chain reaction, and DNA- or RNA-based next-generation sequencing. For patients with TRK fusion cancer, TRK inhibition is an important therapeutic target. Following the FDA approval of the selective TRK inhibitor, larotrectinib, as well as the ongoing development of multi-kinase inhibitors with activity in TRK fusion cancer, testing for NTRK gene fusions should become part of the standard diagnostic process. In this review we discuss the biology of NTRK gene fusions, and we present a testing algorithm to aid detection of these gene fusions in clinical practice and guide treatment decisions.


Asunto(s)
Neoplasias/genética , Factores de Crecimiento Nervioso/genética , Proteínas de Fusión Oncogénica/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/genética , Algoritmos , Humanos , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptor trkA/antagonistas & inhibidores , Receptor trkA/genética , Receptor trkB/antagonistas & inhibidores , Receptor trkB/genética , Receptor trkC/antagonistas & inhibidores , Receptor trkC/genética
6.
J Clin Invest ; 129(6): 2222-2236, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30869655

RESUMEN

Neurofascin-155 (Nfasc155) is an essential glial cell adhesion molecule expressed in paranodal septate-like junctions of peripheral and central myelinated axons. The genetic deletion of Nfasc155 results in the loss of septate-like junctions and in conduction slowing. In humans, IgG4 antibodies against Nfasc155 are implicated in the pathogenesis of chronic inflammatory demyelinating polyneuropathy (CIDP). These antibodies are associated with an aggressive onset, a refractoriness to intravenous immunoglobulin, and tremor of possible cerebellar origin. Here, we examined the pathogenic effects of patient-derived anti-Nfasc155 IgG4. These antibodies did not inhibit the ability of Nfasc155 to complex with its axonal partners contactin-1/CASPR1 or induce target internalization. Passive transfer experiments revealed that IgG4 antibodies target Nfasc155 on Schwann cell surface, and diminished Nfasc155 protein levels and prevented paranodal complex formation in neonatal animals. In adult animals, chronic intrathecal infusions of antibodies also induced the loss of Nfasc155 and of paranodal specialization and resulted in conduction alterations in motor nerves. These results indicate that anti-Nfasc155 IgG4 perturb conduction in absence of demyelination, validating the existence of paranodopathy. These results also shed light on the mechanisms regulating protein insertion at paranodes.


Asunto(s)
Axones/inmunología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Inmunoglobulina G/farmacología , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Polineuropatías , Polirradiculoneuropatía , Animales , Axones/patología , Moléculas de Adhesión Celular/inmunología , Enfermedad Crónica , Femenino , Células HEK293 , Humanos , Inmunoglobulina G/inmunología , Masculino , Neuronas Motoras/inmunología , Neuronas Motoras/patología , Factores de Crecimiento Nervioso/inmunología , Polineuropatías/tratamiento farmacológico , Polineuropatías/inmunología , Polineuropatías/patología , Polirradiculoneuropatía/tratamiento farmacológico , Polirradiculoneuropatía/inmunología , Polirradiculoneuropatía/patología , Ratas , Ratas Endogámicas Lew , Células de Schwann/inmunología , Células de Schwann/patología
7.
Vet Rec ; 184(1): 23, 2019 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-30368458

RESUMEN

Nerve growth factor (NGF) is essential for the survival of sensory and sympathetic neurons during development. However, in the adult, NGF and its interaction with tropomyosin receptor kinase A receptor (TrkA) has been found to play a critical role in nociception and nervous system plasticity in pain conditions. Thus, various monoclonal antibody (mAb) therapies targeting this pathway have been investigated in the development of new pharmacotherapies for chronic pain. Although none of the mAbs against NGF are yet approved for use in humans, they look very promising for the effective control of pain. Recently, species-specific anti-NGF mAbs for the management of osteoarthritis (OA)-associated pain in dogs and cats has been developed, and early clinical trials have been conducted. Anti-NGF therapy looks to be both very effective and very promising as a novel therapy against chronic pain in dogs and cats. This review outlines the mechanism of action of NGF, the role of NGF in osteoarthritis, research in rodent OA models and the current status of the development of anti-NGF mAbs in humans. Furthermore, we describe and discuss the recent development of species-specific anti-NGF mAbs for the treatment of OA-associated pain in veterinary medicine.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedades de los Gatos/terapia , Enfermedades de los Perros/terapia , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Osteoartritis/veterinaria , Manejo del Dolor/veterinaria , Animales , Gatos , Perros , Osteoartritis/complicaciones , Osteoartritis/terapia , Dolor/etiología , Dolor/veterinaria , Ensayos Clínicos Controlados Aleatorios como Asunto
8.
Biochem Biophys Res Commun ; 505(4): 1077-1083, 2018 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-30314698

RESUMEN

Perineural invasion (PNI) potentially increases the risk of relapse and abdominal pain in patients with pancreatic ductal adenocarcinoma (PDAC). However, the underlying mechanisms of PNI of PDAC is incompletely revealed. Our study aimed to investigate roles of miR-429 in modulating PNI in PDAC. We found that miR-429 was downregulated in PDAC cancer tissues and was profoundly decreased in tissues with PNI. It was reduced in nine of the ten examined pancreatic cancer cell lines. MiR-429 mimics restored its cellular expressions in MIA PaCa-2 and BxCP3 cells and significantly suppressed cell viability and invasion of the cancer cells. The online bioinformatic software predicted that neurotrophin-3 (NT-3) was a potential target gene of miR-429. It was showed that NT-3 mRNA elevated in PC cancer tissues, especially in patients presenting PNI. MiR-429 upregulation substantially suppressed the NT-3 mRNA and secretion in cancer cells. Also, the dual luciferase reporter assays confirmed the interaction between miR-429 and NT-3. When co-culturing the two PDAC cells with PC-12 cells, the invaded cell counts significantly increased comparing with the sole culture of cancer cells. However, miR-429 mimic transfection or NT-3 blocking retarded the cancer invasion in the co-culture system. Besides, we found that cancer cells conditioned medium (CM) treatment significantly increased the neurite outgrowth percentage in PC-12 cells, which was suppressed by culturing with CM from miR-429 mimics-transfected cells. In the CM cultured PC-12 cells, NT-3 receptor TrkC as well as pain-related proteins TRPV1 and TRPV2 significantly elevated. Collectively, miR-429 potentially suppressed neurotrophin-3 to alleviate PNI of PDAC.


Asunto(s)
MicroARNs/farmacología , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Humanos , MicroARNs/genética , Factores de Crecimiento Nervioso/metabolismo , Neurotrofina 3 , Células PC12 , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Ratas , Células Tumorales Cultivadas
9.
Chem Pharm Bull (Tokyo) ; 66(1): 78-83, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29311515

RESUMEN

Two new secoiridoid glycosides, obtusifolisides A and B (1, 2), together with 7 known secoiridoid glycosides (3-9) were isolated from the twigs of Ligustrum obtusifolium. The chemical structures of new compounds were determined by a spectroscopic data analysis, including one and two dimensional (1D-, 2D)-NMR, High resolution-MS, and experiments involving chemical reactions. The isolated secoiridoid glycosides were evaluated for their anti-inflammatory effects in lipopolysaccharide (LPS)-stimulated BV-2 murine microglia cells. Compounds 2, 5, 6, 8, and 9 significantly reduced the production of nitric oxide (NO), with IC50 values of 5.45, 11.17, 14.62, 15.45, and 14.96 µM, respectively. None of the compounds were toxic to the cells. Additionally, we evaluated the neuroprotective effects of compounds 1-9 on nerve growth factor (NGF) induction in a C6 rat glioma cell line. Compounds 2 and 6 upregulated NGF secretion to 155.56±7.16%, and 139.35±11.65%, respectively, without significant cell toxicity.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Glicósidos Iridoides/farmacología , Ligustrum/química , Fármacos Neuroprotectores/farmacología , Tallos de la Planta/química , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/aislamiento & purificación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glioma/tratamiento farmacológico , Glioma/metabolismo , Glicósidos Iridoides/química , Glicósidos Iridoides/aislamiento & purificación , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Conformación Molecular , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/metabolismo , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/aislamiento & purificación , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Ratas , Relación Estructura-Actividad
10.
Clin Sci (Lond) ; 132(2): 213-229, 2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29263137

RESUMEN

Ongoing efforts to remove pathological inflammatory stimuli are crucial for the protection of endothelial cells in diabetes. Nerve injury-induced protein 1 (Ninj1) is an adhesion molecule that not only contributes to inflammation but also regulates the apoptosis of endothelial cells. In the present study, Ninj1 was found highly expressed in endothelial cells in Type 2 diabetic mice and increased in high-glucose (HG) cultured HUVECs. Furthermore, we found that Ninj1 levels are up-regulated in endothelial cells in clinical specimens of diabetic patients when compared with nondiabetic tissues, indicating a biological correlation between Ninj1 and endothelial pathophysiology in diabetic condition. Functional blocking of Ninj1 promoted endothelial tube formation and eNOS phosphorylation in the HG condition. Additionally, blocking Ninj1 inhibited the activation of caspase-3 and increased the Bcl-2/Bax ratio, thus inhibiting HUVECs apoptosis induced by HG. HG-induced ROS overproduction, p38 MAPK and NF-κB activation, and the overexpression of VCAM-1, ICAM-1, MCP-1, and IL-6 genes were ameliorated after Ninj1 was blocked. Using the signaling pathway inhibitor LY294002, we found that Bcl-2 expression and eNOS phosphorylation after Ninj1 blockade were regulated via PI3K/Akt signaling pathway. The in vivo endothelial contents, α-SMA+PECAM-1+ vascular numbers, and blood perfusion in the hindlimb were markedly up-regulated after Ninj1 was blocked. According to our findings, functional blocking of Ninj1 shows protective effects on diabetic endothelial cells both in vitro and in vivo Thus, we consider Ninj1 to be a potential therapeutic target for preventing endothelial dysfunction in diabetes mellitus.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliales/efectos de los fármacos , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Animales , Moléculas de Adhesión Celular Neuronal/inmunología , Moléculas de Adhesión Celular Neuronal/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Masculino , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Factores de Crecimiento Nervioso/inmunología , Factores de Crecimiento Nervioso/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos
11.
Cereb Cortex ; 28(1): 395-410, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29136108

RESUMEN

GABAergic synapses in the basolateral amygdala (BLA) play an important role in fear memory generation. We have previously reported that reduction in GABAergic synapses innervating specifically at the axon initial segment (AIS) of principal neurons of BLA, by neurofascin (NF) knockdown, impairs fear extinction. BLA is bidirectionally connected with the medial prefrontal cortex (mPFC), which is a key region involved in extinction of acquired fear memory. Here, we showed that reducing AIS GABAergic synapses within the BLA leads to impairment of synaptic plasticity in the BLA-mPFC pathway, as well as in the ventral subiculum (vSub)-mPFC pathway, which is independent of BLA involvement. The results suggest that the alteration within the BLA subsequently resulted in a form of trans-regional metaplasticity in the mPFC. In support of that notion, we observed that NF knockdown induced a severe deficit in behavioral flexibility as measured by reversal learning. Interestingly, reversal learning similar to extinction learning is an mPFC-dependent behavior. In agreement with that, measurement of the immediate-early gene, c-Fos immunoreactivity after reversal learning was reduced in the mPFC and BLA, supporting further the notion that the BLA GABAergic manipulation resulted in trans-regional metaplastic alterations within the mPFC.


Asunto(s)
Segmento Inicial del Axón/fisiología , Complejo Nuclear Basolateral/fisiología , Plasticidad Neuronal/fisiología , Corteza Prefrontal/fisiología , Sinapsis/fisiología , Vicia faba/metabolismo , Animales , Ansiedad/patología , Ansiedad/fisiopatología , Segmento Inicial del Axón/efectos de los fármacos , Segmento Inicial del Axón/patología , Complejo Nuclear Basolateral/citología , Complejo Nuclear Basolateral/efectos de los fármacos , Complejo Nuclear Basolateral/patología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Condicionamiento Psicológico/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Hipocampo/citología , Hipocampo/patología , Hipocampo/fisiología , Masculino , Memoria/fisiología , Actividad Motora/fisiología , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Vías Nerviosas/citología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/patología , Vías Nerviosas/fisiología , Plasticidad Neuronal/efectos de los fármacos , Corteza Prefrontal/citología , Corteza Prefrontal/patología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Aprendizaje Inverso/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/patología
12.
Cell Physiol Biochem ; 44(4): 1411-1424, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29186716

RESUMEN

Background/ Aims: This study was performed to reveal signaling pathways exploited by pigment epithelium-derived factor (PEDF) derived from retinal (glial) Müller cells to protect retinal ganglion cells (RGCs) from cell death. METHODS: The survival of RGCs was determined in the presence of conditioned culture media (MCM) from or in co-cultures with Müller cells. The significance of PEDF-induced STAT3 activation was evaluated in viability assays and using Western blotting analyses and siRNA-transfected cells. RESULTS: Secreted mediators of Müller cells increased survival of RGCs under normoxia or hypoxia to a similar degree as of PEDF- or IL-6-exposed cells. PEDF and MCM induced an increased STAT3 activation in RGCs and R28 cells, and neutralization of PEDF in MCM attenuated STAT3 activation. Inhibition of STAT3 reduced PEDF-promoted survival of RGCs. Similar to IL-6, PEDF induced STAT3 activation, acting in a dose-dependent manner via the PEDF receptor (PEDF-R) encoded by the PNPLA2 gene. Ablation of PEDF-R attenuated MCM-induced STAT3 activation and compromised the viability of PEDF-exposed R28 cells. CONCLUSIONS: Müller cells are an important source of PEDF, which promotes RGC survival through STAT3 activation and, at least in part, via PEDF-R. Enhancing the secretory function of Müller cells may be useful to promote RGC survival in retinal neurodegenerative diseases.


Asunto(s)
Células Ependimogliales/metabolismo , Proteínas del Ojo/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Factor de Transcripción STAT3/metabolismo , Serpinas/metabolismo , Animales , Hipoxia de la Célula , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Óxidos S-Cíclicos/farmacología , Células Ependimogliales/citología , Proteínas del Ojo/antagonistas & inhibidores , Proteínas del Ojo/genética , Proteínas del Ojo/farmacología , Interleucina-6/farmacología , Lipasa/antagonistas & inhibidores , Lipasa/genética , Lipasa/metabolismo , Ratones , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/farmacología , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Receptores de Neuropéptido/metabolismo , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Serpinas/genética , Serpinas/farmacología , Transducción de Señal/efectos de los fármacos
13.
Am J Med Sci ; 354(3): 299-309, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28918838

RESUMEN

BACKGROUND: Midkine (MDK) is a tumor-promoting factor that is often overexpressed in various human carcinomas, and the role of MDK has not yet been fully investigated in prostate cancer stem cells. MATERIALS AND METHODS: Prostate cancer CD133+ stem cells (PCSCs) were isolated from human castration-resistant PC3 cells. PCSCs were treated with different concentrations of MDK inhibitor, iMDK, for 24-72 hours. The IC50 values were determined by the MTT test. Endogenous MDK messenger RNA expression was knocked down by small interfering RNA. Quantitative reverse transcription polymerase chain reaction, Western blot analyses and image-based cytometry were used to investigate apoptosis and cell cycle progression as well as their underlying molecular mechanisms. Cell migration was evaluated by the wound healing test. RESULTS: iMDK caused dose- and time-dependent inhibition of PCSC survival. Similar growth inhibition was also obtained by small interfering RNA-mediated knockdown of endogenous MDK expression. iMDK was shown to preferentially induce cell cycle arrest at the S and G2/M phases. Suppressed PCSC growth was also accompanied by increases in p53 and the cell cycle inhibitor p21 genes. Combinatorial treatment of iMDK with docetaxel significantly inhibited cell proliferation versus either of the agents used alone. Inhibition of MDK expression strongly suppressed the migration of PCSCs compared to untreated and docetaxel-treated cells. iMDK and the knockdown of MDK decreased p-Akt and significantly upregulated the expression of PI3K/phosphatase/tensin homolog. CONCLUSIONS: Our data indicate that MDK plays a crucial role in controlling PCSC proliferation and migration. Therefore, suppression of endogenous expression of MDK would, in combination with traditional chemotherapy drugs, be a potential treatment for PCSCs.


Asunto(s)
Antígeno AC133/inmunología , Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cumarinas/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Neoplasias de la Próstata/patología , Tiazoles/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Midkina , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Factores de Crecimiento Nervioso/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , ARN Interferente Pequeño/genética , Factores de Tiempo
14.
Biofactors ; 43(6): 760-769, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28817221

RESUMEN

The incidence of neurological disorders is growing in developed countries together with increased lifespan. Nowadays, there are still no effective treatments for neurodegenerative pathologies, which make necessary to search for new therapeutic agents. Natural products, most of them used in traditional medicine, are considered promising alternatives for the treatment of neurodegenerative diseases. Honokiol is a natural bioactive phenylpropanoid compound, belonging to the class of neolignan, found in notable amounts in the bark of Magnolia tree, and has been reported to exert diverse pharmacological properties including neuroprotective activities. Honokiol can permeate the blood brain barrier and the blood-cerebrospinal fluid to increase its bioavailability in neurological tissues. Diverse studies have provided evidence on the neuroprotective effect of honokiol in the central nervous system, due to its potent antioxidant activity, and amelioration of the excitotoxicity mainly related to the blockade of glutamate receptors and reduction in neuroinflammation. In addition, recent studies suggest that honokiol can attenuate neurotoxicity exerted by abnormally aggregated Aß in Alzheimer's disease. The present work summarizes what is currently known concerning the neuroprotective effects of honokiol and its potential molecular mechanisms of action, which make it considered as a promising agent in the treatment and management of neurodegenerative diseases. © 2017 BioFactors, 43(6):760-769, 2017.


Asunto(s)
Antioxidantes/farmacología , Compuestos de Bifenilo/farmacología , Sistema Nervioso Central/efectos de los fármacos , Lignanos/farmacología , Magnolia/química , Enfermedades Neurodegenerativas/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/inmunología , Antioxidantes/aislamiento & purificación , Antioxidantes/farmacocinética , Productos Biológicos/química , Transporte Biológico , Compuestos de Bifenilo/aislamiento & purificación , Compuestos de Bifenilo/farmacocinética , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Citocinas/antagonistas & inhibidores , Citocinas/genética , Citocinas/inmunología , Regulación de la Expresión Génica , Humanos , Lignanos/aislamiento & purificación , Lignanos/farmacocinética , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/inmunología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/aislamiento & purificación , Fármacos Neuroprotectores/farmacocinética , Corteza de la Planta/química , Receptores de Glutamato/genética , Receptores de Glutamato/inmunología
15.
J Biol Chem ; 292(36): 14977-14988, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28698383

RESUMEN

Cytokine-induced endoplasmic reticulum (ER) stress is one of the molecular mechanisms underlying pancreatic ß-cell demise in type 1 diabetes. Thrombospondin 1 (THBS1) was recently shown to promote ß-cell survival during lipotoxic stress. Here we show that ER-localized THBS1 is cytoprotective to rat, mouse, and human ß-cells exposed to cytokines or thapsigargin-induced ER stress. THBS1 confers cytoprotection by maintaining expression of mesencephalic astrocyte-derived neutrotrophic factor (MANF) in ß-cells and thereby prevents the BH3-only protein BIM (BCL2-interacting mediator of cell death)-dependent triggering of the mitochondrial pathway of apoptosis. Prolonged exposure of ß-cells to cytokines or thapsigargin leads to THBS1 and MANF degradation and loss of this prosurvival mechanism. Approaches that sustain intracellular THBS1 and MANF expression in ß-cells should be explored as a cytoprotective strategy in type 1 diabetes.


Asunto(s)
Inflamación/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Trombospondina 1/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Estrés Oxidativo , Tapsigargina/farmacología
16.
Drugs ; 77(13): 1377-1387, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28660479

RESUMEN

Although there is an unmet need for pain medications that are both effective and safe, virtually no novel analgesics have been approved over the past two decades. In view of both experimental and clinical evidence of a major role for nerve growth factor (NGF) in the generation and maintenance of a wide range of pain states, the clinical development of humanised anti-nerve growth factor monoclonal antibodies (anti-NGF mAbs) aroused particular interest. However, the US Food and Drug Administration (FDA) placed a clinical hold on anti-NGF mAb clinical studies in late 2010, first because of reports of serious joint-related adverse events, and afterwards because of sympathetic nervous system safety concerns. The development programmes of tanezumab and fasinumab resumed after the FDA lifted its hold in March 2015, whereas other anti-NGF mAbs were dropped by their sponsors. This article provides an updated review on the analgesic efficacy and safety of anti-NGF agents based on data from fully published studies and public information from websites, and discusses the possible future role of these agents in managing chronic pain. The efficacy of anti-NGF mAbs was highly variable depending on the chronic pain condition studied. The most consistent and convincing results were obtained in patients with symptomatic osteoarthritis of the knee and/or hip. Conversely, studies in non-specific lower back pain and peripheral neuropathic pain generated mixed results. Finally, there was no conclusive evidence of the effectiveness of anti-NGF mAbs in cancer pain and urological chronic pelvic pain syndromes. Treatment-emergent adverse events were similar across anti-NGF mAbs, thus being suggestive of 'class-specific effects'. Although most patients tolerated anti-NGF agents well, neurosensory symptoms occurred frequently, and some patients developed new or worsened peripheral neuropathies. However, the most problematic safety issue was rapidly destructive arthropathies, leading to joint replacement surgery. To date, the aetiologies of joint-related side effects and their pathophysiology have not been clearly elucidated. However, some risk factors have been identified, such as higher doses of anti-NGF mAbs and longer drug exposure, concurrent nonsteroidal anti-inflammatory drug use and pre-existing subchondral insufficiency fractures. Taken together, the present data suggest that low-dose anti-NGF mABs may exhibit a favourable risk-benefit ratio in selected patients with certain chronic pain conditions, especially symptomatic osteoarthritis.


Asunto(s)
Analgésicos/farmacología , Anticuerpos Monoclonales/farmacología , Dolor Crónico/tratamiento farmacológico , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Analgésicos/efectos adversos , Analgésicos/química , Analgésicos/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Humanos , Osteoartritis/tratamiento farmacológico
17.
Anticancer Res ; 37(4): 1687-1696, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28373430

RESUMEN

BACKGROUND/AIM: Ninjurin1 is a 17-kDa membrane protein that is highly expressed in circulating tumor cells (CTCs) obtained from locally-advanced prostate cancer patients. As CTCs are implicated in the initiation of distant metastasis, we examined the potential contribution of Ninjurin1 to the motility of prostate cancer cells. MATERIALS AND METHODS: Ninjurin1 expression was evaluated in CTCs harvested from seven locally advanced patients with no metastatic hallmarks using real-time polymerase chain reaction (PCR). The role of Ninjurin1 in cell motility was investigated using small interfering RNA (siRNA), neutralizing antibodies against Ninjurin1 and Ninjurin1-overexpressing adenoviruses. RESULTS: Ninjurin1 was ranked as the most significantly up-regulated adhesion protein identified by RNA-Seq analysis. Both Ninjurin1 down-regulation by siRNA and neutralizing antibodies blocking Ninjurin1 homophilic interactions effectively inhibited cell motility. In contrast, cell motility was enhanced in prostate cancer cells infected with adenovirus enabling Ninjurin1 overexpression. CONCLUSION: Ninjurin1-neutralizing antibodies or Ninjurin1-targeting siRNA merit further development for patients with metastatic potential.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Células Neoplásicas Circulantes/patología , Factores de Crecimiento Nervioso/metabolismo , Neoplasias de la Próstata/patología , Apoptosis , Western Blotting , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/genética , Proliferación Celular , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Células Neoplásicas Circulantes/metabolismo , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Regulación hacia Arriba
18.
Vitam Horm ; 104: 459-473, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28215304

RESUMEN

Neurotrophins (NTs) have been implicated in generation and modulation of nociceptive pathways. Change in NTs levels is associated with painful conditions and neurological diseases such as migraine. Currently, it is generally recognized that migraine headaches result from the activation and sensitization of trigeminal sensory afferent fibers leading to neuropeptides release such as calcitonin gene-related peptide (CGRP) and substance P (SP). This triggers an inflammatory cascade causing a neurogenic inflammation. The agents responsible for trigeminal activation and release of neuropeptides are still unclear. It is known that the transient receptor potential vanilloid receptor-1 (TRPV1) is an important mediator of CGRP and SP release. TRPV1 is closely associated with tyrosine receptors kinases (Trk), which are NTs receptors. NTs can act on TRPV1 increasing its sensitivity to painful stimuli, therefore predisposing to hyperalgesia. Upregulation of ion channels and pain receptors in dorsal root ganglion neurons may be alternative mechanisms by which NTs contribute to pain development. Only a few studies have been performed to investigate the role of NTs in migraine. These studies have reported changes in NTs levels in migraine patients either during the migraine attack or in free-headache periods.


Asunto(s)
Encéfalo/metabolismo , Trastornos Migrañosos/metabolismo , Modelos Neurológicos , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/agonistas , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Drogas en Investigación/farmacología , Drogas en Investigación/uso terapéutico , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/inmunología , Ganglios Espinales/metabolismo , Humanos , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/inmunología , Trastornos Migrañosos/fisiopatología , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Inflamación Neurogénica/etiología , Inflamación Neurogénica/prevención & control , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/inmunología , Neuronas Aferentes/metabolismo , Dolor Nociceptivo/etiología , Dolor Nociceptivo/prevención & control , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/metabolismo
19.
Vitam Horm ; 104: 497-531, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28215306

RESUMEN

In this chapter, we parse the literature on neurotrophins that have been implicated in the pathogenesis of perineural invasion (PNI) in select lineage-unrelated malignancies. We also detail evidence linking neurotrophins and their receptors (TrkA, RET, p75NGFR, and NCAM) to the pathogenesis of PNI in desmoplastic melanoma and cutaneous squamous cell carcinoma-both malignancies with an established propensity for PNI. Lastly, the clinical potential of neurotrophins as receptors for targeted therapies is explored.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Melanoma/metabolismo , Modelos Neurológicos , Factores de Crecimiento Nervioso/metabolismo , Nervios Periféricos/metabolismo , Receptores de Factor de Crecimiento Nervioso/agonistas , Neoplasias Cutáneas/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Drogas en Investigación/farmacología , Drogas en Investigación/uso terapéutico , Humanos , Melanoma/diagnóstico , Melanoma/tratamiento farmacológico , Melanoma/patología , Terapia Molecular Dirigida , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Nervios Periféricos/efectos de los fármacos , Nervios Periféricos/patología , Pronóstico , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/metabolismo , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Células de Schwann/patología , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología
20.
Pharm Dev Technol ; 22(3): 426-435, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27575893

RESUMEN

Multiple sclerosis (MS) is a chronic central nervous system (CNS) inflammation. Efficient drug delivery to brain is however hampered by blood-brain barrier (BBB). In order to have highly efficient and safe delivery of drugs to brain, solid lipid nanoparticles (SLNs) have indicated promising potentials as smart carriers that can pass the blood-brain barrier and deliver therapeutic biomolecules to the brain. In this study, PEGylated SLNs surface modified using anti-Contactin-2 or anti-Neurofascin, two axo-glial-glycoprotein antigens located in node of Ranvier, were prepared. These targeting moieties are considered as the main targets of autoimmune reaction in MS. The targeted SLNs were then characterized and their in vitro release profile together with their cell viability and uptake were studied. Their brain uptakes were also probed following injections in MS-induced mice. It was found that the targeted PEGylated SLNs had no significant cytotoxicity on U87MG cells although their cellular uptake was increased 4- and 8-fold when surface modified with anti-Contactin-2 or anti-Neurofascin, respectively, compared to control. Brain uptake results demonstrated higher uptake of surface-modified SLNs in the brain tissue compared with the PEGylated SLNs. The results of this report will help scientist to design more efficient nanocarriers for treatment of MS.


Asunto(s)
Antiinflamatorios/administración & dosificación , Encéfalo/metabolismo , Moléculas de Adhesión Celular/antagonistas & inhibidores , Contactina 2/antagonistas & inhibidores , Portadores de Fármacos/química , Metilprednisolona/administración & dosificación , Nanopartículas/química , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Animales , Antiinflamatorios/farmacocinética , Antiinflamatorios/uso terapéutico , Anticuerpos Monoclonales/química , Encéfalo/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Humanos , Metilprednisolona/farmacocinética , Metilprednisolona/uso terapéutico , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Electrónica de Rastreo , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...