Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
1.
Gene ; 838: 146729, 2022 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-35835402

RESUMEN

Two therapeutic agents targeting p75NTR pathways have been recently developed to alleviate retinopathy and bladder dysfunction in diabetes mellitus (DM), namely the small molecule p75NTR antagonist THX-B and a monoclonal antibody (mAb) that neutralizes the receptor ligand proNGF. We herein explore these two components in the context of diabetic kidney disease (DKD). Streptozotocin-injected mice were treated for 4 weeks with THX-B or anti-proNGF mAb. Kidneys were taken for quantification of microRNAs and mRNAs by RT-qPCR and for detection of proteins by immunohistochemistry, immunoblotting and ELISA. Blood was sampled to measure plasma levels of urea, creatinine, and albumin. DM led to increases in plasma concentrations of urea and creatinine and decreases in plasma albumin. Receptor p75NTR was expressed in kidneys and its expression was decreased by DM. All these changes were reversed by THX-B treatment while the effect of mAb was less pronounced. MicroRNAs tightly linked to DKD (miR-21-5p, miR-214-3p and miR-342-3p) were highly expressed in diabetic kidneys compared to healthy ones. Also, miR-146a, a marker of kidney inflammation, and mRNA levels of Fn-1 and Nphs, two markers of fibrosis and inflammation, were elevated in DM. Treatments with THX-B or mAb partially or completely reduced the expression of the aforementioned microRNAs and mRNAs. P75NTR antagonism and proNGF mAb might constitute new therapeutic tools to treat or slow down the progression of kidney disease in DM, along with other diabetic related complications. The translational potential of these strategies is currently being investigated.


Asunto(s)
Complicaciones de la Diabetes , Diabetes Mellitus , Nefropatías Diabéticas , MicroARNs , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Animales , Biomarcadores , Creatinina , Nefropatías Diabéticas/tratamiento farmacológico , Inflamación , Ratones , MicroARNs/genética , Factor de Crecimiento Nervioso/metabolismo , Estreptozocina
2.
Front Immunol ; 12: 606861, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33643311

RESUMEN

Platelets and neurons share many similarities including comparable secretory granule types with homologous calcium-dependent secretory mechanisms as well as internalization, sequestration and secretion of many neurotransmitters. Thus, platelets present a high potential to be used as peripheral biomarkers to reflect neuronal pathologies. The brain-derived neurotrophic factor (BDNF) acts as a neuronal growth factor involved in learning and memory through the binding of two receptors, the tropomyosin receptor kinase B (TrkB) and the 75 kDa pan-neurotrophic receptor (p75NTR). In addition to its expression in the central nervous system, BDNF is found in much greater quantities in blood circulation, where it is largely stored within platelets. Levels 100- to 1,000-fold those of neurons make platelets the most important peripheral reservoir of BDNF. This led us to hypothesize that platelets would express canonical BDNF receptors, i.e., TrkB and p75NTR, and that the receptors on platelets would bear significant resemblance to the ones found in the brain. However, herein we report discrepancies regarding detection of these receptors using antibody-based assays, with antibodies displaying important tissue-specificity. The currently available antibodies raised against TrkB and p75NTR should therefore be used with caution to study platelets as models for neurological disorders. Rigorous characterization of antibodies and bioassays appears critical to understand the interplay between platelet and neuronal biology of BDNF.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/inmunología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/inmunología , Receptor trkB/antagonistas & inhibidores , Receptor trkB/inmunología , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/inmunología , Especificidad de Anticuerpos/inmunología , Biomarcadores , Plaquetas/inmunología , Plaquetas/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Glicosilación , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Transporte de Proteínas , Receptor trkB/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo
3.
Eur J Pharmacol ; 895: 173882, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33482180

RESUMEN

Fabry disease (FD) is an X-linked metabolic storage disorder arising from the deficiency of lysosomal α-galactosidase A, which leads to the gradual accumulation of glycosphingolipids, mainly globotriaosylceramide (Gb3), throughout the body. Pain in the extremities is an early symptom of FD; however, the underlying pathophysiological mechanisms remain unknown. α-Galactosidase A knockout animals exhibit nociceptive behaviors, with enhanced expression levels of several ion channels. These characteristics are observed in animals treated with nerve growth factor (NGF). Here, we aimed to elucidate the potential of NGF signaling as a cause of FD-associated pain, using intraplantar Gb3-treated mice displaying mechanical allodynia. Treatment with a neutralizing antibody against a precursor of NGF (proNGF) or its receptor, p75 neurotrophin receptor (p75NTR), resulted in the recovery from Gb3-induced pain. Conversely, anti-NGF and anti-tropomyosin receptor kinase A antibodies failed to exert analgesic effects. Gb3 injection had no effects on the expression levels of proNGF and p75NTR in the plantar skin and dorsal root ganglia, suggesting that Gb3 activates the pain pathway, possibly mediated through functional up-regulation of proNGF-p75NTR signaling. Furthermore, by pharmacological approaches using a protein kinase A (PKA) inhibitor and a cholesterol-removing agent, we found that p75NTR-phosphorylating PKA and lipid rafts for phosphorylated p75NTR translocation were required for Gb3-induced pain. These results suggest that acute exposure to Gb3 induces mechanical allodynia via activation of the proNGF-p75NTR pathway, which involves lipid rafts and PKA. Our findings provide new pathological insights into FD-associated pain, and suggest the need to develop therapeutic interventions targeting proNGF-p75NTR signaling.


Asunto(s)
Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Umbral del Dolor , Precursores de Proteínas/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Piel/metabolismo , Trihexosilceramidas , Analgésicos/farmacología , Animales , Anticuerpos Neutralizantes/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Ganglios Espinales/fisiopatología , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Hiperalgesia/prevención & control , Masculino , Microdominios de Membrana/metabolismo , Ratones Endogámicos C57BL , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Umbral del Dolor/efectos de los fármacos , Precursores de Proteínas/antagonistas & inhibidores , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Transducción de Señal
4.
Female Pelvic Med Reconstr Surg ; 27(7): 457-461, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32740468

RESUMEN

OBJECTIVE: To investigate the expression levels of nerve growth factor (NGF) and its receptors TrkA and p75NTR in the anterior vaginal wall of postmenopausal patients with pelvic organ prolapse (POP). METHODS: The tissues of anterior vaginal wall of the patients (n = 31) with POP and patients (n = 16) with nonpelvic floor dysfunction were collected during the operation. The expressions of NGF, TrkA, and p75NTR were detected by real-time quantitative polymerase chain reaction, immunohistochemistry and Western blot. RESULTS: The expression levels of mRNA and protein of NGF and its receptors in vaginal anterior wall tissues of postmenopausal POP patients were significantly decrease compared with those of the control group. The ratio of p75NTR/TrkA expression in POP patients was significantly increase compared with that in the control group and was proportional to the degree of prolapse. CONCLUSIONS: The decreased expression of NGF and its receptors p75NTR and TrkA in vaginal anterior wall tissue of postmenopausal POP patients and the change of the ratio of 2 receptors may be related to the occurrence and development of POP.


Asunto(s)
Factor de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Prolapso de Órgano Pélvico/metabolismo , Receptor trkA/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Anciano , Femenino , Humanos , Persona de Mediana Edad , Posmenopausia , ARN Mensajero , Reacción en Cadena en Tiempo Real de la Polimerasa , Vagina/inmunología
5.
Behav Brain Res ; 396: 112905, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32926907

RESUMEN

The precursor of brain derived neurotrophic factor (proBDNF) and its receptor p75NTR are upregulated in depressive patients and chronic stress-induced depressive animals, suggesting that activation of p75NTR signalling may underlie pathogenesis of depression. In the present study we hypothesize that the blockade of p75NTR may have therapeutic effect on depressive mice under chronic stress. The treatment of mice with the recombinant fusion protein of p75NTR extracellular domain and fragment C of immunoglobulin (p75ECD-Fc) significantly reduced the immobility time in the forced swim test and tail suspension test, and increased the time spent in the central zone in the open field test in mice exposed to chronic unpredictable mild stress (CUMS). p75ECD-Fc treatment also significantly increased the length and density of neuronal dendritic spines in the dentate gyrus and amygdala. Our data indicate that blocking p75NTR signalling can alleviate depressive and anxiety-like behaviours of chronically stressed mice and improve the dendritic spinogenesis of neurons under stress.


Asunto(s)
Depresión/tratamiento farmacológico , Depresión/fisiopatología , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/administración & dosificación , Estrés Psicológico/fisiopatología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Espinas Dendríticas/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Depresión/etiología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Proyectos Piloto , Distribución Aleatoria , Método Simple Ciego , Estrés Psicológico/complicaciones
6.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33351787

RESUMEN

Asymmetric cell division (ACD) enables the maintenance of a stem cell population while simultaneously generating differentiated progeny. Cancer stem cells (CSCs) undergo multiple modes of cell division during tumor expansion and in response to therapy, yet the functional consequences of these division modes remain to be determined. Using a fluorescent reporter for cell surface receptor distribution during mitosis, we found that ACD generated a daughter cell with enhanced therapeutic resistance and increased coenrichment of EGFR and neurotrophin receptor (p75NTR) from a glioblastoma CSC. Stimulation of both receptors antagonized differentiation induction and promoted self-renewal capacity. p75NTR knockdown enhanced the therapeutic efficacy of EGFR inhibition, indicating that coinheritance of p75NTR and EGFR promotes resistance to EGFR inhibition through a redundant mechanism. These data demonstrate that ACD produces progeny with coenriched growth factor receptors, which contributes to the generation of a more therapeutically resistant CSC population.


Asunto(s)
División Celular Asimétrica , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Resistencia a Antineoplásicos , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Antígeno AC133/metabolismo , Neoplasias Encefálicas/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Autorrenovación de las Células , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Humanos , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo
7.
Med Oncol ; 37(11): 106, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-33135102

RESUMEN

Small bowel cancers are rare tumors with an incidence 50-100-fold less than colorectal cancer. These tumors carry a poor prognosis. Owing to its rarity, treatment of this disease, particularly in its advanced stages, has not been optimized and is derived mainly from treatment regimens for colorectal cancer. Based on recent studies bevacizumab, an antibody directed against vascular endothelial growth factor and used in the management of metastatic CRC, has been added to treatment guidelines for metastatic small bowel adenocarcinoma. We investigate in this review the evidence behind other targeted treatments that may be beneficial in the treatment of metastatic small bowel adenocarcinoma. These are agents against EGFR, VEGFR-2, HER2, and NTRK as well as immune checkpoint inhibitors. The last class of drugs appears to hold the greatest promise based on the preponderance of evidence supporting its use. However, overall data remains sparse. Results of studies currently underway will be valuable in shedding more light on the management of this aggressive cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Neoplasias Intestinales/tratamiento farmacológico , Bevacizumab/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Terapia Molecular Dirigida , Receptor ErbB-2/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
8.
Nat Commun ; 11(1): 3946, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32770055

RESUMEN

Melanomas can switch to a dedifferentiated cell state upon exposure to cytotoxic T cells. However, it is unclear whether such tumor cells pre-exist in patients and whether they can be resensitized to immunotherapy. Here, we chronically expose (patient-derived) melanoma cell lines to differentiation antigen-specific cytotoxic T cells and observe strong enrichment of a pre-existing NGFRhi population. These fractions are refractory also to T cells recognizing non-differentiation antigens, as well as to BRAF + MEK inhibitors. NGFRhi cells induce the neurotrophic factor BDNF, which contributes to T cell resistance, as does NGFR. In melanoma patients, a tumor-intrinsic NGFR signature predicts anti-PD-1 therapy resistance, and NGFRhi tumor fractions are associated with immune exclusion. Lastly, pharmacologic NGFR inhibition restores tumor sensitivity to T cell attack in vitro and in melanoma xenografts. These findings demonstrate the existence of a stable and pre-existing NGFRhi multitherapy-refractory melanoma subpopulation, which ought to be eliminated to revert intrinsic resistance to immunotherapeutic intervention.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Melanoma/tratamiento farmacológico , Proteínas del Tejido Nervioso/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Linfocitos T Citotóxicos/inmunología , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Masculino , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , RNA-Seq , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Linfocitos T Citotóxicos/metabolismo , Escape del Tumor/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Diabetologia ; 63(9): 1932-1946, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32699962

RESUMEN

AIMS/HYPOTHESIS: Although 80% of diabetic patients will suffer from voiding difficulties and urinary symptoms, defined as diabetic voiding dysfunction (DVD), therapeutic targets and treatment options are limited. We hypothesise that the blockade of the pro-nerve growth factor (NGF)/p75 neurotrophin receptor (p75NTR) axis by an anti-proNGF monoclonal antibody or by a small molecule p75NTR antagonist (THX-B) can restore bladder remodelling (represented by bladder weight) in an animal model of DVD. Secondary outcomes of the study include improvements in bladder compliance, contractility and morphology, as well as in voiding behaviour, proNGF/NGF balance and TNF-α expression. METHODS: In a streptozotocin-induced mouse model of diabetes, diabetic mice received either a blocking anti-proNGF monoclonal antibody or a p75NTR antagonist small molecule as weekly systemic injections for 4 weeks. Animals were tested at baseline (at 2 weeks of diabetes induction), and after 2 and 4 weeks of treatment. Outcomes measured were voiding function with voiding spot assays and cystometry. Bladders were assessed by histological, contractility and protein expression assays. RESULTS: Diabetic mice showed features of DVD as early as 2 weeks after diabetes diagnosis (baseline) presented by hypertrophy, reduced contractility and abnormal cystometric parameters. Following treatment initiation, a twofold increase (p < 0.05) in untreated diabetic mouse bladder weight and thickness compared with non-diabetic controls was observed, and this change was reversed by p75NTR antagonism (37% reduction in bladder weight compared with untreated diabetic mice [95% CI 14%, 60%]) after 4 weeks of treatment. However, blocking proNGF did not help to reverse bladder hypertrophy. While diabetic mice had significantly worse cystometric parameters and contractile responses than non-diabetic controls, proNGF antagonism normalised bladder compliance (0.007 [Q1-Q3; 0.006-0.009] vs 0.015 [Q1-Q3; 0.014-0.029] ml/cmH2O in untreated diabetic mice, representing 62% reduction [95% CI 8%, 110%], p < 0.05) and contractility to KCl, carbachol and electrical field stimulation (p < 0.05 compared with the diabetic group) after 2 weeks of treatment. These effects were not observed after 4 weeks of treatment with proNGF antagonist. p75NTR antagonism did not show important improvements in cystometric parameters after 2 weeks of treatment. Slightly improved bladder compliance (0.01 [Q1-Q3; 0.009-0.012] vs 0.013 [Q1-Q3; 0.011-0.016] ml/cmH2O for untreated diabetic mice) was seen in the p75NTR antagonist-treated group after 4 weeks of treatment with significantly stabilised contractile responses to KCl, carbachol and electric field stimulation (p < 0.05 for each) compared with diabetic mice. Bladder dysfunction observed in diabetic mice was associated with a significant increase in bladder proNGF/NGF ratio (3.1 [±1.2] vs 0.26 [±0.04] ng/pg in control group, p < 0.05 at week 2 of treatment) and TNF-α (p < 0.05). The proNGF/NGF ratio was partially reduced (about 60% reduction) with both treatments (1.03 [±0.6] ng/pg for proNGF antibody-treated group and 1.4 [±0.76] ng/pg for p75NTR blocker-treated group after 2 weeks of treatment), concomitant with a significant decrease in the bladder levels of TNF-α (p < 0.05), despite persistent hyperglycaemia. CONCLUSIONS/INTERPRETATION: Our findings indicate that blockade of proNGF and the p75NTR receptor in diabetes can impede the development and progression of DVD. The reported improvements in morphological and functional features in our DVD model validates the proNGF/p75NTR axis as a potential therapeutic target in this pathology. Graphical abstract.


Asunto(s)
Complicaciones de la Diabetes/fisiopatología , Diabetes Mellitus Experimental/fisiopatología , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Precursores de Proteínas/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Vejiga Urinaria/fisiopatología , Trastornos Urinarios/fisiopatología , Animales , Anticuerpos Monoclonales/farmacología , Adaptabilidad , Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Ratones , Contracción Muscular , Músculo Liso/fisiopatología , Tamaño de los Órganos , Purinas/farmacología , Receptor de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Trastornos Urinarios/metabolismo
10.
ASN Neuro ; 12: 1759091420930865, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32493127

RESUMEN

The p75 neurotrophin receptor (p75NTR) can regulate multiple cellular functions including proliferation, survival, and apoptotic cell death. The p75NTR is widely expressed in the developing brain and is downregulated as the nervous system matures, with only a few neuronal subpopulations retaining expression into adulthood. However, p75NTR expression is induced following damage to the adult brain, including after traumatic brain injury, which is a leading cause of mortality and disability worldwide. A major consequence of traumatic brain injury is the progressive neuronal loss that continues secondary to the initial trauma, which ultimately contributes to cognitive decline. Understanding mechanisms governing this progressive neuronal death is key to developing targeted therapeutic strategies to provide neuroprotection and salvage cognitive function. In this study, we demonstrate that a cortical impact injury to the sensorimotor cortex elicits p75NTR expression in apoptotic neurons in the injury penumbra, confirming previous studies. To establish whether preventing p75NTR induction or blocking the ligands would reduce the extent of secondary neuronal cell death, we used a noninvasive intranasal strategy to deliver either siRNA to block the induction of p75NTR, or function-blocking antibodies to the ligands pro-nerve growth factor and pro-brain-derived neurotrophic factor. We demonstrate that either preventing the induction of p75NTR or blocking the proneurotrophin ligands provides neuroprotection and preserves sensorimotor function.


Asunto(s)
Apoptosis/fisiología , Lesiones Traumáticas del Encéfalo/metabolismo , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/biosíntesis , Administración Intranasal/métodos , Animales , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/psicología , Muerte Celular/fisiología , Técnicas de Silenciamiento del Gen/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , ARN Interferente Pequeño/administración & dosificación , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores
11.
Nanotechnology ; 31(30): 305707, 2020 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-32235073

RESUMEN

Cancer stem cells (CSCs) are considered to maintain the vitality of tumor cell populations through self-renewal and infinite proliferation, but their accessibility is still under investigation. In addition, CSCs are more resistant to chemotherapy and radiotherapy compared with common tumor cells. This study aimed to develop a kind of novel and feasible nanomaterial for targeted photothermal ablation of osteosarcoma stem cells, which could be a promising anticancer strategy. The osteosarcoma stem cells were extracted by serum-free culture and we further verified the stem cell properties. We evaluated the expression of CD271 by flow cytometry. PEGylated multifunctional hollow gold nanospheres (HGNs) were prepared based on CD271 monoclonal antibody. Bifunctional SH-PEG-COOH was used to facilitate the covalent linkage between HGNs and antibody. The efficient uptake and distribution of the functionalized HGNs were investigated using ICP-MS and TEM. Morphological studies and quantitative apoptosis evaluation were performed to detect the effect of photothermal therapy (PTT). Afterwards, we explored the possible mechanism by which PTT induced targeted killing of cancer stem cells. Osteosarcoma cells isolated from serum-free culture were detected to show stem cell properties. CD271 was found to be a potential novel surface marker for osteosarcoma stem cells. By conjugating with CD271 monoclonal antibody, these biomimetic nanoparticles can be targeted and absorbed by osteosarcoma stem cells. HGNs-PEG-CD271 achieved excellent cell viability inhibition compared with non-targeted PEGylated HGNs upon near-infrared (NIR) laser irradiation. The mechanism of targeted killing may be related to the apoptosis pathway and DNA double-strand injuries. CD271 was considered to be a surface biomarker for osteosarcoma stem cells. Functionalized HGNs based on CD271 antibody exhibited excellent potential for targeted PTT, which may be a promising strategy for osteosarcoma treatment.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias Óseas/metabolismo , Oro/química , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Osteosarcoma/metabolismo , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Antineoplásicos Inmunológicos/química , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Neoplasias Óseas/genética , Neoplasias Óseas/terapia , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Humanos , Nanosferas , Células Madre Neoplásicas/efectos de los fármacos , Osteosarcoma/genética , Osteosarcoma/terapia , Terapia Fototérmica
12.
Theranostics ; 10(4): 1649-1677, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32042328

RESUMEN

Rationale: Autophagy in Schwann cells (SCs) is crucial for myelin debris degradation and clearance following peripheral nerve injury (PNI). Nerve growth factor (NGF) plays an important role in reconstructing peripheral nerve fibers and promoting axonal regeneration. However, it remains unclear if NGF effect in enhancing nerve regeneration is mediated through autophagic clearance of myelin debris in SCs. Methods: In vivo, free NGF solution plus with/without pharmacological inhibitors were administered to a rat sciatic nerve crush injury model. In vitro, the primary Schwann cells (SCs) and its cell line were cultured in normal medium containing NGF, their capable of swallowing or clearing degenerated myelin was evaluated through supplement of homogenized myelin fractions. Results: Administration of exogenous NGF could activate autophagy in dedifferentiated SCs, accelerate myelin debris clearance and phagocytosis, as well as promote axon and myelin regeneration at early stage of PNI. These NGF effects were effectively blocked by autophagy inhibitors. In addition, inhibition of the p75 kD neurotrophin receptor (p75NTR) signal or inactivation of the AMP-activated protein kinase (AMPK) also inhibited the NGF effect as well. Conclusions: NGF effect on promoting early nerve regeneration is closely associated with its accelerating autophagic clearance of myelin debris in SCs, which probably regulated by the p75NTR/AMPK/mTOR axis. Our studies thus provide strong support that NGF may serve as a powerful pharmacological therapy for peripheral nerve injuries.


Asunto(s)
Autofagia/efectos de los fármacos , Vaina de Mielina/metabolismo , Factor de Crecimiento Nervioso/farmacología , Regeneración Nerviosa/efectos de los fármacos , Células de Schwann/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/farmacología , Animales , Autofagia/fisiología , Línea Celular , Humanos , Masculino , Ratones , Ratones Noqueados , Vaina de Mielina/fisiología , Factor de Crecimiento Nervioso/administración & dosificación , Factor de Crecimiento Nervioso/metabolismo , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/farmacología , Traumatismos de los Nervios Periféricos/metabolismo , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Ratas , Ratas Wistar , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Células de Schwann/fisiología , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
13.
Life Sci ; 241: 117102, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31790691

RESUMEN

Peripheral nerve injuries are common conditions that often lead to dysfunctions. Although much knowledge exists on the several factors that mediate the complex biological process involved in peripheral nerve regeneration, there is a lack of effective treatments that ensure full functional recovery. Naringenin (NA) is the most abundant flavanone found in citrus fruits and it has promising neuroprotective, anti-inflammatory and antioxidant effects. This study aimed to enhance peripheral nerve regeneration using an inclusion complex containing NA and hydroxypropyl-ß-cyclodextrin (HPßCD), named NA/HPßCD. A mouse sciatic nerve crush model was used to evaluate the effects of NA/HPßCD on nerve regeneration. Sensory and motor parameters, hyperalgesic behavior and the sciatic functional index (SFI), respectively, improved with NA treatment. Western blot analysis revealed that the levels of p75NTR ICD and p75NTR full length as well phospho-JNK/total JNK ratios were preserved by NA treatment. In addition, NA treatment was able to decrease levels of caspase 3. The concentrations of TNF-α and IL-1ß were decreased in the lumbar spine, on the other hand there was an increase in IL-10. NA/HPßCD presented a better overall morphological profile but it was not able to increase the number of myelinated fibers. Thus, NA was able to enhance nerve regeneration, and NA/HPßCD decreased effective drug doses while maintaining the effect of the pure drug, demonstrating the advantage of using the complex over the pure compound.


Asunto(s)
2-Hidroxipropil-beta-Ciclodextrina/farmacología , Flavanonas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Regeneración Nerviosa/efectos de los fármacos , Nervio Ciático/fisiología , Animales , Hiperalgesia/tratamiento farmacológico , Interleucina-10/metabolismo , Masculino , Ratones , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Regeneración Nerviosa/fisiología , Dimensión del Dolor , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/metabolismo , Recuperación de la Función , Nervio Ciático/efectos de los fármacos , Nervio Ciático/lesiones , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
Lancet Oncol ; 21(2): 271-282, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31838007

RESUMEN

BACKGROUND: Entrectinib is a potent inhibitor of tropomyosin receptor kinase (TRK) A, B, and C, which has been shown to have anti-tumour activity against NTRK gene fusion-positive solid tumours, including CNS activity due to its ability to penetrate the blood-brain barrier. We present an integrated efficacy and safety analysis of patients with metastatic or locally advanced solid tumours harbouring oncogenic NTRK1, NTRK2, and NTRK3 gene fusions treated in three ongoing, early-phase trials. METHODS: An integrated database comprised the pivotal datasets of three, ongoing phase 1 or 2 clinical trials (ALKA-372-001, STARTRK-1, and STARTRK-2), which enrolled patients aged 18 years or older with metastatic or locally advanced NTRK fusion-positive solid tumours who received entrectinib orally at a dose of at least 600 mg once per day in a capsule. All patients had an Eastern Cooperative Oncology Group performance status of 0-2 and could have received previous anti-cancer therapy (except previous TRK inhibitors). The primary endpoints, the proportion of patients with an objective response and median duration of response, were evaluated by blinded independent central review in the efficacy-evaluable population (ie, patients with NTRK fusion-positive solid tumours who were TRK inhibitor-naive and had received at least one dose of entrectinib). Overall safety evaluable population included patients from STARTRK-1, STARTRK-2, ALKA-372-001, and STARTRK-NG (NCT02650401; treating young adult and paediatric patients [aged ≤21 years]), who received at least one dose of entrectinib, regardless of tumour type or gene rearrangement. NTRK fusion-positive safety evaluable population comprised all patients who have received at least one dose of entrectinib regardless of dose or follow-up. These ongoing studies are registered with ClinicalTrials.gov, NCT02097810 (STARTRK-1) and NCT02568267 (STARTRK-2), and EudraCT, 2012-000148-88 (ALKA-372-001). FINDINGS: Patients were enrolled in ALKA-372-001 from Oct 26, 2012, to March 27, 2018; in STARTRK-1 from Aug 7, 2014, to May 10, 2018; and in STARTRK-2 from Nov 19, 2015 (enrolment is ongoing). At the data cutoff date for this analysis (May 31, 2018) the efficacy-evaluable population comprised 54 adults with advanced or metastatic NTRK fusion-positive solid tumours comprising ten different tumour types and 19 different histologies. Median follow-up was 12.9 months (IQR 8·77-18·76). 31 (57%; 95% CI 43·2-70·8) of 54 patients had an objective response, of which four (7%) were complete responses and 27 (50%) partial reponses. Median duration of response was 10 months (95% CI 7·1 to not estimable). The most common grade 3 or 4 treatment-related adverse events in both safety populations were increased weight (seven [10%] of 68 patients in the NTRK fusion-positive safety population and in 18 [5%] of 355 patients in the overall safety-evaluable population) and anaemia (8 [12%] and 16 [5%]). The most common serious treatment-related adverse events were nervous system disorders (three [4%] of 68 patients and ten [3%] of 355 patients). No treatment-related deaths occurred. INTERPRETATION: Entrectinib induced durable and clinically meaningful responses in patients with NTRK fusion-positive solid tumours, and was well tolerated with a manageable safety profile. These results show that entrectinib is a safe and active treatment option for patients with NTRK fusion-positive solid tumours. These data highlight the need to routinely test for NTRK fusions to broaden the therapeutic options available for patients with NTRK fusion-positive solid tumours. FUNDING: Ignyta/F Hoffmann-La Roche.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Biomarcadores de Tumor/genética , Fusión Génica , Indazoles/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/genética , Anciano , Antineoplásicos/efectos adversos , Benzamidas/efectos adversos , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Femenino , Humanos , Indazoles/efectos adversos , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Persona de Mediana Edad , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/mortalidad , Neoplasias/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Receptor trkA/antagonistas & inhibidores , Receptor trkA/genética , Receptor trkB/antagonistas & inhibidores , Receptor trkB/genética , Receptor trkC/antagonistas & inhibidores , Receptor trkC/genética , Factores de Tiempo , Resultado del Tratamiento
15.
ESMO Open ; 4(Suppl 2): e000612, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31803506

RESUMEN

NTRK fusions are found at low frequencies (commonly <1%) in a range of common tumour types and at high frequencies (up to or greater than 90%) in rare cancer types (secretory breast carcinoma, mammary analogue secretory carcinoma and infantile fibrosarcoma). The fusions typically occur in a mutually exclusive fashion with other strong mitogenic drivers, and it is of significant importance to identify patients in order to offer transformative treatment with TRK inhibitors. Larotractinib or entrectinib have resulted in fast and durable response with few and reversible adverse events. Even though on-target resistance may occur, second generation TRK inhibitors are in development and have shown promising activity. Diagnostic strategies must be applied, considering available assays and specific tumour types.


Asunto(s)
Biomarcadores de Tumor/antagonistas & inhibidores , Fusión Génica , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Benzamidas/farmacología , Benzamidas/uso terapéutico , Biomarcadores de Tumor/genética , Pruebas Genéticas , Humanos , Indazoles/farmacología , Indazoles/uso terapéutico , Neoplasias/diagnóstico , Neoplasias/genética , Selección de Paciente , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirazoles/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Receptores de Factor de Crecimiento Nervioso/genética , Factores de Tiempo , Resultado del Tratamiento
16.
Neurochem Int ; 129: 104498, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31278975

RESUMEN

The present work aimed to explore the innovative hypothesis that different transcript/protein variants of a pro-neurotrophin may generate different biological outcomes in a cellular system. Nerve growth factor (NGF) is important in the development and progression of neurodegenerative and cancer conditions. Mature NGF (mNGF) originates from a precursor, proNGF, produced in mouse in two major variants, proNGF-A and proNGF-B. Different receptors bind mNGF and proNGF, generating neurotrophic or neurotoxic outcomes. It is known that dysregulation in the proNGF/mNGF ratio and in NGF-receptors expression affects brain homeostasis. To date, however, the specific roles of the two major proNGF variants remain unexplored. Here we attempted a first characterization of the possible differential effects of proNGF-A and proNGF-B on viability, differentiation and endogenous ngf gene expression in the PC12 cell line. We also investigated the differential involvement of NGF receptors in the actions of proNGF. We found that native mouse mNGF, proNGF-A and proNGF-B elicited different effects on PC12 cell survival and differentiation. Only mNGF and proNGF-A promoted neurotrophic responses when all NGF receptors are exposed at the cell surface. Tropomyosine receptor kinase A (TrkA) blockade inhibited cell differentiation, regardless of which NGF was added to culture media. Only proNGF-A exerted a pro-survival effect when TrkA was inhibited. Conversely, proNGF-B exerted differentiative effects when the p75 neurotrophin receptor (p75NTR) was antagonized. Stimulation with NGF variants differentially regulated the autocrine production of distinct proNgf mRNA. Overall, our findings suggest that mNGF and proNGF-A may elicit similar neurotrophic effects, not necessarily linked to activation of the same NGF-receptor, while the action of proNGF-B may be determined by the NGF-receptors balance. Thus, the proposed involvement of proNGF/NGF on the development and progression of neurodegenerative and tumor conditions may depend on the NGF-receptors balance, on specific NGF trancript expression and on the proNGF protein variant ratio.


Asunto(s)
Factor de Crecimiento Nervioso/farmacología , Células PC12/efectos de los fármacos , Precursores de Proteínas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso , Isoformas de Proteínas/farmacología , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/genética , Proteoma , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Receptor trkA/antagonistas & inhibidores , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Especificidad de la Especie
17.
ACS Chem Neurosci ; 10(1): 412-423, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30117729

RESUMEN

Early brain injury (EBI) after aneurysmal subarachnoid hemorrhage (SAH) contributes to high morbidity and mortality. Although it is well recognized that acute neuroinflammation reaction is one of the most important triggers of EBI, pharmacotherapy proved to be clinically effective against the initiating of neuroinflammation after SAH is lacking. The resident microglia and infiltrated peripheral monocyte are two main types of immune cells in central nervous system (CNS) and control the inflammation process in brain after SAH. But the time course and relative contributions of these two immune cell activations after SAH are unknown. The p75 neurotrophin receptor (p75NTR), member of TNF receptor superfamily, expresses on infiltrated peripheral monocytes and suppresses their proinflammatory action after brain insults. But the p75NTR expression on resident microglia in vivo is rarely explored and their function keeps elusive. Therefore, we designed this study to investigate the time course of resident microglia activation and peripheral monocyte infiltration, as well as the microglial expression of p75NTR by using CX3C-chemokine receptor 1 (Cx3cr1) and chemokine receptor 2 (Ccr2) double transgenic mice (Cx3cr1GFP/+Ccr2RFP/+) after SAH. The results showed activated microglia was observed in cortex as early as 24 h and further increased at 48 and 72 h post SAH, while the infiltrated monocyte was not found until 72h. In addition, activated microglia expressed p75NTR acutely and p75NTR specific antagonist TAT-Pep5 significantly reduced microglia activation, neuroinflammation and EBI from 24 to 72 h. Together, these data suggest that the early neuroinflammation reaction might be initiated and intensified mainly by resident microglia rather than infiltrated monocyte at least in the first 48 h after SAH and p75NTR blockading by TAT-Pep5P might alleviate EBI through mediating microglial activation.


Asunto(s)
Lesiones Encefálicas/metabolismo , Microglía/metabolismo , Monocitos/metabolismo , Fármacos Neuroprotectores/farmacología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Hemorragia Subaracnoidea/metabolismo , Animales , Lesiones Encefálicas/etiología , Lesiones Encefálicas/prevención & control , Femenino , Productos del Gen tat/farmacología , Productos del Gen tat/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Monocitos/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Distribución Aleatoria , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/tratamiento farmacológico
18.
Int J Mol Sci ; 19(11)2018 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-30453504

RESUMEN

The assembly and maintenance of cilia depend on intraflagellar transport (IFT) proteins, which play an important role in development and homeostasis. IFT80 is a newly defined IFT protein and partial mutation of IFT80 in humans causes diseases such as Jeune asphyxiating thoracic dystrophy (JATD) and short rib polydactyly (SRP) type III, both characterized by abnormal skeletal development. However, the role and mechanism of IFT80 in the invasion of gastric cancer is unknown. We established SGC-7901 and MKN-45 gastric cancer cell lines that stably overexpressed IFT80, as verified by quantitative reverse transcription-PCR, Western blot, and immunofluorescence. Matrix metalloproteinase-9 (MMP9) plays an important role in tumor invasion, and its expression was assessed by quantitative reverse transcription-PCR, Western blotting, and immunofluorescence. The invasion ability of IFT80 on SGC-7901 and MKN-45 cells was examined by the Matrigel invasion assay. The relationship between p75NGFR, and the p75NGFR antagonists, PD90780 and IFT80, were detected by quantitative reverse transcription-PCR and Western blotting. We first detected an IFT80 expression pattern, and found that IFT80 was highly expressed in gastric cancer clinical samples. Overexpression of IFT80 in the gastric cancer cell lines, SGC-7901 and MKN-45, led to lengthening cilia. Additionally, overexpression of IFT80 significantly improved proliferation and invasion, but inhibited apoptosis, in gastric cancer cells. We further found that overexpression of IFT80 increased p75NGFR and MMP9 mRNA and protein expression. Treatment with the p75NGFR antagonist PD90780 inhibited the increased invasion ability resulting from overexpression of IFT80 in SGC-7901 and MKN-45 gastric cancer cells. Thus, these results suggest that IFT80 plays an important role in invasion of gastric cancer through regulating the ift80/p75NGFR/MMP9 signal pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cilios/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 9 de la Matriz/genética , Invasividad Neoplásica , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Quinazolinas/farmacología , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/genética , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/genética
19.
Cell Chem Biol ; 25(12): 1485-1494.e5, 2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30293939

RESUMEN

Small molecules offer powerful ways to alter protein function. However, most proteins in the human proteome lack small-molecule probes, including the large class of non-catalytic transmembrane receptors, such as death receptors. We hypothesized that small molecules targeting the interfaces between transmembrane domains (TMDs) in receptor complexes may induce conformational changes that alter receptor function. Applying this concept in a screening assay, we identified a compound targeting the TMD of death receptor p75NTR that induced profound conformational changes and receptor activity. The compound triggered apoptotic cell death dependent on p75NTR and JNK activity in neurons and melanoma cells, and inhibited tumor growth in a melanoma mouse model. Due to their small size and crucial role in receptor activation, TMDs represent attractive targets for small-molecule manipulation of receptor function.


Asunto(s)
Antineoplásicos/farmacología , Melanoma/tratamiento farmacológico , Melanoma/patología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antineoplásicos/química , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Melanoma/metabolismo , Ratones , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
20.
Exp Cell Res ; 371(1): 122-129, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30092219

RESUMEN

Tumor hypoxia is associated with several features of aggressive glioma growth, including migration, invasion, and stemness. Most of the cellular adaptation to hypoxia is mediated by the hypoxia-inducible factors HIF-1α and HIF-2α, but regulation of these factors by both oxygen-dependent and -independent mechanisms in brain tumors is only partially understood. Here, we show that the p75 neurotrophin receptor (p75NTR) is stabilized at hypoxia in murine glioma in vivo, as well as in primary human glioma cultures in vitro. Expression of p75NTR resulted in increased stabilization of HIF-1α and HIF-2α, and RNAi or pharmacologic targeting of p75NTR diminished HIF stabilization and HIF-dependent signaling at hypoxia. Consequentially, p75NTR inhibition resulted in decreased migration, invasion, and stemness in response to hypoxia, suggesting that p75NTR is a central regulator of hypoxia-induced glioma aggressiveness. Together, our findings support the literature that identifies p75NTR as a potential therapeutic target in brain tumors.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Hipoxia/genética , Receptores de Factor de Crecimiento Nervioso/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Glioma/metabolismo , Glioma/patología , Humanos , Hipoxia/metabolismo , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Cultivo Primario de Células , Estabilidad Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...