Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
J Neurosci ; 41(42): 8710-8724, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34507952

RESUMEN

We report that the neurotrophin receptor p75 contributes to sensory neuron survival through the regulation of cholesterol metabolism in Schwann cells. Selective deletion of p75 in mouse Schwann cells of either sex resulted in a 30% loss of dorsal root ganglia (DRG) neurons and diminished thermal sensitivity. P75 regulates Schwann cell cholesterol biosynthesis in response to BDNF, forming a co-receptor complex with ErbB2 and activating ErbB2-mediated stimulation of sterol regulatory element binding protein 2 (SREBP2), a master regulator of cholesterol synthesis. Schwann cells lacking p75 exhibited decreased activation of SREBP2 and a reduction in 7-dehydrocholesterol (7-DHC) reductase (DHCR7) expression, resulting in accumulation of the neurotoxic intermediate, 7-dehyrocholesterol in the sciatic nerve. Restoration of DHCR7 in p75 null Schwann cells in mice significantly attenuated DRG neuron loss. Together, these results reveal a mechanism by which the disruption of lipid metabolism in glial cells negatively influences sensory neuron survival, which has implications for a wide range of peripheral neuropathies.SIGNIFICANCE STATEMENT Although expressed in Schwann cells, the role of p75 in myelination has remained unresolved in part because of its dual expression in sensory neurons that Schwann cells myelinate. When p75 was deleted selectively among Schwann cells, myelination was minimally affected, while sensory neuron survival was reduced by 30%. The phenotype is mainly due to dysregulation of cholesterol biosynthesis in p75-deficient Schwann cells, leading to an accumulation of neurotoxic cholesterol precursor, 7-dehydrocholesterol (7-DHC). Mechanism-wise, we discovered that in response to BDNF, p75 recruits and activates ErbB2 independently of ErbB3, thereby stimulating the master regulator, sterol regulatory element binding protein 2 (SREBP2). These results together highlight a novel role of p75 in Schwann cells in regulating DRG neuron survival by orchestrating proper cholesterol metabolism.


Asunto(s)
Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Células de Schwann/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Supervivencia Celular/fisiología , Células Cultivadas , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Células de Schwann/ultraestructura , Células Receptoras Sensoriales/ultraestructura
2.
Am J Physiol Heart Circ Physiol ; 314(3): H415-H423, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29101167

RESUMEN

Cardiac sympathetic nerves stimulate heart rate and force of contraction. Myocardial infarction (MI) leads to the loss of sympathetic nerves within the heart, and clinical studies have indicated that sympathetic denervation is a risk factor for arrhythmias and cardiac arrest. Two distinct types of denervation have been identified in the mouse heart after MI caused by ischemia-reperfusion: transient denervation of peri-infarct myocardium and sustained denervation of the infarct. Sustained denervation is linked to increased arrhythmia risk, but it is not known whether acute nerve loss in peri-infarct myocardium also contributes to arrhythmia risk. Peri-infarct sympathetic denervation requires the p75 neurotrophin receptor (p75NTR), but removal of p75NTR alters the pattern of sympathetic innervation in the heart and increases spontaneous arrhythmias. Therefore, we targeted the p75NTR coreceptor sortilin and the p75NTR-induced protease tumor necrosis factor-α-converting enzyme/A disintegrin and metalloproteinase domain 17 (TACE/ADAM17) to selectively block peri-infarct denervation. Sympathetic nerve density was quantified using immunohistochemistry for tyrosine hydroxylase. Genetic deletion of sortilin had no effect on the timing or extent of axon degeneration, but inhibition of TACE/ADAM17 with the protease inhibitor marimastat prevented the loss of axons from viable myocardium. We then asked whether retention of nerves in peri-infarct myocardium had an impact on cardiac electrophysiology 3 days after MI using ex vivo optical mapping of transmembrane potential and intracellular Ca2+. Preventing acute denervation of viable myocardium after MI did not significantly alter cardiac electrophysiology or Ca2+ handling, suggesting that transient denervation at this early time point has minimal impact on arrhythmia risk. NEW & NOTEWORTHY Sympathetic denervation after myocardial infarction is a risk factor for arrhythmias. We asked whether transient loss of nerves in viable myocardium contributed to arrhythmia risk. We found that targeting protease activity could prevent acute peri-infarct denervation but that it did not significantly alter cardiac electrophysiology or Ca2+ handling 3 days after myocardial infarction.


Asunto(s)
Arritmias Cardíacas/etiología , Corazón/inervación , Infarto del Miocardio/complicaciones , Miocardio/patología , Sistema Nervioso Simpático/fisiopatología , Proteína ADAM17/metabolismo , Potenciales de Acción , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Señalización del Calcio , Modelos Animales de Enfermedad , Frecuencia Cardíaca , Preparación de Corazón Aislado , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Sistema Nervioso Simpático/metabolismo , Factores de Tiempo , Supervivencia Tisular
3.
J Alzheimers Dis ; 59(3): 941-949, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28697556

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is pathologically known for the amyloid-ß (Aß) deposition, neurofibrillary tangles, and neuronal loss in the brain. The precursor of brain-derived neurotrophic factor (proBDNF) before proteolysis has opposing functions to its mature form in neuronal survival and neurite growth. However, the role of proBDNF in the pathogenesis of AD remains unclear. OBJECTIVE: To investigate the effects of proBDNF on neurons in vitro, and on learning and memory impairment and brain Aß production in a transgenic AD mouse model (APPswePS1dE9). METHODS: We here examined the effects of proBDNF on the viability (MTT assay) and neurite growth (morphologic measurement) of the primary neurons in vitro. After the intracerebroventricular injection of adeno-associated virus-proBDNF (AAV-proBDNF), we then investigated the learning and memory impairment (Morris water maze) and Aß deposition in the brains of the AD mice. RESULTS: The results showed that proBDNF could inhibit neuronal viability and neurite growth in vitro, enhance Aß levels, and accelerate its deposition in the brain, which was consistent with the learning and memory impairment of AD mice, likely dependent on the membrane receptor of p75NTR. CONCLUSIONS: Our findings suggest that proBDNF may exert a crucially negative effect during AD pathogenesis andprogression.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/metabolismo , Discapacidades para el Aprendizaje/metabolismo , Trastornos de la Memoria/metabolismo , Precursores de Proteínas/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Hipocampo/citología , Inyecciones Intraventriculares , Discapacidades para el Aprendizaje/etiología , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Ratones , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Presenilina-1/genética , Precursores de Proteínas/uso terapéutico , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Transducción Genética
4.
PLoS One ; 11(10): e0165586, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27792755

RESUMEN

Perineural invasion (PNI) is thought to be one of the factors responsible for the high rate of tumor recurrence after surgery and the pain generation associated with pancreatic cancer. Signaling via the nerve growth factor (NGF) pathway between pancreatic cancer cells and the surrounding nerves has been implicated in PNI, and increased levels of these proteins have been correlated to poor prognosis. In this study, we examine the molecular mechanism of the NGF signaling pathway in PNI in pancreatic cancer. We show that knocking down NGF or its receptors, TRKA and p75NTR, or treatment with GW441756, a TRKA kinase inhibitor, reduces the proliferation and migration of pancreatic cancer cells in vitro. Furthermore, pancreatic cancer cells migrate towards dorsal root ganglia (DRG) in a co-culture assay, indicating a paracrine NGF signaling between the DRGs and pancreatic cancer cells. Knocking down the expression of NGF pathway proteins or inhibiting the activity of TRKA by GW441756 reduced the migratory ability of Mia PaCa2 towards the DRGs. Finally, blocking NGF signaling by NGF neutralizing antibodies or GW441756 inhibited the neurite formation in PC-12 cells in response to conditioned media from pancreatic cancer cells, indicating a reciprocal signaling pathway between the pancreatic cancer cells and nerves. Our results indicate that NGF signaling pathway provides a potential target for developing molecularly targeted therapies to decrease PNI and reduce pain generation. Since there are several TRKA antagonists currently in early clinical trials they could now be tested in the clinical situation of pancreatic cancer induced pain.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Sistema Nervioso/patología , Neoplasias Pancreáticas/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Ganglios Espinales/patología , Técnicas de Inactivación de Genes , Humanos , Indoles/farmacología , Invasividad Neoplásica , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Receptor trkA/antagonistas & inhibidores , Receptor trkA/deficiencia , Receptor trkA/genética , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética
5.
Mol Psychiatry ; 20(11): 1301-10, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25917367

RESUMEN

In Alzheimer's disease (AD), neurodegenerative signals such as amyloid-beta (Aß) and the precursors of neurotrophins, outbalance neurotrophic signals, causing synaptic dysfunction and neurodegeneration. The neurotrophin receptor p75 (p75NTR) is a receptor of Aß and mediates Aß-induced neurodegenerative signals. The shedding of its ectodomain from the cell surface is physiologically regulated; however, the function of the diffusible p75NTR ectodomain (p75ECD) after shedding remains largely not known. Here, we show that p75ECD levels in cerebrospinal fluid and in the brains of Alzheimer's patients and amyloid-beta precursor protein (APP)/PS1 transgenic mice were significantly reduced, due to inhibition of the sheddase-tumor necrosis factor-alpha-converting enzyme by Aß. Restoration of p75ECD to the normal level by brain delivery of the gene encoding human p75ECD before or after Aß deposition in the brain of APP/PS1 mice reversed the behavioral deficits and AD-type pathologies, such as Aß deposit, apoptotic events, neuroinflammation, Tau phosphorylation and loss of dendritic spine, neuronal structures and synaptic proteins. Furthermore, p75ECD can also reduce amyloidogenesis by suppressing ß-secretase expression and activities. Our data demonstrate that p75ECD is a physiologically neuroprotective molecule against Aß toxicity and would be a novel therapeutic target and biomarker for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Encéfalo/patología , Proteínas del Tejido Nervioso/química , Estructura Terciaria de Proteína/fisiología , Receptores de Factor de Crecimiento Nervioso/química , Proteínas ADAM/metabolismo , Proteína ADAM17 , Factores de Edad , Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Precursor de Proteína beta-Amiloide/genética , Animales , Apoptosis/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Estudios de Casos y Controles , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/terapia , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Presenilina-1/genética , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Proteínas Recombinantes/uso terapéutico , Transducción Genética
6.
J Invest Dermatol ; 135(3): 786-795, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25330297

RESUMEN

CD271 is the low-affinity neurotrophin (p75NTR) receptor that belongs to the tumor necrosis factor receptor superfamily. Because in human epidermis, CD271 is predominantly expressed in transit-amplifying (TA) cells, we evaluated the role of this receptor in keratinocyte differentiation and in the transition from keratinocyte stem cells (KSCs) to progeny. Calcium induced an upregulation of CD271 in subconfluent keratinocytes, which was prevented by CD271 small interfering RNA. Furthermore, CD271 overexpression provoked the switch of KSCs to TA cells, whereas silencing CD271 induced TA cells to revert to a KSC phenotype, as shown by the expression of ß1-integrin and by the increased clonogenic ability. CD271(+) keratinocytes sorted from freshly isolated TA cells expressed more survivin and keratin 15 (K15) compared with CD271(-) cells and displayed a higher proliferative capacity. Early differentiation markers and K15 were more expressed in the skin equivalent generated from CD271(+) TA than from those derived from CD271(-) TA cells. By contrast, late differentiation markers were more expressed in skin equivalents from CD271(-) than in reconstructs from CD271(+) TA cells. Finally, skin equivalents originated from CD271(-) TA cells displayed a psoriatic phenotype. These results indicate that CD271 is critical for keratinocyte differentiation and regulates the transition from KSCs to TA cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Epidérmicas , Queratinocitos/citología , Proteínas del Tejido Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Células Madre/citología , Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Epidermis/efectos de los fármacos , Epidermis/metabolismo , Humanos , Técnicas In Vitro , Proteínas Inhibidoras de la Apoptosis/metabolismo , Queratina-15/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Fenotipo , Psoriasis/patología , ARN Interferente Pequeño/farmacología , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Survivin
7.
Cancer Chemother Pharmacol ; 73(2): 271-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24253178

RESUMEN

PURPOSE: Neuroblastoma is the most common extracranial solid tumor of childhood. The retinoic acid analogue, fenretinide (4-hydroxyphenyl retinamide; 4-HPR), induces apoptosis in neuroblastoma cells in vitro and is currently in clinical trials for children with refractory neuroblastoma. We have previously shown that expression of the p75 neurotrophin receptor (p75NTR) enhances apoptosis induction and mitochondrial accumulation of reactive oxygen species by 4-HPR in neuroblastoma cells. We now examine the signaling events that underlie this effect. METHODS: Systematic examination of pro- and anti-apoptotic signaling effectors was performed by Western blot. Specific inhibitors of JNK phosphorylation and scavengers of mitochondrial reactive oxygen species were used to demonstrate the roles of these phenomena in the enhancement of fenretinide efficacy. RESULTS: The present studies demonstrate that enhancement of 4-HPR-induced apoptosis by p75NTR is dependent upon p38MAPK phosphorylation, JNK phosphorylation, caspase 3 activation, Akt cleavage, and decreased Akt phosphorylation. In addition, treatment with 4-HPR results in upregulation of MKK4 and MEKK1, and phosphorylation of MKK3/6. Efforts to enhance the efficacy of 4-HPR and to identify those tumors most likely to respond to it might exploit these effectors of 4-HPR-induced apoptosis. CONCLUSIONS: Pharmacological agents that enhance MKK4 or MEKK1 expression or JNK expression or phosphorylation may enhance efficacy of 4-HPR in neuroblastomas that do not express high levels of p75NTR.


Asunto(s)
Antineoplásicos/farmacología , Fenretinida/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuroblastoma/tratamiento farmacológico , Receptores de Factor de Crecimiento Nervioso/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , MAP Quinasa Quinasa 4/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteína Oncogénica v-akt/metabolismo , Fosforilación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Transducción de Señal/efectos de los fármacos , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Cell Death Dis ; 4: e579, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23559013

RESUMEN

The p75 neurotrophin receptor (p75(NTR)) is a known mediator of ß-amyloid (Aß)-induced neurotoxicity implicated in Alzheimer's disease (AD). Here, we demonstrate that death receptor 6 (DR6) binds to p75(NTR) and is a component of the p75(NTR) signaling complex responsible for Aß-induced cortical neuron death. Cortical neurons isolated from either DR6 or p75(NTR) null mice are resistant to Aß-induced neurotoxicity. Blocking DR6 function in cortical neurons by anti-DR6 antibodies that block the binding of DR6 to p75(NTR) receptor complex or by a dominant negative DR6 construct lacking the cytoplasmic signaling death domain attenuates Aß-induced caspase 3 activation and cell death. DR6 expression is upregulated in AD cortex and correlates with elevated neuronal death. Targeting the disruption of the DR6/p75(NTR) complex to prevent Aß cytotoxicity represents a new approach for the treatment of neurodegenerative disorders such as AD.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores de Factor de Crecimiento Nervioso/genética , Receptores del Factor de Necrosis Tumoral/genética , Péptidos beta-Amiloides/farmacología , Animales , Anticuerpos/farmacología , Caspasa 3/genética , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Cultivo Primario de Células , Unión Proteica , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores del Factor de Necrosis Tumoral/deficiencia , Transducción de Señal/efectos de los fármacos
9.
J Neurochem ; 125(3): 386-98, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23350698

RESUMEN

The expression of the neurotrophins and their receptors is essential for peripheral nervous system development and myelination. We have previously demonstrated that brain-derived neurotrophic factor (BDNF) exerts contrasting influences upon Schwann cell myelination in vitro - promoting myelination via neuronally expressed p75NTR, but inhibiting myelination via neuronally expressed TrkB. We have generated a small peptide called cyclo-dPAKKR that structurally mimics the region of BDNF that binds p75NTR. Here, we have investigated whether utilizing cyclo-dPAKKR to selectively target p75NTR is an approach that could exert a unified promyelinating response. Like BDNF, cyclo-dPAKKR promoted myelination of nerve growth factor-dependent neurons in vitro, an effect dependent on the neuronal expression of p75NTR. Importantly, cyclo-dPAKKR also significantly promoted the myelination of tropomyosin-related kinase receptor B-expressing neurons in vitro, whereas BDNF exerted a significant inhibitory effect. This indicated that while BDNF exerted a contrasting influence upon the myelination of distinct subsets of dorsal root ganglion (DRG) neurons in vitro, cyclo-dPAKKR uniformly promoted their myelination. Local injection of cyclo-dPAKKR adjacent to the developing sciatic nerve in vivo significantly enhanced myelin protein expression and significantly increased the number of myelinated axons. These results demonstrate that cyclo-dPAKKR promotes peripheral myelination in vitro and in vivo, suggesting it is a strategy worthy of further investigation for the treatment of peripheral demyelinating diseases.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/química , Factor Neurotrófico Derivado del Encéfalo/farmacología , Vaina de Mielina/metabolismo , Péptidos/farmacología , Nervio Ciático/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Ganglios Espinales/citología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/efectos de los fármacos , Neurregulinas , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Factor de Crecimiento Nervioso/deficiencia , Células de Schwann , Nervio Ciático/efectos de los fármacos
10.
Arterioscler Thromb Vasc Biol ; 32(12): e149-60, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23065828

RESUMEN

OBJECTIVE: The p75 neurotrophin receptor (p75(NTR)) contributes to diabetes mellitus-induced defective postischemic neovascularization. The interleukin-33 receptor ST2 is expressed as transmembrane (ST2L) and soluble (sST2) isoforms. Here, we studied the following: (1) the impact of p75(NTR) in the healing of ischemic and diabetic calf wounds; (2) the link between p75(NTR) and ST2; and (3) circulating sST2 levels in critical limb ischemia (CLI) patients. METHODS AND RESULTS: Diabetes mellitus was induced in p75(NTR) knockout (p75KO) mice and wild-type (WT) littermates by streptozotocin. Diabetic and nondiabetic p75KO and WT mice received left limb ischemia induction and a full-thickness wound on the ipsilateral calf. Diabetes mellitus impaired wound closure and angiogenesis and increased ST2 expression in WT, but not in p75KO wounds. In cultured endothelial cells, p75(NTR) promoted ST2 (both isoforms) expression through p38(MAPK)/activating transcription factor 2 pathway activation. Next, sST2 was measured in the serum of patients with CLI undergoing either revascularization or limb amputation and in the 2 nondiabetic groups (with CLI or nonischemic individuals). Serum sST2 increased in diabetic patients with CLI and was directly associated with higher mortality at 1 year from revascularization. CONCLUSIONS: p75(NTR) inhibits the healing of ischemic lower limb wounds in diabetes mellitus and promotes ST2 expression. Circulating sST2 predicts mortality in diabetic CLI patients.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus/mortalidad , Isquemia/fisiopatología , Extremidad Inferior/irrigación sanguínea , Proteínas del Tejido Nervioso/fisiología , Receptores de Superficie Celular/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Factor de Crecimiento Nervioso/fisiología , Factor de Transcripción Activador 2/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores/metabolismo , Células Cultivadas , Complicaciones de la Diabetes/complicaciones , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Humanos , Proteína 1 Similar al Receptor de Interleucina-1 , Isquemia/etiología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas del Tejido Nervioso/farmacología , Valor Predictivo de las Pruebas , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Estreptozocina/efectos adversos , Cicatrización de Heridas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Biol Psychiatry ; 71(1): 75-83, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21978521

RESUMEN

BACKGROUND: Stress is causally associated with anxiety. Although the underlying cellular mechanisms are not well understood, the basal forebrain cholinergic neurons have been implicated in stress response. p75(NTR) is a panneurotrophin receptor expressed almost exclusively in basal forebrain cholinergic neurons in adult brain. This study investigated whether and how p75(NTR), via regulation of the cholinergic system and hippocampal synaptic plasticity, influences stress-related behaviors. METHODS: We used a combination of slice electrophysiology, behavioral analyses, pharmacology, in vivo microdialysis, and neuronal activity mapping to assess the role of p75(NTR) in mood and stress-related behaviors and its underlying cellular and molecular mechanisms. RESULTS: We show that acute stress enables hippocampal long-term depression (LTD) in adult wild-type mice but not in mice lacking p75(NTR). The p75(NTR) mutant mice also exhibit two distinct behavioral impairments: baseline anxiety-like behavior and a deficit in coping with and recovering from stressful situations. Blockade of stress-enabled LTD with a GluA2-derived peptide impaired stress recovery without affecting baseline anxiety. Pharmacological manipulations of cholinergic transmission mimicked the p75(NTR) perturbation in both baseline anxiety and responses to acute stress. Finally, we show evidence of misregulated cholinergic signaling in animals with p75(NTR) deletion. CONCLUSIONS: Our results suggest that loss of p75(NTR) leads to changes in hippocampal cholinergic signaling, which may be involved in regulation of stress-enabled hippocampal LTD and in modulating behaviors related to stress and anxiety.


Asunto(s)
Acetilcolina/metabolismo , Ansiedad/genética , Hipocampo/fisiopatología , Depresión Sináptica a Largo Plazo/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Estrés Psicológico/genética , Adaptación Psicológica/efectos de los fármacos , Adaptación Psicológica/fisiología , Análisis de Varianza , Animales , Ansiedad/patología , Ansiedad/fisiopatología , Biofisica , Colinérgicos/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Depresión Sináptica a Largo Plazo/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microdiálisis , Microinyecciones , N-Metilaspartato/farmacología , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Factor de Crecimiento Nervioso/deficiencia , Estrés Psicológico/patología , Estrés Psicológico/fisiopatología , Factores de Tiempo
13.
Mol Cell ; 44(3): 476-90, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-22055192

RESUMEN

Homeostatic control of oxygen availability allows cells to survive oxygen deprivation. Although the transcription factor hypoxia-inducible factor 1α (HIF-1α) is the main regulator of the hypoxic response, the upstream mechanisms required for its stabilization remain elusive. Here, we show that p75 neurotrophin receptor (p75(NTR)) undergoes hypoxia-induced γ-secretase-dependent cleavage to provide a positive feed-forward mechanism required for oxygen-dependent HIF-1α stabilization. The intracellular domain of p75(NTR) directly interacts with the evolutionarily conserved zinc finger domains of the E3 RING ubiquitin ligase Siah2 (seven in absentia homolog 2), which regulates HIF-1α degradation. p75(NTR) stabilizes Siah2 by decreasing its auto-ubiquitination. Genetic loss of p75(NTR) dramatically decreases Siah2 abundance, HIF-1α stabilization, and induction of HIF-1α target genes in hypoxia. p75(NTR-/-) mice show reduced HIF-1α stabilization, vascular endothelial growth factor (VEGF) expression, and neoangiogenesis after retinal hypoxia. Thus, hypoxia-induced intramembrane proteolysis of p75(NTR) constitutes an apical oxygen-dependent mechanism to control the magnitude of the hypoxic response.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/metabolismo , Procesamiento Proteico-Postraduccional , Receptores de Factor de Crecimiento Nervioso/metabolismo , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Hipoxia de la Célula , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Células 3T3 NIH , Complejo de la Endopetidasa Proteasomal/metabolismo , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Receptores de Factor de Crecimiento Nervioso/química , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Neovascularización Retiniana/metabolismo , Factores de Tiempo , Transfección , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Auton Neurosci ; 164(1-2): 13-9, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21646052

RESUMEN

Cardiac sympathetic neurons stimulate heart rate and the force of contraction through release of norepinephrine. Nerve growth factor modulates sympathetic transmission through activation of TrkA and p75NTR. Nerve growth factor plays an important role in post-infarct sympathetic remodeling. We used mice lacking p75NTR to examine the effect of altered nerve growth factor signaling on sympathetic neuropeptide expression, cardiac norepinephrine, and ventricular function after myocardial infarction. Infarct size was similar in wildtype and p75NTR-/- mice after ischemia-reperfusion surgery. Likewise, mRNAs encoding vasoactive intestinal peptide, galanin, and pituitary adenylate cyclase activating peptides were identical in wildtype and p75NTR-/- cardiac sympathetic neurons, as was expression of the TrkA neurotrophin receptor. Norepinephrine content was elevated in the base of the p75NTR-/- ventricle compared to wildtype, but levels were identical below the site of occlusion. Left ventricular pressure, dP/dt(MAX), and dP/dt(MIN) were measured under isoflurane anesthesia 3 and 7 days after surgery. Ventricular pressure decreased significantly 3 days after infarction, and deficits in dP/dt(MAX) were revealed by stimulating beta receptors with dobutamine and release of endogenous norepinephrine with tyramine. dP/dt(MIN) was not altered by genotype or surgical group. Few differences were observed between genotypes 3 days after surgery, in contrast to low pressure and dP/dt(MAX) previously reported in control p75NTR-/- animals. Seven days after surgery ventricular pressure and dP/dt(MAX) were significantly lower in p75NTR-/- hearts compared to WT hearts. Thus, the lack of p75NTR did not enhance cardiac function after myocardial infarction.


Asunto(s)
Ventrículos Cardíacos/inervación , Ventrículos Cardíacos/fisiopatología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Norepinefrina/metabolismo , Receptores de Factor de Crecimiento Nervioso/fisiología , Fibras Simpáticas Posganglionares/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Ventrículos Cardíacos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Norepinefrina/biosíntesis , Norepinefrina/fisiología , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Fibras Simpáticas Posganglionares/metabolismo , Regulación hacia Arriba/genética
15.
J Neurosci ; 31(6): 2292-304, 2011 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21307265

RESUMEN

Accumulation of toxic amyloid-ß (Aß) in the cerebral cortex and hippocampus is a major pathological feature of Alzheimer's disease (AD). The neurotrophin receptor p75NTR has been proposed to mediate Aß-induced neurotoxicity; however, its role in the development of AD remains to be clarified. The p75NTR/ExonIII-/- mice and APPSwe/PS1dE9 mice were crossed to generate transgenic AD mice with deletion of p75NTR gene. In APPSwe/PS1dE9 transgenic mice, p75NTR expression was localized in the basal forebrain neurons and degenerative neurites in neocortex, increased with aging, and further activated by Aß accumulation. Deletion of the p75NTR gene in APPSwe/PS1dE9 mice reduced soluble Aß levels in the brain and serum, but increased the accumulation of insoluble Aß and Aß plaque formation. There was no change in the levels of amyloid precursor protein (APP) and its proteolytic derivatives, or α-, ß-, and γ-secretase activities, or in levels of BACE1, neprilysin (NEP), and insulin-degrading enzyme (IDE) proteins. Aß production by cortical neurons of APPSwe/PS1dE9 mice was reduced by deletion of p75NTR gene in vitro. Recombinant extracellular domain of p75NTR attenuated the oligomerization and fibrillation of synthetic Aß(42) peptide in vitro, and reduced local Aß plaques after hippocampus injection in vivo. In addition, deletion of p75NTR attenuated microgliosis but increased the microhemorrhage profiles in the brain. The deletion of p75NTR did not significantly change the cognitive function of the mice up to the age of 9 months. Our data suggest that p75NTR plays a critical role in regulating Aß levels by both increasing Aß production and attenuating its aggregation, and they caution that a therapeutic intervention simply reducing p75NTR may exacerbate AD pathology.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Regulación de la Expresión Génica/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Factores de Edad , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Conducta Animal , Encéfalo/citología , Humanos , Insulisina/metabolismo , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Mutación/genética , Neprilisina/metabolismo , Neuronas/metabolismo , Presenilina-1/genética , Receptores de Factor de Crecimiento Nervioso/deficiencia
16.
Br J Pharmacol ; 162(5): 1186-201, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21091650

RESUMEN

BACKGROUND AND PURPOSE: Pancreatitis represents a life-threatening inflammatory condition where leucocytes, cytokines and vascular endothelium contribute to the development of the inflammatory disease. The glucocorticoid-induced tumour necrosis factor (TNF) receptor family-related protein (GITR) is a costimulatory molecule for T lymphocytes, modulates innate and adaptive immune system and has been found to participate in a variety of immune responses and inflammatory processes. Our purpose was to verify whether inhibition of GITR triggering results in a better outcome in experimental pancreatitis. EXPERIMENTAL APPROACH: In male GITR knock-out (GITR(-/-)) and GITR(+/+) mice on Sv129 background, acute pancreatitis was induced after i.p. administration of cerulein. Other experimental groups of GITR(+/+) mice were also treated with different doses of Fc-GITR fusion protein (up to 6.25 µg·mouse⁻¹), given by implanted mini-osmotic pump. Clinical score and pro-inflammatory parameters were evaluated. KEY RESULTS: A less acute pancreatitis was found in GITR(-/-) mice than in GITR(+/+) mice, with marked differences in oedema, neutrophil infiltration, pancreatic dysfunction and injury. Co-treatment of GITR(+/+) mice with cerulein and Fc-GITR fusion protein (6.25 µg·mouse⁻¹) decreased the inflammatory response and tissue injury, compared with treatment with cerulein alone. Inhibition of GITR triggering was found to modulate activation of nuclear factor κB as well as the production of TNF-α, interleukin-1ß, inducible nitric oxide synthase, nitrotyrosine, poly-ADP-ribose, intercellular adhesion molecule-1 and P-selectin. CONCLUSIONS AND IMPLICATIONS: The GITR-GITR ligand system is crucial to the development of acute pancreatitis in mice. Our results also suggest that the Fc-GITR fusion protein could be useful in the treatment of acute pancreatitis.


Asunto(s)
Pancreatitis/etiología , Receptores de Factor de Crecimiento Nervioso/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Animales , Apoptosis , Ceruletida/toxicidad , Edema/etiología , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Proteínas I-kappa B/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/metabolismo , Ligandos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Inhibidor NF-kappaB alfa , Infiltración Neutrófila , Óxido Nítrico Sintasa de Tipo II/metabolismo , Selectina-P/metabolismo , Pancreatitis/patología , Pancreatitis/fisiopatología , Pancreatitis/prevención & control , Poli(ADP-Ribosa) Polimerasas/metabolismo , Receptores de Factor de Crecimiento Nervioso/administración & dosificación , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Receptores del Factor de Necrosis Tumoral/administración & dosificación , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Linfocitos T/fisiología , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
J Immunol ; 186(1): 275-83, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21106849

RESUMEN

Treatment of tumor-bearing mice with a stimulatory Ab to glucocorticoid-induced TNFR family-related receptor (GITR) has previously been shown to elicit protective T cell responses against poorly immunogenic tumors. However, the role of GITR stimulation on CD8 T cells and the nature of tumor rejection Ags have yet to be determined. In this study, we show that a stimulatory mAb to GITR (clone DTA-1) acts directly on CD8 T cells, but not on CD4(+)CD25(+) regulatory T (T(reg)) cells, in B16 tumor-bearing mice to induce concomitant immunity against secondary B16 tumors, as well as protective memory following surgical excision of the primary tumor. Melanoma growth itself induced GITR expression on tumor-specific CD8 T cells, providing a mechanism whereby these cells may respond to stimulatory anti-GITR. Unexpectedly, in contrast to T(reg) cell depletion therapy with anti-CD4, GITR stimulation induced very weak CD8 T cell responses to melanocyte differentiation Ags expressed by the tumor, and did not induce autoimmune vitiligo. Accordingly, GITR-stimulated hosts that were primed with B16 melanoma rejected B16, but not the unrelated JBRH melanoma, indicating that tumor rejection Ags are tumor-specific rather than shared. In support of this, we show that GITR stimulation induces CD8 T cell responses to a tumor-specific Ag, and that these responses are of higher functional avidity compared with those induced by T(reg) cell depletion. We conclude that stimulation of GITR on effector CD8 T cells results in high-avidity T cell responses to tumor-specific Ags, thereby inducing potent antitumor immunity in the absence of autoimmunity.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Epítopos de Linfocito T/inmunología , Activación de Linfocitos/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Linfocitos T CD8-positivos/patología , Adhesión Celular/inmunología , Línea Celular Tumoral , Femenino , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Depleción Linfocítica , Masculino , Melanoma Experimental/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/inmunología , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/inmunología , Linfocitos T Reguladores/inmunología
19.
Nature ; 466(7302): 133-7, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20596026

RESUMEN

The question of whether tumorigenic cancer stem cells exist in human melanomas has arisen in the last few years. Here we show that in melanomas, tumour stem cells (MTSCs, for melanoma tumour stem cells) can be isolated prospectively as a highly enriched CD271(+) MTSC population using a process that maximizes viable cell transplantation. The tumours sampled in this study were taken from a broad spectrum of sites and stages. High-viability cells isolated by fluorescence-activated cell sorting and re-suspended in a matrigel vehicle were implanted into T-, B- and natural-killer-deficient Rag2(-/-)gammac(-/-) mice. The CD271(+) subset of cells was the tumour-initiating population in 90% (nine out of ten) of melanomas tested. Transplantation of isolated CD271(+) melanoma cells into engrafted human skin or bone in Rag2(-/-)gammac(-/-) mice resulted in melanoma; however, melanoma did not develop after transplantation of isolated CD271(-) cells. We also show that in mice, tumours derived from transplanted human CD271(+) melanoma cells were capable of metastatsis in vivo. CD271(+) melanoma cells lacked expression of TYR, MART1 and MAGE in 86%, 69% and 68% of melanoma patients, respectively, which helps to explain why T-cell therapies directed at these antigens usually result in only temporary tumour shrinkage.


Asunto(s)
Melanoma/metabolismo , Melanoma/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Animales , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/metabolismo , Trasplante Óseo , Huesos/patología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Humanos , Neoplasias Pulmonares/secundario , Antígenos Específicos del Melanoma , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/trasplante , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Cresta Neural/citología , Cresta Neural/patología , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Piel/patología , Trasplante de Piel , Trasplante Heterólogo/patología
20.
J Immunol ; 182(12): 7490-500, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494272

RESUMEN

Glucocorticoid-induced TNF receptor family-related protein (GITR) is expressed on activated and regulatory T cells, but its role on these functionally opposing cell types is not fully understood. Here we describe that transgenic expression of GITR's unique ligand (GITRL) induces a prominent increase of both effector and regulatory CD4(+) T cells, but not CD8(+) T cells. Regulatory T cells from GITRL transgenic mice are phenotypically activated and retain their suppressive capacity. The accumulation of effector and regulatory T cells is not due to enhanced differentiation of naive T cells, but is a direct result of increased proliferation. Functional consequences of increased numbers of both regulatory and effector T cells were tested in an autoimmune model and show that GITR stimulation is protective, as it significantly delays disease induction. These data indicate that GITR regulates the balance between regulatory and effector CD4(+) T cells by enhancing proliferation of both populations in parallel.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Células Cultivadas , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Humanos , Memoria Inmunológica/inmunología , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA