Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Neurobiol Dis ; 97(Pt B): 169-178, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27063797

RESUMEN

The therapeutic potential of neurotrophic factors has been recognized for decades, with clinical trials in human neurodegenerative diseases extending back at least 25years. While improvements in clinical dosing paradigms have reduced the side effects commonly seen in the earlier trials, efficacy has remained a serious disappointment (reviewed in Bartus and Johnson, 2016). This lengthy clinical effort stands in contrast to robust effects consistently achieved from different neurotrophic factors in a variety of animal models of neurodegeneration. This review discusses the prevailing assumption and supporting data that the major reason for the disappointing efficacy of past clinical trials is related to suboptimal dosing methods. It is concluded that while further improvements in dosing parameters might be useful, a much greater problem centers around a number of specific morphologic and functional changes in neurons in human neurodegenerative disease that mitigate the ability of neurotrophic factors to exert their effects. Moreover, the biological substrate which neurotrophic factors depend upon to exert their effects continues to erode as time progresses, due to the progressive nature of these diseases. For this reason, most of the empirically-supported reasons contributing to the weak neurotrophic responses in human patients can be mitigated by enrolling less severely advanced cases. It is further concluded that recent clinical trials of neurotrophic factors have generated important evidence that shifts risk: benefit assessments to support enrolling earlier-stage patients. While the Alzheimer's field has begun to shift attention toward much earlier-stage (even prodromal) patients in trials intended to modify disease progression, other neurodegenerative diseases (e.g., Parkinson's, ALS and possibly HD) must now consider similar changes in approach.


Asunto(s)
Factores de Crecimiento Nervioso/administración & dosificación , Enfermedades Neurodegenerativas/terapia , Fármacos Neuroprotectores/administración & dosificación , Animales , Ensayos Clínicos como Asunto , Humanos , Factores de Crecimiento Nervioso/genética
2.
Neurobiol Dis ; 97(Pt B): 156-168, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27063798

RESUMEN

Over the past 25years, about 3 dozen clinical reports have been published regarding the safety and possible efficacy of neurotrophic factors in patients with various neurodegenerative diseases. This effort involved a half dozen different neurotrophic factors, using at least 5 different general delivery approaches for ALS (amyolateral sclerosis), peripheral neuropathies, PD (Parkinson's disease) and AD (Alzheimer's disease). While none of these efforts have yet produced efficacy data sufficiently robust or reliable to establish neurotrophic factors as treatments for any human disease, the obstacles encountered and novel information reported, when viewed collectively, provide important insight to help future efforts. Three consistent themes emerge from these publications: (1) unexpected and undesirable side effects, at times serious, have plagued many efforts to deliver neurotrophic factors to humans; (2) the magnitude and consistency of clinical benefit has been disappointing; (3) by far that most consistently proposed reason for the side effects and poor efficacy has been inadequate dosing and delivery. This paper reviews and attempts to synthesize the available data derived from clinical tests of neurotrophic factors for neurodegenerative diseases. The obstacles encountered, the solutions attempted, and the lessons learned are discussed. The vast majority of solutions have involved changes in dosing paradigms and dose levels, which has primarily led to improved safety outcomes. However, lack of adequate efficacy remains a significant issue. While current efforts continue to focus exclusively on still-further changes in dosing parameters, a review of available data argues that it may now be the time to ask whether other, non-dose-related variables should be given more serious consideration as being responsible for the great divide that exists between the robust effects seen in animal models and the relatively weak effects seen in human neurodegenerative patients. Foremost among these appears to be the severe degeneration seen in the majority of patients enrolled in past and current trials testing neurotrophic factors in humans. A companion paper (Bartus and Johnson, 2016), reviews the contemporary data and concludes that compelling empirical evidence already exists for enrolling earlier-stage subjects as likely essential to achieving more robust and reliable benefit.


Asunto(s)
Factores de Crecimiento Nervioso/administración & dosificación , Enfermedades Neurodegenerativas/terapia , Fármacos Neuroprotectores/administración & dosificación , Animales , Ensayos Clínicos como Asunto , Humanos , Factores de Crecimiento Nervioso/genética
3.
J Neurosci ; 33(43): 17095-107, 2013 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-24155314

RESUMEN

The loss of dopaminergic neurons in the substantia nigra pars compacta (SNc) and consequent depletion of striatal dopamine are known to underlie the motor deficits observed in Parkinson's disease (PD). Adaptive changes in dopaminergic terminals and in postsynaptic striatal neurons can compensate for significant losses of striatal dopamine, resulting in preservation of motor behavior. In addition, compensatory changes independent of striatal dopamine have been proposed based on PD therapies that modulate nondopaminergic circuits within the basal ganglia. We used a genetic strategy to selectively destroy dopaminergic neurons in mice during development to determine the necessity of these neurons for the maintenance of normal motor behavior in adult and aged mice. We find that loss of 90% of SNc dopaminergic neurons and consequent depletion of >95% of striatal dopamine does not result in changes in motor behavior in young-adult or aged mice as evaluated by an extensive array of motor behavior tests. Treatment of aged mutant mice with the dopamine receptor antagonist haloperidol precipitated motor behavior deficits in aged mutant mice, indicating that <5% of striatal dopamine is sufficient to maintain motor function in these mice. We also found that mutant mice exhibit an exaggerated response to l-DOPA compared with control mice, suggesting that preservation of motor function involves sensitization of striatal dopamine receptors. Our results indicate that congenital loss of dopaminergic neurons induces remarkable adaptions in the nigrostriatal system where limited amounts of dopamine in the dorsal striatum can maintain normal motor function.


Asunto(s)
Dopamina/metabolismo , Neuronas Dopaminérgicas/patología , Marcha , Animales , Toxina Diftérica/genética , Toxina Diftérica/toxicidad , Dopamina/deficiencia , Antagonistas de Dopamina/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/deficiencia , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Haloperidol/farmacología , Levodopa/farmacología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/patología , Ratones , Ratones Transgénicos , Mutación
4.
Neurobiol Dis ; 58: 38-48, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23631873

RESUMEN

This paper reassesses the currently accepted viewpoint that targeting the terminal fields (i.e. striatum) of degenerating nigrostriatal dopamine neurons with neurotrophic factors in Parkinson's disease (PD) is sufficient for achieving an optimal neurotrophic response. Recent insight indicating that PD is an axonopathy characterized by axonal transport deficits prompted this effort. We tested whether a significantly greater neurotrophic response might be induced in SN neurons when the neurotrophic factor neurturin (NRTN) is also targeted to the substantia nigra (SN), compared to the more conventional, striatum-only target. While recognizing the importance of maintaining the integrity of nigrostriatal fibers and terminals (especially for achieving optimal function), we refocused attention to the fate of SN neurons. Under conditions of axonal degeneration and neuronal transport deficits, this component of the nigrostriatal system is most vulnerable to the lack of neurotrophic exposure following striatal-only delivery. Given the location of repair genes induced by neurotrophic factors, achieving adequate neurotrophic exposure to the SN neurons is essential for an optimal neurotrophic response, while the survival of these neurons is essential to the very survival of the fibers. Two separate studies were performed using the 6-OHDA model of nigrostriatal degeneration, in conjunction with delivery of the viral vector AAV2-NRTN (CERE-120) to continuously express NRTN to either striatum or nigra alone or combined striatal/nigral exposure, including conditions of ongoing axonopathy. These studies provide additional insight for reinterpreting past animal neurotrophic/6-OHDA studies conducted under conditions where axon transport deficiencies were generally not accounted for, which suggested that targeting the striatum was both necessary and sufficient. The current data demonstrate that delivering NRTN directly to the SN produces 1) expanded NRTN distribution within the terminal field and cell bodies of targeted nigrostriatal neurons, 2) enhanced intracellular neurotrophic factor signaling in the nigrostriatal neurons, and 3) produced greater numbers of surviving dopamine neurons against 6-OHDA-induced toxicity, particularly under the conditions of active axonopathy. Thus, these data provide empirical support that targeting the SN with neurotrophic factors (in addition to striatum) may help enhance the neurotrophic response in midbrain neurons, particularly under conditions of active neurodegeneration which occurs in PD patients.


Asunto(s)
Adenoviridae/genética , Cuerpo Estriado/metabolismo , Factores de Crecimiento Nervioso/administración & dosificación , Enfermedades Neurodegenerativas/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Sustancia Negra/metabolismo , Adrenérgicos/toxicidad , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Vectores Genéticos/fisiología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/metabolismo , Oxidopamina/toxicidad , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa
5.
J Neurosci ; 30(11): 3983-94, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20237269

RESUMEN

Small unmyelinated sensory neurons classified as nociceptors are divided into two subpopulations based on phenotypic differences, including expression of neurotrophic factor receptors. Approximately half of unmyelinated nociceptors express the NGF receptor TrkA, and half express the GDNF family ligand (GFL) receptor Ret. The function of NGF/TrkA signaling in the TrkA population of nociceptors has been extensively studied, and NGF/TrkA signaling is a well established mediator of pain. The GFLs are analgesic in models of neuropathic pain emphasizing the importance of understanding the physiological function of GFL/Ret signaling in nociceptors. However, perinatal lethality of Ret-null mice has precluded the study of the physiological role of GFL/Ret signaling in the survival, maintenance, and function of nociceptors in viable mice. We deleted Ret exclusively in nociceptors by crossing nociceptor-specific Na(v)1.8 Cre and Ret conditional mice to produce Ret-Na(v)1.8 conditional knock-out (CKO) mice. Loss of Ret exclusively in nociceptors results in a reduction in nociceptor number and size, indicating that Ret signaling is important for the survival and trophic support of these cells. Ret-Na(v)1.8 CKO mice exhibit reduced epidermal innervation but normal central projections. In addition, Ret-Na(v)1.8 CKO mice have increased sensitivity to cold and increased formalin-induced pain, demonstrating that Ret signaling modulates the function of nociceptors in vivo. Enhanced inflammation-induced pain may be mediated by decreased prostatic acid phosphatase (PAP), as PAP levels are markedly reduced in Ret-Na(v)1.8 CKO mice. The results of this study identify the physiological role of endogenous Ret signaling in the survival and function of nociceptors.


Asunto(s)
Nociceptores/fisiología , Proteínas Proto-Oncogénicas c-ret/fisiología , Transducción de Señal/fisiología , Animales , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Femenino , Formaldehído/administración & dosificación , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.8 , Nociceptores/citología , Dimensión del Dolor/métodos , Proteínas Proto-Oncogénicas c-ret/deficiencia , Proteínas Proto-Oncogénicas c-ret/genética , Transducción de Señal/genética , Canales de Sodio/deficiencia , Canales de Sodio/genética
6.
Neurobiol Dis ; 44(1): 38-52, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21704161

RESUMEN

Recent analyses of autopsied brains from subjects previously administered AAV2-neurturin (NRTN) gene transfer argues that optimizing the effects of neurotrophic factors in Parkinson's disease (PD) likely requires delivery to both the degenerating cell bodies (in substantia nigra) and their terminals (in striatum). Prior to implementing this novel dosing paradigm in humans, we conducted eight nonclinical experiments with three general objectives: (1) evaluate the feasibility, safety and effectiveness of targeting the substantia nigra (SN) with AAV2-NRTN, (2) better understand and appraise recent warnings of serious weight loss that might occur with targeting the SN with neurotrophic factors, and (3) define an appropriate dose of AAV2-NRTN that should safely and effectively cover the SN in PD patients. Toward these ends, we first determined SN volume for rats, monkeys and humans, and employed these values to calculate comparable dose equivalents for each species by scaling each dose, based on relative SN volume. Using this information, we next injected AAV2-GFP to monkey SN to quantify AAV2-vector distribution and confirm reasonable SN coverage. We then selected and administered a ~200-fold range of AAV2-NRTN doses (and a single AAV2-GDNF dose) to rat SN, producing a wide range of protein expression. In contrast to recent warnings regarding nigra targeting, no dose produced any serious side effects or toxicity, though we replicated the modest reduction in weight gain reported by others with the highest AAV2-NRTN and the AAV2-GDNF dose. A dose-related increase in NRTN expression was seen, with the lower doses limiting NRTN to the peri-SN and the highest dose producing mistargeted NRTN well outside the SN. We then demonstrated that the reduction in weight gain following excessive-doses can be dissociated from NRTN in the targeted SN, and is linked to mistargeted NRTN in the diencephalon. We also showed that prior destruction of the dopaminergic SN neurons via 6-OHDA had no impact on the weight loss phenomenon, further dissociating neurotrophic exposure to the SN as the culprit for weight changes. Finally, low AAV2-NRTN doses provided significant neuroprotection against 6-OHDA toxicity, establishing a wide therapeutic index for nigral targeting. These data support targeting the SN with AAV2-NRTN in PD patients, demonstrating that properly targeted and scaled AAV2-NRTN provides safe and effective NRTN expression. They also provided the means to define an appropriate human-equivalent dose for proceeding into an ongoing clinical trial, using empirically-based scaling to account for marked differences in SN volume between species.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Neurturina/metabolismo , Enfermedad de Parkinson/terapia , Sustancia Negra/metabolismo , Animales , Conducta Animal/fisiología , Dieta , Dosificación de Gen , Marcación de Gen , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Proteínas Fluorescentes Verdes , Inmunohistoquímica , Masculino , Neuronas/metabolismo , Neurturina/efectos adversos , Ratas , Ratas Sprague-Dawley , Medición de Riesgo , Tirosina 3-Monooxigenasa/metabolismo , Aumento de Peso/genética , Aumento de Peso/fisiología , Pérdida de Peso/fisiología
7.
Mov Disord ; 26(1): 27-36, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21322017

RESUMEN

BACKGROUND: AAV2-neurturin (CERE-120) is designed to deliver the neurotrophic-factor, neurturin, to the striatum to restore and protect degenerating nigrostriatal neurons in Parkinson's disease (PD). A common hypothesis is that following expression in the striatum, neurotrophic-factors like neurturin (NRTN) will be transported from degenerating terminals to their cell bodies in the substantia nigra pars compacta (SNc). METHODS: We tested this concept using immunohistochemistry, comparing the bioactivity of AAV2-neurturin in brains of PD patients versus those of nonhuman primates similarly treated. RESULTS: NRTN-immunostaining in the targeted striatum was seen in all PD cases (mean putaminal coverage: ∼15% by volume); comparable expression was observed in young, aged, and parkinsonian monkeys. In the SNc cell bodies, however, only rare evidence of neurturin was seen in PD, while ample evidence of intense nigral-NRTN was observed in all monkeys. NRTN-expression was associated with occasional, sparse TH-induction in the striatum of PD, but nothing apparent in the SNc. In primates, NRTN produced robust TH-induction throughout the nigrostriatal neurons. DISCUSSION: These data provide the first evidence that gene therapy can increase expression of a neurotrophic-factor deep in the PD brain and that clear but modest enhancement of degenerating neurons can be induced. They also provide important insight regarding deficiencies in the status of nigrostriatal neurons in advanced PD, suggesting that serious axon-transport deficits reduced the bioactivity of AAV2-NRTN by limiting the protein exposed to the cell body. Thus, future efforts using neurotrophic-factors to treat neurodegenerative diseases will need to target both the terminal fields and the cell bodies of degenerating neurons to assure maximal benefit is achieved.


Asunto(s)
Cuerpo Estriado/metabolismo , Terapia Genética/métodos , Intoxicación por MPTP/terapia , Neurturina/uso terapéutico , Enfermedad de Parkinson/terapia , Anciano , Animales , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Lateralidad Funcional , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Intoxicación por MPTP/inducido químicamente , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/patología , Macaca mulatta , Masculino , Persona de Mediana Edad , Neurturina/genética , Neurturina/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Tirosina 3-Monooxigenasa/metabolismo
8.
J Cell Biol ; 170(3): 401-11, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16061693

RESUMEN

c-Jun is induced in many neuronal death paradigms. A critical step in c-Jun regulation involves phosphorylation of Ser63/Ser73 located in the NH2-terminal transactivation domain. To determine the importance of this phosphorylation for neuronal apoptosis, we analyzed the sympathetic neurons of mice carrying a mutant c-Jun gene that lacks Ser63/Ser73 phosphorylation sites (jun aa). Trophic factor-deprivation or DNA damage-induced death was significantly delayed in jun aa/aa neurons. Neuronal c-Jun induction was only partially inhibited, demonstrating that phosphorylation of Ser63/73 is not required for c-Jun activation. The inductions of proapoptotic BH3-only proteins, Bim and PUMA/Bbc3, were delayed during neuronal apoptosis in mutant neurons. These results demonstrate that NH2-terminal c-Jun phosphorylation is important, but not necessary, for the induction of proapoptotic genes and neuronal apoptosis. Thus, additional JNK substrates may be critical for neuronal death. As potential mediators, we identified additional nuclear MLK/JNK substrates, including Nup214 subunit of the nuclear pore complex.


Asunto(s)
Apoptosis/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/fisiología , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/fisiología , Animales , Proteínas Reguladoras de la Apoptosis , Proteína 11 Similar a Bcl2 , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Daño del ADN , Activación Enzimática , Ganglios Espinales/citología , Regulación de la Expresión Génica , Genes jun , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Factor de Crecimiento Nervioso/deficiencia , Fosforilación , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
9.
J Neurosci ; 28(16): 4123-35, 2008 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-18417692

RESUMEN

The glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) [GDNF, NRTN (neurturin), ARTN (artemin), and PSPN (persephin)] interact with GDNF family receptors (GFRalphas) and activate intracellular signaling through the Ret receptor tyrosine kinase. To characterize the role of Ret signaling in retinal activity, we examined Ret hypomorphic and Ret conditional mice using electroretinography. We found that aberrant Ret function resulted in markedly diminished scotopic and photopic responses. Using mice deficient in individual GFLs, we found that only NRTN deficiency led to reduced retinal activity. To determine the potential target cell type for NRTN, we examined the retinal expression of its coreceptors (GFRalpha1 and GFRalpha2) and Ret using mice expressing fluorescence reporter enhanced green fluorescent protein from their respective loci. We found robust GFRalpha1 and Ret expression in horizontal, amacrine, and ganglion cells, whereas GFRalpha2 expression was only detected in a subset of amacrine and ganglion cells. In contrast to previous studies, no expression of GFRalpha1, GFRalpha2, or Ret was detected in photoreceptors or Müller cells, suggesting that these cells are not directly affected by Ret. Finally, detailed morphologic analyses of retinas from NRTN- and Ret-deficient mice demonstrated a reduction in normal horizontal cell dendrites and axons, abnormal extensions of horizontal cell and bipolar cell processes into the outer nuclear layer, and mislocalized synaptic complexes. These anatomic abnormalities indicate a possible basis for the abnormal retinal activity in the Ret and NRTN mutant mice.


Asunto(s)
Neurturina/fisiología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Retina/fisiología , Animales , Ratones , Ratones Noqueados , Neurturina/genética , Proteínas Proto-Oncogénicas c-ret/deficiencia , Proteínas Proto-Oncogénicas c-ret/genética , Retina/citología , Retina/crecimiento & desarrollo , Transducción de Señal/genética , Transducción de Señal/fisiología
10.
J Cell Biol ; 157(5): 771-81, 2002 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-12021257

RESUMEN

Opening of the permeability transition pore (PTP) has been implicated as an important mitochondrial event that occurs during apoptosis. We examined the role of the PTP in the well-characterized cell death of rat sympathetic neurons deprived of nerve growth factor (NGF) in vitro. Removal of NGF causes these neurons to undergo either a classic apoptotic cell death or, when treated with a broad-spectrum caspase inhibitor such as boc-aspartyl(OMe)-fluoromethylketone (BAF), a delayed, nonapoptotic cell death. The PTP inhibitor, cyclosporin A (CsA), blocked commitment-to-die in the presence of BAF, as defined by the ability of NGF readdition to rescue cells, but had little effect on commitment-to-die in the absence of BAF. CsA did not have trophic effects on BAF-saved cells, but did block the decrease in mitochondrial membrane potential. These data suggest that PTP opening is a critical event in caspase-independent, nonapoptotic (but not caspase-dependent, apoptotic) death of NGF-deprived rat sympathetic neurons.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclosporina/farmacología , Inhibidores Enzimáticos/farmacología , Mitocondrias/metabolismo , Neuronas/citología , Animales , Apoptosis/fisiología , Inhibidores de la Calcineurina , Inhibidores de Caspasas , Caspasas/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/fisiología , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Factor de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Ganglio Cervical Superior/citología
11.
J Cell Biol ; 162(2): 245-56, 2003 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-12876275

RESUMEN

Mitochondrial release of cytochrome c in apoptotic cells activates caspases, which execute apoptotic cell death. However, the events themselves that culminate in caspase activation can have deleterious effects because caspase inhibitor-saved cells ultimately die in a caspase-independent manner. To determine what events may underlie this form of cell death, we examined bioenergetic changes in sympathetic neurons deprived of NGF in the presence of a broad-spectrum caspase inhibitor, boc-aspartyl-(OMe)-fluoromethylketone. Here, we report that NGF-deprived, boc-aspartyl-(OMe)-fluoromethylketone-saved neurons rely heavily on glycolysis for ATP generation and for survival. Second, the activity of F0F1 contributes to caspase-independent death, but has only a minor role in the maintenance of mitochondrial membrane potential, which is maintained primarily by electron transport. Third, permeability transition pore inhibition by cyclosporin A attenuates NGF deprivation-induced loss of mitochondrial proteins, suggesting that permeability transition pore opening may have a function in regulating the degradation of mitochondria after cytochrome c release. Identification of changes in caspase inhibitor-saved cells may provide the basis for rational strategies to augment the effectiveness of the therapeutic use of postmitochondrial interventions.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Factor de Crecimiento Nervioso/fisiología , Neuronas/efectos de los fármacos , Adenosina Trifosfato/análisis , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/fisiología , Células Cultivadas , Grupo Citocromo c/metabolismo , Transporte de Electrón/efectos de los fármacos , Glucólisis/efectos de los fármacos , Potenciales de la Membrana , Mitocondrias/metabolismo , Mitocondrias/fisiología , Modelos Biológicos , Factor de Crecimiento Nervioso/farmacología , Neuronas/metabolismo , Neuronas/ultraestructura , Fosforilación Oxidativa/efectos de los fármacos , ATPasas de Translocación de Protón/efectos de los fármacos , Ratas
12.
J Cell Biol ; 157(3): 441-53, 2002 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-11980919

RESUMEN

Trophic factor deprivation (TFD)-induced apoptosis in sympathetic neurons requires macromolecular synthesis-dependent BAX translocation, cytochrome c (cyt c) release, and caspase activation. Here, we report the contributions of other intrinsic and extrinsic pathway signals to these processes. Sympathetic neurons expressed all antiapoptotic BCL-2 proteins examined, yet expressed only certain BH3-only and multidomain proapoptotic BCL-2 family members. All coexpressed proapoptotic proteins did not, however, exhibit functional redundancy or compensatory expression, at least in the Bax-/-, Bak-/-, Bim-/-, Bid-/-, and Bad-/- neurons examined. Although the subcellular distribution or posttranslational modification of certain BCL-2 proteins changed with TFD, neither transcriptional nor posttranslational mechanisms regulated the expression or subcellular localization of BID, BAD, or BAK in this paradigm. Despite modest induction of Fas and FasL expression, Fas-mediated signaling did not contribute to TFD-induced apoptosis in sympathetic neurons. Similar findings were obtained with K+ withdrawal-induced apoptosis in cerebellar granule neurons, a model for activity-dependent neuronal survival in the CNS. Thus, expression alone does not guarantee functional redundancy (or compensation) among BCL-2 family members, and, at least in some cells, extrinsic pathway signaling and certain BH3-only proteins (i.e., BID and BAD) do not contribute to BAX-dependent cyt c release or apoptosis caused by TFD.


Asunto(s)
Apoptosis , Neuronas/citología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Animales , Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Grupo Citocromo c/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Factores de Crecimiento Nervioso/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ratas , Sistema Nervioso Simpático/citología , Proteína Destructora del Antagonista Homólogo bcl-2 , Proteína Letal Asociada a bcl
13.
Neuron ; 33(2): 261-73, 2002 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-11804573

RESUMEN

During postnatal development, sympathetic neurons lose their dependence upon NGF for survival but continue to require NGF for soma and process growth and for development of a mature neurotransmitter phenotype. Although c-Ret is expressed in sympathetic neurons during this period, its function in these transitional processes is unclear. The level of Ret phosphorylation markedly increased with postnatal age in SCG neurons in vitro and in vivo. Postnatal Ret phosphorylation did not require either GFLs or GFR(alpha) coreceptors. Instead, NGF promoted age-dependent Ret phosphorylation with delayed kinetics both in vitro and in vivo. Functionally, maximal NGF-dependent trophism of mature sympathetic neurons required Ret, but not GFR(alpha) coreceptors. Therefore, NGF promotes phosphorylation of the heterologous RTK Ret resulting in augmented growth, metabolism, and gene expression.


Asunto(s)
Proteínas de Drosophila , Factor de Crecimiento Nervioso/fisiología , Factores de Crecimiento Nervioso/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Transducción de Señal/fisiología , Sistema Nervioso Simpático/fisiología , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Animales Recién Nacidos/metabolismo , Supervivencia Celular/fisiología , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Neurturina , Fosforilación , Proteínas Proto-Oncogénicas c-ret , Ratas , Ratas Sprague-Dawley , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/metabolismo , Sistema Nervioso Simpático/citología
14.
Neuron ; 44(4): 623-36, 2004 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-15541311

RESUMEN

The GDNF family ligands signal through a receptor complex composed of a ligand binding subunit, GFRalpha, and a signaling subunit, the RET tyrosine kinase. GFRalphas are expressed not only in RET-expressing cells, but also in cells lacking RET. A body of evidence suggests that RET-independent GFRalphas are important for (1) modulation of RET signaling in a non-cell-autonomous fashion (trans-signaling) and (2) regulation of NCAM function. To address the physiological significance of these roles, we generated mice specifically lacking RET-independent GFRalpha1. These mice exhibited no deficits in regions where trans-signaling has been implicated in vitro, including enteric neurons, motor neurons, kidney, and regenerating nerves. Furthermore, no abnormalities were found in the olfactory bulb, which requires proper NCAM function for its formation and is putatively a site of GDNF-GFRalpha-NCAM signaling. Thus RET-independent GFRalpha1 is dispensable for organogenesis and nerve regeneration in vivo, indicating that trans-signaling and GFRalpha-dependent NCAM signaling play a minor role physiologically.


Asunto(s)
Regeneración Nerviosa/fisiología , Organogénesis/fisiología , Animales , Sistema Nervioso Central/embriología , Sistema Nervioso Entérico/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Riñón/embriología , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Músculo Esquelético/inervación , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Bulbo Olfatorio/metabolismo , Sistema Nervioso Periférico/embriología , Sistema Nervioso Periférico/patología , Proteínas Proto-Oncogénicas , Proteínas Proto-Oncogénicas c-ret , Proteínas Tirosina Quinasas Receptoras
15.
Neuron ; 38(6): 899-914, 2003 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-12818176

RESUMEN

Trophic factor deprivation (TFD) activates c-Jun N-terminal kinases (JNKs), culminating in coordinate AP1-dependent transactivation of the BH3-only BCL-2 proteins BIM(EL) and HRK, which in turn are critical for BAX-dependent cytochrome c release, caspase activation, and apoptosis. Here, we report that TFD caused not only induction but also phosphorylation of BIM(EL). Mitochondrially localized JNKs but not upstream activators, like mixed-lineage kinases (MLKs) or mitogen-activated protein kinase kinases (MKKs), specifically phosphorylated BIM(EL) at Ser65, potentiating its proapoptotic activity. Inhibition of the JNK pathway attenuated BIM(EL) expression, prevented BIM(EL) phosphorylation, and abrogated TFD-induced apoptosis. Conversely, activation of this pathway promoted BIM(EL) expression and phosphorylation, causing BIM- and BAX-dependent cell death. Thus, JNKs regulate the proapoptotic activity of BIM(EL) during TFD, both transcriptionally and posttranslationally.


Asunto(s)
Apoptosis , Proteínas Portadoras/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas de la Membrana , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neuronas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Proto-Oncogénicas/fisiología , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis , Proteína 11 Similar a Bcl2 , Proteínas Portadoras/química , Proteínas Portadoras/genética , Células Cultivadas , Cerebelo , Activación Enzimática , Regulación de la Expresión Génica , Sueros Inmunes/farmacología , MAP Quinasa Quinasa 4 , Ratones , Mitocondrias/enzimología , Datos de Secuencia Molecular , Mutagénesis , Factor de Crecimiento Nervioso/inmunología , Factor de Crecimiento Nervioso/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Serina/genética , Transducción de Señal , Relación Estructura-Actividad , Ganglio Cervical Superior , Transfección , Proteína X Asociada a bcl-2
16.
J Neurosci ; 26(10): 2777-87, 2006 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-16525057

RESUMEN

The receptor tyrosine kinase (RTK) Ret is activated by the formation of a complex consisting of ligands such as glial cell line-derived neurotrophic factor (GDNF) and glycerophosphatidylinositol-anchored coreceptors termed GFRalphas. During activation, Ret translocates into lipid rafts, which is critical for functional responses to GDNF. We found that Ret was rapidly ubiquitinated and degraded in sympathetic neurons when activated with GDNF, but, unlike other RTKs that are trafficked to lysosomes for degradation, Ret was degraded predominantly by the proteasome. After GDNF stimulation, the majority of ubiquitinated Ret was located outside of lipid rafts and Ret was lost predominantly from nonraft membrane domains. Consistent with the predominance of Ret degradation outside of rafts, disruption of lipid rafts in neurons did not alter either the GDNF-dependent ubiquitination or degradation of Ret. GDNF-mediated survival of sympathetic neurons was inhibited by lipid raft depletion, and this inhibitory effect of raft disruption on GDNF-mediated survival was reversed if Ret degradation was blocked via proteasome inhibition. Therefore, lipid rafts sequester Ret away from the degradation machinery located in nonraft membrane domains, such as Cbl family E3 ligases, thereby sustaining Ret signaling.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Microdominios de Membrana/metabolismo , Neuronas/citología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-ret/fisiología , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Western Blotting/métodos , Supervivencia Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Ganglios Simpáticos/citología , Inmunoprecipitación/métodos , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oligopéptidos/farmacología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-ret/clasificación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Sales de Tetrazolio , Tiazoles , Factores de Tiempo , Ubiquitina/metabolismo
17.
J Neurosci ; 26(43): 11230-8, 2006 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-17065462

RESUMEN

Glial cell-line derived neurotrophic factor (GDNF)-mediated RET tyrosine kinase signaling is implicated in the survival of several PNS and CNS neuronal populations that are important in the pathogenesis of several disorders including Parkinson's disease and drug addiction. However, it has been difficult to study these processes and the physiological importance of this pathway in adult mice because of the neonatal lethality of Gdnf and Ret null mice. We report successful creation of RET conditional reporter mice to investigate postnatal physiologic roles of RET and monitor the fate of RET-expressing cell types. To delete RET specifically in dopaminergic neurons and determine the physiologic requirement of RET in the maintenance of substantia nigra compacta (SNC) and ventral tegmental area (VTA), we bred the RET conditional mice with mice that specifically express Cre from the dopamine transporter (Dat) locus. A detailed morphometric and biochemical analysis including dopaminergic neuron number and size in SNC and VTA, and fiber density in the striatum and nucleus accumbens, and dopamine levels indicate that RET is not required for providing global trophic support to midbrain dopaminergic neurons in adult mice. Furthermore, RET deficiency in these neurons does not cause major sensorimotor abnormalities. Hence our results support the idea that RET signaling is not critical for the normal physiology of the SNC and VTA in adult mice.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/fisiología , Mesencéfalo/fisiología , Neuronas/fisiología , Proteínas Proto-Oncogénicas c-ret/fisiología , Animales , Dopamina/fisiología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/deficiencia , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Transducción de Señal/fisiología , Sustancia Negra/citología , Sustancia Negra/fisiología , Área Tegmental Ventral/citología , Área Tegmental Ventral/fisiología
18.
Trends Neurosci ; 25(8): 412-7, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12127758

RESUMEN

Lipid rafts are plasma membrane microdomains rich in cholesterol and sphingolipids, which provide a particularly ordered lipid environment. Rafts are enriched in glycosylphosphatidylinositol (GPI)-anchored proteins, as well as proteins involved in signal transduction and intracellular trafficking. In neurons, lipid rafts act as platforms for the signal transduction initiated by several classes of neurotrophic factors, including neurotrophins and glial-derived neurotrophic factor (GDNF)-family ligands. Emerging evidence also indicates that such rafts are important for neuronal cell adhesion, axon guidance and synaptic transmission. Thus, lipid rafts are structurally unique components of plasma membranes, crucial for neural development and function.


Asunto(s)
Sistema Nervioso Central/metabolismo , Lípidos de la Membrana/metabolismo , Microdominios de Membrana/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Animales , Adhesión Celular/fisiología , Sistema Nervioso Central/ultraestructura , Conos de Crecimiento/metabolismo , Conos de Crecimiento/ultraestructura , Humanos , Factores de Crecimiento Nervioso/metabolismo , Neuronas/ultraestructura , Neurotransmisores/metabolismo , Transmisión Sináptica/fisiología
19.
J Neurosci ; 22(18): 8018-27, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12223555

RESUMEN

Sympathetic neuronal apoptosis after nerve growth factor (NGF) deprivation requires the activation of two events: a protein synthesis-dependent, Bax-dependent release of mitochondrial cytochrome c and a protein synthesis-independent, Bax-independent development of competence. Unlike in most cells, cytosolic cytochrome c is not sufficient to induce cell death in NGF-maintained sympathetic neurons but can do so in neurons that have developed competence. We report that cytosolic cytochrome c-induced apoptosis in competent sympathetic neurons is completely dependent on caspase-9. In addition, the neuroprotective agents KCl and chlorophenylthio-cAMP are potent inhibitors of the development-of-competence pathway in NGF-deprived sympathetic neurons. We also find that the development of competence is reversible. Readdition of NGF reverses competence, and neurons can regain their resistance to cytosolic cytochrome c. Importantly, we examined the mechanism of development of competence and report that the inability of cytochrome c to activate caspases in NGF-maintained sympathetic neurons can be overcome with exogenous Smac that inhibits the inhibitor of apoptosis (IAP) family of proteins. Microinjection of cytochrome c and Smac, but neither alone, induces rapid cell death in NGF-maintained neurons. These data suggest that development of competence may be the result of the loss of the function of one or more members of the IAP family of caspase inhibitors that is needed before cytochrome c can activate caspases and induce cell death in neurons.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/farmacología , Caspasas/metabolismo , Proteínas Mitocondriales/farmacología , Neuronas/efectos de los fármacos , Proteínas , Proteínas Proto-Oncogénicas c-bcl-2 , Sistema Nervioso Simpático/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Caspasa 9 , Caspasas/deficiencia , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Grupo Citocromo c/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/metabolismo , Proteínas Inhibidoras de la Apoptosis , Proteínas de Insectos/antagonistas & inhibidores , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Microinyecciones , Factor de Crecimiento Nervioso/farmacología , Neuronas/citología , Neuronas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Cloruro de Potasio/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/efectos de los fármacos , Estaurosporina/farmacología , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/metabolismo , Proteína X Asociada a bcl-2
20.
J Neurosci ; 22(10): 3953-62, 2002 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12019314

RESUMEN

The mechanisms by which trophic factors bring about spinal motor neuron (MN) survival and regulate their number during development are not well understood. We have developed an organotypic slice culture model for the in vitro study of the trophic requirements and cell death pathways in MNs of postnatal day 1-2 mice. Both lateral motor column (LMC) and medial motor column (MMC) neurons died within 72 hr when grown in serum-free medium without trophic factors. Brain-derived neurotrophic factor, ciliary neurotrophic factor, and 8-(4-chlorophenylthio)-cAMP promoted the survival of a proportion of the neurons, but glial cell line-derived neurotrophic factor (GDNF) was the most effective trophic factor, supporting approximately 60% of MNs for 1 week in culture. Homozygous deficiency for bax, a proapoptotic member of the Bcl-2 family, saved the same proportion of neurons as GDNF, suggesting that GDNF alone was sufficient to maintain all "rescuable" MNs for at least 1 week. Analysis of MN survival in GFRalpha-1(-/-) mice demonstrated that the trophic effect of GDNF was completely mediated by its preferred coreceptor, GDNF family receptor alpha-1 (GFRalpha-1). None of the other GDNF family ligands supported significant MN survival, suggesting that there is little ligand-coreceptor cross talk within the slice preparation. Although MN subtypes can be clearly defined by both anatomical distribution and ontogenetic specification, the pattern of trophic factor responsiveness of neurons from the MMC was indistinguishable from that seen in the LMC. Thus, in contrast to all other factors and drugs studied to date, GDNF is likely to be a critical trophic agent for all early postnatal MN populations.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , AMP Cíclico/análogos & derivados , Proteínas de Drosophila , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/fisiología , Factores de Crecimiento Nervioso , Proteínas del Tejido Nervioso/farmacología , Proteínas Proto-Oncogénicas c-bcl-2 , Médula Espinal/fisiología , Animales , Animales Recién Nacidos , Apoptosis , Factor Neurotrófico Derivado del Encéfalo/farmacología , Supervivencia Celular/fisiología , Factor Neurotrófico Ciliar/farmacología , Medio de Cultivo Libre de Suero/farmacología , AMP Cíclico/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Homocigoto , Técnicas In Vitro , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Mutantes , Neuronas Motoras/citología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ret , Proteínas Tirosina Quinasas Receptoras/deficiencia , Proteínas Tirosina Quinasas Receptoras/metabolismo , Médula Espinal/citología , Tionucleótidos/farmacología , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA