Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 97(Pt B): 169-178, 2017 01.
Article in English | MEDLINE | ID: mdl-27063797

ABSTRACT

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.


Subject(s)
Nerve Growth Factors/administration & dosage , Neurodegenerative Diseases/therapy , Neuroprotective Agents/administration & dosage , Animals , Clinical Trials as Topic , Humans , Nerve Growth Factors/genetics
2.
Neurobiol Dis ; 97(Pt B): 156-168, 2017 01.
Article in English | MEDLINE | ID: mdl-27063798

ABSTRACT

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.


Subject(s)
Nerve Growth Factors/administration & dosage , Neurodegenerative Diseases/therapy , Neuroprotective Agents/administration & dosage , Animals , Clinical Trials as Topic , Humans , Nerve Growth Factors/genetics
3.
J Neurosci ; 33(43): 17095-107, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24155314

ABSTRACT

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.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/pathology , Gait , Animals , Diphtheria Toxin/genetics , Diphtheria Toxin/toxicity , Dopamine/deficiency , Dopamine Antagonists/pharmacology , Dopamine Plasma Membrane Transport Proteins/deficiency , Dopamine Plasma Membrane Transport Proteins/genetics , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Haloperidol/pharmacology , Levodopa/pharmacology , Mesencephalon/drug effects , Mesencephalon/pathology , Mice , Mice, Transgenic , Mutation
4.
Neurobiol Dis ; 58: 38-48, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23631873

ABSTRACT

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.


Subject(s)
Adenoviridae/genetics , Corpus Striatum/metabolism , Nerve Growth Factors/administration & dosage , Neurodegenerative Diseases/prevention & control , Neuroprotective Agents/administration & dosage , Substantia Nigra/metabolism , Adrenergic Agents/toxicity , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Genetic Vectors/physiology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neuroprotective Agents/metabolism , Oxidopamine/toxicity , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase
5.
Neurobiol Dis ; 44(1): 38-52, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21704161

ABSTRACT

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.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Neurturin/metabolism , Parkinson Disease/therapy , Substantia Nigra/metabolism , Animals , Behavior, Animal/physiology , Diet , Gene Dosage , Gene Targeting , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/genetics , Green Fluorescent Proteins , Immunohistochemistry , Male , Neurons/metabolism , Neurturin/adverse effects , Rats , Rats, Sprague-Dawley , Risk Assessment , Tyrosine 3-Monooxygenase/metabolism , Weight Gain/genetics , Weight Gain/physiology , Weight Loss/physiology
6.
Mov Disord ; 26(1): 27-36, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21322017

ABSTRACT

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.


Subject(s)
Corpus Striatum/metabolism , Genetic Therapy/methods , MPTP Poisoning/therapy , Neurturin/therapeutic use , Parkinson Disease/therapy , Aged , Animals , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Functional Laterality , Gene Expression Regulation/drug effects , Humans , MPTP Poisoning/chemically induced , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Macaca mulatta , Male , Middle Aged , Neurturin/genetics , Neurturin/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Tyrosine 3-Monooxygenase/metabolism
7.
J Neurosci ; 30(11): 3983-94, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-20237269

ABSTRACT

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.


Subject(s)
Nociceptors/physiology , Proto-Oncogene Proteins c-ret/physiology , Signal Transduction/physiology , Animals , Cell Survival/genetics , Cell Survival/physiology , Female , Formaldehyde/administration & dosage , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , NAV1.8 Voltage-Gated Sodium Channel , Nociceptors/cytology , Pain Measurement/methods , Proto-Oncogene Proteins c-ret/deficiency , Proto-Oncogene Proteins c-ret/genetics , Signal Transduction/genetics , Sodium Channels/deficiency , Sodium Channels/genetics
9.
J Neurosci ; 28(16): 4123-35, 2008 Apr 16.
Article in English | MEDLINE | ID: mdl-18417692

ABSTRACT

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.


Subject(s)
Neurturin/physiology , Proto-Oncogene Proteins c-ret/metabolism , Retina/physiology , Animals , Mice , Mice, Knockout , Neurturin/genetics , Proto-Oncogene Proteins c-ret/deficiency , Proto-Oncogene Proteins c-ret/genetics , Retina/cytology , Retina/growth & development , Signal Transduction/genetics , Signal Transduction/physiology
10.
J Neurochem ; 102(4): 1184-94, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17663753

ABSTRACT

Glial cell derived neurotrophic factor (GDNF)-dependent receptor tyrosine kinase RET activity is required for proper development of the nervous system and genitourinary tract. Loss-of-function mutations in RET are associated with enteric nervous system abnormalities (Hirschsprung disease) and renal deficits (Potter's syndrome), whereas activating mutations lead to hereditary cancer syndromes (multiple endocrine neoplasia type 2A and type 2B). RET activation is crucial for the proper regulation of a variety of cellular processes including cell migration, proliferation and neurite outgrowth. By analyzing a series of RET mutants we found that Y1062 was critical for stimulating GDNF-mediated proliferation as well as proliferation stimulated by GDNF-independent oncogenic forms of RET. Studies using small interfering RNA driven by lentivirus to knock-down expression of particular adaptor proteins that interact with RET phospho-Y1062, demonstrated that only Src-homology 2 and growth factor receptor binding protein 2 were necessary for RET-mediated proliferation by wild type and oncogenic forms of RET. Interestingly, we discovered that Y1062 was also required for GDNF-stimulated neurite outgrowth. However, small interfering RNAs to either Src-homology 2 or growth factor receptor binding protein 2 or a panel of other adaptor proteins known to interact with RET Y1062 were incapable of blocking GDNF-stimulated neurite formation, indicating that differential use of intracellular adaptors is responsible for regulating alternative RET-stimulated cellular events such as proliferation versus a differentiation response like neurite outgrowth.


Subject(s)
Cell Proliferation , GRB2 Adaptor Protein/metabolism , Neurites/physiology , Proto-Oncogene Proteins c-ret/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Transformed , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Activation/genetics , Fibroblasts/cytology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Mutation/physiology , Neurites/drug effects , PC12 Cells , Proto-Oncogene Proteins c-ret/genetics , RNA, Small Interfering/pharmacology , Rats , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tyrosine/genetics
11.
J Neurochem ; 100(5): 1169-76, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17241133

ABSTRACT

Nerve growth factor (NGF) is required for the trophic maintenance of postnatal sympathetic neurons. A significant portion of the growth-promoting activity of NGF is from NGF-dependent phosphorylation of the heterologous receptor tyrosine kinase, Ret. We found that NGF applied selectively to distal axons of sympathetic neurons maintained in compartmentalized cultures activated Ret located in these distal axons. Inhibition of either proteasomal or lysosomal degradation pathways mimicked the effect of NGF on Ret activation. Likewise, NGF inhibited the degradation of Ret induced by glial cell line-derived neurotrophic factor-dependent activation, a process that requires ubiquitination and proteasomal degradation. NGF induced the accumulation of autophosphorylated Ret predominantly in the plasma membrane, in contrast to GDNF, which promoted the internalization of activated Ret. An accretion of monoubiquitinated, but not polyubiquitinated, Ret occurred in NGF-treated neurons, in contrast to glial cell line-derived neurotrophic factor that promoted the robust polyubiquitination of Ret. Thus, NGF stimulates Ret activity in mature sympathetic neurons by inhibiting the ongoing ubiquitin-mediated degradation of Ret before its internalization and polyubiquitination.


Subject(s)
Nerve Growth Factor/physiology , Neurons/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Superior Cervical Ganglion/cytology , Ubiquitin/metabolism , Animals , Axons/metabolism , Cell Membrane/metabolism , Cells, Cultured , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Lysosomes/metabolism , Phosphorylation , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Rats
12.
J Neurosci ; 26(43): 11230-8, 2006 Oct 25.
Article in English | MEDLINE | ID: mdl-17065462

ABSTRACT

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.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/physiology , Mesencephalon/physiology , Neurons/physiology , Proto-Oncogene Proteins c-ret/physiology , Animals , Dopamine/physiology , Dopamine Plasma Membrane Transport Proteins/deficiency , Dopamine Plasma Membrane Transport Proteins/genetics , Glial Cell Line-Derived Neurotrophic Factor/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Signal Transduction/physiology , Substantia Nigra/cytology , Substantia Nigra/physiology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology
13.
J Neurochem ; 99(1): 218-25, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16911582

ABSTRACT

In neurons, DNA damage induces protein synthesis-dependent apoptosis mediated by the mitochondrial intrinsic cell-death pathway. Signal transduction cascades activated by genotoxic stress upstream of the mitochondria are largely unknown. We identified protein kinase D (PKD) as one of the earliest markers of neuronal DNA damage. Phosphorylation of the PKD-activation domain could be detected within 15 min of genotoxic stress and was concurrent with ataxia telangiectasia-mutated (ATM) activation. PKD stimulation was selective to DNA damage and did not occur with other stress stimuli examined. In vivo, both young and adult rats showed increased levels of phosphorylated PKD in neuronal tissues after injection of DNA-toxin etoposide. These results indicate that PKD activation is an early neuronal response to DNA damage, suggesting that signaling downstream of PKD may be critical for neuronal survival after genotoxic stress.


Subject(s)
DNA Damage , Neurons/pathology , Protein Kinase C/metabolism , Animals , Animals, Newborn , Ataxia Telangiectasia Mutated Proteins , Biomarkers/metabolism , Cell Cycle Proteins/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Enzyme Activation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Sympathetic Nervous System/physiology , Tumor Suppressor Proteins/metabolism
14.
J Neurosci ; 26(10): 2777-87, 2006 Mar 08.
Article in English | MEDLINE | ID: mdl-16525057

ABSTRACT

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.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/metabolism , Membrane Microdomains/metabolism , Neurons/cytology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-ret/physiology , Signal Transduction/physiology , Animals , Animals, Newborn , Blotting, Western/methods , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Activation/drug effects , Ganglia, Sympathetic/cytology , Immunoprecipitation/methods , Membrane Proteins/metabolism , Nerve Growth Factor/pharmacology , Neurons/drug effects , Neurons/metabolism , Oligopeptides/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Proto-Oncogene Proteins c-ret/classification , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tetrazolium Salts , Thiazoles , Time Factors , Ubiquitin/metabolism
15.
Genes Dev ; 20(3): 321-33, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452504

ABSTRACT

Molecular mechanisms that lead to congenital anomalies of kidneys and the lower urinary tract (CAKUT) are poorly understood. To elucidate the molecular basis for signaling specificity of GDNF-mediated RET signaling in kidney development, we characterized mice that exclusively express either the human RET9 or RET51 isoform, or express these isoforms with individual mutations in docking tyrosines for PTB and SH2-domain-containing adaptors Src (Y981), PLCgamma (Y1015), and Shc (Y1062). Our results provide evidence for differential and isoform-specific roles of these docking sites in murine kidney development. Homozygous Ret(RET9) and Ret(RET51) mice were viable and show normally developed kidneys, indicating redundant roles of human RET isoforms in murine kidney development. In the context of the RET51 isoform, only mutation of the docking Tyr 1015 (Y1015F) resulted in severe renal anomalies. These included bilateral megaureters and multicystic kidneys that were caused by supernumerary ureteric buds that fail to separate from the wolffian duct as well as decreased branching morphogenesis. Similar kidney and ureter defects were observed in RET9(Y1015F) mice that contain the Y1015F mutation in the RET9 isoform. Interestingly, loss of RET9(Y1062)-mediated AKT/MAPK activation resulted in renal agenesis or kidney rudiments, whereas mutation of this residue in RET51 had no obvious effect on AKT/MAPK activity and renal development. These results reveal novel roles of key RET-dependent signaling pathways in embryonic kidney development and provide murine models and new insights into the molecular basis for CAKUT.


Subject(s)
Kidney/embryology , Proto-Oncogene Proteins c-ret/physiology , Tyrosine/metabolism , Animals , Apoptosis , Gonads/physiology , Humans , Kidney/abnormalities , Kidney/growth & development , Mice , Mice, Mutant Strains , Morphogenesis/genetics , Morphogenesis/physiology , Mutation , Phenylalanine/genetics , Phenylalanine/metabolism , Phospholipase C gamma/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Signal Transduction/genetics , Tyrosine/genetics , Ureter/physiology
16.
J Cell Biol ; 170(3): 401-11, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061693

ABSTRACT

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.


Subject(s)
Apoptosis/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/physiology , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/physiology , Animals , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , DNA Damage , Enzyme Activation , Ganglia, Spinal/cytology , Gene Expression Regulation , Genes, jun , JNK Mitogen-Activated Protein Kinases/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutation , Nerve Growth Factor/deficiency , Phosphorylation , Protein Subunits/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
18.
Neurobiol Dis ; 19(1-2): 183-93, 2005.
Article in English | MEDLINE | ID: mdl-15837573

ABSTRACT

The study tested the hypothesis that transplantation of embryonic stem (ES) cells into rat cortex after a severe focal ischemia would promote structural repair and functional recovery. Overexpression of the human anti-apoptotic gene bcl-2 in ES cells was tested for increasing survival and differentiation of transplanted cells and promoting functional benefits. Mouse ES cells, pretreated with retinoic acid to induce differentiation down neural lineages, were transplanted into the post-infarct brain cavity of adult rats 7 days after 2-h occlusion of the middle cerebral artery (MCA). Over 1-8 weeks after transplantation, the lesion cavity filled with ES cell-derived cells that expressed markers for neurons, astrocytes, oligodendrocytes, and endothelial cells. ES cell-derived neurons exhibited dendrite outgrowth and formed a neuropil. ES cell-transplanted animals exhibited enhanced functional recovery on neurological and behavioral tests, compared to control animals injected with adult mouse cortical cells or vehicle. Furthermore, transplantation with ES cells overexpressing Bcl-2 further increased the survival of transplanted ES cells, neuronal differentiation, and functional outcome. This study supports that ES cell transplantation and gene modification may have values for enhancing recovery after stroke.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Ischemic Attack, Transient/physiopathology , Ischemic Attack, Transient/therapy , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Recovery of Function/physiology , Stem Cell Transplantation/methods , Animals , Cell Line , Ischemic Attack, Transient/metabolism , Male , Mice , Proto-Oncogene Proteins c-bcl-2/physiology , Rats , Rats, Wistar , Stem Cells/metabolism
19.
Exp Neurol ; 191 Suppl 1: S68-79, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15629763

ABSTRACT

The carotid body is a neural crest-derived neuroendocrine organ that detects the oxygen level in blood and regulates ventilation. Unlike many other neural crest derivatives, the trophic factors mediating survival and differentiation of neuroendocrine cells of the carotid body are unknown. Given that many neural crest derivatives rely on the glial cell line-derived neurotrophic factor (GDNF) family of ligands (GFLs) for survival and function, we undertook an analysis of the carotid body as a potential site of GFL action. RET and GDNF family receptor alphas (GFRalpha) 1-3 are expressed in the developing carotid body as detected by RT-PCR and immunocytochemistry. mRNA for GDNF, and artemin (ARTN) were also present. In vitro, treatment with GDNF, neurturin (NRTN), or ARTN, individually or in combination, produced an increase in the number and length of processes of the Type-I glomus cells of the carotid body [embryonic day-17 (E17) rats]. However, GFLs alone or in combination had no effect on glomus cell survival in either postnatal day-1 (P1) or E17 carotid body cultures. These results suggest that one or more GFLs may have a role in carotid body function. In addition, the results of this study suggest that endogenous or exogenous GFLs may enhance target innervation by carotid body transplants.


Subject(s)
Carotid Body/metabolism , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Carotid Body/cytology , Carotid Body/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Ligands , Nerve Growth Factors/pharmacology , Neurites/drug effects , Neurturin , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine 3-Monooxygenase/biosynthesis
20.
Exp Neurol ; 191(1): 65-76, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15589513

ABSTRACT

Embryonic stem (ES) cells are genetically manipulable pluripotential cells that can be differentiated in vitro into neurons, oligodendrocytes, and astrocytes. Given their potential utility as a source of replacement cells for the injured nervous system and the likelihood that transplantation interventions might include co-application of growth factors, we examined the effects of neurotrophin and GDNF family ligands on the survival and excitotoxic vulnerability of ES cell-derived neurons (ES neurons) grown in vitro. ES cells were differentiated down a neural lineage in vitro using the 4-/4+ protocol (Bain et al., Dev Biol 168:342-57, 1995). RT-PCR demonstrated expression of receptors for neurotrophins and GDNF family ligands in ES neural lineage cells. Neuronal expression of GFRalpha1, GFRalpha2, and ret was confirmed by immunocytochemistry. Exposure to 30-100 ng/ml GDNF or neurturin (NRTN) resulted in activation of ret. Addition of NT-3 and GDNF did not increase cell division but did increase the number of neurons in the cultures 7 days after plating. Pretreatment with NT-3 enhanced the vulnerability of ES neurons to NMDA-induced death (100 microM NMDA for 10 min) and enhanced the NMDA-induced increase in neuronal [Ca2+]i, but did not alter expression of NMDA receptor subunits NR2A or NR2B. In contrast, pretreatment with GDNF reduced the vulnerability of ES neurons to NMDA-induced death while modestly enhancing the NMDA-induced increase in neuronal [Ca2+]i. These findings demonstrate that the response of ES-derived neurons to neurotrophins and GDNF family ligands is largely similar to that of other cultured central neurons.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Nerve Growth Factors/physiology , Neurons/drug effects , Neurons/physiology , Stem Cells/drug effects , Stem Cells/physiology , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Embryo, Mammalian , Glial Cell Line-Derived Neurotrophic Factor , Ligands , Mice , N-Methylaspartate/pharmacology , Nerve Growth Factors/metabolism , Neurons/cytology , Receptors, Nerve Growth Factor/biosynthesis , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...