Skip to main content
Erschienen in: Molecular Neurodegeneration 1/2012

Open Access 01.12.2012 | Research article

Wld S but not Nmnat1 protects dopaminergic neurites from MPP+neurotoxicity

verfasst von: Jo Ann V Antenor-Dorsey, Karen L O'Malley

Erschienen in: Molecular Neurodegeneration | Ausgabe 1/2012

Abstract

Background

The Wld S mouse mutant ("Wallerian degeneration-slow") delays axonal degeneration in a variety of disorders including in vivo models of Parkinson's disease. The mechanisms underlying Wld S -mediated axonal protection are unclear, although many studies have attributed Wld S neuroprotection to the NAD+-synthesizing Nmnat1 portion of the fusion protein. Here, we used dissociated dopaminergic cultures to test the hypothesis that catalytically active Nmnat1 protects dopaminergic neurons from toxin-mediated axonal injury.

Results

Using mutant mice and lentiviral transduction of dopaminergic neurons, the present findings demonstrate that Wld S but not Nmnat1, Nmnat3, or cytoplasmically-targeted Nmnat1 protects dopamine axons from the parkinsonian mimetic N-methyl-4-phenylpyridinium (MPP+). Moreover, NAD+ synthesis is not required since enzymatically-inactive Wld S still protects. In addition, NAD+ by itself is axonally protective and together with Wld S is additive in the MPP+ model.

Conclusions

Our data suggest that NAD+ and Wld S act through separate and possibly parallel mechanisms to protect dopamine axons. As MPP+ is thought to impair mitochondrial function, these results suggest that Wld S might be involved in preserving mitochondrial health or maintaining cellular metabolism.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1750-1326-7-5) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JAD participated in experimental design, carried out all the experiments described and drafted the manuscript. KOM was involved in the design of experiments and production of the manuscript. Both authors participated in revising and editing the final manuscript. The final manuscript was read and approved by both authors.

Background

Parkinson's disease (PD) is the second most common neurodegenerative disorder in the U.S., affecting 1-2% of people over the age of 55. Characterized by loss of dopaminergic neurons in the substantia nigra (SN) [1, 2], the cardinal motor symptoms of PD include resting tremor, bradykinesia, rigidity, and abnormal gait [3, 4]. Another characteristic of PD is its late onset and progressive nature. Symptoms appear after 50-70% [5, 6] of striatal dopamine has been depleted and 30-50% [7, 8] of the nigral dopaminergic cells have died. Such studies suggest that the extent of striatal dopamine depletion is better correlated with the severity of PD symptoms than the loss of dopaminergic neurons in the SN [7].
Data from PD-linked genetic mutations also support the notion that axonal pathology and/or dysfunction occurs prior to the loss of dopaminergic cell bodies. For example, α-synuclein pathology is seen in neurites before it is observed in PD-associated cell bodies [3, 9]. α-synuclein mutants accumulate in the cell soma when overexpressed in cortical neurons, suggesting impaired axonal transport as well [10]. Moreover, transgenic models expressing the PD-linked mutant gene leucine rich repeat kinase 2 (LRRK2) also exhibit pronounced axonal loss and pathology prior to cell body loss [11]. In addition, genetic mutations in other PD-linked genes such as Parkin, an E3 ligase [12], and PINK1 (PTEN-induced putative kinase 1 protein) a mitochondrially-targeted kinase, also alter axonal transport [13, 14]. Collectively, these findings have led to the idea that nigral neurons degenerate through a "dying back" axonopathy where degeneration starts in the distal axon and proceeds over time towards the cell body.
Environmental toxins known to mimic PD such as rotenone and MPP+ also disrupt axonal function. These factors not only inhibit mitochondrial Complex I activity, but also de-polymerize microtubules leading to axon fragmentation and decreased synaptic function [1517]. Moreover, MPP+ can directly inhibit axon transport in the squid axoplasm [18] and DA neurons [19]. Thus, results from PD-associated environmental and genetic factors support an early, critical role for axonal impairment in PD.
Recent data suggest that the Wallerian degeneration slow fusion protein (Wld S ) can delay axonal degeneration about 10-fold from a wide variety of genetic and toxin-inducing stimuli in the peripheral nervous system [20]. Wld S also blocks axon degeneration in several central nervous system (CNS) models of degeneration including animal models of PD [21, 22]. For example, we previously found that Wld S rescues 85% of dopaminergic axons for at least 7 days post MPTP treatment in vivo [23]. Because no other mutation or drug protects axons as robustly as Wld S , understanding how the Wld S fusion protein is able to prevent axon degeneration is the first step towards defining an intervention that would leave axons intact.
Wld S is a chimeric protein composed of the first 70 amino acids of the ubiquitination factor E4b (Ube4b) followed by an 18-amino acid linker region and then the entire coding sequence for nicotinamide mononucleotide adenylyltransferase (Nmnat1), a nicotinamide adenine dinucleotide (NAD+) synthesizing enzyme [24, 25]. Most studies suggest that catalytically active Nmnat1 is necessary for axonal protection [26, 27], hence, exogenous addition of NAD+ has been reported to delay Wallerian degeneration in response to axotomy in dorsal root ganglion (DRG) cells [28]. In Drosophila, however, the picture is more complex in that Avery et al. [29], showed that Nmnat enzymatic activity is required following axotomy whereas Zhai et al. [30] found that Nmnat does not need its catalytic domain to protect axons. In this model [30], as well as in a new study demonstrating that Nmnat also protects dendrites [31], Nmnat exhibits a separate chaperone-like activity which protects axons and dendrites [30, 32].
Inasmuch as most studies have been done in peripheral model systems and because we have previously shown that Wld S protects dopaminergic terminal fields from MPTP in vivo, we used a dissociated midbrain culture system to determine the mechanism of Wld S -mediated neurite protection in dopamine neurons. Here, we show that, regardless of its enzymatic activity, the entire Wld S sequence is needed for the Wld S ' neuroprotective phenotype in dopaminergic neurons. Our data also illustrate that NAD+ has a neuroprotective effect that is different from Wld S -mediated protection.

Results

Wld S protects cell bodies and neurites from MPP+

Previously we have shown that dopaminergic terminal fields but not cell bodies of Wld S mice are protected against MPTP injury [23]. To confirm and extend these observations in a more tractable system, we utilized dissociated cultures of midbrain neurons in which 1-5% of the total cells plated are dopaminergic [33]. Results show that cultures from Wld S mice exhibited significant protection of neurites not seen in wild type cultures after MPP+ treatment (Figure 1A, C). Moreover, dopaminergic cell death from MPP+ treatment was also attenuated in Wld S cultures, unlike those seen in vivo (Figure 1A, B). Thus Wld S can effectively protect neurites (dendrites and axons) as well as cell bodies from known PD mimetics in vitro.

Cytoplasmic Wld S protects cell bodies and neurites against MPP+

Recent studies have reported that the localization of Wld S influences its neuroprotective effect. Babetto et. al. have reported that a cytoplasmic version of the Wld S protein (cyto Wld S ) confers a higher level of protection than the native form of Wld S [34]. In addition, Sasaki et. al. have reported that cytoplasmic Nmnat1 (cyto Nmnat1) and Nmnat3, which is primarily localized in the mitochondria, also confer a higher level of protection than Nmnat1 [35, 36]. To test whether cytoplasmic localization of Wld S rescued or changed the level of protection seen with nuclear Wld S , primary dopaminergic neurons were prepared from wild type and cyto Wld S mice, treated with MPP+, and analyzed as described. Results show that cyto Wld S mice exhibited a similar level of protection of dopaminergic cell bodies and neurites as seen in Wld S mice (Figure 2). Therefore, as reported for peripheral model systems and certain CNS paradigms [37], redirecting most of WldS from the nucleus to the cytoplasm protects processes equally well.

Nmnat1 does not protect against MPP+toxicity

Many studies, especially in peripheral model systems, have shown that Nmnat1 can at least partially mimic the effects of Wld S [26, 27]. To determine whether this is true in dopaminergic neurons, we transduced primary midbrain cultures from wild type animals with either GFP, Nmnat1, the 70 amino acid fragment of Ube4b encoded within the Wld S gene, or the entire Wld S coding region using lentiviral vectors expressing GFP [27] (Figure 3). We also tested the effects of Nmnat3, cytoplasmic Nmnat1, and enzymatically inactive Nmnat1 (Nmnat1 (W170A)) [28, 38] (Figure 3D, E and Additional File 1 Figure S1C, D). Immunofluorescence and western blotting was done to confirm that transductions led to similar expression levels in dissociated cultures (Figure 3B, C, Additional File 1 Figure S1B, D-E). Despite equivalent levels of transgene expression, only neurites transduced with the entire coding sequence of Wld S were protected from MPP+ injury (Figure 3D, E).
Because many studies have suggested that Nmnat and in particular cyto Nmnat or axonally targeted Nmnat can be as effective as Wld S in protecting axons from mechanical or toxic insults, we used DRG cultures as a positive control [34, 39]. Consistent with those studies, both Wld S and cyto Nmnat rescued DRG neurites from the neurotoxin, vincristine, whereas the GFP-only and inactive Wld S virus did not (Figure 4). Taken together, these data confirm previous results showing that cyto Nmnat is necessary and sufficient to save DRG neurites. In contrast, only Wld S but not cyto Nmnat, Nmnat1, or Nmnat3 was able to protect dopaminergic neurons from the neurotoxic effects of MPP+.

Inactive Wld S protects cell bodies and neurites against MPP+

To corroborate the hypothesis that Nmnat1 does not protect dopaminergic neurons from MPP+, we transduced dissociated primary midbrain cultures with enzymatically inactive Wld S (W258A; [27]). In contrast to our own results in DRG cultures (Figure 4) as well as results published by others using this same construct [27], the inactive Wld S plasmid was as effective as NAD+-synthesizing Wld S animals in protecting dopaminergic cell bodies and neurites against MPP+ injury (Figure 5). Therefore, the entire Wld S chimeric protein, but not its NAD+-synthesizing activity, is required for neuroprotection of dopaminergic neurons.

NAD+ protects cell bodies and neurites against MPP+

Previous studies have shown that NAD+ itself can be neuroprotective [27]. Although Nmnat1 by itself did not recapitulate the neuroprotective effect of Wld S on dopaminergic neurons, we tested whether NAD+ or one of its precursors (Figure 6A) rescued cell bodies or neurites from MPP+ treatment. Therefore, dissociated dopaminergic wild type cultures were pretreated with either 1 mM of NAD+, nicotinamide mononucleotide (NMN), or nicotinic acid mononucleotide (NaMN) 24 hours before MPP+ treatment. Both NAD+ and NMN but not NaMN protected cell bodies and neurites against MPP+ toxicity (Figure 6B, C). These findings together with the results showing that catalytically-inactive Wld S was able to protect dopamine neurons (Figure 5) but catalytically active Nmnat did not (Figure 3F) suggest that different pathways are being invoked in response to MPP+ toxicity.

Sirt1 is not responsible for the NAD-mediated protection of cell bodies and neurites against MPP+

Previous studies in DRG neurons have attributed the protective phenotype of Wld S to its action on the Nmnat1-NAD+-Sirt1 pathway [27]. To test the involvement of Sirt1, we prepared dissociated dopaminergic cultures from Sirt1 knockout mice. Following 24 hour pretreatment with 1 mM NAD+ or vehicle control, cultures were exposed to MPP+. Consistent with the notion that NAD+ is not acting through Sirt1 but rather through a different mechanism, NAD+ protected cell bodies and neurites in Sirt1 knockout cultures from MPP+ toxicity (Figure 7).

NAD+ and Wld S effects are additive

The data described above suggest that Wld S is acting through a separate possibly parallel pathway from that of NAD+ in dopaminergic neurons. If so, then adding NAD+ to Wld S cultures will enhance the neuroprotective phenotype of Wld S . To see whether the NAD+ effect overlapped with Wld S or was additive, dissociated dopaminergic cultures were prepared from wild type and Wld S mice and pre-treated with and without NAD+ as previously described. Both NAD+ and Wld S alone exhibited similar levels of cell body and neurite protection (Figure 8). However, NAD+ together with Wld S generated significantly higher levels of protection suggesting this is an additive process (Figure 8). To test whether these effects were maximal, additional cultures were treated with 5 mM NAD+; no significant differences in neuroprotection were observed when compared with the lower dose of 1 mM NAD+ (Figure 8D). These findings demonstrate that NAD+ and Wld S are additive in the MPP+ model suggesting that they are acting through separate and possibly parallel neuroprotective mechanisms.

Discussion

The mechanism(s) by which Wld S protects axons is still unclear. Peripheral model studies underscore the role of Nmnat and its product, NAD+, in protecting axons from various injuries whereas few central nervous system studies have been done. Using cellular, molecular and pharmacological tools, the present findings show that the chimeric Wld S gene product plays a critical role in protecting dopaminergic processes, one not dependent upon Nmnat activity. Specifically, neither Nmnat, cytoplasmically-targeted Nmnat, nor Nmnat 3 were able to prevent toxicity associated with the dopaminergic toxin MPP+ whereas, akin to previous reports [27, 28], cyto Nmnat protected DRG axons from known axonal toxins. In contrast, Wld S , cytoplasmically-expressed Wld S , and Wld S with an inactive Nmnat domain, all significantly protected dopaminergic neurites from toxin-mediated loss. Despite the inability of Nmnat to protect dopamine processes, NAD+ and its precursor, Nmn, were neuroprotective. As Wld S and NAD+ were additive in this model system, current results suggest that these protectants act through separate, possibly parallel pathways. This is in agreement with previous findings by Wishart et. al. (2008) [40] showing that Wld S increases expression of cell cycle-related genes through both NAD+-dependent and independent pathways. Thus, NAD+ or its derivatives as well as Wld S and its targets protect dopamine processes and may aid in the development of therapeutics preserving the connections and circuitry important in PD.
The role of Nmnat and NAD+ in recapitulating the full effect of Wld S has been controversial. In vitro studies have shown that overexpression of Nmnat1 by itself protects axons from many mechanical, genetic or toxin-induced injuries [20, 41]. In contrast, transgenic animals expressing nuclear Nmnat1 did not replicate the effects of Wld S [42, 43] whereas cytoplasmically [39] or axonally targeted Nmnat1 [34] were equally if not more effective. Thus, site of action plays a role in Nmnat1's effectiveness [20]. These data together with findings showing that the first 16 N-terminal amino acids of the Wld S gene product are required for full Wld S protection [26], possibly by redistributing enough Wld S to cytoplasmic or axonal compartments, are consistent with the notion that both the N-terminal portion of Wld S and Nmnat1 are necessary for full axonal protection [20].
The importance of Nmnat catalytic activity is reflected in several mutational studies in which Nmnat's active sites have been disrupted and neuroprotection was lost [26, 27, 29, 43]. Moreover, NAD+ itself and/or some of its biosynthetic precursors, protect against axonal degeneration in peripheral model systems as well as in experimental autoimmune encephalomyelitis (EAE); [28, 44], ischemia [45, 46], Alzheimer's disease [47], and PD [4850]. In at least one study however, addition of NAD+ was not effective [42]. Moreover, Drosophila Nmnat (dNmnat) did not require enzymatic activity for axon protection against insults such as excitotoxicity, polyglutamine-induced dysfunction, or mechanical injury [32] leading to the suggestion that dNmnat may perform a chaperone-like function [30]. Indeed, structural studies of various Nmnats have revealed characteristic similarities to known chaperones such as UspA and Hsp100 [51]. Consistent with this notion, dNmnat was recently shown to function as a stress protein in response to heat shock, hypoxia, and the mitochondrial Complex I toxin, paraquat [52]. However, in dopaminergic neurons, Nmnat1 does not seem to function as either an axonal protectant or a chaperone.
Studies have indicated that MPP+ can block electron transport by acting at the same site as the Complex I inhibitor, rotenone, leading to the production of free radical species and a loss of ATP production [5355]. MPP+ affects other processes as well including the rapid release of dopamine from vesicular stores [56, 57]; depolymerization of microtubules [16, 58]; induction of autophagy [19, 59], and the rapid loss of mitochondrial membrane potential and reduction in mitochondrial motility in dopamine axons [19]. Since many of these effects involve mitochondrial function, conceivably the Wld S gene product is involved in preserving mitochondrial health or maintaining homeostatic control. Recently, Barrientos et al. reported that Wld S is able to regulate the mitochondrial permeability transition pore (PTP) preventing, amongst other things, calcium release, ATP loss, oxidative stress and release of proteins involved in axonal degeneration [60]. This is consistent with Wishart et al. (2007) showing that synaptosomes isolated from Wld S versus wild type animals expressed higher levels of various mitochondrial proteins including the PTP protein, VDAC2 [61]. Barrientos et al. suggested that Wld S is part of a regulatory cascade that also involves JNK activation upstream of PTP opening [60]. Although JNK is a known regulator of axon degeneration in DRGs [62], it has been reported to not play a role in 6-OHDA-mediated degeneration of the striatum [7]. In addition, we have recently showed that the JNK inhibitor, SP600125, did not prevent MPP+ effects on dopaminergic mitochondria [19]. Thus diverse, unknown, regulatory steps appear to mediate Wld S effects in dopamine axons.
Given its role as a ubiquitous cofactor, NAD+ influences many cellular decisions such as DNA damage repair [63] and transcriptional regulation and differentiation [64]. Earlier studies suggested that increased NAD+ levels led to SIRT activation which, in turn, activated a transcription factor that induced genes involved in neuroprotection [27, 32]. Although an attractive hypothesis, subsequent studies using SIRT1 knock out animals did not support this notion for DRG neurons [65], or as in the present study, for dopaminergic neurons (Figure 8).
Unlike what we have reported in vivo [23], dopaminergic cell death from MPP+ treatment was also attenuated in Wld S cultures (Figure 1). Given that Wld S is known to protect axons and synapses from injury, it is possible that it can also indirectly protect cell bodies. Similar indirect effects on cell bodies have been reported by Gillingwater et. al. (2004) in both the caudate nucleus and hippocampus of Wld S mice following transient global ischemia [66]. More recently, a cytoplasm-targeted Nmnat transgenic mouse protected cell bodies and processes from NMDA-mediated excitotoxicity [37]. Authors of the latter study speculate that Nmnat can potentially influence a common pathway, albeit one not tied to caspase 3 activation, in certain neurons. Perhaps a similar pathway is activated in other systems as well since Wld S also protects motoneuron cell bodies in a mouse model of progressive motor neuropathy [67].
Why are results in dopamine neurons different than other systems? Because many of the studies published have been performed in peripheral model systems with dramatically over-expressed protein, there may be neuronal-specific or expression level-related effects that might account for the differences. For example, Wld S has shown protection in several central nervous system models, but few have been further tested with only Nmnat1 even in dissociated neuronal models [37]. Then too, dopaminergic axons may have intrinsic differences that contribute to the Wld S effect. For instance, dopamine neurons have fewer [68], smaller and slower mitochondria than non-dopaminergic neurons [19]. Moreover, dopamine neurons produce a neurotransmitter prone to oxidation [69], exhibit a greater dependence on L-type Ca2+ channels with subunits that result in deleterious amounts of intracellular calcium and ensuing mitochondrial dysfunction [70], and extend long, thin lightly-myelinated processes which are selectively vulnerable in PD [71]. This suggests that dopaminergic neurons may be more vulnerable to insults that affect mitochondrial function. Given that enzymatically inactive Wld S is able to protect dopaminergic, but not DRG neurons, it is possible that Wld S also protects mitochondria in a manner independent of its NAD+-synthesizing ability. This as yet unknown function of Wld S may be unmasked in dopaminergic neurons due to their unique phenotype. In contrast, NAD+ may contribute more to Wld S -mediated protection in non-DA neurons.

Conclusions

In support of our previous in vivo study showing that Wld S protects dopaminergic terminal fields from MPTP, the current results demonstrate in dissociated dopamine cultures that the entire Wld S sequence is needed for axonal protection, regardless of its NAD+-synthesizing activity. Indeed, NAD+ and Wld S act through separate, possibly parallel, mechanisms to protect dopamine axons. As MPP+ is thought to impair mitochondrial function, in agreement with other studies, our results suggest that Wld S might be involved in preserving mitochondrial health or maintaining cellular metabolism. Given that Parkinson's disease is the second most common neurodegenerative disorder, our findings support the idea that studies expanding therapeutic efforts towards maintaining connections as well as saving the cell body will help in developing better interventions for PD.

Materials and methods

Cell culture and toxin treatment

For primary midbrain cultures, the ventral mesencephalon was removed from embryonic day 14 (E14) murine embryos as previously described [33, 72]. Wild-type (C57/Bl6) and Wld S (C57Bl/OlaHsd-WldS) mice were ordered from Harlan (Bichester, UK). Sirt1 knockout mice were obtained from Dr. Christian Sheline (Louisiana State University - Health Science Center, New Orleans, LA). Cyto Wld S mice were obtained from Dr. Michael Coleman (Babraham Institute, UK) [73]. Animals were treated in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals. All procedures were approved by the Washington University School of Medicine animal experimentation committee. Plates were pre-coated overnight with 0.2 mg/ml poly-D-lysine (Sigma-Aldrich, St. Louis, MO). Cells were plated at a density of approximately 125,000 cells/cm2 and maintained in serum-free Neurobasal medium (Invitrogen, Carlsbad, CA) supplemented with 1× B27 supplement (Invitrogen), 0.5 mM L-glutamine (Sigma-Aldrich), and 0.01 μg/ml streptomycin plus 100 U penicillin. Half of the culture medium was replaced with fresh Neurobasal medium after 5 days in vitro (DIV). Cultures were pretreated with 1 mM NAD+, 1 mM NMN, 1 mM nicotinic acid mononucleotide (NaMN), or a comparable volume of vehicle 24 hours before toxin treatment. Cultures were treated with either 1 μM 1-methyl-4-phenylpyridinium (MPP+), the active metabolite of MPTP or vehicle on DIV 7. Dorsal root ganglion (DRG) cells were obtained from E14 murine embryos as previously described [74]. Cells were plated on coverslips precoated with 0.1 mg/ml poly-L-ornithine (Invitrogen) and 32 μg/ml laminin-1 (Invitrogen) and maintained in DRG media which consisted of Eagle Minimal Essential Media (Invitrogen) supplemented with chick embryo extract (Invitrogen), 10% fetal calf serum (Invitrogen), 50 ng/ml Nerve Growth Factor (Harlan Biosciences, Madison, WI) and 50 U/ml penicillin-50 g/ml streptomycin. Half of the culture medium was replaced with fresh DRG medium after DIV 5. After transduction with lentivirus on DIV 2, DRG cultures were treated with 0.4 μM vincristine or vehicle on DIV 7. NAD+, NMN, NaMN, MPP+, and vincristine were all obtained from Sigma-Aldrich.

Lentiviral infection of dopaminergic neurons

The lentiviral expression plasmids FUGW, FCIV-WldS, FCIV-Nmnat1, FCIV-Ube4b, FCIV-Nmnat3, FCIV- Nmnat1(W170A), FCIV-cytNmnat1, and FCIV-WldS(W258A) were obtained from Dr. Jeffrey Milbrandt (Washington University, Saint Louis). Lentiviruses expressing transgenes were generated by the Hope Center for Neurological Disorders Viral Core (Washington University, Saint Louis). For infection of DRG and primary midbrain neurons, 50 μl lentivirus (105 infectious units/μl) was added to the well of a 7-mm dish containing approximately 70,000 neurons on DIV 2. Transduced primary midbrain and DRG neurons were treated with MPP+ and vincristine, respectively, on DIV 7. Viral infection and transgene expression was monitored using the GFP reporter via fluorescent microscopy.

Immunocytochemistry

Primary dopaminergic cultures and DRGs were plated in 7 mm microwell plates (MatTek Corp., Ashland, MA). Cells treated with MPP+ were fixed with 4% paraformaldehyde (PFA) in PBS after 48 hours. Cultures were stained with sheep polyclonal anti-tyrosine hydroxylase (TH) (Novus Biologicals, Littleton, CO) and Cy3 α-sheep (Molecular Probes, Carlsbad, CA). Localization of cytoplasmic Wld S was confirmed using rabbit Wld S antibody (gift of M.P. Coleman) and Alexa488 α-rabbit (Molecular Probes). TH+ cells and neurites were counted using unbiased stereological methods (Stereo Investigator, MicroBrightField, Williston, VT). DRG cultures treated with vincristine were subsequently stained with mouse acetylated tubulin (Sigma-Aldrich) and Cy3 α-mouse (Molecular Probes). Neurites were counted as described above. All images were acquired by confocal microscopy (Olympus Fluoview 500, Olympus, Center Valley, PA) and processed in ImageJ (NIH).

Western Blotting

Primary midbrain cultures were plated in 48-well plates and transduced with the transgene of interest as described above. Lysates were collected in RIPA buffer (150 mM NaCl, 1% Nonidet P-40, 0.5% NaDoc, 0.1% SDS, 50 mM Tris pH 8.0) with protease inhibitor mixture (Roche, Mannheim, Germany) and incubated on ice for 30 minutes. Insoluble cell debris was removed by centrifugation and the protein concentration of each cell lysate was determined by Bradford protein assay (BioRad, Hercules, CA). Equal amounts of protein were run on SDS-polyacrylamide gels and transferred to polyvinylidene diflouride (PVDF) membranes (BioRad). PVDF membranes were probed with either rabbit Wld S antibody or chicken polyclonal anti-GFP antibody (Aves Labs, Tigard, OR). As a control, PVDF membranes were also probed with goat polyclonal anti-HRP60 antibody (Santa Cruz Biotechnology, Santa Cruz, CA). The secondary antibodies used were either a HRP-linked rabbit antibody or HRP-linked anti-chicken antibody and a HRP-linked anti-goat antibody (Jackson Immunoresearch, West Grove, PA). Membranes were developed with enhanced chemiluminescence (Amersham Biosciences), imaged with either a Storm PhosphorImager (Molecular Dynamics) or a ChemiDoc XRS System (Bio-Rad, Hercules, CA) and band intensities were determined using ImageQuant software (Amersham Biosciences).

Quantification of Cells and Neurites

TH+ cells and neurites were counted using unbiased stereological methods [75] (Stereo Investigator (MicroBrightfield, Williston, VT), in combination with a Zeiss Axioplan2 microscope (Thornwood, NY) and an Optronics Microfire camera. The number of counting sites necessary to achieve a coefficient of error < 0.1 was determined by preliminary experiments. The total number of TH+ cell bodies was calculated using the Fractionator function on Stereo Investigator by dividing the estimated number of cells by the estimated volume (μm3) of the dish sampled. Using the Petrimetrics function on Stereo Investigator, TH+ neurites intersecting the boundary of the Petrimetric probe were counted. Neurite length was derived by dividing the total estimated neurite length (μm) by the estimated volume (μm3) of the dish sampled

Statistical analysis

GraphPad Prism software (San Diego, CA) was used for statistical analysis. All data was collected from a minimum of three independent experiments done in triplicate. The significance of effects between control and experimental conditions was determined by a Student t-test or one-way ANOVA with Bonferroni Multiple Comparisons tests.

Acknowledgements

This work was supported by National Institutes of Health Grants NS39084 (K.L.O.) and National Institutes of Health Neuroscience Blueprint Core Grant NS057105 to Washington University. This work was also supported by the Bakewell Family Foundation. We thank Steven K. Harmon for technical support and Drs. Michael Coleman, Jeffrey Milbrandt, Christian Sheline, Valeria Cavalli and Jeong Sook Kim-Han for materials and helpful discussions.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JAD participated in experimental design, carried out all the experiments described and drafted the manuscript. KOM was involved in the design of experiments and production of the manuscript. Both authors participated in revising and editing the final manuscript. The final manuscript was read and approved by both authors.
Literatur
1.
Zurück zum Zitat Dauer W, Przedborski S: Parkinson's disease: mechanisms and models. Neuron. 2003, 39: 889-909. 10.1016/S0896-6273(03)00568-3.CrossRefPubMed Dauer W, Przedborski S: Parkinson's disease: mechanisms and models. Neuron. 2003, 39: 889-909. 10.1016/S0896-6273(03)00568-3.CrossRefPubMed
2.
Zurück zum Zitat Blum D, Torch S, Lambeng N, Nissou M, Benabid AL, Sadoul R, Verna JM: Molecular pathways involved in the neurotoxicity of 6-OHDA, dopamine and MPTP: contribution to the apoptotic theory in Parkinson's disease. Prog Neurobiol. 2001, 65: 135-172. 10.1016/S0301-0082(01)00003-X.CrossRefPubMed Blum D, Torch S, Lambeng N, Nissou M, Benabid AL, Sadoul R, Verna JM: Molecular pathways involved in the neurotoxicity of 6-OHDA, dopamine and MPTP: contribution to the apoptotic theory in Parkinson's disease. Prog Neurobiol. 2001, 65: 135-172. 10.1016/S0301-0082(01)00003-X.CrossRefPubMed
3.
Zurück zum Zitat Braak H, Ghebremedhin E, Rub U, Bratzke H, Del Tredici K: Stages in the development of Parkinson's disease-related pathology. Cell Tissue Res. 2004, 318: 121-134. 10.1007/s00441-004-0956-9.CrossRefPubMed Braak H, Ghebremedhin E, Rub U, Bratzke H, Del Tredici K: Stages in the development of Parkinson's disease-related pathology. Cell Tissue Res. 2004, 318: 121-134. 10.1007/s00441-004-0956-9.CrossRefPubMed
4.
Zurück zum Zitat Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F: Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci. 1973, 20: 415-455. 10.1016/0022-510X(73)90175-5.CrossRefPubMed Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F: Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci. 1973, 20: 415-455. 10.1016/0022-510X(73)90175-5.CrossRefPubMed
5.
Zurück zum Zitat Scherman D, Desnos C, Darchen F, Pollak P, Javoy-Agid F, Agid Y: Striatal dopamine deficiency in Parkinson's disease: role of aging. Ann Neurol. 1989, 26: 551-557. 10.1002/ana.410260409.CrossRefPubMed Scherman D, Desnos C, Darchen F, Pollak P, Javoy-Agid F, Agid Y: Striatal dopamine deficiency in Parkinson's disease: role of aging. Ann Neurol. 1989, 26: 551-557. 10.1002/ana.410260409.CrossRefPubMed
6.
Zurück zum Zitat Riederer P, Wuketich S: Time course of nigrostriatal degeneration in parkinson's disease. A detailed study of influential factors in human brain amine analysis. J Neural Transm. 1976, 38: 277-301. 10.1007/BF01249445.CrossRefPubMed Riederer P, Wuketich S: Time course of nigrostriatal degeneration in parkinson's disease. A detailed study of influential factors in human brain amine analysis. J Neural Transm. 1976, 38: 277-301. 10.1007/BF01249445.CrossRefPubMed
7.
Zurück zum Zitat Cheng HC, Ulane CM, Burke RE: Clinical progression in Parkinson disease and the neurobiology of axons. Ann Neurol. 2010, 67: 715-725. 10.1002/ana.21995.PubMedCentralCrossRefPubMed Cheng HC, Ulane CM, Burke RE: Clinical progression in Parkinson disease and the neurobiology of axons. Ann Neurol. 2010, 67: 715-725. 10.1002/ana.21995.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Fearnley JM, Lees AJ: Ageing and Parkinson's disease: substantia nigra regional selectivity. Brain. 1991, 114 (Pt 5): 2283-2301.CrossRefPubMed Fearnley JM, Lees AJ: Ageing and Parkinson's disease: substantia nigra regional selectivity. Brain. 1991, 114 (Pt 5): 2283-2301.CrossRefPubMed
9.
Zurück zum Zitat Kramer ML, Schulz-Schaeffer WJ: Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J Neurosci. 2007, 27: 1405-1410. 10.1523/JNEUROSCI.4564-06.2007.CrossRefPubMed Kramer ML, Schulz-Schaeffer WJ: Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J Neurosci. 2007, 27: 1405-1410. 10.1523/JNEUROSCI.4564-06.2007.CrossRefPubMed
10.
Zurück zum Zitat Saha AR, Hill J, Utton MA, Asuni AA, Ackerley S, Grierson AJ, Miller CC, Davies AM, Buchman VL, Anderton BH, Hanger DP: Parkinson's disease alpha-synuclein mutations exhibit defective axonal transport in cultured neurons. J Cell Sci. 2004, 117: 1017-1024. 10.1242/jcs.00967.CrossRefPubMed Saha AR, Hill J, Utton MA, Asuni AA, Ackerley S, Grierson AJ, Miller CC, Davies AM, Buchman VL, Anderton BH, Hanger DP: Parkinson's disease alpha-synuclein mutations exhibit defective axonal transport in cultured neurons. J Cell Sci. 2004, 117: 1017-1024. 10.1242/jcs.00967.CrossRefPubMed
11.
Zurück zum Zitat Li Y, Liu W, Oo TF, Wang L, Tang Y, Jackson-Lewis V, Zhou C, Geghman K, Bogdanov M, Przedborski S, Beal MF, Burke RE, Li C: Mutant LRRK2(R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson's disease. Nat Neurosci. 2009, 12: 826-828. 10.1038/nn.2349.PubMedCentralCrossRefPubMed Li Y, Liu W, Oo TF, Wang L, Tang Y, Jackson-Lewis V, Zhou C, Geghman K, Bogdanov M, Przedborski S, Beal MF, Burke RE, Li C: Mutant LRRK2(R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson's disease. Nat Neurosci. 2009, 12: 826-828. 10.1038/nn.2349.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N: Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998, 392: 605-608. 10.1038/33416.CrossRefPubMed Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N: Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998, 392: 605-608. 10.1038/33416.CrossRefPubMed
13.
Zurück zum Zitat Weihofen A, Thomas KJ, Ostaszewski BL, Cookson MR, Selkoe DJ: Pink1 forms a multiprotein complex with Miro and Milton, linking Pink1 function to mitochondrial trafficking. Biochemistry. 2009, 48: 2045-2052. 10.1021/bi8019178.PubMedCentralCrossRefPubMed Weihofen A, Thomas KJ, Ostaszewski BL, Cookson MR, Selkoe DJ: Pink1 forms a multiprotein complex with Miro and Milton, linking Pink1 function to mitochondrial trafficking. Biochemistry. 2009, 48: 2045-2052. 10.1021/bi8019178.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Mortiboys H, Thomas KJ, Koopman WJ, Klaffke S, Abou-Sleiman P, Olpin S, Wood NW, Willems PH, Smeitink JA, Cookson MR, Bandmann O: Mitochondrial function and morphology are impaired in parkin-mutant fibroblasts. Ann Neurol. 2008, 64: 555-565. 10.1002/ana.21492.PubMedCentralCrossRefPubMed Mortiboys H, Thomas KJ, Koopman WJ, Klaffke S, Abou-Sleiman P, Olpin S, Wood NW, Willems PH, Smeitink JA, Cookson MR, Bandmann O: Mitochondrial function and morphology are impaired in parkin-mutant fibroblasts. Ann Neurol. 2008, 64: 555-565. 10.1002/ana.21492.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Ren Y, Liu W, Jiang H, Jiang Q, Feng J: Selective vulnerability of dopaminergic neurons to microtubule depolymerization. J Biol Chem. 2005, 280: 34105-34112. 10.1074/jbc.M503483200.CrossRefPubMed Ren Y, Liu W, Jiang H, Jiang Q, Feng J: Selective vulnerability of dopaminergic neurons to microtubule depolymerization. J Biol Chem. 2005, 280: 34105-34112. 10.1074/jbc.M503483200.CrossRefPubMed
16.
Zurück zum Zitat Cappelletti G, Pedrotti B, Maggioni MG, Maci R: Microtubule assembly is directly affected by MPP(+)in vitro. Cell Biol Int. 2001, 25: 981-984. 10.1006/cbir.2001.0772.CrossRefPubMed Cappelletti G, Pedrotti B, Maggioni MG, Maci R: Microtubule assembly is directly affected by MPP(+)in vitro. Cell Biol Int. 2001, 25: 981-984. 10.1006/cbir.2001.0772.CrossRefPubMed
17.
Zurück zum Zitat Cartelli D, Ronchi C, Maggioni MG, Rodighiero S, Giavini E, Cappelletti G: Microtubule dysfunction precedes transport impairment and mitochondria damage in MPP+ -induced neurodegeneration. J Neurochem. 2010, 115: 247-258. 10.1111/j.1471-4159.2010.06924.x.CrossRefPubMed Cartelli D, Ronchi C, Maggioni MG, Rodighiero S, Giavini E, Cappelletti G: Microtubule dysfunction precedes transport impairment and mitochondria damage in MPP+ -induced neurodegeneration. J Neurochem. 2010, 115: 247-258. 10.1111/j.1471-4159.2010.06924.x.CrossRefPubMed
18.
Zurück zum Zitat Morfini G, Pigino G, Opalach K, Serulle Y, Moreira JE, Sugimori M, Llinas RR, Brady ST: 1-Methyl-4-phenylpyridinium affects fast axonal transport by activation of caspase and protein kinase C. Proc Natl Acad Sci USA. 2007, 104: 2442-2447. 10.1073/pnas.0611231104.PubMedCentralCrossRefPubMed Morfini G, Pigino G, Opalach K, Serulle Y, Moreira JE, Sugimori M, Llinas RR, Brady ST: 1-Methyl-4-phenylpyridinium affects fast axonal transport by activation of caspase and protein kinase C. Proc Natl Acad Sci USA. 2007, 104: 2442-2447. 10.1073/pnas.0611231104.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Kim-Han JS, Antenor-Dorsey JA, O'Malley KL: The Parkinsonian Mimetic, MPP+, Specifically Impairs Mitochondrial Transport in Dopamine Axons. J Neurosci. 2011, 31: 7212-7221. 10.1523/JNEUROSCI.0711-11.2011.PubMedCentralCrossRefPubMed Kim-Han JS, Antenor-Dorsey JA, O'Malley KL: The Parkinsonian Mimetic, MPP+, Specifically Impairs Mitochondrial Transport in Dopamine Axons. J Neurosci. 2011, 31: 7212-7221. 10.1523/JNEUROSCI.0711-11.2011.PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Coleman MP, Freeman MR: Wallerian degeneration, wld(s), and nmnat. Annu Rev Neurosci. 2010, 33: 245-267. 10.1146/annurev-neuro-060909-153248.CrossRefPubMed Coleman MP, Freeman MR: Wallerian degeneration, wld(s), and nmnat. Annu Rev Neurosci. 2010, 33: 245-267. 10.1146/annurev-neuro-060909-153248.CrossRefPubMed
21.
Zurück zum Zitat Mi W, Beirowski B, Gillingwater TH, Adalbert R, Wagner D, Grumme D, Osaka H, Conforti L, Arnhold S, Addicks K, Wada K, Ribchester RR, Coleman MP: The slow Wallerian degeneration gene, WldS, inhibits axonal spheroid pathology in gracile axonal dystrophy mice. Brain. 2005, 128: 405-416.CrossRefPubMed Mi W, Beirowski B, Gillingwater TH, Adalbert R, Wagner D, Grumme D, Osaka H, Conforti L, Arnhold S, Addicks K, Wada K, Ribchester RR, Coleman MP: The slow Wallerian degeneration gene, WldS, inhibits axonal spheroid pathology in gracile axonal dystrophy mice. Brain. 2005, 128: 405-416.CrossRefPubMed
22.
Zurück zum Zitat Sajadi A, Schneider BL, Aebischer P: Wlds-mediated protection of dopaminergic fibers in an animal model of Parkinson disease. Curr Biol. 2004, 14: 326-330.CrossRefPubMed Sajadi A, Schneider BL, Aebischer P: Wlds-mediated protection of dopaminergic fibers in an animal model of Parkinson disease. Curr Biol. 2004, 14: 326-330.CrossRefPubMed
23.
Zurück zum Zitat Hasbani DM, O'Malley KL: Wld(S) mice are protected against the Parkinsonian mimetic MPTP. Exp Neurol. 2006, 202: 93-99. 10.1016/j.expneurol.2006.05.017.CrossRefPubMed Hasbani DM, O'Malley KL: Wld(S) mice are protected against the Parkinsonian mimetic MPTP. Exp Neurol. 2006, 202: 93-99. 10.1016/j.expneurol.2006.05.017.CrossRefPubMed
24.
Zurück zum Zitat Mack TG, Reiner M, Beirowski B, Mi W, Emanuelli M, Wagner D, Thomson D, Gillingwater T, Court F, Conforti L, Fernando FS, Tarlton A, Andressen C, Addicks K, Magni G, Ribchester RR, Perry VH, Coleman MP: Wallerian degeneration of injured axons and synapses is delayed by a Ube4b/Nmnat chimeric gene. Nat Neurosci. 2001, 4: 1199-1206.CrossRefPubMed Mack TG, Reiner M, Beirowski B, Mi W, Emanuelli M, Wagner D, Thomson D, Gillingwater T, Court F, Conforti L, Fernando FS, Tarlton A, Andressen C, Addicks K, Magni G, Ribchester RR, Perry VH, Coleman MP: Wallerian degeneration of injured axons and synapses is delayed by a Ube4b/Nmnat chimeric gene. Nat Neurosci. 2001, 4: 1199-1206.CrossRefPubMed
25.
Zurück zum Zitat Coleman MP, Conforti L, Buckmaster EA, Tarlton A, Ewing RM, Brown MC, Lyon MF, Perry VH: An 85-kb tandem triplication in the slow Wallerian degeneration (Wlds) mouse. Proc Natl Acad Sci USA. 1998, 95: 9985-9990. 10.1073/pnas.95.17.9985.PubMedCentralCrossRefPubMed Coleman MP, Conforti L, Buckmaster EA, Tarlton A, Ewing RM, Brown MC, Lyon MF, Perry VH: An 85-kb tandem triplication in the slow Wallerian degeneration (Wlds) mouse. Proc Natl Acad Sci USA. 1998, 95: 9985-9990. 10.1073/pnas.95.17.9985.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Conforti L, Wilbrey A, Morreale G, Janeckova L, Beirowski B, Adalbert R, Mazzola F, Di Stefano M, Hartley R, Babetto E, Smith T, Gilley J, Billington RA, Genazzani AA, Ribchester RR, Magni G, Coleman M: Wld S protein requires Nmnat activity and a short N-terminal sequence to protect axons in mice. J Cell Biol. 2009, 184: 491-500. 10.1083/jcb.200807175.PubMedCentralCrossRefPubMed Conforti L, Wilbrey A, Morreale G, Janeckova L, Beirowski B, Adalbert R, Mazzola F, Di Stefano M, Hartley R, Babetto E, Smith T, Gilley J, Billington RA, Genazzani AA, Ribchester RR, Magni G, Coleman M: Wld S protein requires Nmnat activity and a short N-terminal sequence to protect axons in mice. J Cell Biol. 2009, 184: 491-500. 10.1083/jcb.200807175.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Araki T, Sasaki Y, Milbrandt J: Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science. 2004, 305: 1010-1013. 10.1126/science.1098014.CrossRefPubMed Araki T, Sasaki Y, Milbrandt J: Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science. 2004, 305: 1010-1013. 10.1126/science.1098014.CrossRefPubMed
28.
Zurück zum Zitat Sasaki Y, Araki T, Milbrandt J: Stimulation of nicotinamide adenine dinucleotide biosynthetic pathways delays axonal degeneration after axotomy. J Neurosci. 2006, 26: 8484-8491. 10.1523/JNEUROSCI.2320-06.2006.CrossRefPubMed Sasaki Y, Araki T, Milbrandt J: Stimulation of nicotinamide adenine dinucleotide biosynthetic pathways delays axonal degeneration after axotomy. J Neurosci. 2006, 26: 8484-8491. 10.1523/JNEUROSCI.2320-06.2006.CrossRefPubMed
29.
Zurück zum Zitat Avery MA, Sheehan AE, Kerr KS, Wang J, Freeman MR: WldS requires Nmnat1 enzymatic activity and N16-VCP interactions to suppress Wallerian degeneration. J Cell Biol. 2009, 184: 501-513. 10.1083/jcb.200808042.PubMedCentralCrossRefPubMed Avery MA, Sheehan AE, Kerr KS, Wang J, Freeman MR: WldS requires Nmnat1 enzymatic activity and N16-VCP interactions to suppress Wallerian degeneration. J Cell Biol. 2009, 184: 501-513. 10.1083/jcb.200808042.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Zhai RG, Zhang F, Hiesinger PR, Cao Y, Haueter CM, Bellen HJ: NAD synthase NMNAT acts as a chaperone to protect against neurodegeneration. Nature. 2008, 452: 887-891. 10.1038/nature06721.PubMedCentralCrossRefPubMed Zhai RG, Zhang F, Hiesinger PR, Cao Y, Haueter CM, Bellen HJ: NAD synthase NMNAT acts as a chaperone to protect against neurodegeneration. Nature. 2008, 452: 887-891. 10.1038/nature06721.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat Wen Y, Parrish JZ, He R, Zhai RG, Kim MD: Nmnat exerts neuroprotective effects in dendrites and axons. Mol Cell Neurosci. 2011 Wen Y, Parrish JZ, He R, Zhai RG, Kim MD: Nmnat exerts neuroprotective effects in dendrites and axons. Mol Cell Neurosci. 2011
32.
Zurück zum Zitat Zhai RG, Cao Y, Hiesinger PR, Zhou Y, Mehta SQ, Schulze KL, Verstreken P, Bellen HJ: Drosophila NMNAT maintains neural integrity independent of its NAD synthesis activity. PLoS Biol. 2006, 4: e416-10.1371/journal.pbio.0040416.PubMedCentralCrossRefPubMed Zhai RG, Cao Y, Hiesinger PR, Zhou Y, Mehta SQ, Schulze KL, Verstreken P, Bellen HJ: Drosophila NMNAT maintains neural integrity independent of its NAD synthesis activity. PLoS Biol. 2006, 4: e416-10.1371/journal.pbio.0040416.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Lotharius J, Dugan LL, O'Malley KL: Distinct mechanisms underlie neurotoxin-mediated cell death in cultured dopaminergic neurons. J Neurosci. 1999, 19: 1284-1293.PubMed Lotharius J, Dugan LL, O'Malley KL: Distinct mechanisms underlie neurotoxin-mediated cell death in cultured dopaminergic neurons. J Neurosci. 1999, 19: 1284-1293.PubMed
34.
Zurück zum Zitat Babetto E, Beirowski B, Janeckova L, Brown R, Gilley J, Thomson D, Ribchester RR, Coleman MP: Targeting NMNAT1 to axons and synapses transforms its neuroprotective potency in vivo. J Neurosci. 2010, 30: 13291-13304. 10.1523/JNEUROSCI.1189-10.2010.CrossRefPubMed Babetto E, Beirowski B, Janeckova L, Brown R, Gilley J, Thomson D, Ribchester RR, Coleman MP: Targeting NMNAT1 to axons and synapses transforms its neuroprotective potency in vivo. J Neurosci. 2010, 30: 13291-13304. 10.1523/JNEUROSCI.1189-10.2010.CrossRefPubMed
35.
Zurück zum Zitat Sasaki Y, Milbrandt J: Axonal degeneration is blocked by nicotinamide mononucleotide adenylyltransferase (Nmnat) protein transduction into transected axons. J Biol Chem. 2010, 285: 41211-41215. 10.1074/jbc.C110.193904.PubMedCentralCrossRefPubMed Sasaki Y, Milbrandt J: Axonal degeneration is blocked by nicotinamide mononucleotide adenylyltransferase (Nmnat) protein transduction into transected axons. J Biol Chem. 2010, 285: 41211-41215. 10.1074/jbc.C110.193904.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Press C, Milbrandt J: Nmnat delays axonal degeneration caused by mitochondrial and oxidative stress. J Neurosci. 2008, 28: 4861-4871. 10.1523/JNEUROSCI.0525-08.2008.PubMedCentralCrossRefPubMed Press C, Milbrandt J: Nmnat delays axonal degeneration caused by mitochondrial and oxidative stress. J Neurosci. 2008, 28: 4861-4871. 10.1523/JNEUROSCI.0525-08.2008.PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Verghese PB, Sasaki Y, Yang D, Stewart F, Sabar F, Finn MB, Wroge CM, Mennerick S, Neil JJ, Milbrandt J, Holtzman DM: Nicotinamide mononucleotide adenylyl transferase 1 protects against acute neurodegeneration in developing CNS by inhibiting excitotoxic-necrotic cell death. Proc Natl Acad Sci USA. 2011, 108: 19054-19059. 10.1073/pnas.1107325108.PubMedCentralCrossRefPubMed Verghese PB, Sasaki Y, Yang D, Stewart F, Sabar F, Finn MB, Wroge CM, Mennerick S, Neil JJ, Milbrandt J, Holtzman DM: Nicotinamide mononucleotide adenylyl transferase 1 protects against acute neurodegeneration in developing CNS by inhibiting excitotoxic-necrotic cell death. Proc Natl Acad Sci USA. 2011, 108: 19054-19059. 10.1073/pnas.1107325108.PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Sasaki Y, Vohra BP, Lund FE, Milbrandt J: Nicotinamide mononucleotide adenylyl transferase-mediated axonal protection requires enzymatic activity but not increased levels of neuronal nicotinamide adenine dinucleotide. J Neurosci. 2009, 29: 5525-5535. 10.1523/JNEUROSCI.5469-08.2009.PubMedCentralCrossRefPubMed Sasaki Y, Vohra BP, Lund FE, Milbrandt J: Nicotinamide mononucleotide adenylyl transferase-mediated axonal protection requires enzymatic activity but not increased levels of neuronal nicotinamide adenine dinucleotide. J Neurosci. 2009, 29: 5525-5535. 10.1523/JNEUROSCI.5469-08.2009.PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Sasaki Y, Vohra BP, Baloh RH, Milbrandt J: Transgenic mice expressing the Nmnat1 protein manifest robust delay in axonal degeneration in vivo. J Neurosci. 2009, 29: 6526-6534. 10.1523/JNEUROSCI.1429-09.2009.PubMedCentralCrossRefPubMed Sasaki Y, Vohra BP, Baloh RH, Milbrandt J: Transgenic mice expressing the Nmnat1 protein manifest robust delay in axonal degeneration in vivo. J Neurosci. 2009, 29: 6526-6534. 10.1523/JNEUROSCI.1429-09.2009.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Wishart TM, Pemberton HN, James SR, McCabe CJ, Gillingwater TH: Modified cell cycle status in a mouse model of altered neuronal vulnerability (slow Wallerian degeneration; Wlds). Genome Biol. 2008, 9: R101-10.1186/gb-2008-9-6-r101.PubMedCentralCrossRefPubMed Wishart TM, Pemberton HN, James SR, McCabe CJ, Gillingwater TH: Modified cell cycle status in a mouse model of altered neuronal vulnerability (slow Wallerian degeneration; Wlds). Genome Biol. 2008, 9: R101-10.1186/gb-2008-9-6-r101.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Yan T, Feng Y, Zheng J, Ge X, Zhang Y, Wu D, Zhao J, Zhai Q: Nmnat2 delays axon degeneration in superior cervical ganglia dependent on its NAD synthesis activity. Neurochem Int. 2010, 56: 101-106. 10.1016/j.neuint.2009.09.007.CrossRefPubMed Yan T, Feng Y, Zheng J, Ge X, Zhang Y, Wu D, Zhao J, Zhai Q: Nmnat2 delays axon degeneration in superior cervical ganglia dependent on its NAD synthesis activity. Neurochem Int. 2010, 56: 101-106. 10.1016/j.neuint.2009.09.007.CrossRefPubMed
42.
Zurück zum Zitat Conforti L, Fang G, Beirowski B, Wang MS, Sorci L, Asress S, Adalbert R, Silva A, Bridge K, Huang XP, Magni G, Glass JD, Coleman MP: NAD(+) and axon degeneration revisited: Nmnat1 cannot substitute for Wld(S) to delay Wallerian degeneration. Cell Death Differ. 2007, 14: 116-127. 10.1038/sj.cdd.4401944.CrossRefPubMed Conforti L, Fang G, Beirowski B, Wang MS, Sorci L, Asress S, Adalbert R, Silva A, Bridge K, Huang XP, Magni G, Glass JD, Coleman MP: NAD(+) and axon degeneration revisited: Nmnat1 cannot substitute for Wld(S) to delay Wallerian degeneration. Cell Death Differ. 2007, 14: 116-127. 10.1038/sj.cdd.4401944.CrossRefPubMed
43.
Zurück zum Zitat Yahata N, Yuasa S, Araki T: Nicotinamide mononucleotide adenylyltransferase expression in mitochondrial matrix delays Wallerian degeneration. J Neurosci. 2009, 29: 6276-6284. 10.1523/JNEUROSCI.4304-08.2009.CrossRefPubMed Yahata N, Yuasa S, Araki T: Nicotinamide mononucleotide adenylyltransferase expression in mitochondrial matrix delays Wallerian degeneration. J Neurosci. 2009, 29: 6276-6284. 10.1523/JNEUROSCI.4304-08.2009.CrossRefPubMed
44.
Zurück zum Zitat Kaneko S, Wang J, Kaneko M, Yiu G, Hurrell JM, Chitnis T, Khoury SJ, He Z: Protecting axonal degeneration by increasing nicotinamide adenine dinucleotide levels in experimental autoimmune encephalomyelitis models. J Neurosci. 2006, 26: 9794-9804. 10.1523/JNEUROSCI.2116-06.2006.CrossRefPubMed Kaneko S, Wang J, Kaneko M, Yiu G, Hurrell JM, Chitnis T, Khoury SJ, He Z: Protecting axonal degeneration by increasing nicotinamide adenine dinucleotide levels in experimental autoimmune encephalomyelitis models. J Neurosci. 2006, 26: 9794-9804. 10.1523/JNEUROSCI.2116-06.2006.CrossRefPubMed
45.
Zurück zum Zitat Yang J, Klaidman LK, Chang ML, Kem S, Sugawara T, Chan P, Adams JD: Nicotinamide therapy protects against both necrosis and apoptosis in a stroke model. Pharmacol Biochem Behav. 2002, 73: 901-910. 10.1016/S0091-3057(02)00939-5.CrossRefPubMed Yang J, Klaidman LK, Chang ML, Kem S, Sugawara T, Chan P, Adams JD: Nicotinamide therapy protects against both necrosis and apoptosis in a stroke model. Pharmacol Biochem Behav. 2002, 73: 901-910. 10.1016/S0091-3057(02)00939-5.CrossRefPubMed
46.
Zurück zum Zitat Sakakibara Y, Mitha AP, Ayoub IA, Ogilvy CS, Maynard KI: Delayed treatment with nicotinamide (vitamin B3) reduces the infarct volume following focal cerebral ischemia in spontaneously hypertensive rats, diabetic and non-diabetic Fischer 344 rats. Brain Res. 2002, 931: 68-73. 10.1016/S0006-8993(02)02263-1.CrossRefPubMed Sakakibara Y, Mitha AP, Ayoub IA, Ogilvy CS, Maynard KI: Delayed treatment with nicotinamide (vitamin B3) reduces the infarct volume following focal cerebral ischemia in spontaneously hypertensive rats, diabetic and non-diabetic Fischer 344 rats. Brain Res. 2002, 931: 68-73. 10.1016/S0006-8993(02)02263-1.CrossRefPubMed
47.
Zurück zum Zitat Demarin V, Podobnik SS, Storga-Tomic D, Kay G: Treatment of Alzheimer's disease with stabilized oral nicotinamide adenine dinucleotide: a randomized, double-blind study. Drugs Exp Clin Res. 2004, 30: 27-33.PubMed Demarin V, Podobnik SS, Storga-Tomic D, Kay G: Treatment of Alzheimer's disease with stabilized oral nicotinamide adenine dinucleotide: a randomized, double-blind study. Drugs Exp Clin Res. 2004, 30: 27-33.PubMed
48.
Zurück zum Zitat Birkmayer JG, Vrecko C, Volc D, Birkmayer W: Nicotinamide adenine dinucleotide (NADH)--a new therapeutic approach to Parkinson's disease. Comparison of oral and parenteral application. Acta Neurol Scand Suppl. 1993, 146: 32-35.PubMed Birkmayer JG, Vrecko C, Volc D, Birkmayer W: Nicotinamide adenine dinucleotide (NADH)--a new therapeutic approach to Parkinson's disease. Comparison of oral and parenteral application. Acta Neurol Scand Suppl. 1993, 146: 32-35.PubMed
49.
Zurück zum Zitat Birkmayer JG: Coenzyme nicotinamide adenine dinucleotide: new therapeutic approach for improving dementia of the Alzheimer type. Ann Clin Lab Sci. 1996, 26: 1-9.PubMed Birkmayer JG: Coenzyme nicotinamide adenine dinucleotide: new therapeutic approach for improving dementia of the Alzheimer type. Ann Clin Lab Sci. 1996, 26: 1-9.PubMed
50.
Zurück zum Zitat Kuhn W, Muller T, Winkel R, Danielczik S, Gerstner A, Hacker R, Mattern C, Przuntek H: Parenteral application of NADH in Parkinson's disease: clinical improvement partially due to stimulation of endogenous levodopa biosynthesis. J Neural Transm. 1996, 103: 1187-1193. 10.1007/BF01271203.CrossRefPubMed Kuhn W, Muller T, Winkel R, Danielczik S, Gerstner A, Hacker R, Mattern C, Przuntek H: Parenteral application of NADH in Parkinson's disease: clinical improvement partially due to stimulation of endogenous levodopa biosynthesis. J Neural Transm. 1996, 103: 1187-1193. 10.1007/BF01271203.CrossRefPubMed
51.
Zurück zum Zitat Zhai RG, Rizzi M, Garavaglia S: Nicotinamide/nicotinic acid mononucleotide adenylyltransferase, new insights into an ancient enzyme. Cell Mol Life Sci. 2009, 66: 2805-2818. 10.1007/s00018-009-0047-x.CrossRefPubMed Zhai RG, Rizzi M, Garavaglia S: Nicotinamide/nicotinic acid mononucleotide adenylyltransferase, new insights into an ancient enzyme. Cell Mol Life Sci. 2009, 66: 2805-2818. 10.1007/s00018-009-0047-x.CrossRefPubMed
52.
Zurück zum Zitat Ali YO, McCormack R, Darr A, Zhai RG: Nicotinamide Mononucleotide Adenylyltransferase Is a Stress Response Protein Regulated by the Heat Shock Factor/Hypoxia-inducible Factor 1α Pathway. J Biol Chem. 2011, 286: 19089-19099. 10.1074/jbc.M111.219295.PubMedCentralCrossRefPubMed Ali YO, McCormack R, Darr A, Zhai RG: Nicotinamide Mononucleotide Adenylyltransferase Is a Stress Response Protein Regulated by the Heat Shock Factor/Hypoxia-inducible Factor 1α Pathway. J Biol Chem. 2011, 286: 19089-19099. 10.1074/jbc.M111.219295.PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Nicklas WJ, Vyas I, Heikkila RE: Inhibition of NADH-linked oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. Life Sci. 1985, 36: 2503-2508. 10.1016/0024-3205(85)90146-8.CrossRefPubMed Nicklas WJ, Vyas I, Heikkila RE: Inhibition of NADH-linked oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. Life Sci. 1985, 36: 2503-2508. 10.1016/0024-3205(85)90146-8.CrossRefPubMed
54.
Zurück zum Zitat Ramsay RR, Krueger MJ, Youngster SK, Gluck MR, Casida JE, Singer TP: Interaction of 1-methyl-4-phenylpyridinium ion (MPP+) and its analogs with the rotenone/piericidin binding site of NADH dehydrogenase. J Neurochem. 1991, 56: 1184-1190. 10.1111/j.1471-4159.1991.tb11409.x.CrossRefPubMed Ramsay RR, Krueger MJ, Youngster SK, Gluck MR, Casida JE, Singer TP: Interaction of 1-methyl-4-phenylpyridinium ion (MPP+) and its analogs with the rotenone/piericidin binding site of NADH dehydrogenase. J Neurochem. 1991, 56: 1184-1190. 10.1111/j.1471-4159.1991.tb11409.x.CrossRefPubMed
55.
Zurück zum Zitat Ramsay RR, Salach JI, Singer TP: Uptake of the neurotoxin 1-methyl-4-phenylpyridine (MPP+) by mitochondria and its relation to the inhibition of the mitochondrial oxidation of NAD+-linked substrates by MPP+. Biochem Biophys Res Commun. 1986, 134: 743-748. 10.1016/S0006-291X(86)80483-1.CrossRefPubMed Ramsay RR, Salach JI, Singer TP: Uptake of the neurotoxin 1-methyl-4-phenylpyridine (MPP+) by mitochondria and its relation to the inhibition of the mitochondrial oxidation of NAD+-linked substrates by MPP+. Biochem Biophys Res Commun. 1986, 134: 743-748. 10.1016/S0006-291X(86)80483-1.CrossRefPubMed
56.
Zurück zum Zitat Lotharius J, O'Malley KL: The parkinsonism-inducing drug 1-methyl-4-phenylpyridinium triggers intracellular dopamine oxidation. A novel mechanism of toxicity. J Biol Chem. 2000, 275: 38581-38588.CrossRefPubMed Lotharius J, O'Malley KL: The parkinsonism-inducing drug 1-methyl-4-phenylpyridinium triggers intracellular dopamine oxidation. A novel mechanism of toxicity. J Biol Chem. 2000, 275: 38581-38588.CrossRefPubMed
57.
Zurück zum Zitat Chang GD, Ramirez VD: The mechanism of action of MPTP and MPP+ on endogenous dopamine release from the rat corpus striatum superfused in vitro. Brain Res. 1986, 368: 134-140. 10.1016/0006-8993(86)91050-4.CrossRefPubMed Chang GD, Ramirez VD: The mechanism of action of MPTP and MPP+ on endogenous dopamine release from the rat corpus striatum superfused in vitro. Brain Res. 1986, 368: 134-140. 10.1016/0006-8993(86)91050-4.CrossRefPubMed
58.
Zurück zum Zitat Cappelletti G, Surrey T, Maci R: The parkinsonism producing neurotoxin MPP+ affects microtubule dynamics by acting as a destabilising factor. FEBS Lett. 2005, 579: 4781-4786. 10.1016/j.febslet.2005.07.058.CrossRefPubMed Cappelletti G, Surrey T, Maci R: The parkinsonism producing neurotoxin MPP+ affects microtubule dynamics by acting as a destabilising factor. FEBS Lett. 2005, 579: 4781-4786. 10.1016/j.febslet.2005.07.058.CrossRefPubMed
59.
Zurück zum Zitat Zhu JH, Horbinski C, Guo F, Watkins S, Uchiyama Y, Chu CT: Regulation of autophagy by extracellular signal-regulated protein kinases during 1-methyl-4-phenylpyridinium-induced cell death. Am J Pathol. 2007, 170: 75-86. 10.2353/ajpath.2007.060524.PubMedCentralCrossRefPubMed Zhu JH, Horbinski C, Guo F, Watkins S, Uchiyama Y, Chu CT: Regulation of autophagy by extracellular signal-regulated protein kinases during 1-methyl-4-phenylpyridinium-induced cell death. Am J Pathol. 2007, 170: 75-86. 10.2353/ajpath.2007.060524.PubMedCentralCrossRefPubMed
60.
Zurück zum Zitat Barrientos SA, Martinez NW, Yoo S, Jara JS, Zamorano S, Hetz C, Twiss JL, Alvarez J, Court FA: Axonal degeneration is mediated by the mitochondrial permeability transition pore. J Neurosci. 2011, 31: 966-978. 10.1523/JNEUROSCI.4065-10.2011.PubMedCentralCrossRefPubMed Barrientos SA, Martinez NW, Yoo S, Jara JS, Zamorano S, Hetz C, Twiss JL, Alvarez J, Court FA: Axonal degeneration is mediated by the mitochondrial permeability transition pore. J Neurosci. 2011, 31: 966-978. 10.1523/JNEUROSCI.4065-10.2011.PubMedCentralCrossRefPubMed
61.
Zurück zum Zitat Wishart TM, Paterson JM, Short DM, Meredith S, Robertson KA, Sutherland C, Cousin MA, Dutia MB, Gillingwater TH: Differential proteomics analysis of synaptic proteins identifies potential cellular targets and protein mediators of synaptic neuroprotection conferred by the slow Wallerian degeneration (Wlds) gene. Mol Cell Proteomics. 2007, 6: 1318-1330. 10.1074/mcp.M600457-MCP200.PubMedCentralCrossRefPubMed Wishart TM, Paterson JM, Short DM, Meredith S, Robertson KA, Sutherland C, Cousin MA, Dutia MB, Gillingwater TH: Differential proteomics analysis of synaptic proteins identifies potential cellular targets and protein mediators of synaptic neuroprotection conferred by the slow Wallerian degeneration (Wlds) gene. Mol Cell Proteomics. 2007, 6: 1318-1330. 10.1074/mcp.M600457-MCP200.PubMedCentralCrossRefPubMed
62.
Zurück zum Zitat Miller BR, Press C, Daniels RW, Sasaki Y, Milbrandt J, DiAntonio A: A dual leucine kinase-dependent axon self-destruction program promotes Wallerian degeneration. Nat Neurosci. 2009, 12: 387-389. 10.1038/nn.2290.PubMedCentralCrossRefPubMed Miller BR, Press C, Daniels RW, Sasaki Y, Milbrandt J, DiAntonio A: A dual leucine kinase-dependent axon self-destruction program promotes Wallerian degeneration. Nat Neurosci. 2009, 12: 387-389. 10.1038/nn.2290.PubMedCentralCrossRefPubMed
63.
Zurück zum Zitat Surjana D, Halliday GM, Damian DL: Role of nicotinamide in DNA damage, mutagenesis, and DNA repair. J Nucleic Acids 2010. 2010 Surjana D, Halliday GM, Damian DL: Role of nicotinamide in DNA damage, mutagenesis, and DNA repair. J Nucleic Acids 2010. 2010
64.
Zurück zum Zitat Hisahara S, Chiba S, Matsumoto H, Horio Y: Transcriptional regulation of neuronal genes and its effect on neural functions: NAD-dependent histone deacetylase SIRT1 (Sir2α). J Pharmacol Sci. 2005, 98: 200-204. 10.1254/jphs.FMJ05001X2.CrossRefPubMed Hisahara S, Chiba S, Matsumoto H, Horio Y: Transcriptional regulation of neuronal genes and its effect on neural functions: NAD-dependent histone deacetylase SIRT1 (Sir2α). J Pharmacol Sci. 2005, 98: 200-204. 10.1254/jphs.FMJ05001X2.CrossRefPubMed
65.
Zurück zum Zitat Wang J, Zhai Q, Chen Y, Lin E, Gu W, McBurney MW, He Z: A local mechanism mediates NAD-dependent protection of axon degeneration. J Cell Biol. 2005, 170: 349-355. 10.1083/jcb.200504028.PubMedCentralCrossRefPubMed Wang J, Zhai Q, Chen Y, Lin E, Gu W, McBurney MW, He Z: A local mechanism mediates NAD-dependent protection of axon degeneration. J Cell Biol. 2005, 170: 349-355. 10.1083/jcb.200504028.PubMedCentralCrossRefPubMed
66.
Zurück zum Zitat Gillingwater TH, Haley JE, Ribchester RR, Horsburgh K: Neuroprotection after transient global cerebral ischemia in Wld(s) mutant mice. J Cereb Blood Flow Metab. 2004, 24: 62-66.CrossRefPubMed Gillingwater TH, Haley JE, Ribchester RR, Horsburgh K: Neuroprotection after transient global cerebral ischemia in Wld(s) mutant mice. J Cereb Blood Flow Metab. 2004, 24: 62-66.CrossRefPubMed
67.
Zurück zum Zitat Ferri A, Sanes JR, Coleman MP, Cunningham JM, Kato AC: Inhibiting axon degeneration and synapse loss attenuates apoptosis and disease progression in a mouse model of motoneuron disease. Curr Biol. 2003, 13: 669-673. 10.1016/S0960-9822(03)00206-9.CrossRefPubMed Ferri A, Sanes JR, Coleman MP, Cunningham JM, Kato AC: Inhibiting axon degeneration and synapse loss attenuates apoptosis and disease progression in a mouse model of motoneuron disease. Curr Biol. 2003, 13: 669-673. 10.1016/S0960-9822(03)00206-9.CrossRefPubMed
68.
Zurück zum Zitat Liang CL, Wang TT, Luby-Phelps K, German DC: Mitochondria mass is low in mouse substantia nigra dopamine neurons: implications for Parkinson's disease. Exp Neurol. 2007, 203: 370-380. 10.1016/j.expneurol.2006.08.015.CrossRefPubMed Liang CL, Wang TT, Luby-Phelps K, German DC: Mitochondria mass is low in mouse substantia nigra dopamine neurons: implications for Parkinson's disease. Exp Neurol. 2007, 203: 370-380. 10.1016/j.expneurol.2006.08.015.CrossRefPubMed
69.
Zurück zum Zitat Hastings TG: The role of dopamine oxidation in mitochondrial dysfunction: implications for Parkinson's disease. J Bioenerg Biomembr. 2009, 41: 469-472. 10.1007/s10863-009-9257-z.CrossRefPubMed Hastings TG: The role of dopamine oxidation in mitochondrial dysfunction: implications for Parkinson's disease. J Bioenerg Biomembr. 2009, 41: 469-472. 10.1007/s10863-009-9257-z.CrossRefPubMed
70.
Zurück zum Zitat Surmeier DJ, Guzman JN, Sanchez-Padilla J, Goldberg JA: What causes the death of dopaminergic neurons in Parkinson's disease?. Prog Brain Res. 2010, 183: 59-77.CrossRefPubMed Surmeier DJ, Guzman JN, Sanchez-Padilla J, Goldberg JA: What causes the death of dopaminergic neurons in Parkinson's disease?. Prog Brain Res. 2010, 183: 59-77.CrossRefPubMed
71.
Zurück zum Zitat Dickson DW, Fujishiro H, Orr C, DelleDonne A, Josephs KA, Frigerio R, Burnett M, Parisi JE, Klos KJ, Ahlskog JE: Neuropathology of non-motor features of Parkinson disease. Parkinsonism Relat Disord. 2009, 15 (Suppl 3): S1-5.CrossRefPubMed Dickson DW, Fujishiro H, Orr C, DelleDonne A, Josephs KA, Frigerio R, Burnett M, Parisi JE, Klos KJ, Ahlskog JE: Neuropathology of non-motor features of Parkinson disease. Parkinsonism Relat Disord. 2009, 15 (Suppl 3): S1-5.CrossRefPubMed
72.
Zurück zum Zitat Bernstein AI, Garrison SP, Zambetti GP, O'Malley KL: 6-OHDA generated ROS induces DNA damage and p53- and PUMA-dependent cell death. Mol Neurodegener. 2011, 6: 2-10.1186/1750-1326-6-2.PubMedCentralCrossRefPubMed Bernstein AI, Garrison SP, Zambetti GP, O'Malley KL: 6-OHDA generated ROS induces DNA damage and p53- and PUMA-dependent cell death. Mol Neurodegener. 2011, 6: 2-10.1186/1750-1326-6-2.PubMedCentralCrossRefPubMed
73.
Zurück zum Zitat Beirowski B, Babetto E, Gilley J, Mazzola F, Conforti L, Janeckova L, Magni G, Ribchester RR, Coleman MP: Non-nuclear Wld(S) determines its neuroprotective efficacy for axons and synapses in vivo. J Neurosci. 2009, 29: 653-668. 10.1523/JNEUROSCI.3814-08.2009.CrossRefPubMed Beirowski B, Babetto E, Gilley J, Mazzola F, Conforti L, Janeckova L, Magni G, Ribchester RR, Coleman MP: Non-nuclear Wld(S) determines its neuroprotective efficacy for axons and synapses in vivo. J Neurosci. 2009, 29: 653-668. 10.1523/JNEUROSCI.3814-08.2009.CrossRefPubMed
74.
Zurück zum Zitat Brown JA, Wysolmerski RB, Bridgman PC: Dorsal root ganglion neurons react to semaphorin 3A application through a biphasic response that requires multiple myosin II isoforms. Mol Biol Cell. 2009, 20: 1167-1179. 10.1091/mbc.E08-01-0065.PubMedCentralCrossRefPubMed Brown JA, Wysolmerski RB, Bridgman PC: Dorsal root ganglion neurons react to semaphorin 3A application through a biphasic response that requires multiple myosin II isoforms. Mol Biol Cell. 2009, 20: 1167-1179. 10.1091/mbc.E08-01-0065.PubMedCentralCrossRefPubMed
75.
Zurück zum Zitat Magni G, Amici A, Emanuelli M, Orsomando G, Raffaelli N, Ruggieri S: Enzymology of NAD+ homeostasis in man. Cell Mol Life Sci. 2004, 61: 19-34. 10.1007/s00018-003-3161-1.CrossRefPubMed Magni G, Amici A, Emanuelli M, Orsomando G, Raffaelli N, Ruggieri S: Enzymology of NAD+ homeostasis in man. Cell Mol Life Sci. 2004, 61: 19-34. 10.1007/s00018-003-3161-1.CrossRefPubMed
Metadaten
Titel
Wld S but not Nmnat1 protects dopaminergic neurites from MPP+neurotoxicity
verfasst von
Jo Ann V Antenor-Dorsey
Karen L O'Malley
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
Molecular Neurodegeneration / Ausgabe 1/2012
Elektronische ISSN: 1750-1326
DOI
https://doi.org/10.1186/1750-1326-7-5

Weitere Artikel der Ausgabe 1/2012

Molecular Neurodegeneration 1/2012 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.