Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2005

Open Access 01.12.2005 | Research

Signaling pathways mediating a selective induction of nitric oxide synthase II by tumor necrosis factor alpha in nerve growth factor-responsive cells

verfasst von: Michael S Thomas, WenRu Zhang, Paivi M Jordan, H Uri Saragovi, Giulio Taglialatela

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2005

Abstract

Background

Inflammation and oxidative stress play a critical role in neurodegeneration associated with acute and chronic insults of the nervous system. Notably, affected neurons are often responsive to and dependent on trophic factors such as nerve growth factor (NGF). We previously showed in NGF-responsive PC12 cells that tumor necrosis factor alpha (TNFα) and NGF synergistically induce the expression of the free-radical producing enzyme inducible nitric oxide synthase (iNOS). We proposed that NGF-responsive neurons might be selectively exposed to iNOS-mediated oxidative damage as a consequence of elevated TNFα levels. With the aim of identifying possible therapeutic targets, in the present study we investigated the signaling pathways involved in NGF/TNFα-promoted iNOS induction.

Methods

Western blotting, RT-PCR, transcription factor-specific reporter gene systems, mutant cells lacking the low affinity p75NTR NGF receptor and transfections of TNFα/NGF chimeric receptors were used to investigate signalling events associated with NGF/TNFα-promoted iNOS induction in PC12 cells.

Results

Our results show that iNOS expression resulting from NGF/TNFα combined treatment can be elicited in PC12 cells. Mutant PC12 cells lacking p75NTR did not respond, suggesting that p75NTR is required to mediate iNOS expression. Furthermore, cells transfected with chimeric TNFα/NGF receptors demonstrated that the simultaneous presence of both p75NTR and TrkA signaling is necessary to synergize with TNFα to mediate iNOS expression. Lastly, our data show that NGF/TNFα-promoted iNOS induction requires activation of the transcription factor nuclear factor kappa B (NF-κB).

Conclusion

Collectively, our in vitro model suggests that cells bearing both the high and low affinity NGF receptors may display increased sensitivity to TNFα in terms of iNOS expression and therefore be selectively at risk during acute (e.g. neurotrauma) or chronic (e.g. neurodegenerative diseases) conditions where high levels of pro-inflammatory cytokines in the nervous system occur pathologically. Our results also suggest that modulation of NFκB-promoted transcription of selective genes could serve as a potential therapeutic target to prevent neuroinflammation-induced neuronal damage.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1742-2094-2-19) contains supplementary material, which is available to authorized users.

Competing interests

The author(s) declare they have no competing interests.

Authors' contributions

MST participated in the conception and design of the study, carried out the bulk of experiments, performed data analysis, and drafted the manuscript. PMJ participated in study design especially with regards to the IGF experiments. WZ participated in study design and coordination and provided the expertise for RTPCR and withdrawal experiments. HUS sub-cloned the PC12p75NTR(-) cells and participated in study design and result interpretation of experiments involving these cells. GT participated in conception, study design, coordination and helped to draft and review the manuscript. All authors read and approved the final manuscript.
Abkürzungen
AraC
cytosine β-D-arabinofuranoside
AD
Alzheimers disease
BDNF
brain derived neurotrophic factor
BFCN
basal forebrain cholinergic neurons
CNS
central nervous system
CRE
cyclic-AMP response element
GDNF
glial derived neurotrophic factor
IGF
insulin-like growth factor
IL-1β
interleukin-1beta
iNOS
inducible nitric oxide synthase
MAPK
mitogen activated protein kinase
NF-κB
nuclear factor kappa B
NGF
nerve growth factor
NO
nitric oxide
nNOS
neuronal nitric oxide synthase
NTR
neurotrophin receptor
PC12
pheochromocytoma
PCN
penicillin
PDTC
pyrrolidinedithyocarbamate
PSI
proteosome inhibitor
SDS
sodium dodecylsulfate
SEAP
secreted alkaline phosphatase
S.E.M
standard error of the mean
Strep
streptomycin
TNFα
tumor necrosis factor alpha
TrkA
troponin-like receptor kinase A
TTBS
tris-buffered saline with tween 20

Background

Neuroinflammation is thought to play a prominent role in neurodegeneration associated with a variety of acute and chronic insults in both the central (CNS) and peripheral (PNS) nervous system [1, 2]. Examples of neurotraumatic or neurodegenerative conditions where the occurrence or role of neuroinflammation has been documented include peripheral nerve injury [36], acute and chronic spinal cord injury [711], traumatic brain injury [1214], stroke [1517], amyotrophic lateral sclerosis (ALS, [1820] and Alzheimer Disease (AD, [2124].
Neurons susceptible to neuroinflammatory insults are often dependent for their survival on target derived neurotrophic factors such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF) or glia-derived neurotrophic factor (GDNF). The same neurodegenerative conditions have also been associated with the presence of damaging high levels of free radical species leading to pathological oxidative stress [25]. For example, inflammatory involvement in AD pathogenesis has been proposed partly based on observations of increased levels of the pro-inflammatory cytokines tumor necrosis factor alpha (TNFα) and interleukin-1 beta (IL-1β) in cerebrospinal fluid and brain cortex of AD patients [26, 27]. Additionally, among the most affected neurons in AD are the basal forebrain cholinergic neurons (BFCN, [2830]), which rely upon trophic support by target-derived NGF [31, 32]. Furthermore, there is strong evidence for the presence of oxidative damage in the AD brain [3336]. Similarly, neuronal damage following acute spinal cord injury or peripheral nerve injury has been shown to involve a neuroinflammatory as well as oxidative stress component [1, 8, 10, 11, 3739], and traumatic head injury is also known to be associated with increased circulating concentrations of inflammatory cytokines and reduced numbers of basal forebrain cholinergic neurons [13, 4042].
Thus, there seems to be an intimate relationship between pro-inflammatory cytokines, oxidative stress and trophic factors that underscores the neuropathological consequences of extrinsic (e.g. traumatic) or intrinsic (e.g. disease-related) injury to the nervous system. Our previous work has shown that in NGF-responsive rat pheochromocytoma (PC12) cells TNFα induces expression of the free radical nitric oxide (NO) synthesizing enzyme NOS II (iNOS) only in the presence of NGF acting through its high affinity receptor TrkA [43]. Indeed, perturbed levels of NOS and NO-derived oxidative damage have been reported in both acute and chronic neurodegenerative conditions [25], including spinal cord injury [4446], stroke [47, 48] and AD [4953]. However, TNFα alone has not been shown to be an effective inducer of human iNOS promoter activity [54] or of rat cortical iNOS expression when administered intracerebroventricularly [55]. Nonetheless, TNFα has been shown to contribute to the death of NGF-dependent neurons in vitro [56] and in vivo [57, 58]. Therefore, our previous results suggest the attractive idea that one mechanism through which increased levels of TNFα affect certain trophic factor-responsive neurons may involve NO-derived oxidative damage brought about by a synergistic induction of iNOS. Understanding the molecular mechanisms mediating the synergistic NGF/TNFα-promoted induction of iNOS may thus provide novel therapeutic targets for the prevention of certain neurodegenerative events associated with acute or chronic injury of the nervous system.
Here we report that a reversible expression of iNOS, produced in PC12 cells by simultaneous exposure to NGF and TNFα, requires the simultaneous presence of both the low-affinity p75NTR and the high-affinity TrkA NGF receptors. Furthermore, using specific inhibitors and a reporter gene assay, we show that such synergistic effect of the combined NGF/TNFα treatment is mediated by the transcription factor nuclear factor kappa B (NF-κB).

Methods

Materials

All routine reagents and chemicals were obtained from Sigma-Aldrich (St Louis, MO, USA), except where noted otherwise. Recombinant human and rat TNF
https://static-content.springer.com/image/art%3A10.1186%2F1742-2094-2-19/MediaObjects/12974_2005_Article_43_Equa_HTML.gif
and rat IGF were obtained from R&D Systems, Minneapolis, MN, USA, purified mouse NGF from Harlan Bioproducts, Indianapolis, IN, USA, and pyrrolidine dithiocarmbamate (PDTC), the octapeptide proteasome inhibitor (PSI), PD98059, K252a and 1400 W from Calbiochem, San Diego, CA, USA.

Clonal cell lines

Stock cultures of rat pheochromocytoma cells (PC12; a kind gift of Dr. Lloyd Greene, Columbia University, New York, NY, USA) and PC12 cells lacking the low affinity p75NTR NGF receptor were maintained in 75 cm2 tissue culture flasks in 10 ml RPMI-1640 culture medium supplemented with 5% heat inactivated fetal bovine serum in a humidified cell incubator at 37°C kept at a 5% CO2 atmosphere. Half of the medium was replaced every other day and the cells were split once a week to maintain cell viability.

Expression vectors

Transient transfection of cells was performed by a liposomal packaging system. Briefly, 1.2 pmol of expression vector were mixed with DMRIE-C (Life Technologies, Carlsbad, CA, USA) in a 1:3 DNA to liposome ratio. The DNA/liposomes were diluted in 400 μl serum free transfection medium (Optimem) and then added to approximately 100,000 cells in a 12 well cell culture plate. The cells were allowed to take up the liposomal DNA for 3 hours before being washed and returned to cell culture medium. Cells were allowed to recover for 24 hours before any treatments. The cDNA coding for chimeric proteins bearing the extracellular domain of the TNFR1 receptor and the transmembrane and cytosolic domains of the NGF receptors (either p75NTR or TrkA) was a kind gift from Dr. Eric Shooter and prepared as described [77], (Stanford University, Palo Alto, Ca, USA). The p-SEAP expression vector, containing the SEAP gene under NF-kB, AP1 or CRE enhancer control, was purchased from Clontech (Palo Alto, CA, USA). Conditioned medium from cells transfected with the SEAP reporter vectors was assayed for alkaline phosphatase by sampling the medium and using the chemiluminescent Great EscAPe SEAP assay (Clontech, Palo Alto, CA, USA), according to manufacturer's instructions.

Western blot analysis

Cells were lysed using an SDS-based lysis buffer (2% SDS, 5 mM EDTA, 50 mM Tris, 1 mM each of DTT, PMSF and protease inhibitor cocktail). Following an ice-cold PBS wash, cells were lysed with SDS lysis buffer and the sonicated briefly before clarifying by centrifugation at 20,000 g for 20 minutes at 4°C. After centrifugation the supernatant was collected and protein content was measured using the standard BCA protein assay (Pierce, Rockford, IL, USA). Protein extracts (40 μg) were diluted in 6X sample buffer and loaded onto a 6% SDS-polyacrylamide gel. Gels were run for one hour at 100 V and then were transferred to a nitrocellulose membrane overnight at 25 V. All incubations were at room temperature in 0.5% Tween in Tris buffered saline (TTBS). The membranes were blocked for one hour in 5% milk in TTBS. Primary monoclonal anti-iNOS (Signal Transduction Laboratories, San Diego, CA, USA) or polyclonal anti-TNFR1 (Santa Cruz Biotechnology, Santa Cruz, CA, USA) were diluted in 2.5% milk in TTBS at 1:1000 and membranes were incubated with the antibody for one hour at room temperature. Membranes were washed three times for ten minutes each in TTBS before incubating for one hour with a horseradish-peroxidase secondary antibody (BioRad, Hercules, CA, USA) at 1:7500 in 2.5% milk in TTBS. Finally, membranes were washed again in TTBS three times for ten minutes each. Immunoreactive bands were visualized by a chemiluminescent western blot detection kit (Amersham Biosciences, Piscatay, NJ, USA) according to manufacturer's instructions. Images were captured using a 12 bit monochrome camera (UVP, Upland, CA, USA).

Reverse transcriptase polymerase chain reaction assay

Total RNA was extracted with Trizol Extraction Kit (Gibco BRL, San Diego, CA, USA) according to manufacturer's instructions. One μg of total RNA from each sample was applied to Ready-to-go RT-PCR Beads (Amersham Biosciences, Piscatay, NJ, USA) and used to complete the amplification protocol according to manufacturer's instructions. Primer sequences for rat iNOS were as follows; forward 5'-CAC GGA GAA CAG AGT TGG-3' and reverse 5'-GGA ACA CAG TAA TGG CCG ACC-3'. Amplified samples were run on agarose gels and stained with ethidium bromide. Images were captured using a 12 bit monochrome camera (UVP, Upland, CA, USA).

Flow cytometry

One μg of antibody against TrkA or p75NTR (Santa Cruz Biotechnology, Santa Cruz, CA, USA) was labeled with Zenon Rabbit IgG labeling kit from Molecular Probes (Eugene, OR) according to manufacturer's instructions and incubated for 1 hr with the cells in suspension. After incubation, labeled cells were visualized and quantified using a Becton Dickinson FACS Vantage Flow Cytometer set at appropriate instrument parameters.

Statistical analysis

Where appropriate, data were expressed as mean +/- standard error of the mean (S.E.M.), and analyzed by student unpaired two-tailed t test with significance set at p < 0.05.

Results

Combined NGF and TNFα induce iNOS message and protein

The upper panel of figure 1 shows a western blot detecting iNOS in PC12 cells treated simultaneously with 10 ng/ml NGF and 10 ng/ml TNFα in the presence or absence of 50 nM K252a, an inhibitor of phosphorylative events associated with tyrosine kinase receptor activation that has been shown to block the function of the high affinity NGF receptor TrkA [61]. There was a marked induction of iNOS expression only in cells simultaneously treated with NGF and TNFα, while neither treatment alone elicited any effect. Furthermore, K252a completely abolished NGF/TNFα-promoted iNOS induction, suggesting that TrkA function is essential to mediate it. As shown in the lower panel of figure 1, along with increased protein levels there was also an induction of iNOS mRNA in PC12 cells treated with NGF and TNFα but not in cells treated with either factor alone.

NGF and TNFα are both required for sustained iNOS expression

Figure 2A shows western blots detecting iNOS in cells treated with increasing concentrations of NGF (top panel) or TNFα (bottom panel), in the presence or absence of a fixed amount of TNFα or NGF, respectively. Either factor was ineffective when added alone at any of the concentrations tested. However, there was a marked dose-response increase in iNOS expression when increasing concentrations of NGF or TNFα were added in the presence of a fixed amount of TNFα or NGF, respectively. Figure 2B shows a representative western blot detecting iNOS expression in cells continuously treated with NGF and TNFα as compared to cells in which the combined treatment was withdrawn after 24 hr. The expression of iNOS returned to basal, undetectable, levels between 24 and 48 hr after withdrawal of both TNFα and NGF. Furthermore, as shown in figure 2C, withdrawal of either NGF or TNFα alone was sufficient to abolish iNOS expression induced by the combined treatment, both at the protein (top panel) and mRNA level (bottom panel). To exclude the involvement of unknown serum factors, NGF/TNFα-promoted induction of iNOS was determined in cells cultured for 24 hr in serum free or in defined medium N2 (Figure 2D). There was a detectable iNOS induction in both serum free- and defined medium-cultured cells, although much reduced in serum free conditions, which is predictable as PC12 cells do not survive for longer periods of time (24–48 hrs) in the absence of serum or N2 supplements. Since insulin is present in both serum and the N2 supplement, and can activate the insulin-like growth factor (IGF) receptor, we asked whether TNFα may synergize with IGF, which is also present in serum, to induce iNOS expression. The results shown in Figure 2E indicate that this is not the case.

TNFα/NGF-mediated iNOS expression is independent of NOS enzymatic activity

In order to determine whether the enzymatic activity of iNOS may play a role in sustaining TNFα/NGF-promoted signaling we pretreated PC12 cells with two NOS inhibitors prior to TNFα/NGF tretament. Pretreatment with N(G)-nitro-L-arginine methyl ester (L-NAME) did not affect expression of iNOS induced by the NGF/ TNFα combined treatment (Figure 3A). The same result was observed if a more specific inhibitor of iNOS (1400 W) was used instead of L-NAME (Figure 3B). Concentrations of 1400 W used here have been previously shown to be effective in inhibiting selectively iNOS activity in PC12 cells by others [78]. These results suggest that sustained iNOS expression in response of the combined NGF/TNFα treatment is independent of NOS enzymatic activity.

NGF/TNFα promoted iNOS induction requires the transcription factor NF-κB

Figure 4 shows results from PC12 cells transiently transfected with a secreted alkaline phosphatase reporter gene construct (SEAP) promoted by enhancer sequences specific for nuclear factor kappa B (NF-κB), activator protein 1 (AP-1), cAMP-responsive element (CRE) or Tal (non-inducible control). Twenty-four hr after transfection cells were treated with 10 ng/ml each of TNFα and NGF (alone or combined) and SEAP released in the culture medium (an index of endogenous transcription factor activation) was assayed 3 hr and 12 hr later. At 3 hr, cells treated with TNFα showed a significant increase in NF-κB activity but not AP-1 or CRE. Cells treated with NGF alone showed at 3 hr no significant increase in NF-κB, AP1 or CRE activity. When cells were exposed to the combined NGF/ TNFα treatment, there was a robust increase in NF-κB activity that was significantly higher than the response induced by the individual treatment with TNFα. On the other hand, neither AP-1 nor CRE activity were significantly affected by the combined NGF/ TNFα treatment. At 12 hr, both TNFα and NGF/TNFα combined treatments significantly increased NF-κB activity, but were not statistically significantly different. NGF-treated cells showed a significant increase in AP-1 and CRE activity at 12 hr, while NF-κB activity was not affected. As a result, there was also a significant increase in AP-1 and CRE activity elicited by the NGF/TNFα combined treatment at 12 hr. Neither NGF nor TNFα (alone or combined) elicited any effect on the control reporter construct Tal, either at 3 or 12 hr.
Involvement of NF-κB was further explored by determining the extent to which pharmacological inhibition of NF-κB would block NGF/TNFα-promoted iNOS induction in PC12 cells. As shown in figure 5A, treatment of PC12 cells with either pyrrolidine di-thio-carbamate (PDTC) or the octapeptide proteasome inhibitor PSI (two effective NF-κB inhibitors that have distinct mechanisms of action [8, 6365], completely abolished NGF/ TNFα-promoted iNOS induction. In this experiment, PD98059, a selective MAPK inhibitor, was used as a negative control. Both NF-κB inhibitors effectively blocked NF-κB-mediated transcriptional activity as determined by SEAP reporter gene assay (Figure 5B), whereas PD98059 had no effect. However, PD98059 completely blocked NGF-promoted neurite outgrowth (Figure 5C), an event that in PC12 cells is dependent on MAPK activation [66]. Furthermore, consistent with the results reported in Figure 4, inhibition of NOS activity by L-NAME did not affect NFκB activation by NGF/TNFα combined treatment (Figure 5D).

NGF/TNFα-promoted iNOS induction requires the simultaneous presence of both the p75NTR and TrkA NGF receptors

Next, we subcloned a PC12 mutant cell line (PC12p75NTR (-)) that lacks p75NTR expression while retaining TrkA at levels comparable with wild type PC12 cells (Figure 6A). NF-κB activity was not significantly increased by the NGF/TNFα combined treatment over the levels induced by TNFα alone in PC12p75NTR (-) (Figure 6B). Consistent with this finding, PC12p75NTR (-) cells exposed to the combined NGF/TNFα treatment did not show any induction of iNOS expression as compared to the parent cell line (Figure 6C). It is important to note that the PC12p75NTR (-) cells used here express TNFα receptor type 1 (TNFR1) at levels comparable (or even higher) than wild type PC12 cells (Figure 6D). Therefore lack of iNOS induction by the NGF/TNFα combined treatment in these cells cannot be ascribed to lack of TNFα responsiveness (as can also be appreciated by the NFκB response induced by TNFα alone shown in figure 6B).
The results obtained in PC12p75NTR(-) would suggest that p75NTR is essential to mediate iNOS induction by the combined TNFα/NGF treatment while the results obtained using K252a (Figure 1) would suggest a prominent role for TrkA. In order to ultimately ascertain the relative role of the two NGF receptors in mediating TNFα/NGF-promoted iNOS induction we made use of PC12 cells transiently transfected with expression vectors coding for chimeric TNFα/NGF receptors constructed as described by Rovelli et al. [77]. These constructs bear the ligand binding domain from the human TNFR1 and the signal transduction domain from rat NGF receptors, either TrkA or p75NTR. Previously, it has been shown that transfection with these chimeras allows for TNF-promoted NGF signaling [77]. Figure 7 shows a western blot detecting iNOS in PC12 cells individually or simultaneously transfected with chimeric TNFα receptors bearing the intracellular domain of p75NTR (p55/p75NTR) or TrkA (p55/TrkA). Transfected cells were then treated either with TNFα and NGF alone, or with both TNFα and NGF. As expected, the combined TNFα/NGF treatment induced a robust expression of iNOS in these PC12 cells, regardless of the presence of any transfected expression vector. As also expected, NGF alone did not elicit iNOS expression in any of the transfected cells. Similarly, TNFα alone did not induce iNOS in cells transfected with either p55/p75NTR or p55/TrkA chimeric receptors. However, TNFα promptly induced iNOS expression in cells transfected with both p55/p75NTR and p55/TrkA chimeric receptors.

Discussion

The work presented here stems from our original observation that iNOS expression and subsequent NO production can be synergistically induced by NGF and TNFα in a TrkA-dependent manner in PC12 cells [43]. Our present results investigated the signalling pathways involved. Since we consistently observed a higher iNOS expression if NGF is added simultaneously to TNFα, we propose that iNOS expression was induced selectively in NGF-responsive cells. These results do not allow us to rule out the possibility that intermediate factors induced by TNFα or NGF may play a role in sensitizing indirectly cells to NGF or TNFα, respectively. However, the results shown in Figure 2 seem to exclude such a possibility. Indeed, while withdrawal of NGF and/or TNFα allows for a prompt ablation of iNOS expression (Figure 2B), neither NGF nor TNFα alone is sufficient to sustain iNOS expression following withdrawal of TNFα or NGF (Figure 2C). These observations suggest that the simultaneous and continuous presence of both factors is required to sustain iNOS induction/expression and that cell sensitization through a priming mechanism seems unlikely. Nonetheless, other researchers have attributed increased TNFα toxicity in PC12 cells to NGF-induced differentiation [67]. However, our results seem to exclude that differentiation of PC12 cells may have played a role. First, in our experimental conditions iNOS expression occurs as early as 3 hr after the exposure to the combined NGF/TNFα treatment [43], earlier than any morphological differentiation induced by NGF. Second, while blockade of NGF-induced differentiation by the MAPK inhibitor PD98059 (Figure 5C, [68]) had no effect on NGF/TNFα-promoted iNOS expression (Figure 5A), blockade of NFκB did not affect NGF-induced differentiation (Figure 5C) but completely inhibited iNOS expression.
In the present study we also report that induction and maintenance of iNOS expression by the combined NGF/TNFα treatment requires continuous de novo iNOS mRNA synthesis, presumably due to transcription factor regulation. Indeed, abolishing iNOS enzymatic activity had no effect on NGF/TNFα-promoted iNOS induction (Figure 4A,B). Therefore, the involvement of positive feedback due to NO seems unlikely. On the other hand, analysis of transcriptional activity of NF-κB, AP-1 and CRE revealed that NF-κB most likely mediates synergistic iNOS induction by TNFα and NGF. Since iNOS induction can be observed as early as 3 hr after NGF/TNFα combined treatment in PC12 cells [43], the results shown in figure 5 suggest that NF-κB is the only transcription factor among those tested here that is responsive to the simultaneous treatment with TNFα and NGF in a fashion consistent with induction of iNOS expression. In fact, while TNFα alone induced NFκB at 3 hr, this induction was significantly lower than the one promoted by the combined NGF/TNFα treatment. Whether the extent to which NFκB is activated or whether qualitative differences in NFκB subunit composition in response to TNFα as compared to NGF/TNFα treatment may play a role in inducing iNOS expression remains to be established. Nonetheless, inhibition of NF-κB completely inhibited iNOS induction while inhibition of MAPK was ineffective (Figure 5A). Lastly, inhibition of NOS activity failed to block NGF/TNFα-promoted NFκB activation, thus further supporting the idea that targeting NO may acutely ameliorate associated oxidative stress, but could not represent the most comprehensive approach to achieve a long term correction of these events.
Previous studies indicated that NGF can induce NF-κB by acting through the low affinity p75NTR receptor [70]. Thus, involvement of NF-κB in mediating NGF/TNFα combined effects would suggest a role for p75NTR. Indeed, we found that mutant PC12 cells that lack expression of the p75NTR receptor failed to respond in terms of iNOS expression when simultaneously treated with NGF and TNFα. Consistent with this finding, in PC12 cell mutants lacking p75NTR expression NF-κB activity was not induced by the combined NGF/TNFα treatment above the levels observed in cells treated with TNFα alone (Figure 6B).
That PC12 cells bearing only the TrkA receptor failed to respond the combined NGF/TNFα treatment suggests that signaling from p75NTR in combination with TNFα is necessary to induce iNOS expression. On the other hand, our previous work illustrated the importance of TrkA-associated signaling in mediating NGF/TNFα-promoted induction of iNOS [43] (see also figure 1). These results are only apparently in contrast. Indeed, in an admittedly artificial system making use of chimeric constructs we observed that only in the presence of both TNFα-responsive NGF receptor signaling can TNFα promote iNOS expression when added alone. Whether this is a consequence of simultaneous but independent signaling of both types of NGF receptors [79] or recruitment of intracellular signalling elements uniquely driven by the simultaneous activation of both NGF receptors' signaling domains remains to be investigated. On the other hand, these results exclude the possibility that the combined action of TNFα and NGF may derive from yet undescribed interaction(s) of the extracellular domains of their respective receptors following ligand binding.
Thus, our combined results would indicate that there exists a specific pathway involving NF-κB and requiring the simultaneous expression or both types of NGF receptors that is synergistically induced by TNFα and NGF to promote expression of iNOS. This is of particular interest given that neuron types expressing both TrkA and p75NTR receptors are limited and known to be affected in neurodegenerative conditions where neuroinflammation and pro-inflammatory cytokines have been shown to play a significant role. Notably, simultaneous expression of TrkA and p75NTR in the CNS is mostly restricted to the BFCN that are known to be particularly affected in AD. Indeed, others have also described signaling pathways that require the simultaneous expression of both TrkA and p75NTR [71, 72] as well as the convergence of TrkA and p75NTR-mediated signaling impinging upon NF-κB [73]. Recent reports in neurons of TNF-promoted signaling occurring selectively in the presence of the glutamate agonist NMDA [4] illustrate the importance of considering the signaling "context" when studying the effects of cytokine treatment.
Overall, our data indicate the possibility that a convergence between NGF-promoted trophic signaling and TNFα could selectively endanger NGF-responsive neurons under conditions of neuroinflammation because of a synergistic action between TNFα and NGF to induce iNOS expression. For example, TNFα overexpressing transgenic mice show selective neurodegeneration of NGF-responsive basal forebrain cholinergic neurons [57] and direct TNFα administration in the brain of mice results in an impairment of basal forebrain cholinergic function [58]. However, whether induction of iNOS and subsequent oxidative damage may play a role in these two models remains to be determined [80].

Conclusion

TNFα and NGF, via concerted signaling events involving NFκB transcriptional activity and targeting NGF-responsive cells bearing both the high and low affinity NGF receptors, converge to stimulate de novo transcription of iNOS. Our present results are relevant to neurodegenerative conditions such as AD [22, 74], stroke [17, 75], ALS [20, 76] and spinal chord injury [8, 10] where neuroinflammation and high levels of pro-inflammatory cytokines have been shown to play a significant role and proposed as therapeutic targets.

Acknowledgements

This work was supported in part by a research development grant by the UTMB Sealy Endowed Fund for Biomedical Research. Michael Thomas is supported by an NIEHS training grant pre-doctoral fellowship from T32 ES007254 and the UTMB Sealy Center for Aging pre-doctoral fellowship.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​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 author(s) declare they have no competing interests.

Authors' contributions

MST participated in the conception and design of the study, carried out the bulk of experiments, performed data analysis, and drafted the manuscript. PMJ participated in study design especially with regards to the IGF experiments. WZ participated in study design and coordination and provided the expertise for RTPCR and withdrawal experiments. HUS sub-cloned the PC12p75NTR(-) cells and participated in study design and result interpretation of experiments involving these cells. GT participated in conception, study design, coordination and helped to draft and review the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Floyd RA: Neuroinflammatory processes are important in neurodegenerative diseases: an hypothesis to explain the increased formation of reactive oxygen and nitrogen species as major factors involved in neurodegenerative disease development. Free Radic Biol Med. 1999, 26: 1346-55. 10.1016/S0891-5849(98)00293-7.CrossRefPubMed Floyd RA: Neuroinflammatory processes are important in neurodegenerative diseases: an hypothesis to explain the increased formation of reactive oxygen and nitrogen species as major factors involved in neurodegenerative disease development. Free Radic Biol Med. 1999, 26: 1346-55. 10.1016/S0891-5849(98)00293-7.CrossRefPubMed
2.
Zurück zum Zitat McGeer PL, McGeer EG: Innate immunity, local inflammation, and degenerative disease. Sci Aging Knowledge Environ. 2002, 2002: re3.CrossRefPubMed McGeer PL, McGeer EG: Innate immunity, local inflammation, and degenerative disease. Sci Aging Knowledge Environ. 2002, 2002: re3.CrossRefPubMed
3.
Zurück zum Zitat Creange A, Lefaucheur JP, Authier FJ, Gherardi RK: Cytokines and peripheral neuropathies. Revue Neurologique. 1998, 154: 208-216.PubMed Creange A, Lefaucheur JP, Authier FJ, Gherardi RK: Cytokines and peripheral neuropathies. Revue Neurologique. 1998, 154: 208-216.PubMed
4.
Zurück zum Zitat Sung YJ, Ambron RT: Pathways that elicit long-term changes in gene expression in nociceptive neurons following nerve injury: contributions to neuropathic pain. Neurol Res. 2004, 26: 195-203. 10.1179/016164104225013761.CrossRefPubMed Sung YJ, Ambron RT: Pathways that elicit long-term changes in gene expression in nociceptive neurons following nerve injury: contributions to neuropathic pain. Neurol Res. 2004, 26: 195-203. 10.1179/016164104225013761.CrossRefPubMed
5.
Zurück zum Zitat Chandross KJ: Nerve injury and inflammatory cytokines modulate gap junctions in the peripheral nervous system. Glia. 1998, 24: 21-31. 10.1002/(SICI)1098-1136(199809)24:1<21::AID-GLIA3>3.0.CO;2-3.CrossRefPubMed Chandross KJ: Nerve injury and inflammatory cytokines modulate gap junctions in the peripheral nervous system. Glia. 1998, 24: 21-31. 10.1002/(SICI)1098-1136(199809)24:1<21::AID-GLIA3>3.0.CO;2-3.CrossRefPubMed
6.
Zurück zum Zitat Stoll G, Jander S, Myers RR: Degeneration and regeneration of the peripheral nervous system: from Augustus Waller's observations to neuroinflammation. J Peripher Nerv Syst. 2002, 7: 13-27. 10.1046/j.1529-8027.2002.02002.x.CrossRefPubMed Stoll G, Jander S, Myers RR: Degeneration and regeneration of the peripheral nervous system: from Augustus Waller's observations to neuroinflammation. J Peripher Nerv Syst. 2002, 7: 13-27. 10.1046/j.1529-8027.2002.02002.x.CrossRefPubMed
7.
Zurück zum Zitat Norenberg MD, Smith J, Marcillo A: The pathology of human spinal cord injury: defining the problems. J Neurotrauma. 2004, 21: 429-40. 10.1089/089771504323004575.CrossRefPubMed Norenberg MD, Smith J, Marcillo A: The pathology of human spinal cord injury: defining the problems. J Neurotrauma. 2004, 21: 429-40. 10.1089/089771504323004575.CrossRefPubMed
8.
Zurück zum Zitat La Rosa G, Cardali S, Genovese T, Conti A, Di Paola R, La Torre D, Cacciola F, Cuzzocrea S: Inhibition of the nuclear factor-kappaB activation with pyrrolidine dithiocarbamate attenuating inflammation and oxidative stress after experimental spinal cord trauma in rats. J Neurosurg Spine. 2004, 1: 311-21.CrossRefPubMed La Rosa G, Cardali S, Genovese T, Conti A, Di Paola R, La Torre D, Cacciola F, Cuzzocrea S: Inhibition of the nuclear factor-kappaB activation with pyrrolidine dithiocarbamate attenuating inflammation and oxidative stress after experimental spinal cord trauma in rats. J Neurosurg Spine. 2004, 1: 311-21.CrossRefPubMed
9.
Zurück zum Zitat Popovich PG, Jones TB: Manipulating neuroinflammatory reactions in the injured spinal cord: back to basics. Trends Pharmacol Sci. 2003, 24: 13-7. 10.1016/S0165-6147(02)00006-8.CrossRefPubMed Popovich PG, Jones TB: Manipulating neuroinflammatory reactions in the injured spinal cord: back to basics. Trends Pharmacol Sci. 2003, 24: 13-7. 10.1016/S0165-6147(02)00006-8.CrossRefPubMed
10.
Zurück zum Zitat Hausmann ON: Post-traumatic inflammation following spinal cord injury. Spinal Cord. 2003, 41: 369-78. 10.1038/sj.sc.3101483.CrossRefPubMed Hausmann ON: Post-traumatic inflammation following spinal cord injury. Spinal Cord. 2003, 41: 369-78. 10.1038/sj.sc.3101483.CrossRefPubMed
11.
Zurück zum Zitat Bareyre FM, Schwab ME: Inflammation, degeneration and regeneration in the injured spinal cord: insights from DNA microarrays. Trends Neurosci. 2003, 26: 555-63. 10.1016/j.tins.2003.08.004.CrossRefPubMed Bareyre FM, Schwab ME: Inflammation, degeneration and regeneration in the injured spinal cord: insights from DNA microarrays. Trends Neurosci. 2003, 26: 555-63. 10.1016/j.tins.2003.08.004.CrossRefPubMed
12.
Zurück zum Zitat Bayir H, Kochanek PM, Clark RS: Traumatic brain injury in infants and children: mechanisms of secondary damage and treatment in the intensive care unit. Crit Care Clin. 2003, 19: 529-49. 10.1016/S0749-0704(03)00014-9.CrossRefPubMed Bayir H, Kochanek PM, Clark RS: Traumatic brain injury in infants and children: mechanisms of secondary damage and treatment in the intensive care unit. Crit Care Clin. 2003, 19: 529-49. 10.1016/S0749-0704(03)00014-9.CrossRefPubMed
13.
Zurück zum Zitat Morganti-Kossmann MC, Rancan M, Stahel PF, Kossmann T: Inflammatory response in acute traumatic brain injury: a double-edged sword. Curr Opin Crit Care. 2002, 8: 101-5. 10.1097/00075198-200204000-00002.CrossRefPubMed Morganti-Kossmann MC, Rancan M, Stahel PF, Kossmann T: Inflammatory response in acute traumatic brain injury: a double-edged sword. Curr Opin Crit Care. 2002, 8: 101-5. 10.1097/00075198-200204000-00002.CrossRefPubMed
14.
Zurück zum Zitat Lenzlinger PM, Morganti-Kossmann MC, Laurer HL, McIntosh TK: The duality of the inflammatory response to traumatic brain injury. Mol Neurobiol. 2001, 24: 169-81. 10.1385/MN:24:1-3:169.CrossRefPubMed Lenzlinger PM, Morganti-Kossmann MC, Laurer HL, McIntosh TK: The duality of the inflammatory response to traumatic brain injury. Mol Neurobiol. 2001, 24: 169-81. 10.1385/MN:24:1-3:169.CrossRefPubMed
15.
Zurück zum Zitat Sundararajan S, Landreth GE: Antiinflammatory properties of PPARgamma agonists following ischemia. Drug News Perspect. 2004, 17: 229-36. 10.1358/dnp.2004.17.4.829049.CrossRefPubMed Sundararajan S, Landreth GE: Antiinflammatory properties of PPARgamma agonists following ischemia. Drug News Perspect. 2004, 17: 229-36. 10.1358/dnp.2004.17.4.829049.CrossRefPubMed
16.
Zurück zum Zitat Dirnagl U: Inflammation in stroke: the good, the bad, and the unknown. Ernst Schering Res Found Workshop. 2004, 87-99. Dirnagl U: Inflammation in stroke: the good, the bad, and the unknown. Ernst Schering Res Found Workshop. 2004, 87-99.
17.
Zurück zum Zitat Danton GH, Dietrich WD: Inflammatory mechanisms after ischemia and stroke. J Neuropathol Exp Neurol. 2003, 62: 127-36.PubMed Danton GH, Dietrich WD: Inflammatory mechanisms after ischemia and stroke. J Neuropathol Exp Neurol. 2003, 62: 127-36.PubMed
18.
Zurück zum Zitat Consilvio C, Vincent AM, Feldman EL: Neuroinflammation, COX-2, and ALS – a dual role?. Exp Neurol. 2004, 187: 1-10. 10.1016/j.expneurol.2003.12.009.CrossRefPubMed Consilvio C, Vincent AM, Feldman EL: Neuroinflammation, COX-2, and ALS – a dual role?. Exp Neurol. 2004, 187: 1-10. 10.1016/j.expneurol.2003.12.009.CrossRefPubMed
19.
Zurück zum Zitat Pompl PN, Ho L, Bianchi M, McManus T, Qin W, Pasinetti GM: A therapeutic role for cyclooxygenase-2 inhibitors in a transgenic mouse model of amyotrophic lateral sclerosis. Faseb J. 2003, 17: 725-7.PubMed Pompl PN, Ho L, Bianchi M, McManus T, Qin W, Pasinetti GM: A therapeutic role for cyclooxygenase-2 inhibitors in a transgenic mouse model of amyotrophic lateral sclerosis. Faseb J. 2003, 17: 725-7.PubMed
20.
Zurück zum Zitat McGeer PL, McGeer EG: Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve. 2002, 26: 459-70. 10.1002/mus.10191.CrossRefPubMed McGeer PL, McGeer EG: Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve. 2002, 26: 459-70. 10.1002/mus.10191.CrossRefPubMed
21.
Zurück zum Zitat Cacquevel M, Lebeurrier N, Cheenne S, Vivien D: Cytokines in neuroinflammation and Alzheimer's disease. Curr Drug Targets. 2004, 5: 529-34. 10.2174/1389450043345308.CrossRefPubMed Cacquevel M, Lebeurrier N, Cheenne S, Vivien D: Cytokines in neuroinflammation and Alzheimer's disease. Curr Drug Targets. 2004, 5: 529-34. 10.2174/1389450043345308.CrossRefPubMed
22.
Zurück zum Zitat McGeer EG, McGeer PL: Inflammatory processes in Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry. 2003, 27: 741-9. 10.1016/S0278-5846(03)00124-6.CrossRefPubMed McGeer EG, McGeer PL: Inflammatory processes in Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry. 2003, 27: 741-9. 10.1016/S0278-5846(03)00124-6.CrossRefPubMed
23.
Zurück zum Zitat Gupta A, Pansari K: Inflammation and Alzheimer's disease. Int J Clin Pract. 2003, 57: 36-9.PubMed Gupta A, Pansari K: Inflammation and Alzheimer's disease. Int J Clin Pract. 2003, 57: 36-9.PubMed
24.
Zurück zum Zitat McGeer PL, McGeer EG: Local neuroinflammation and the progression of Alzheimer's disease. J Neurovirol. 2002, 8: 529-38. 10.1080/13550280290100969.CrossRefPubMed McGeer PL, McGeer EG: Local neuroinflammation and the progression of Alzheimer's disease. J Neurovirol. 2002, 8: 529-38. 10.1080/13550280290100969.CrossRefPubMed
25.
Zurück zum Zitat Emerit J, Edeas M, Bricaire F: Neurodegenerative diseases and oxidative stress. Biomed Pharmacother. 2004, 58: 39-46. 10.1016/j.biopha.2003.11.004.CrossRefPubMed Emerit J, Edeas M, Bricaire F: Neurodegenerative diseases and oxidative stress. Biomed Pharmacother. 2004, 58: 39-46. 10.1016/j.biopha.2003.11.004.CrossRefPubMed
26.
Zurück zum Zitat Tarkowski E, Liljeroth AM, Minthon L, Tarkowski A, Wallin A, Blennow K: Cerebral pattern of pro- and anti-inflammatory cytokines in dementias. Brain Res Bull. 2003, 61: 255-60. 10.1016/S0361-9230(03)00088-1.CrossRefPubMed Tarkowski E, Liljeroth AM, Minthon L, Tarkowski A, Wallin A, Blennow K: Cerebral pattern of pro- and anti-inflammatory cytokines in dementias. Brain Res Bull. 2003, 61: 255-60. 10.1016/S0361-9230(03)00088-1.CrossRefPubMed
27.
Zurück zum Zitat Tarkowski E, Andreasen N, Tarkowski A, Blennow K: Intrathecal inflammation precedes development of Alzheimer's disease. J Neurol Neurosurg Psychiatry. 2003, 74: 1200-5. 10.1136/jnnp.74.9.1200.PubMedCentralCrossRefPubMed Tarkowski E, Andreasen N, Tarkowski A, Blennow K: Intrathecal inflammation precedes development of Alzheimer's disease. J Neurol Neurosurg Psychiatry. 2003, 74: 1200-5. 10.1136/jnnp.74.9.1200.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat McGeer PL, McGeer EG, Suzuki J, Dolman CE, Nagai T: Aging, Alzheimer's disease, and the cholinergic system of the basal forebrain. Neurology. 1984, 34: 741-5.CrossRefPubMed McGeer PL, McGeer EG, Suzuki J, Dolman CE, Nagai T: Aging, Alzheimer's disease, and the cholinergic system of the basal forebrain. Neurology. 1984, 34: 741-5.CrossRefPubMed
29.
Zurück zum Zitat Rinne JO, Paljarvi L, Rinne UK: Neuronal size and density in the nucleus basalis of Meynert in Alzheimer's disease. J Neurol Sci. 1987, 79: 67-76. 10.1016/0022-510X(87)90260-7.CrossRefPubMed Rinne JO, Paljarvi L, Rinne UK: Neuronal size and density in the nucleus basalis of Meynert in Alzheimer's disease. J Neurol Sci. 1987, 79: 67-76. 10.1016/0022-510X(87)90260-7.CrossRefPubMed
30.
Zurück zum Zitat Vogels OJ, Broere CA, ter Laak HJ, ten Donkelaar HJ, Nieuwenhuys R, Schulte BP: Cell loss and shrinkage in the nucleus basalis Meynert complex in Alzheimer's disease. Neurobiology of Aging. 1990, 11: 3-13. 10.1016/0197-4580(90)90056-6.CrossRefPubMed Vogels OJ, Broere CA, ter Laak HJ, ten Donkelaar HJ, Nieuwenhuys R, Schulte BP: Cell loss and shrinkage in the nucleus basalis Meynert complex in Alzheimer's disease. Neurobiology of Aging. 1990, 11: 3-13. 10.1016/0197-4580(90)90056-6.CrossRefPubMed
31.
Zurück zum Zitat Hefti F: Nerve growth factor promotes survival of septal cholinergic neurons after fimbrial transections. J Neurosci. 1986, 6: 2155-62.PubMed Hefti F: Nerve growth factor promotes survival of septal cholinergic neurons after fimbrial transections. J Neurosci. 1986, 6: 2155-62.PubMed
32.
Zurück zum Zitat Hartikka J, Hefti F: Development of septal cholinergic neurons in culture: plating density and glial cells modulate effects of NGF on survival, fiber growth, and expression of transmitter-specific enzymes. J Neurosci. 1988, 8: 2967-85.PubMed Hartikka J, Hefti F: Development of septal cholinergic neurons in culture: plating density and glial cells modulate effects of NGF on survival, fiber growth, and expression of transmitter-specific enzymes. J Neurosci. 1988, 8: 2967-85.PubMed
33.
Zurück zum Zitat Perry G, Castellani RJ, Smith MA, Harris PL, Kubat Z, Ghanbari K, Jones PK, Cordone G, Tabaton M, Wolozin B: Oxidative damage in the olfactory system in Alzheimer's disease. Acta Neuropathol (Berl). 2003, 106: 552-6. 10.1007/s00401-003-0761-7.CrossRef Perry G, Castellani RJ, Smith MA, Harris PL, Kubat Z, Ghanbari K, Jones PK, Cordone G, Tabaton M, Wolozin B: Oxidative damage in the olfactory system in Alzheimer's disease. Acta Neuropathol (Berl). 2003, 106: 552-6. 10.1007/s00401-003-0761-7.CrossRef
34.
Zurück zum Zitat Butterfield DA, Boyd-Kimball D, Castegna A: Proteomics in Alzheimer's disease: insights into potential mechanisms of neurodegeneration. J Neurochem. 2003, 86: 1313-27. 10.1046/j.1471-4159.2003.01948.x.CrossRefPubMed Butterfield DA, Boyd-Kimball D, Castegna A: Proteomics in Alzheimer's disease: insights into potential mechanisms of neurodegeneration. J Neurochem. 2003, 86: 1313-27. 10.1046/j.1471-4159.2003.01948.x.CrossRefPubMed
35.
Zurück zum Zitat Giasson BI, Ischiropoulos H, Lee VM, Trojanowski JQ: The relationship between oxidative/nitrative stress and pathological inclusions in Alzheimer's and Parkinson's diseases. Free Radic Biol Med. 2002, 32: 1264-75. 10.1016/S0891-5849(02)00804-3.CrossRefPubMed Giasson BI, Ischiropoulos H, Lee VM, Trojanowski JQ: The relationship between oxidative/nitrative stress and pathological inclusions in Alzheimer's and Parkinson's diseases. Free Radic Biol Med. 2002, 32: 1264-75. 10.1016/S0891-5849(02)00804-3.CrossRefPubMed
36.
Zurück zum Zitat Perry G, Nunomura A, Hirai K, Takeda A, Aliev G, Smith MA: Oxidative damage in Alzheimer's disease: the metabolic dimension. Int J Dev Neurosci. 2000, 18: 417-21. 10.1016/S0736-5748(00)00006-X.CrossRefPubMed Perry G, Nunomura A, Hirai K, Takeda A, Aliev G, Smith MA: Oxidative damage in Alzheimer's disease: the metabolic dimension. Int J Dev Neurosci. 2000, 18: 417-21. 10.1016/S0736-5748(00)00006-X.CrossRefPubMed
37.
Zurück zum Zitat Gold BG, Udina E, Bourdette D, Navarro X: Neuroregenerative and neuroprotective actions of neuroimmunophilin compounds in traumatic and inflammatory neuropathies. Neurol Res. 2004, 26: 371-80. 10.1179/016164104225013734.CrossRefPubMed Gold BG, Udina E, Bourdette D, Navarro X: Neuroregenerative and neuroprotective actions of neuroimmunophilin compounds in traumatic and inflammatory neuropathies. Neurol Res. 2004, 26: 371-80. 10.1179/016164104225013734.CrossRefPubMed
38.
Zurück zum Zitat Rogerio F, Teixeira SA, de Rezende AC, de Sa RC, de Souza Queiroz L, De Nucci G, Muscara MN, Langone F: Superoxide dismutase isoforms 1 and 2 in lumbar spinal cord of neonatal rats after sciatic nerve transection and melatonin treatment. Brain Res Dev Brain Res. 2005, 154: 217-25. 10.1016/j.devbrainres.2004.10.017.CrossRefPubMed Rogerio F, Teixeira SA, de Rezende AC, de Sa RC, de Souza Queiroz L, De Nucci G, Muscara MN, Langone F: Superoxide dismutase isoforms 1 and 2 in lumbar spinal cord of neonatal rats after sciatic nerve transection and melatonin treatment. Brain Res Dev Brain Res. 2005, 154: 217-25. 10.1016/j.devbrainres.2004.10.017.CrossRefPubMed
39.
Zurück zum Zitat Kim HK, Park SK, Zhou JL, Taglialatela G, Chung K, Coggeshall RE, Chung JM: Reactive oxygen species (ROS) play an important role in a rat model of neuropathic pain. Pain. 2004, 111: 116-24. 10.1016/j.pain.2004.06.008.CrossRefPubMed Kim HK, Park SK, Zhou JL, Taglialatela G, Chung K, Coggeshall RE, Chung JM: Reactive oxygen species (ROS) play an important role in a rat model of neuropathic pain. Pain. 2004, 111: 116-24. 10.1016/j.pain.2004.06.008.CrossRefPubMed
40.
Zurück zum Zitat van Beek J, Elward K, Gasque P: Activation of complement in the central nervous system: roles in neurodegeneration and neuroprotection. Ann N Y Acad Sci. 2003, 992: 56-71.CrossRefPubMed van Beek J, Elward K, Gasque P: Activation of complement in the central nervous system: roles in neurodegeneration and neuroprotection. Ann N Y Acad Sci. 2003, 992: 56-71.CrossRefPubMed
41.
Zurück zum Zitat Murdoch I, Perry EK, Court JA, Graham DI, Dewar D: Cortical cholinergic dysfunction after human head injury. J Neurotrauma. 1998, 15: 295-305.CrossRefPubMed Murdoch I, Perry EK, Court JA, Graham DI, Dewar D: Cortical cholinergic dysfunction after human head injury. J Neurotrauma. 1998, 15: 295-305.CrossRefPubMed
42.
Zurück zum Zitat Murdoch I, Nicoll JA, Graham DI, Dewar D: Nucleus basalis of Meynert pathology in the human brain after fatal head injury. J Neurotrauma. 2002, 19: 279-84. 10.1089/08977150252807018.CrossRefPubMed Murdoch I, Nicoll JA, Graham DI, Dewar D: Nucleus basalis of Meynert pathology in the human brain after fatal head injury. J Neurotrauma. 2002, 19: 279-84. 10.1089/08977150252807018.CrossRefPubMed
43.
Zurück zum Zitat Macdonald NJ, Taglialatela G: Tumor necrosis factor alpha and nerve growth factor synergistically induce iNOS in pheochromocytoma cells. Neuroreport. 2000, 11: 3453-3456.CrossRefPubMed Macdonald NJ, Taglialatela G: Tumor necrosis factor alpha and nerve growth factor synergistically induce iNOS in pheochromocytoma cells. Neuroreport. 2000, 11: 3453-3456.CrossRefPubMed
44.
Zurück zum Zitat Isaksson J, Farooque M, Olsson Y: Improved functional outcome after spinal cord injury in iNOS-deficient mice. Spinal Cord. 2004 Isaksson J, Farooque M, Olsson Y: Improved functional outcome after spinal cord injury in iNOS-deficient mice. Spinal Cord. 2004
45.
Zurück zum Zitat Urushitani M, Shimohama S, Kihara T, Sawada H, Akaike A, Ibi M, Inoue R, Kitamura Y, Taniguchi T, Kimura J: Mechanism of selective motor neuronal death after exposure of spinal cord to glutamate: Involvement of glutamate induced nitric oxide in motor neuron toxicity and nonmotor neuron protection. Annals of Neurology. 1998, 44: 796-807. 10.1002/ana.410440514.CrossRefPubMed Urushitani M, Shimohama S, Kihara T, Sawada H, Akaike A, Ibi M, Inoue R, Kitamura Y, Taniguchi T, Kimura J: Mechanism of selective motor neuronal death after exposure of spinal cord to glutamate: Involvement of glutamate induced nitric oxide in motor neuron toxicity and nonmotor neuron protection. Annals of Neurology. 1998, 44: 796-807. 10.1002/ana.410440514.CrossRefPubMed
46.
Zurück zum Zitat Diaz-Ruiz A, Ibarra A, Perez-Severiano F, Guizar-Sahagun G, Grijalva I, Rios C: Constitutive and inducible nitric oxide synthase activities after spinal cord contusion in rats. Neurosci Lett. 2002, 319: 129-32. 10.1016/S0304-3940(01)02540-X.CrossRefPubMed Diaz-Ruiz A, Ibarra A, Perez-Severiano F, Guizar-Sahagun G, Grijalva I, Rios C: Constitutive and inducible nitric oxide synthase activities after spinal cord contusion in rats. Neurosci Lett. 2002, 319: 129-32. 10.1016/S0304-3940(01)02540-X.CrossRefPubMed
47.
Zurück zum Zitat Parmentier-Batteur S, Bohme GA, Lerouet D, Zhou-Ding L, Beray V, Margaill I, Plotkine M: Antisense oligodeoxynucleotide to inducible nitric oxide synthase protects against transient focal cerebral ischemia-induced brain injury. Journal of Cerebral Blood Flow & Metabolism. 2001, 21: 15-21. 10.1097/00004647-200101000-00003.CrossRef Parmentier-Batteur S, Bohme GA, Lerouet D, Zhou-Ding L, Beray V, Margaill I, Plotkine M: Antisense oligodeoxynucleotide to inducible nitric oxide synthase protects against transient focal cerebral ischemia-induced brain injury. Journal of Cerebral Blood Flow & Metabolism. 2001, 21: 15-21. 10.1097/00004647-200101000-00003.CrossRef
48.
Zurück zum Zitat Sarchielli P, Galli F, Floridi A, Gallai V: Relevance of protein nitration in brain injury: a key pathophysiological mechanism in neurodegenerative, autoimmune, or inflammatory CNS diseases and stroke. Amino Acids. 2003, 25: 427-36. 10.1007/s00726-003-0028-6.CrossRefPubMed Sarchielli P, Galli F, Floridi A, Gallai V: Relevance of protein nitration in brain injury: a key pathophysiological mechanism in neurodegenerative, autoimmune, or inflammatory CNS diseases and stroke. Amino Acids. 2003, 25: 427-36. 10.1007/s00726-003-0028-6.CrossRefPubMed
49.
Zurück zum Zitat Luth HJ, Munch G, Arendt T: Aberrant expression of NOS isoforms in Alzheimer's disease is structurally related to nitrotyrosine formation. Brain Res. 2002, 953: 135-43. 10.1016/S0006-8993(02)03280-8.CrossRefPubMed Luth HJ, Munch G, Arendt T: Aberrant expression of NOS isoforms in Alzheimer's disease is structurally related to nitrotyrosine formation. Brain Res. 2002, 953: 135-43. 10.1016/S0006-8993(02)03280-8.CrossRefPubMed
50.
Zurück zum Zitat Luth HJ: Expression of endothelial and inducible NOS-isoforms is increased in Alzheimer's disease, in APP23 transgenic mice and after experimental brain lesion in rat: evidence for an induction by amyloid pathology. Brain Research. 2001, 913: 57-67. 10.1016/S0006-8993(01)02758-5.CrossRefPubMed Luth HJ: Expression of endothelial and inducible NOS-isoforms is increased in Alzheimer's disease, in APP23 transgenic mice and after experimental brain lesion in rat: evidence for an induction by amyloid pathology. Brain Research. 2001, 913: 57-67. 10.1016/S0006-8993(01)02758-5.CrossRefPubMed
51.
Zurück zum Zitat Law A, Gauthier S, Quirion R: Say NO to Alzheimer's disease: the putative links between nitric oxide and dementia of the Alzheimer's type. Brain Res Brain Res Rev. 2001, 35: 73-96. 10.1016/S0165-0173(00)00051-5.CrossRefPubMed Law A, Gauthier S, Quirion R: Say NO to Alzheimer's disease: the putative links between nitric oxide and dementia of the Alzheimer's type. Brain Res Brain Res Rev. 2001, 35: 73-96. 10.1016/S0165-0173(00)00051-5.CrossRefPubMed
52.
Zurück zum Zitat de la Monte SM, Lu BX, Sohn YK, Etienne D, Kraft J, Ganju N, Wands JR: Aberrant expression of nitric oxide synthase III in Alzheimer's disease: relevance to cerebral vasculopathy and neurodegeneration. Neurobiology of Aging. 2000, 21: 309-19. 10.1016/S0197-4580(99)00108-6.CrossRefPubMed de la Monte SM, Lu BX, Sohn YK, Etienne D, Kraft J, Ganju N, Wands JR: Aberrant expression of nitric oxide synthase III in Alzheimer's disease: relevance to cerebral vasculopathy and neurodegeneration. Neurobiology of Aging. 2000, 21: 309-19. 10.1016/S0197-4580(99)00108-6.CrossRefPubMed
53.
Zurück zum Zitat Lee S, Zhao ML, Hirano A, Dickson D: Inducible nitric oxide synthase immunoreactivity in the Alzheimer disease hippocampus: association with Hirano bodies, neurofibrillary tangles, and senile plaques. Journal of Neuropathology and Experimental Neurology. 1999, 58: 1163-1169.CrossRefPubMed Lee S, Zhao ML, Hirano A, Dickson D: Inducible nitric oxide synthase immunoreactivity in the Alzheimer disease hippocampus: association with Hirano bodies, neurofibrillary tangles, and senile plaques. Journal of Neuropathology and Experimental Neurology. 1999, 58: 1163-1169.CrossRefPubMed
54.
Zurück zum Zitat Darville MI, Eizirik DL: Regulation by cytokines of the inducible nitric oxide synthase promoter in insulin-producing cells. Diabetologia. 1998, 41: 1101-8. 10.1007/s001250051036.CrossRefPubMed Darville MI, Eizirik DL: Regulation by cytokines of the inducible nitric oxide synthase promoter in insulin-producing cells. Diabetologia. 1998, 41: 1101-8. 10.1007/s001250051036.CrossRefPubMed
55.
Zurück zum Zitat Kong GY, Peng ZC, Costanzo C, Kristensson K, Bentivoglio M: Inducible nitric oxide synthase expression elicited in the mouse brain by inflammatory mediators circulating in the cerebrospinal fluid. Brain Res. 2000, 878: 105-18. 10.1016/S0006-8993(00)02716-5.CrossRefPubMed Kong GY, Peng ZC, Costanzo C, Kristensson K, Bentivoglio M: Inducible nitric oxide synthase expression elicited in the mouse brain by inflammatory mediators circulating in the cerebrospinal fluid. Brain Res. 2000, 878: 105-18. 10.1016/S0006-8993(00)02716-5.CrossRefPubMed
56.
Zurück zum Zitat Barker V, Middleton G, Davey F, Davies AM: TNFalpha contributes to the death of NGF-dependent neurons during development. Nature Neuroscience. 2001, 4: 1194-8. 10.1038/nn755.CrossRefPubMed Barker V, Middleton G, Davey F, Davies AM: TNFalpha contributes to the death of NGF-dependent neurons during development. Nature Neuroscience. 2001, 4: 1194-8. 10.1038/nn755.CrossRefPubMed
57.
Zurück zum Zitat Aloe L, Fiore M, Probert L, Turrini P, Tirassa P: Overexpression of tumour necrosis factor alpha in the brain of transgenic mice differentially alters nerve growth factor levels and choline acetyltransferase activity. Cytokine. 1999, 11: 45-54. 10.1006/cyto.1998.0397.CrossRefPubMed Aloe L, Fiore M, Probert L, Turrini P, Tirassa P: Overexpression of tumour necrosis factor alpha in the brain of transgenic mice differentially alters nerve growth factor levels and choline acetyltransferase activity. Cytokine. 1999, 11: 45-54. 10.1006/cyto.1998.0397.CrossRefPubMed
58.
Zurück zum Zitat Wenk GL, McGann K, Hauss-Wegrzyniak B, Rosi S: The toxicity of tumor necrosis factor-alpha upon cholinergic neurons within the nucleus basalis and the role of norepinephrine in the regulation of inflammation: implications for alzheimer's disease. Neuroscience. 2003, 121: 719-29. 10.1016/S0306-4522(03)00545-1.CrossRefPubMed Wenk GL, McGann K, Hauss-Wegrzyniak B, Rosi S: The toxicity of tumor necrosis factor-alpha upon cholinergic neurons within the nucleus basalis and the role of norepinephrine in the regulation of inflammation: implications for alzheimer's disease. Neuroscience. 2003, 121: 719-29. 10.1016/S0306-4522(03)00545-1.CrossRefPubMed
59.
Zurück zum Zitat Kiss JZ, Wang C, Olive S, Rougon G, Lang J, Baetens D, Harry D, Pralong WF: Activity-dependent mobilization of the adhesion molecule polysialic NCAM to the cell surface of neurons and endocrine cells. Embo J. 1994, 13: 5284-92.PubMedCentralPubMed Kiss JZ, Wang C, Olive S, Rougon G, Lang J, Baetens D, Harry D, Pralong WF: Activity-dependent mobilization of the adhesion molecule polysialic NCAM to the cell surface of neurons and endocrine cells. Embo J. 1994, 13: 5284-92.PubMedCentralPubMed
60.
Zurück zum Zitat Stoppini L, Buchs PA, Muller D: A simple method for organotypic cultures of nervous tissue. Journal of Neuroscience Methods. 1991, 37: 173-82. 10.1016/0165-0270(91)90128-M.CrossRefPubMed Stoppini L, Buchs PA, Muller D: A simple method for organotypic cultures of nervous tissue. Journal of Neuroscience Methods. 1991, 37: 173-82. 10.1016/0165-0270(91)90128-M.CrossRefPubMed
61.
Zurück zum Zitat Ohmichi M, Decker SJ, Pang L, Saltiel AR: Inhibition of the cellular actions of nerve growth factor by staurosporine and K252A results from the attenuation of the activity of the trk tyrosine kinase. Biochemistry. 1992, 31: 4034-9. 10.1021/bi00131a019.CrossRefPubMed Ohmichi M, Decker SJ, Pang L, Saltiel AR: Inhibition of the cellular actions of nerve growth factor by staurosporine and K252A results from the attenuation of the activity of the trk tyrosine kinase. Biochemistry. 1992, 31: 4034-9. 10.1021/bi00131a019.CrossRefPubMed
62.
Zurück zum Zitat Pappas TC, Decorti F, Macdonald NJ, Neet KE, Taglialatela G: Tumour necrosis factor-alpha- vs. growth factor deprivation-promoted cell death: different receptor requirements for mediating nerve growth factor-promoted rescue. Aging Cell. 2003, 2: 83-92. 10.1046/j.1474-9728.2003.00039.x.CrossRefPubMed Pappas TC, Decorti F, Macdonald NJ, Neet KE, Taglialatela G: Tumour necrosis factor-alpha- vs. growth factor deprivation-promoted cell death: different receptor requirements for mediating nerve growth factor-promoted rescue. Aging Cell. 2003, 2: 83-92. 10.1046/j.1474-9728.2003.00039.x.CrossRefPubMed
63.
Zurück zum Zitat Taglialatela G, Robinson R, Perez-Polo JR: Inhibition of nuclear factor kappa B (NFkappaB) activity induces nerve growth factor-resistant apoptosis in PC12 cells. Journal of Neuroscience Research. 1997, 47: 155-62. 10.1002/(SICI)1097-4547(19970115)47:2<155::AID-JNR4>3.0.CO;2-E.CrossRefPubMed Taglialatela G, Robinson R, Perez-Polo JR: Inhibition of nuclear factor kappa B (NFkappaB) activity induces nerve growth factor-resistant apoptosis in PC12 cells. Journal of Neuroscience Research. 1997, 47: 155-62. 10.1002/(SICI)1097-4547(19970115)47:2<155::AID-JNR4>3.0.CO;2-E.CrossRefPubMed
64.
Zurück zum Zitat Taglialatela G, Kaufmann JA, Trevino A, Perez-Polo JR: Central nervous system DNA fragmentation induced by the inhibition of nuclear factor kappa B. Neuroreport. 1998, 9: 489-93.CrossRefPubMed Taglialatela G, Kaufmann JA, Trevino A, Perez-Polo JR: Central nervous system DNA fragmentation induced by the inhibition of nuclear factor kappa B. Neuroreport. 1998, 9: 489-93.CrossRefPubMed
65.
Zurück zum Zitat Traenckner EB, Wilk S, Baeuerle PA: A proteasome inhibitor prevents activation of NF-kappa B and stabilizes a newly phosphorylated form of I kappa B-alpha that is still bound to NF-kappa B. Embo J. 1994, 13: 5433-41.PubMedCentralPubMed Traenckner EB, Wilk S, Baeuerle PA: A proteasome inhibitor prevents activation of NF-kappa B and stabilizes a newly phosphorylated form of I kappa B-alpha that is still bound to NF-kappa B. Embo J. 1994, 13: 5433-41.PubMedCentralPubMed
66.
Zurück zum Zitat Pang L, Sawada T, Decker SJ, Saltiel AR: Inhibition of MAP kinase kinase blocks the differentiation of PC-12 cells induced by nerve growth factor. Journal of Biological Chemistry. 1995, 270: 13585-8. 10.1074/jbc.270.36.21040.CrossRefPubMed Pang L, Sawada T, Decker SJ, Saltiel AR: Inhibition of MAP kinase kinase blocks the differentiation of PC-12 cells induced by nerve growth factor. Journal of Biological Chemistry. 1995, 270: 13585-8. 10.1074/jbc.270.36.21040.CrossRefPubMed
67.
Zurück zum Zitat Mielke K, Herdegen T: Fatal shift of signal transduction is an integral part of neuronal differentiation: JNKs realize TNFalpha-mediated apoptosis in neuronlike, but not naive, PC12 cells. Mol Cell Neurosci. 2002, 20: 211-24. 10.1006/mcne.2002.1132.CrossRefPubMed Mielke K, Herdegen T: Fatal shift of signal transduction is an integral part of neuronal differentiation: JNKs realize TNFalpha-mediated apoptosis in neuronlike, but not naive, PC12 cells. Mol Cell Neurosci. 2002, 20: 211-24. 10.1006/mcne.2002.1132.CrossRefPubMed
68.
Zurück zum Zitat Morooka T, Nishida E: Requirement of P38 Mitogen-Activated Protein Kinase For Neuronal Differentiation in Pc12 Cells. Journal of Biological Chemistry. 1998, 273: 24285-24288. 10.1074/jbc.273.38.24285.CrossRefPubMed Morooka T, Nishida E: Requirement of P38 Mitogen-Activated Protein Kinase For Neuronal Differentiation in Pc12 Cells. Journal of Biological Chemistry. 1998, 273: 24285-24288. 10.1074/jbc.273.38.24285.CrossRefPubMed
69.
Zurück zum Zitat Perez-Polo JR, Foreman PJ, Jackson GR, Shan D, Taglialatela G, Thorpe LW, Werrbach-Perez K: Nerve growth factor and neuronal cell death. Molecular Neurobiology. 1990, 4: 57-91.CrossRefPubMed Perez-Polo JR, Foreman PJ, Jackson GR, Shan D, Taglialatela G, Thorpe LW, Werrbach-Perez K: Nerve growth factor and neuronal cell death. Molecular Neurobiology. 1990, 4: 57-91.CrossRefPubMed
70.
Zurück zum Zitat Carter BD, Kaltschmidt C, Kaltschmidt B, Offenhauser N, Bohm-Matthaei R, Baeuerle PA, Barde YA: Selective activation of NF-kappa B by nerve growth factor through the neurotrophin receptor p75 [see comments]. Science. 1996, 272: 542-5.CrossRefPubMed Carter BD, Kaltschmidt C, Kaltschmidt B, Offenhauser N, Bohm-Matthaei R, Baeuerle PA, Barde YA: Selective activation of NF-kappa B by nerve growth factor through the neurotrophin receptor p75 [see comments]. Science. 1996, 272: 542-5.CrossRefPubMed
71.
Zurück zum Zitat Lad SP, Peterson DA, Bradshaw RA, Neet KE: Individual and combined effects of TrkA and p75NTR nerve growth factor receptors. A role for the high affinity receptor site. J Biol Chem. 2003, 278: 24808-17. 10.1074/jbc.M212270200.CrossRefPubMed Lad SP, Peterson DA, Bradshaw RA, Neet KE: Individual and combined effects of TrkA and p75NTR nerve growth factor receptors. A role for the high affinity receptor site. J Biol Chem. 2003, 278: 24808-17. 10.1074/jbc.M212270200.CrossRefPubMed
72.
Zurück zum Zitat Szutowicz A, Madziar B, Pawelczyk T, Tomaszewicz M, Bielarczyk H: Effects of NGF on acetylcholine, acetyl-CoA metabolism, and viability of differentiated and non-differentiated cholinergic neuroblastoma cells. J Neurochem. 2004, 90: 952-61. 10.1111/j.1471-4159.2004.02556.x.CrossRefPubMed Szutowicz A, Madziar B, Pawelczyk T, Tomaszewicz M, Bielarczyk H: Effects of NGF on acetylcholine, acetyl-CoA metabolism, and viability of differentiated and non-differentiated cholinergic neuroblastoma cells. J Neurochem. 2004, 90: 952-61. 10.1111/j.1471-4159.2004.02556.x.CrossRefPubMed
73.
Zurück zum Zitat Foehr ED, Lin X, O'Mahony A, Geleziunas R, Bradshaw RA, Greene WC: NF-kappa B signaling promotes both cell survival and neurite process formation in nerve growth factor-stimulated PC12 cells. J Neurosci. 2000, 20: 7556-63.PubMed Foehr ED, Lin X, O'Mahony A, Geleziunas R, Bradshaw RA, Greene WC: NF-kappa B signaling promotes both cell survival and neurite process formation in nerve growth factor-stimulated PC12 cells. J Neurosci. 2000, 20: 7556-63.PubMed
74.
Zurück zum Zitat Hoozemans JJ, Veerhuis R, Rozemuller AJ, Eikelenboom P: Non-steroidal anti-inflammatory drugs and cyclooxygenase in Alzheimer's disease. Curr Drug Targets. 2003, 4: 461-8. 10.2174/1389450033490902.CrossRefPubMed Hoozemans JJ, Veerhuis R, Rozemuller AJ, Eikelenboom P: Non-steroidal anti-inflammatory drugs and cyclooxygenase in Alzheimer's disease. Curr Drug Targets. 2003, 4: 461-8. 10.2174/1389450033490902.CrossRefPubMed
75.
Zurück zum Zitat Barone FC, Parsons AA: Therapeutic potential of anti-inflammatory drugs in focal stroke. Expert Opin Investig Drugs. 2000, 9: 2281-306. 10.1517/13543784.9.10.2281.CrossRefPubMed Barone FC, Parsons AA: Therapeutic potential of anti-inflammatory drugs in focal stroke. Expert Opin Investig Drugs. 2000, 9: 2281-306. 10.1517/13543784.9.10.2281.CrossRefPubMed
76.
Zurück zum Zitat Weydt P, Weiss MD, Moller T, Carter GT: Neuro-inflammation as a therapeutic target in amyotrophic lateral sclerosis. Curr Opin Investig Drugs. 2002, 3: 1720-4.PubMed Weydt P, Weiss MD, Moller T, Carter GT: Neuro-inflammation as a therapeutic target in amyotrophic lateral sclerosis. Curr Opin Investig Drugs. 2002, 3: 1720-4.PubMed
77.
Zurück zum Zitat Rovelli G, Heller RA, Canossa M, Shooter EM: Chimeric tumor necrosis factor-TrkA receptors reveal that ligand-dependent activation of the TrkA tyrosine kinase is sufficient for differentiation and survival of PC12 cells. Proc Natl Acad Sci U S A. 1993, 90: 8717-8721.PubMedCentralCrossRefPubMed Rovelli G, Heller RA, Canossa M, Shooter EM: Chimeric tumor necrosis factor-TrkA receptors reveal that ligand-dependent activation of the TrkA tyrosine kinase is sufficient for differentiation and survival of PC12 cells. Proc Natl Acad Sci U S A. 1993, 90: 8717-8721.PubMedCentralCrossRefPubMed
78.
Zurück zum Zitat Jiang H, Koubi D, Zhang L, Kuo J, Rodriguez AI, Jackson Hunter T: Inhibitors of iNOS protects PC12 cells against the apoptosis induced by oxygen and glucose deprivation. Neuroscience Letters. 2005, 375: 59-63. 10.1016/j.neulet.2004.10.067.CrossRefPubMed Jiang H, Koubi D, Zhang L, Kuo J, Rodriguez AI, Jackson Hunter T: Inhibitors of iNOS protects PC12 cells against the apoptosis induced by oxygen and glucose deprivation. Neuroscience Letters. 2005, 375: 59-63. 10.1016/j.neulet.2004.10.067.CrossRefPubMed
79.
Zurück zum Zitat Woo SB, Page J, Saragovi HU, Neet KE: Binding of nerve growth factor (NGF) to Trk receptor chimeras. Faseb Journal. 2004, 18: C273. Woo SB, Page J, Saragovi HU, Neet KE: Binding of nerve growth factor (NGF) to Trk receptor chimeras. Faseb Journal. 2004, 18: C273.
80.
Zurück zum Zitat Floden AM, Li SS, Combs CK: beta-Amyloid-stimulated microglia induce neuron death via synergistic stimulation of tumor necrosis factor alpha and NMDA receptors. Journal of Neuroscience. 2005, 25: 2566-2575. 10.1523/JNEUROSCI.4998-04.2005.CrossRefPubMed Floden AM, Li SS, Combs CK: beta-Amyloid-stimulated microglia induce neuron death via synergistic stimulation of tumor necrosis factor alpha and NMDA receptors. Journal of Neuroscience. 2005, 25: 2566-2575. 10.1523/JNEUROSCI.4998-04.2005.CrossRefPubMed
Metadaten
Titel
Signaling pathways mediating a selective induction of nitric oxide synthase II by tumor necrosis factor alpha in nerve growth factor-responsive cells
verfasst von
Michael S Thomas
WenRu Zhang
Paivi M Jordan
H Uri Saragovi
Giulio Taglialatela
Publikationsdatum
01.12.2005
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2005
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-2-19

Weitere Artikel der Ausgabe 1/2005

Journal of Neuroinflammation 1/2005 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

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.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.