Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2017

Open Access 01.12.2017 | Research

Etiogenic factors present in the cerebrospinal fluid from amyotrophic lateral sclerosis patients induce predominantly pro-inflammatory responses in microglia

verfasst von: Pooja-Shree Mishra, K. Vijayalakshmi, A. Nalini, T. N. Sathyaprabha, B. W. Kramer, Phalguni Anand Alladi, T. R. Raju

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2017

Abstract

Background

Microglial cell-associated neuroinflammation is considered as a potential contributor to the pathophysiology of sporadic amyotrophic lateral sclerosis. However, the specific role of microglia in the disease pathogenesis remains to be elucidated.

Methods

We studied the activation profiles of the microglial cultures exposed to the cerebrospinal fluid from these patients which recapitulates the neurodegeneration seen in sporadic amyotrophic lateral sclerosis. This was done by investigating the morphological and functional changes including the expression levels of prostaglandin E2 (PGE2), cyclooxygenase-2 (COX-2), TNF-α, IL-6, IFN-γ, IL-10, inducible nitric oxide synthase (iNOS), arginase, and trophic factors. We also studied the effect of chitotriosidase, the inflammatory protein found upregulated in the cerebrospinal fluid from amyotrophic lateral sclerosis patients, on these cultures.

Results

We report that the cerebrospinal fluid from amyotrophic lateral sclerosis patients could induce an early and potent response in the form of microglial activation, skewed primarily towards a pro-inflammatory profile. It was seen in the form of upregulation of the pro-inflammatory cytokines and factors including IL-6, TNF-α, iNOS, COX-2, and PGE2. Concomitantly, a downregulation of beneficial trophic factors and anti-inflammatory markers including VEGF, glial cell line-derived neurotrophic factor, and IFN-γ was seen. In addition, chitotriosidase-1 appeared to act specifically via the microglial cells.

Conclusion

Our findings demonstrate that the cerebrospinal fluid from amyotrophic lateral sclerosis patients holds enough cues to induce microglial inflammatory processes as an early event, which may contribute to the neurodegeneration seen in the sporadic amyotrophic lateral sclerosis. These findings highlight the dynamic role of microglial cells in the pathogenesis of the disease, thus suggesting the need for a multidimensional and temporally guarded therapeutic approach targeting the inflammatory pathways for its treatment.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12974-017-1028-x) contains supplementary material, which is available to authorized users.
Abkürzungen
ALS
Amyotrophic lateral sclerosis
ALS-CSF
Cerebrospinal fluid of amyotrophic lateral sclerosis patients
ALS-CSF-MCM
Amyotrophic lateral sclerosis cerebrospinal fluid-exposed microglial cell-conditioned medium
ANOVA
Analysis of variance
CHIT-1
Chitotriosidase-1
CHIT-1-MCM
Chitotriosidase-1-exposed microglial cell-conditioned medium
CNS
Central nervous system
COX-2
Cyclooxygenase-2
DCFDA
Dichlorofluorescin diacetate
FALS
Familial amyotrophic lateral sclerosis
GDNF
Glial cell line-derived neurotrophic factor
GFAP
Glial fibrillary acidic protein
IFN-γ
Interferon-γ
IL-6
Interleukin-6
IL-10
Interleukin-10
iNOS
Inducible nitric oxide synthase
MGC
Multinucleated giant cell
mRNA
Messenger RNA
MS
Multiple sclerosis
MTT
3,4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
MV
Microvesicle
NALS-CSF
CSF of non-ALS patients
NC
Normal control
NO
Nitric oxide
PGE2
Prostaglandin E2
ROS
Reactive oxygen species
SALS
Sporadic amyotrophic lateral sclerosis
SOD1
Superoxide dismutase 1
TAR
Transactive response
TDP-43
43-kDa transactive response DNA-binding protein
TNF-α
Tumor necrosis factor alpha
VEGF
Vascular endothelial growth factor

Background

The exact pathomechanism of neurodegeneration in amyotrophic lateral sclerosis (ALS), esp. the sporadic forms of ALS, remains poorly understood, thus leading to the lack of an effective therapeutic intervention for the disease [1, 2]. Several studies have reported a non-cell autonomous glial involvement in the pathogenesis, and the relevance of innate as well as adaptive immunity in ALS has also been widely discussed [3]. However, the requirement of the dynamic modulatory signals from within the central nervous system (CNS) for the recruitment and regulation of the adaptive immunity across the blood-brain barrier (BBB)/brain-spinal cord barrier (BSCB) further highlights the importance of the resident immune cells of the CNS in the disease pathology [4]. Microglia, the specialized immune cells of the CNS, constantly survey and dynamically regulate the neuronal milieu in the healthy and diseased CNS [5]. Depending on the nature and extent of the insult, microglial cells have been proposed to adopt morphologically and functionally distinct reactive phenotypes. These distinct phenotypes may perform diversified functions ranging from facilitating the pro-inflammatory processes that promote neuroinflammation to inducing an anti-inflammatory process that is engaged in healing and wound repair [6]. A fine balance among these phenotypes is considered to be crucial for a competent surveillance system, and its disruption could lead to the self-propagating chronic neuroinflammation seen in several neurological disorders [7]. However, the typical classification based on microglial polarization into classically (M1) and alternatively (M2) activated microglia is controversial and has been parallelly challenged [8].
Activated microglia have previously been reported in the autopsy samples and animal models of familial ALS (FALS), as well as throughout the symptomatic stages as demonstrated by neuroimaging of the ALS patients [911]. While many studies report microglial activation to be actively neurotoxic in ALS [1214], there are also studies that report the microglial involvement to ne either neuroprotective or having no significant role to play in the event of neurodegeneration in such ALS models [15, 16]. Further, the spatial microglia appeared to affect their activation status in these animal models [17]. Incidentally, majority of these studies were conducted with the transgenic models containing superoxide dismutase 1 (SOD1) mutations that are uncommon in sporadic ALS (SALS) (> 1%) [18]. In recent years, the focus has largely shifted to transgenic models with novel, gene mutations more commonly reported in ALS patients, thus narrowing the gap between animal models and actual disease etiopathogenesis. Of prominence are ubiquitinated cytoplasmic inclusions (~ 98%)/mutations (FALS 5%, SALS 1%) in 43-kDa TAR DNA-binding protein (TDP-43), fused in sarcoma (FUS) (FALS 4%, SALS < 1%) and, most prominently, the hexaneucleotide repeat expansion in C9orf72 (FALS 40%, SALS > 10%) [1921]. However, while these models recapitulate the etiopathogenesis of FALS more effectively, their relevance from the perspective of SALS pathogenesis, which constitutes 90% of the ALS etiology, is still dubious [22]. Although the emergence of these models has opened newer research avenues including aberrant RNA processing and protein degradation pathways, as well as perturbed nucleocytoplasmic transport [21], the neuroinflammatory pathways, esp. in SALS remain unraveled. Moreover, it remains unclear whether the microglial cell-mediated neurotoxicity or protection targeted the mutated protein or mimicked the overall disease pathology. The recent in vitro approaches targeting the induced pluripotent stem cells from SALS patients have been a significant step, but majority of the studies conducted till date have focused on the motor neuron pathology, precluding their relevance to microglial contribution [23, 24].
Therefore, we studied the effect of the disease-related factors circulated in the cerebrospinal fluid of ALS patients (ALS-CSF) on microglia to understand the direct nature of the insult in the SALS pathogenesis. Our hypothesis was based on the assumption that ALS-CSF may provide a mirror to the molecular effectors being produced and propagated in the diseased CNS, which may lead to progression of the disease. The neurodegenerative potential of ALS-CSF has already been documented in a plethora of molecular, electrophysiological, and behavioral studies, and the efficacy of the model in recapitulating the ALS pathology has been suggested [2532]. Our previous investigations that aimed at determining the putative toxic agent(s) by the proteomic analysis of ALS-CSF demonstrated multifold upregulation of chitotriosidase-1 (CHIT-1), an inflammatory protein secreted by activated macrophages [33, 34]. Therefore, we investigated the response of microglia towards the ALS-CSF, as well as towards CHIT-1 in doses comparable to those found in the CSF samples taken for the study [33]. Microglial cultures exposed to ALS-CSF were analyzed for the production of toxic factors including free radicals and nitric oxide release as well as for inflammatory cytokines interleukin-6 (IL-6), interleukin-10 (IL-10), interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α), Cyclooxygenase-2 (COX-2), and prostaglandin E2 (PGE2). In order to determine the trophic modulation in microglia in response to ALS-CSF, the changes in the expression patterns of vascular endothelial growth factor (VEGF) and glial cell line-derived neurotrophic factor (GDNF) were studied.

Methods

Diagnosis, CSF collection, and exposure

Diagnosis of ALS was based on the revised El Escorial criteria [35]. CSF samples from five drug-naive SALS patients (ALS-CSF) (Table 1, Additional file 1) and the age-matched disease control (patients suffering from neurological diseases, except neuroinflammatory and neurodegenerative diseases; NALS-CSF) (Table 2, Additional file 1) were collected after obtaining informed consent in accordance with the institutional human ethics committee guidelines. For ALS-CSF, careful screening of the family history of the patients was done to rule out FALS. The mean duration of illness taken into the consideration for the study was 12 ± 6 months, with mild (40%) to moderate disease severity (60%) and limb (40%), as well as bulbar (60%) onset patterns. The samples thus obtained were snap frozen in liquid nitrogen and stored at − 80 °C until further use.
Table 1
List of the antibodies used in the study
Antibody
Manufacturer
Ratio
Time (h)
Temperature (°C)
Primary antibody
 Anti-arginase rabbit polyclonal
Abcam
1:500
24
4
 Anti-CHIT-1 rabbit polyclonal
Abcam
1:500
24
4
 Anti-COX-2 rabbit polyclonal
Abcam
1:800
24
4
 Anti-GDNF mouse monoclonal
SCBT
1:200
24
4
 Anti-Iba-1 goat polyclonal
Abcam
1:800
24
4
 Anti-IFN-γ goat polyclonal
SCBT
1:200
24
4
 Anti-IL-10 rabbit polyclonal
Abcam
1:500
24
4
 Anti-IL-6 rabbit polyclonal
Abcam
1:500
24
4
 Anti-iNOS rabbit polyclonal
Abcam
1:800
24
4
 Anti-PGE2 rabbit polyclonal
Abcam
1:500
24
4
 Anti-TNF-α mouse monoclonal
Abcam
1:200
24
4
 Anti-VEGF rabbit polyclonal
Abcam
1:500
24
4
Secondary antibody
 Anti-rabbit IgG (FITC-conjugated)
Chemicon
1:200
2
RT
 Anti-rabbit IgG (FITC-conjugated)
Chemicon
1:200
2
RT
 Anti-rabbit IgG (Cy3-conjugated)
Sigma-Aldrich
1:200
2
RT
 Anti-mouse IgG (Cy3-conjugated)
Sigma-Aldrich
1:200
2
RT
 Anti-goat IgG (Cy3-conjugated)
Sigma-Aldrich
1:200
2
RT
 Anti-goat IgG (Cy3-conjugated)
Sigma-Aldrich
1:200
2
RT
 Anti-rabbit IgG (Cy3-conjugated)
Sigma-Aldrich
1:200
2
RT
 Anti-rabbit IgG (FITC-conjugated)
Chemicon
1:200
2
RT
 Anti-rabbit IgG (Cy3-conjugated)
Chemicon
1:200
2
RT
 Anti-rabbit IgG (FITC-conjugated)
Chemicon
1:200
2
RT
 Anti-mouse IgG (Cy3-conjugated)
Sigma-Aldrich
1:200
2
RT
 Anti-rabbit IgG (FITC-conjugated)
Chemicon
1:200
2
RT
Table 2
List of the primers and hydrolysis probes used in the study
Gene
Sense primer
Anti-sense primer
Probe
Arginase
AGCTGAGCAGCTGGACAG
CTCCGATAATCTCTATGGGCTTTGG
AGCAGCAGCAGCAGCAGGAACC
COX-2
CCAACCTCTCCTACTACACCAG
GTTCCTTATTTCCTTTCACACCCATG
CCTTCCTCCTGTGGCTGATGACTGC
GDNF
CGCCGGTAAGAGGCTTCTC
GATAATCTTCGGGCATATTGGAGTC
CGCCCGCCGAAGACCACTCCCT
IFN-γ
GCACAAAGCTGTCAATGAACTCA
CCAGAATCAGCACCGACTCC
CTGTCACCAGAATCTAGCCTAAGGAAGCGG
IL-10
CATGGCCTTGTAGACACCTTTG
CATCGATTTCTCCCCTGTGAGA
TCATTCTTCACCTGCTCCACTGCCTTGCTT
IL-6
TCCAGCCAGTTGCCTTCTTG
TCCTCTGTGAAGTCTCCTCTCC
ACTGATGTTGTTGACAGCCACTGCCTTCC
iNOS
CATCGACCTGGGCTGGAA
CCTCTGGATCTTGACCGTGAG
CGATGTGCTGCCTCTGGTCCTGC
TNF-α
TGGCGTGTTCATCCGTTCTCTA
CTCTGAGGAGTAGACGATA
TGGCGTGTTCATCCGTTCTCTA
VEGF
GAGCAACGTCACTATCGAGATC
GGCTTTGTTCTATCTTTCTTGGTC
TGCCGATCAAACCTCACCAAAGCCA

Study groups

For the experiments involving ALS-CSF exposure, the cultures were supplemented with 10% v/v CSF in Dulbecco’s modified Eagle’s medium (DMEM) and the effects were studied in duplicates or triplicates for individual samples. The study consisted of the following experimental groups:
  • Normal control (NC): cultures propagated in DMEM alone
  • NALS (disease control): cultures exposed to CSF of the NALS (10% v/v in DMEM)
  • ALS: cultures exposed to ALS-CSF (10% v/v in DMEM)
For the experiments related to studying the effect of microglial cell-conditioned media (MCM), the groups were as follows:
  • NC-MCM: NSC-34 cultures grown in the conditioned media from normal control microglial cultures
  • NALS-MCM: NSC-34 cultures grown in the conditioned media from NALS group
  • ALS-MCM: NSC-34 cultures grown in the conditioned media from the ALS group
For the study concerning exposure to CHIT-1, the experimental groups were as follows:
  • NC: cultures propagated in DMEM
  • Buffer control (buffer): cultures exposed to buffer (10% v/v of DMEM)
  • CHIT-1: cultures exposed to CHIT-1 in a dose equivalent to that found in ALS-CSF [33] (18 pg/μl; 10% v/v, DMEM)

Cell culture

Mixed glial cultures and enriched astroglial cultures

Spinal cords of Wistar rat pups (P2–P3) were dissected, freed of meninges, and mechanically triturated in DMEM. The single cell suspension thus obtained was propagated in DMEM with 10% FBS (Gibco BRL), with a seeding density of 2.5 × 104 cells/ml. The enriched primary astroglial cultures were obtained from the mixed glial cultures using a previously described protocol [36]. Both the mixed glial and enriched astroglial cultures were allowed to attain a confluence of 70–80%, following which the exposure to ALS-CSF or CHIT-1, along with appropriate control groups, was carried out.

Microglial cultures

Enriched microglial cultures were obtained using a method by Saura and colleagues to ensure the purity and yield [37]. Briefly, upon reaching confluence (15–20 DIV), the mixed glial cultures were subjected to mild trypsin treatment (trypsin/EDTA DMEM, 1:4) to remove the astroglial monolayer, leaving intact the microglial layer growing beneath the astroglial monolayer. The purity of the microglial cultures was determined to be ≥ 99% using the microglial marker Iba-1, nuclear marker TO-PRO-3, and astroglial marker glial fibrillary acidic protein (GFAP). The enriched cultures were then subjected to the experimental treatments within 24 h.

NSC-34 cultures

The NSC-34 motor neuronal cell line was procured (Cedarlane, Canada) and maintained in accordance with the published protocol [27]. To study the effect of conditioned media, NSC-34 cells were plated at a density of 2.5 × 104 cells/ml and allowed to reach 70–80% confluence. The cultures were then subjected to different experimental conditions.

Immunofluorescence

Expression and localization of CHIT-1 were determined by qualitative observations on immunofluorescently labeled cells. A qualitative, as well as quantitative, approach was taken for the localization and analysis of cytokines, inflammatory markers, and trophic factors [36]. Briefly, mixed glial and enriched microglial cultures were plated onto poly-l-lysine-coated 13-mm circular coverslips (0.1 mg/ml). The cultures were then exposed to the ALS-CSF or CHIT-1 experimental groups and fixed with 4% paraformaldehyde (PFA) for 15 min at RT. Following blocking with 3% bovine serum albumin (BSA), the cultures were subsequently incubated with primary antibodies of interest followed by fluorescently labeled appropriate secondary antibodies (FITC or Cy3) (Table 1). TO-PRO-3-iodide (TOPRO) was used to stain the nucleus. The coverslips were then mounted using PVA-DABCO (Sigma-Aldrich, USA) and viewed under the laser scanning confocal microscope (Leica TCS SL, Germany), with excitation wavelengths at 488, 514, and 633 nm for FITC, Cy3, and TOPRO, respectively. In the case of double immunofluorescence, the antibodies raised in different animals were chosen and emission frequencies were segregated to avoid non-specific overlap of labeling.

Measurement of protein expression

We quantified the cellular expression of proteins of interest in terms of mean fluorescence intensity of individual cells on the images captured by a confocal microscope for each immunolabeled protein, using the inbuilt software of Leica Microsystems [36, 38, 39]. Briefly, 8-bit images were captured at × 20 magnifications with a constant PMT voltage, from randomly selected ten non-overlapping fields on each coverslip. Other parameters like optical zoom, frame and line average, resolution, frame (1024 × 1024), and exposure time were kept constant. The fluorescence intensity was measured on a scale of 0–255, and the immunostained region of each cell (region of interest, ROI) was demarcated using the polyline profile of the software. The area from 20 such cells or ROI, thus randomly marked, was quantified for each image, following which the fluorescence intensities were generated as numerical values commensurate to the staining. Ten such images were quantified for a single replicate of a sample. Five such samples were analyzed in duplicates for each group. Background reduction was applied for each analysis, and the mean fluorescence intensities (MFIs) were then compared between the study groups.

Measurement of soma area

For the measurement of soma area, the contour of cells was carefully drawn using the polyline profile in the same manner as described above. The report was generated for the soma area for each ROI, and the mean area per image was analyzed and compared within the groups.

Morphological transformation studies

After 48 h of ALS-CSF exposure, microglial cultures were observed and quantified for the morphological diversity ranging from ramified/resting to amoeboid/activated cells in randomly chosen fields, by phase contrast, as well as by Iba-1 immunofluorescence. Ten such fields were analyzed in duplicates for each study group, and the results were compared.

Estimation of secreted cytokines

For a temporal analysis, the cultures were exposed to CSF for 12, 24, and 48 h, respectively, and the decanted media from the cultures were used for the estimation. Quantitative analysis of the secreted IL-6, IL-10, IFN-γ, and TNF-α levels was performed using specific rat ELISA kits and according to the manufacturer’s instructions (Ray Biotech, Inc., Georgia, USA).

Glutamate estimation

For glutamate estimation, the medium from the normal controls and each of the subsets exposed to ALS-CSF for 12, 24, and 48 h was collected and centrifuged at 14,000 rpm to remove the cellular debris. Glutamate levels were measured using the glutamate assay kit (MAK004; Sigma-Aldrich, USA). The measurement was based on enzymatic conversion, and the absorbance was read at 450 nm using a colorimetric ELISA microplate plate reader (Tecan 2500 fluorometer, USA).

ROS measurement

After CSF exposure for 48 h, the cultures were treated with dichlorofluorescin diacetate (DCFDA), harvested, and lysed and the resultant fluorescence release was measured at the excitation/emission spectrum of 480/530 nm in the 96-well plate reader (Tecan 2500 fluorometer, USA). Fluorescence/cell was calculated (arbitrary fluorescence unit, AFU) and statistically analyzed. Another set of cultures grown on 13-mm coverslips was incubated with DCFDA at 37 °C for 30 min and directly viewed (excitation = 480 nm; emission = 530 nm) under a confocal laser microscope.

Nitric oxide measurement

Media from different experimental groups were collected as previously discussed. In addition, lysates were prepared from the microglial cultures after 48 h of exposure to ALS-CSF. The level of nitrates was assayed in the samples using a nitric oxide assay kit (AB65328; Abcam) at 540 nm using the colorimetric ELISA microplate reader.

Cell viability and toxicity assay

Microglial cultures and NSC-34 cells seeded on 96-well plates (500 cells/well) were subjected to different experimental conditions, and MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and/or lactate dehydrogenase (LDH) assays were performed on these cultures and/or culture media according to the previously published protocols [27].

Quantitative RT-PCR

Following exposure to CSF for 48 h, total RNA was extracted from the microglial cultures using the RNeasy Plus Mini Kit (Qiagen, USA) and quantified using 1 μl of RNA in a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Inc.). Total RNA (10 ng) was reverse transcribed (RT) with random primers using high-capacity cDNA reverse transcription kit (Applied Biosystems, USA). Amplification was carried out in triplicates using specific primers and hydrolysis probes for the target genes (Eurogentec, Belgium, Table 2), and reaction efficiencies for the target genes were analyzed and compared to 18S messenger RNA (mRNA) (endogenous control) using the standard curve method, so that a PCR efficiency of ~ 100 ± 10% was achieved. The cycle threshold (Ct) values were normalized to the endogenous control 18S mRNA, and the relative fold change was calculated using the comparative CT method (ΔΔCT method) [40].

Statistical analysis

The data on mRNA expression and quantitative immunofluorescence was statistically assessed for significance by one-way ANOVA followed by Tukey’s post hoc test. For ELISA, glutamate, NO, ROS, and MTT assays, the statistical analysis was carried out using Student’s t test. GraphPad Prism was the software used to determine the statistical significance and P values for each experiment.

Results

ALS-CSF activated microglia, and the response was dynamic as well as temporally regulated

Upon exposure to ALS-CSF, the microglial cells showed dynamic structural responses, adopting various morphologies ranging from the ramified to amoeboid morphology (Fig. 1a–d”). The increasing number of cells showing retraction of processes with a highly granular and distinctively enlarged soma suggested the adoption of a motile phagocytic phenotype in response to the etiogenic factors present in ALS-CSF (Fig. 1g, i). The response was specific to ALS-CSF, as the disease controls (NALS) did not induce this phenomenon. Apart from the amoeboid morphology, we also found multinucleated giant microglial cells (MGCs) predominantly in the cultures exposed to ALS-CSF (Fig. 1e–f”). Some cells also showed a disintegrated cytoplasm, resembling the process of cytorrhexis [41] (Fig. 1e–e”, white arrowheads). Increased Iba-1 expression and enhanced MTT reduction in cultures exposed to ALS-CSF for 48 h indicated sustained activation, possibly leading to microglial proliferation (Fig. 1h, j, k).
The activation of microglia in response to ALS-CSF could be seen as early as 12 h (Fig. 2). Further, the activated microglia were predominantly surrounded by Iba-1-immunopositive vesicular structures ranging from a few nanometers to a few micrometers (Fig. 2, arrowheads and arrows, respectively) suggestive of a process akin to microvesicular shedding/release, which showed a subsequent decline at 24 and 48 h. A 24-h exposure to ALS-CSF resulted mostly in amoeboid morphology, while in addition, at 48 h, the cells also adopted a multinucleated morphology or they underwent cytorrhexis (Fig. 1e–f”). To test the functional relevance of microvesicle (MV)-like structures, we performed an analysis of the secretory profiles of microglia exposed to ALS-CSF for 12, 24, and 48 h, respectively. We observed an upregulated release of the pro-inflammatory cytokines, IL-6 and TNF-α, in the media supernatants from the ALS group, as compared to the controls as early as 12 h following exposure (Fig. 2b, c). The difference between the control and experimental groups was maximal at 12 h for TNF-α, which tapered gradually at subsequent time points. The overexpression of glutamate was more prominent following longer exposure (Fig. 2f). Concomitantly, a temporal downregulation was observed in the secretory levels of the anti-inflammatory cytokines IL-10 as well as IFN-γ (Fig. 2d, e). Out of all the time periods studied, a 48-h exposure resulted in a prolonged and sustained microglial activation and exhibited maximum morphological heterogeneity. Hence, a 48-h exposure period was chosen for further studies.

Microglial cells respond to ALS-CSF by adopting a phenotype skewed towards pro-inflammatory pathways along with downregulated trophic support

Molecular analysis of microglial activation revealed an upregulation of ROS and NO levels in cells exposed to ALS-CSF as compared to the normal controls (Fig. 3a–c). Further, the mRNA and protein expression of inducible nitric oxide synthase (iNOS), which is an inducer of pathological NO and a marker of classical activation pathway, was significantly upregulated in cultures exposed to ALS-CSF (Fig. 3d–f). The mRNA and protein expression for arginase, a marker of alternative activation, was downregulated (Fig. 3g–i). Increased iNOS/arginase ratio in response to ALS-CSF exposure for 48 h indicated that the microglial activation was skewed towards the classical pathway (Fig. 3j). Further, the conditioned medium from the microglial cells exposed to ALS-CSF induced cell death in NSC-34 cells, suggesting the existence of neurotoxic potential in activated microglial cells as a result of exposure to ALS-CSF (Fig. 3k–l). We studied the mRNA expression as well as cellular localization of the inflammatory markers after 48 h of ALS-CSF exposure. An upregulation was registered in the mRNA as well as the protein levels of IL-6 (Fig. 4a, e–e”, i) and TNF-α (Fig. 4b, f–f”, j), while IFN-γ mRNA was downregulated with no significant changes observed in its cellular protein expression (Fig. 4c, g–g”, k) in the ALS group as compared to the normal and disease controls. While a significant upregulation was seen in the IL-10 mRNA levels in response to ALS-CSF, the cellular protein expression showed a downregulation, which could explain the decreased secretion of IL-10 in cultures exposed to ALS-CSF (Fig. 4d, h–h”, l).
ALS-CSF also induced changes in inflammatory markers such as COX-2 and PGE2. With regard to mRNA expression of COX-2, three out of five ALS-CSF samples studied showed a significant upregulation of COX-2 mRNA, while two showed downregulation compared to the controls (Fig. 5a, a’). However, all the five ALS-CSF samples showed a significant upregulation of the cellular expression of COX-2 as well as PGE2, when compared to the normal and disease controls (Fig. 5b–e).
Further, ALS-CSF exposure also resulted in a significant downregulation of the mRNA as well as protein expression of the trophic factors, namely GDNF (Fig. 6a, c–c”, e) and VEGF (Fig. 6b, d–d”, f, f”), when compared to the normal and disease controls. This indicated a reduced trophic support provided by the microglial cells along with the neuroinflammatory response to the toxic factors present in the ALS-CSF.

Chitotrisidase-1, the factor prominently upregulated in ALS-CSF, acts selectively on microglial cells

We previously reported a multifold increase in the concentration of CHIT-1 in ALS-CSF and its enhanced expression in microglia in response to ALS-CSF [33]. We also reported the induction of inflammation in astroglia following exposure to ALS-CSF [36]. To further delineate whether the CHIT-1 expression is specific to microglia or also present in activated astrocytes, we studied CHIT-1 expression in the mixed glial cultures exposed to ALS-CSF. While CHIT-1 immunoreactivity was clearly absent in the GFAP-positive astrocytes in both normal controls and ALS group, the non-GFAP-positive cells showed enhanced CHIT-1 expression upon ALS-CSF exposure, thus corroborating our previous findings of microglial cells as the primary source of CHIT-1 in CSF (Fig. 7a–b”).
Further, in order to investigate the direct effects of CHIT-1, we exposed the microglial cells to the concentrations of CHIT-1 similar to that found in ALS-CSF. Exposure to CHIT-1 caused a microglial transformation similar to that seen in the ALS group, with the presence of MV-like structures prominent at 12 h, while other transformations including MGCs and cytorrhexis were found at 48 h (Fig. 7c–c”’). An increase in the MTT reduction was observed in the enriched microglial cultures exposed to CHIT-1, in a manner similar to ALS-CSF, thereby indicating a direct action of CHIT-1 on microglial activation (Fig. 7e). The apparent specificity of CHIT-1 action on microglia was further supported by the absence of any significant effect on the viability of primary astrocytes (Fig. 7e).

Discussion

Microglial activation is a dynamic process and may involve various temporally, physiologically, and/or spatially regulated events that govern the morphological and functional changes observed in the reactive microglia [6, 42, 43]. The pro-inflammatory microglial transformation observed in response to ALS-CSF indicates an inflammatory microenvironment of the CNS in SALS that may be further dynamically regulated by the actively transforming microglia, in a closed, amplifying feedback loop. The ability of the conditioned media from the microglial cultures exposed to ALS-CSF to exert toxicity to the NSC-34 cells corroborates the non-cell autonomous disease propagation in ALS [42]. Recent studies discuss the importance of MV/exosomes in the neuronal spread of TDP-43 proteinopathies in ALS [44, 45]. Studies conducted on the glial MVs released by the reactive astrocytes and glioma cells hint towards a possible engagement of MVs in the intercellular communication [46, 47]. Further, investigations on microglial microvesicular release in neuroinflammation also suggest its participation in disseminating neurotoxicity via proteins and/or microRNAs [48, 49]. The reports of pro-inflammatory nature of microglial MVs, especially at early time points (8 h) as opposed to the anti-inflammatory nature at later stages (> 24 h), support our observations of early pro-inflammatory secretion and concomitantly increased vesicular structures in the ALS-CSF-exposed microglial cultures [50, 51]. Interestingly, at 48 h, ALS-CSF also induced multinucleation and cytorrhexis in the microglial cultures, which hints at dysregulated pathways leading to failed cytokinesis and overactivation-induced self-moderation [52, 53]. Such a phenomenon has also been reported in another study involving the mutant SOD1 mice model, where it is explained as an accidental product of the high-intensity neuroinflammation [41]. Induction of NF-κB, the inflammatory regulator for glial activation, has been documented in the autopsy studies as well as TDP-43 models [5456]. Further, studies involving the C9orf72 pathology suggest towards a potential role of the microglial C9orf72 repeat expansions in exacerbating neuroinflammation, resulting in a loss-of-function endosomal-lysosomal aberration, thus favoring a pro-inflammatory microglial phenotype [57]. These observations highlight the role of neuroinflammation in the disease pathology irrespective of the nature of the etiogenesis.
Upregulation of microglial inflammatory mediators and retraction of the beneficial trophic support demonstrate the ability of ALS-CSF to shift the microglial functionality predominantly towards an activated pro-inflammatory physiology that is able to propagate inflammation by sending inflammatory signals in the circulation. Overexpression of iNOS, along with the downregulation of arginase, may suggest the induction of a pathological switch towards physiologically inflamed or the classically activated (M1) microglia in response to ALS-CSF [58]. This correlated with the increased expression and secretion of IL-6 and TNF-α, as well as a temporal decline in the secretion of the neuroprotective cytokines IL-10 and IFN-γ. However, the upregulation of IL-10 mRNA levels and the differential regulation of COX-2 mRNA in response to different ALS-CSF samples, contrary to their pro-inflammatory protein expression patterns, also depict a conflicting state of activation that may defy the typical M1/M2 polarization. Studies conducted in the mutant SOD1 mice model also suggested a disease-specific microglial profile, where a simultaneous dysregulation was reported in the expression of M1- and M2-specific markers in early stages, with either a slight bias [10] towards the M1 type or none [17] at the later stages. Since in our study the overall findings support the neurotoxic outcomes, we propose that these profiles may arise either as the result of a dynamic, reversible switch between a pro- and anti-inflammatory phenotype or a failed attempt of the repair processes at work.
l-Glutamate release from activated microglia downregulates astroglial glutamate transporter expression [59], a pathological phenomenon well documented in ALS [60]. Apart from this, the elevated microglial glutamate may also exacerbate neurodegeneration through dysregulated gliotransmission via purinergic receptors and Ca2+ wave propagation [61] or induce necrosis and necroptosis as seen in response to ALS-CSF [27, 38]. Alternatively, excess glutamate can also accentuate neuroinflammation by interacting with inflammatory modulators including ROS [62], PGE2 [63], and NO [64], which were found elevated in response to the ALS-CSF. The neurotoxic potential of the conditioned media from microglial cultures exposed to the ALS-CSF (ALS-MCM) corroborated the pro-inflammatory role of the factors predominantly upregulated in ALS-MCM, including IL-6, TNF-α, and NO. Further, the higher quantum of TNF-α levels, specifically in the initial phase, suggests its role as a potent, early initiator of neuroinflammation, much in accordance with the studies conducted in mSOD1 models [65, 66]. Recent literature elucidates the neuroprotective nature of microglial IFN-γ signaling in CNS pathology through the recruitment of immunoregulatory cells including macrophages, and their dysregulation in the neurodegenerative disorders [4, 67, 68]. The downregulation of microglial IFN-γ secretion may thus suggest a reduced anti-inflammatory leucocyte infiltration leading to the aggravation of the disease pathology.
Apart from a reduced trophic support, downregulation of microglial VEGF has been shown to correspond with a phagocytic microglial phenotype [69]. In a similar manner, a study by Matsushita and colleagues also reported a significant downregulation of GDNF in response to endotoxin LPS, suggesting that the reduction in GDNF expression could be a feature of inflammation [70]. These studies highlight the protective roles of both these trophic factors in disease pathogenesis through inhibition of inflammation and provide sufficient evidence to consider their relevance in clinical applications.
These observations clearly suggest that following exposure to ALS-CSF, microglial cells are activated and adopt a toxic phenotype. Additionally, we have provided compelling evidence inflammatory response of the spinal cord-derived microglia to ALS-CSF-mediated insult. Interestingly, in contrast to the early microglial activation, the maximal degenerative changes in the neurons exposed to ALS-CSF were observed at 48 h [27]. Similarly, the first known astroglial response to ALS-CSF in terms of gliosis or cytokine secretion was not observed before 24 h and became prominent at 48 h. The present observations, along with the previously reported, marginally delayed, inflammatory behavior of astrocytes towards ALS-CSF, might suggest a synergistic interplay between microglia and astrocytes in potentiating, sustaining, and aggravating the insult [36].
In addition, the present study attempts to explain the continued failure of anti-inflammatory therapeutic approaches in the clinical trials, despite obvious pathological contribution of inflammation in ALS [71, 72]. Since the alterations observed in the microglial physiology can at best be described as multifactorial with the interplay of components like pro-inflammatory and anti-inflammatory cytokines, PGE2, ROS, and glutamate, mitigating inflammation by blocking specific molecules/pathways would be inefficient. Moreover, the lacuna with employing anti-inflammatory drugs to target inflammation in general lies in the controversial observations of varied effects of these drugs, ranging from amelioration to exacerbation of the disease pathology, in different conditions, thus rendering them unsuitable for therapeutic interventions. For instance, minocycline was reported to be protective in the animal studies during the initial stages, as well as in the clinical trials at specific dosage, and stages of the disease, but had detrimental effects in the later stages or in higher doses [7274]. Hence, it is imperative to thoroughly investigate the mechanism of microglial inflammation in order to pin down the exact pathways affected in ALS, and design a combinatorial approach to target microglial activation and neuroinflammation.
CHIT-1, the protein primarily upregulated in the ALS-CSF, is considered as a marker of chronic activation of macrophages in the peripheral system [75]. Although CHIT-1 has also been implicated in various neurological diseases, its function in the CNS pathology remains least understood. While CHIT-1 is shown to have a pro-inflammatory potential in neurological disorders like stroke [76], its neuroprotective role in the inflammatory conditions like multiple sclerosis has also been suggested [77]. The microglial cell-specific upregulation of CHIT-1 expression in response to ALS-CSF suggests the possibility of neuroinflammatory process in ALS patients, led by the chronically activated microglia. The selective action of CHIT-1 on microglial cells suggests a vicious cycle of neurodegeneration primarily mediated by microglial cells and propagated to others via a cell-to-cell communication and/or circulating fluids. Since the reactive microglia in ALS are derived from the endogenous pool and not from the circulating monocytes [10], the endogenous microglial population may act as the predominant source of CHIT-1 in ALS-CSF, thus rendering them as potential clinical targets.

Conclusion

Based on our findings, we propose that microglial pathology in SALS is morphologically and functionally dynamic, leading to the neurodegeneration through myriad pathways (Fig. 8). It appears to operate by a push-forward mechanism, where microglia are the first to strike, possibly accompanied by astrocytes, thus setting up a neuroinflammatory cascade and eventually leading to accentuated neurodegeneration. In the light of the above, a combinatorial therapeutic approach is warranted, by suppressing the pro-inflammatory pathways and promoting the anti-inflammatory role of microglia.

Acknowledgements

The authors thank Dr. Anu Mary Varghese for her inputs towards the manuscript.

Funding

The study was funded by the Department of Biotechnology (BT/PR5384/MED/30/817/2012) and the Indian Council for Medical Research under the Centre for Advanced Research Initiative. PM was a senior research fellow under the University Grants Commission (UGC)/Council of Scientific and Industrial Research (CSIR) fellowship scheme, Government of India.

Availability of data and materials

Data supporting the conclusions of this article are included within the manuscript.
The human ethics committee of NIMHANS, where the study was conducted, approved the use of human CSF samples for the study, and individual consent was taken prior to CSF collection (Item no. III, SI no. 3.01, Basic Sciences). Animal procedures were approved by the Institutional Animal Ethics Committee (AEC/44/264/NP and AEC/55/343/NP).
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
3.
Zurück zum Zitat Puentes F, Malaspina A, van Noort JM, Amor S. Non-neuronal cells in ALS: role of glial, immune cells and blood-CNS barriers. Brain Pathol. 2016; Puentes F, Malaspina A, van Noort JM, Amor S. Non-neuronal cells in ALS: role of glial, immune cells and blood-CNS barriers. Brain Pathol. 2016;
4.
Zurück zum Zitat Kunis G, Baruch K, Miller O, Schwartz M. Immunization with a myelin-derived antigen activates the brain’s choroid plexus for recruitment of immunoregulatory cells to the CNS and attenuates disease progression in a mouse model of ALS. J Neurosci. 2015;35:6381–93.CrossRefPubMed Kunis G, Baruch K, Miller O, Schwartz M. Immunization with a myelin-derived antigen activates the brain’s choroid plexus for recruitment of immunoregulatory cells to the CNS and attenuates disease progression in a mouse model of ALS. J Neurosci. 2015;35:6381–93.CrossRefPubMed
5.
Zurück zum Zitat Ferreira R, Bernardino L. Dual role of microglia in health and disease: pushing the balance toward repair. Front Cell Neurosci. 2015;9 Ferreira R, Bernardino L. Dual role of microglia in health and disease: pushing the balance toward repair. Front Cell Neurosci. 2015;9
6.
Zurück zum Zitat Graeber MB, Streit WJ. Microglia: biology and pathology. Acta Neuropathol. 2010;119:89–105.CrossRefPubMed Graeber MB, Streit WJ. Microglia: biology and pathology. Acta Neuropathol. 2010;119:89–105.CrossRefPubMed
7.
Zurück zum Zitat Cherry JD, Olschowka JA, O’Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation. 2014;11:98.CrossRefPubMedPubMedCentral Cherry JD, Olschowka JA, O’Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation. 2014;11:98.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Ransohoff RM. A polarizing question: do M1 and M2 microglia exist? Nat Neurosci. 2016;19:987–91.CrossRefPubMed Ransohoff RM. A polarizing question: do M1 and M2 microglia exist? Nat Neurosci. 2016;19:987–91.CrossRefPubMed
9.
Zurück zum Zitat Lloyd CM, Richardson MP, Brooks DJ, Al-Chalabi A, Leigh PN. Extramotor involvement in ALS: PET studies with the GABA(A) ligand [(11)C]flumazenil. Brain. 2000;123(Pt 11):2289–96.CrossRefPubMed Lloyd CM, Richardson MP, Brooks DJ, Al-Chalabi A, Leigh PN. Extramotor involvement in ALS: PET studies with the GABA(A) ligand [(11)C]flumazenil. Brain. 2000;123(Pt 11):2289–96.CrossRefPubMed
10.
Zurück zum Zitat Chiu IM, Morimoto ET, Goodarzi H, Liao JT, O’Keeffe S, Phatnani HP, Muratet M, Carroll MC, Levy S, Tavazoie S, et al. A neurodegeneration-specific gene-expression signature of acutely isolated microglia from an amyotrophic lateral sclerosis mouse model. Cell Rep. 2013;4:385–401.CrossRefPubMedPubMedCentral Chiu IM, Morimoto ET, Goodarzi H, Liao JT, O’Keeffe S, Phatnani HP, Muratet M, Carroll MC, Levy S, Tavazoie S, et al. A neurodegeneration-specific gene-expression signature of acutely isolated microglia from an amyotrophic lateral sclerosis mouse model. Cell Rep. 2013;4:385–401.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Chio A, Pagani M, Agosta F, Calvo A, Cistaro A, Filippi M. Neuroimaging in amyotrophic lateral sclerosis: insights into structural and functional changes. Lancet Neurol. 2014;13:1228–40.CrossRefPubMed Chio A, Pagani M, Agosta F, Calvo A, Cistaro A, Filippi M. Neuroimaging in amyotrophic lateral sclerosis: insights into structural and functional changes. Lancet Neurol. 2014;13:1228–40.CrossRefPubMed
12.
Zurück zum Zitat Liu Y, Hao W, Dawson A, Liu S, Fassbender K. Expression of amyotrophic lateral sclerosis-linked SOD1 mutant increases the neurotoxic potential of microglia via TLR2. J Biol Chem. 2009;284:3691–9.CrossRefPubMed Liu Y, Hao W, Dawson A, Liu S, Fassbender K. Expression of amyotrophic lateral sclerosis-linked SOD1 mutant increases the neurotoxic potential of microglia via TLR2. J Biol Chem. 2009;284:3691–9.CrossRefPubMed
13.
Zurück zum Zitat Boillée S, Cleveland DW. Revisiting oxidative damage in ALS: microglia, Nox, and mutant SOD1. J Clin Invest. 2008;118:474–8.PubMedPubMedCentral Boillée S, Cleveland DW. Revisiting oxidative damage in ALS: microglia, Nox, and mutant SOD1. J Clin Invest. 2008;118:474–8.PubMedPubMedCentral
14.
Zurück zum Zitat Borchelt DR. Amyotrophic lateral sclerosis—are microglia killing motor neurons? N Engl J Med. 2006;355:1611–3.CrossRefPubMed Borchelt DR. Amyotrophic lateral sclerosis—are microglia killing motor neurons? N Engl J Med. 2006;355:1611–3.CrossRefPubMed
15.
Zurück zum Zitat Gowing G, Philips T, Van Wijmeersch B, Audet JN, Dewil M, Van Den Bosch L, Billiau AD, Robberecht W, Julien JP. Ablation of proliferating microglia does not affect motor neuron degeneration in amyotrophic lateral sclerosis caused by mutant superoxide dismutase. J Neurosci. 2008;28:10234–44.CrossRefPubMed Gowing G, Philips T, Van Wijmeersch B, Audet JN, Dewil M, Van Den Bosch L, Billiau AD, Robberecht W, Julien JP. Ablation of proliferating microglia does not affect motor neuron degeneration in amyotrophic lateral sclerosis caused by mutant superoxide dismutase. J Neurosci. 2008;28:10234–44.CrossRefPubMed
16.
Zurück zum Zitat Kawamura MF, Yamasaki R, Kawamura N, Tateishi T, Nagara Y, Matsushita T, Ohyagi Y, Kira J. Impaired recruitment of neuroprotective microglia and T cells during acute neuronal injury coincides with increased neuronal vulnerability in an amyotrophic lateral sclerosis model. Exp Neurol. 2012;234:437–45.CrossRefPubMed Kawamura MF, Yamasaki R, Kawamura N, Tateishi T, Nagara Y, Matsushita T, Ohyagi Y, Kira J. Impaired recruitment of neuroprotective microglia and T cells during acute neuronal injury coincides with increased neuronal vulnerability in an amyotrophic lateral sclerosis model. Exp Neurol. 2012;234:437–45.CrossRefPubMed
17.
Zurück zum Zitat Nikodemova M, Small AL, Smith SM, Mitchell GS, Watters JJ. Spinal but not cortical microglia acquire an atypical phenotype with high VEGF, galectin-3 and osteopontin, and blunted inflammatory responses in ALS rats. Neurobiol Dis. 2014;69:43–53.CrossRefPubMed Nikodemova M, Small AL, Smith SM, Mitchell GS, Watters JJ. Spinal but not cortical microglia acquire an atypical phenotype with high VEGF, galectin-3 and osteopontin, and blunted inflammatory responses in ALS rats. Neurobiol Dis. 2014;69:43–53.CrossRefPubMed
18.
Zurück zum Zitat Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O’Regan JP, Deng HX. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;362:59–62.CrossRefPubMed Rosen DR, Siddique T, Patterson D, Figlewicz DA, Sapp P, Hentati A, Donaldson D, Goto J, O’Regan JP, Deng HX. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;362:59–62.CrossRefPubMed
19.
Zurück zum Zitat Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–3.CrossRefPubMed Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–3.CrossRefPubMed
20.
Zurück zum Zitat Smith BN, Newhouse S, Shatunov A, Vance C, Topp S, Johnson L, Miller J, Lee Y, Troakes C, Scott KM, et al. The C9ORF72 expansion mutation is a common cause of ALS+/-FTD in Europe and has a single founder. Eur J Hum Genet. 2013;21:102–8.CrossRefPubMed Smith BN, Newhouse S, Shatunov A, Vance C, Topp S, Johnson L, Miller J, Lee Y, Troakes C, Scott KM, et al. The C9ORF72 expansion mutation is a common cause of ALS+/-FTD in Europe and has a single founder. Eur J Hum Genet. 2013;21:102–8.CrossRefPubMed
21.
Zurück zum Zitat Ji A-L, Zhang X, Chen W-W, Huang W-J. Genetics insight into the amyotrophic lateral sclerosis/frontotemporal dementia spectrum. J Med Genet. 2017;54:145–54.CrossRefPubMed Ji A-L, Zhang X, Chen W-W, Huang W-J. Genetics insight into the amyotrophic lateral sclerosis/frontotemporal dementia spectrum. J Med Genet. 2017;54:145–54.CrossRefPubMed
23.
Zurück zum Zitat Burkhardt MF, Martinez FJ, Wright S, Ramos C, Volfson D, Mason M, Garnes J, Dang V, Lievers J, Shoukat-Mumtaz U, et al. A cellular model for sporadic ALS using patient-derived induced pluripotent stem cells. Mol Cell Neurosci. 2013;56:355–64.CrossRefPubMedPubMedCentral Burkhardt MF, Martinez FJ, Wright S, Ramos C, Volfson D, Mason M, Garnes J, Dang V, Lievers J, Shoukat-Mumtaz U, et al. A cellular model for sporadic ALS using patient-derived induced pluripotent stem cells. Mol Cell Neurosci. 2013;56:355–64.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Myszczynska M, Ferraiuolo L. New in vitro models to study amyotrophic lateral sclerosis. Brain Pathol. 2016;26:258–65.CrossRefPubMed Myszczynska M, Ferraiuolo L. New in vitro models to study amyotrophic lateral sclerosis. Brain Pathol. 2016;26:258–65.CrossRefPubMed
25.
Zurück zum Zitat Shobha K, Alladi PA, Nalini A, Sathyaprabha TN, Raju TR. Exposure to CSF from sporadic amyotrophic lateral sclerosis patients induces morphological transformation of astroglia and enhances GFAP and S100beta expression. Neurosci Lett. 2010;473:56–61.CrossRefPubMed Shobha K, Alladi PA, Nalini A, Sathyaprabha TN, Raju TR. Exposure to CSF from sporadic amyotrophic lateral sclerosis patients induces morphological transformation of astroglia and enhances GFAP and S100beta expression. Neurosci Lett. 2010;473:56–61.CrossRefPubMed
26.
Zurück zum Zitat Sankaranarayani R, Nalini A, Rao Laxmi T, Raju TR. Altered neuronal activities in the motor cortex with impaired motor performance in adult rats observed after infusion of cerebrospinal fluid from amyotrophic lateral sclerosis patients. Behav Brain Res. 2010;206:109–19.CrossRefPubMed Sankaranarayani R, Nalini A, Rao Laxmi T, Raju TR. Altered neuronal activities in the motor cortex with impaired motor performance in adult rats observed after infusion of cerebrospinal fluid from amyotrophic lateral sclerosis patients. Behav Brain Res. 2010;206:109–19.CrossRefPubMed
27.
Zurück zum Zitat Vijayalakshmi K, Alladi PA, Sathyaprabha TN, Subramaniam JR, Nalini A, Raju TR. Cerebrospinal fluid from sporadic amyotrophic lateral sclerosis patients induces degeneration of a cultured motor neuron cell line. Brain Res. 2009;1263 Vijayalakshmi K, Alladi PA, Sathyaprabha TN, Subramaniam JR, Nalini A, Raju TR. Cerebrospinal fluid from sporadic amyotrophic lateral sclerosis patients induces degeneration of a cultured motor neuron cell line. Brain Res. 2009;1263
28.
Zurück zum Zitat Gunasekaran R, Narayani RS, Vijayalakshmi K, Alladi PA, Shobha K, Nalini A, Sathyaprabha TN, Raju TR. Exposure to cerebrospinal fluid of sporadic amyotrophic lateral sclerosis patients alters Nav1.6 and Kv1.6 channel expression in rat spinal motor neurons. Brain Res. 2009;1255:170–9.CrossRefPubMed Gunasekaran R, Narayani RS, Vijayalakshmi K, Alladi PA, Shobha K, Nalini A, Sathyaprabha TN, Raju TR. Exposure to cerebrospinal fluid of sporadic amyotrophic lateral sclerosis patients alters Nav1.6 and Kv1.6 channel expression in rat spinal motor neurons. Brain Res. 2009;1255:170–9.CrossRefPubMed
29.
Zurück zum Zitat Sharma A, Varghese AM, Vijaylakshmi K, Sumitha R, Prasanna VK, Shruthi S, Chandrasekhar Sagar BK, Datta KK, Gowda H, Nalini A, et al. Cerebrospinal fluid from sporadic amyotrophic lateral sclerosis patients induces mitochondrial and lysosomal dysfunction. Neurochem Res. 2016;41:965–84.CrossRefPubMed Sharma A, Varghese AM, Vijaylakshmi K, Sumitha R, Prasanna VK, Shruthi S, Chandrasekhar Sagar BK, Datta KK, Gowda H, Nalini A, et al. Cerebrospinal fluid from sporadic amyotrophic lateral sclerosis patients induces mitochondrial and lysosomal dysfunction. Neurochem Res. 2016;41:965–84.CrossRefPubMed
30.
Zurück zum Zitat Ding X, Ma M, Teng J, Teng RKF, Zhou S, Yin J, Fonkem E, Huang JH, Wu E, Wang X. Exposure to ALS-FTD-CSF generates TDP-43 aggregates in glioblastoma cells through exosomes and TNTs-like structure. Oncotarget. 2015;6:24178–91.CrossRefPubMedPubMedCentral Ding X, Ma M, Teng J, Teng RKF, Zhou S, Yin J, Fonkem E, Huang JH, Wu E, Wang X. Exposure to ALS-FTD-CSF generates TDP-43 aggregates in glioblastoma cells through exosomes and TNTs-like structure. Oncotarget. 2015;6:24178–91.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Chen Y-Y, Liu X-W, Gong T-X, Zhang Z-Y, Liu Y-E, Zhang Y-B, Xiang L-B, Xia H. Role of platelet derived growth factor (PDGF) in reverting neuronal nuclear and soma size alterations in NSC-34 cells exposed to cerebrospinal fluid from amyotrophic lateral sclerosis patients. Clin Neurol Neurosurg. 2014;120:1–5.CrossRefPubMed Chen Y-Y, Liu X-W, Gong T-X, Zhang Z-Y, Liu Y-E, Zhang Y-B, Xiang L-B, Xia H. Role of platelet derived growth factor (PDGF) in reverting neuronal nuclear and soma size alterations in NSC-34 cells exposed to cerebrospinal fluid from amyotrophic lateral sclerosis patients. Clin Neurol Neurosurg. 2014;120:1–5.CrossRefPubMed
32.
Zurück zum Zitat Gómez-Pinedo U, Galán L, Yañez M, Matias-Guiu J, Valencia C, Guerrero-Sola A, Lopez-Sosa F, Brin JR, Benito-Martin MS, Leon-Espinosa G, et al. Histological changes in the rat brain and spinal cord following prolonged intracerebroventricular infusion of cerebrospinal fluid from amyotrophic lateral sclerosis patients are similar to those caused by the disease. Neurologia. 2016; Gómez-Pinedo U, Galán L, Yañez M, Matias-Guiu J, Valencia C, Guerrero-Sola A, Lopez-Sosa F, Brin JR, Benito-Martin MS, Leon-Espinosa G, et al. Histological changes in the rat brain and spinal cord following prolonged intracerebroventricular infusion of cerebrospinal fluid from amyotrophic lateral sclerosis patients are similar to those caused by the disease. Neurologia. 2016;
33.
Zurück zum Zitat Varghese AM, Sharma A, Mishra P, Vijayalakshmi K, Harsha HC, Sathyaprabha TN, Bharath SM, Nalini A, Alladi PA, Raju TR. Chitotriosidase—a putative biomarker for sporadic amyotrophic lateral sclerosis. Clin Proteomics. 2013;10:19.CrossRefPubMedPubMedCentral Varghese AM, Sharma A, Mishra P, Vijayalakshmi K, Harsha HC, Sathyaprabha TN, Bharath SM, Nalini A, Alladi PA, Raju TR. Chitotriosidase—a putative biomarker for sporadic amyotrophic lateral sclerosis. Clin Proteomics. 2013;10:19.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Chen X, Chen Y, Wei Q, Ou R, Cao B, Zhao B, Shang H-F. Assessment of a multiple biomarker panel for diagnosis of amyotrophic lateral sclerosis. BMC Neurol. 2016;16:173.CrossRefPubMedPubMedCentral Chen X, Chen Y, Wei Q, Ou R, Cao B, Zhao B, Shang H-F. Assessment of a multiple biomarker panel for diagnosis of amyotrophic lateral sclerosis. BMC Neurol. 2016;16:173.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Brooks BR, Miller RG, Swash M, Munsat TL, Diseases WFoNRGoMN. El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000;1:293–9.CrossRefPubMed Brooks BR, Miller RG, Swash M, Munsat TL, Diseases WFoNRGoMN. El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000;1:293–9.CrossRefPubMed
36.
Zurück zum Zitat Mishra PS, Dhull DK, Nalini A, Vijayalakshmi K, Sathyaprabha TN, Alladi PA, Raju TR. Astroglia acquires a toxic neuroinflammatory role in response to the cerebrospinal fluid from amyotrophic lateral sclerosis patients. J Neuroinflammation. 2016;13:212.CrossRefPubMedPubMedCentral Mishra PS, Dhull DK, Nalini A, Vijayalakshmi K, Sathyaprabha TN, Alladi PA, Raju TR. Astroglia acquires a toxic neuroinflammatory role in response to the cerebrospinal fluid from amyotrophic lateral sclerosis patients. J Neuroinflammation. 2016;13:212.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Saura J, Tusell JM, Serratosa J. High-yield isolation of murine microglia by mild trypsinization. Glia. 2003;44:183–9.CrossRefPubMed Saura J, Tusell JM, Serratosa J. High-yield isolation of murine microglia by mild trypsinization. Glia. 2003;44:183–9.CrossRefPubMed
38.
Zurück zum Zitat Vijayalakshmi K, Ostwal P, Sumitha R, Shruthi S, Varghese AM, Mishra P, Manohari SG, Sagar BC, Sathyaprabha TN, Nalini A, et al. Role of VEGF and VEGFR2 receptor in reversal of ALS-CSF induced degeneration of NSC-34 motor neuron cell line. Mol Neurobiol. 2015;51:995–1007.CrossRefPubMed Vijayalakshmi K, Ostwal P, Sumitha R, Shruthi S, Varghese AM, Mishra P, Manohari SG, Sagar BC, Sathyaprabha TN, Nalini A, et al. Role of VEGF and VEGFR2 receptor in reversal of ALS-CSF induced degeneration of NSC-34 motor neuron cell line. Mol Neurobiol. 2015;51:995–1007.CrossRefPubMed
39.
Zurück zum Zitat Dong Z, Wang L, Xu J, Li Y, Zhang Y, Zhang S, Miao J. Promotion of autophagy and inhibition of apoptosis by low concentrations of cadmium in vascular endothelial cells. Toxicol in Vitro. 2009;23:105–10.CrossRefPubMed Dong Z, Wang L, Xu J, Li Y, Zhang Y, Zhang S, Miao J. Promotion of autophagy and inhibition of apoptosis by low concentrations of cadmium in vascular endothelial cells. Toxicol in Vitro. 2009;23:105–10.CrossRefPubMed
40.
Zurück zum Zitat Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-delta delta C(T)) method. Methods. 2001;25:402–8.CrossRefPubMed Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-delta delta C(T)) method. Methods. 2001;25:402–8.CrossRefPubMed
41.
Zurück zum Zitat Fendrick SE, Xue QS, Streit WJ. Formation of multinucleated giant cells and microglial degeneration in rats expressing a mutant Cu/Zn superoxide dismutase gene. J Neuroinflammation. 2007;4:9.CrossRefPubMedPubMedCentral Fendrick SE, Xue QS, Streit WJ. Formation of multinucleated giant cells and microglial degeneration in rats expressing a mutant Cu/Zn superoxide dismutase gene. J Neuroinflammation. 2007;4:9.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Brites D, Vaz AR. Microglia centered pathogenesis in ALS: insights in cell interconnectivity. Front Cell Neurosci. 2014;8 Brites D, Vaz AR. Microglia centered pathogenesis in ALS: insights in cell interconnectivity. Front Cell Neurosci. 2014;8
43.
Zurück zum Zitat Liao B, Zhao W, Beers DR, Henkel JS, Appel SH. Transformation from a neuroprotective to a neurotoxic microglial phenotype in a mouse model of ALS. Exp Neurol. 2012;237:147–52.CrossRefPubMedPubMedCentral Liao B, Zhao W, Beers DR, Henkel JS, Appel SH. Transformation from a neuroprotective to a neurotoxic microglial phenotype in a mouse model of ALS. Exp Neurol. 2012;237:147–52.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Feiler MS, Strobel B, Freischmidt A, Helferich AM, Kappel J, Brewer BM, Li D, Thal DR, Walther P, Ludolph AC, et al. TDP-43 is intercellularly transmitted across axon terminals. J Cell Biol. 2015;211:897–911.CrossRefPubMedPubMedCentral Feiler MS, Strobel B, Freischmidt A, Helferich AM, Kappel J, Brewer BM, Li D, Thal DR, Walther P, Ludolph AC, et al. TDP-43 is intercellularly transmitted across axon terminals. J Cell Biol. 2015;211:897–911.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Iguchi Y, Eid L, Parent M, Soucy G, Bareil C, Riku Y, Kawai K, Takagi S, Yoshida M, Katsuno M, et al. Exosome secretion is a key pathway for clearance of pathological TDP-43. Brain. 2016; Iguchi Y, Eid L, Parent M, Soucy G, Bareil C, Riku Y, Kawai K, Takagi S, Yoshida M, Katsuno M, et al. Exosome secretion is a key pathway for clearance of pathological TDP-43. Brain. 2016;
46.
Zurück zum Zitat Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, Rak J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10:619–24.CrossRefPubMed Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, Rak J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10:619–24.CrossRefPubMed
47.
Zurück zum Zitat Falchi AM, Sogos V, Saba F, Piras M, Congiu T, Piludu M. Astrocytes shed large membrane vesicles that contain mitochondria, lipid droplets and ATP. Histochem Cell Biol. 2013;139:221–31.CrossRefPubMed Falchi AM, Sogos V, Saba F, Piras M, Congiu T, Piludu M. Astrocytes shed large membrane vesicles that contain mitochondria, lipid droplets and ATP. Histochem Cell Biol. 2013;139:221–31.CrossRefPubMed
48.
Zurück zum Zitat Wu B, Huang Y, Braun AL, Tong Z, Zhao R, Li Y, Liu F, Zheng JC. Glutaminase-containing microvesicles from HIV-1-infected macrophages and immune-activated microglia induce neurotoxicity. Mol Neurodegener. 2015;10:61.CrossRefPubMedPubMedCentral Wu B, Huang Y, Braun AL, Tong Z, Zhao R, Li Y, Liu F, Zheng JC. Glutaminase-containing microvesicles from HIV-1-infected macrophages and immune-activated microglia induce neurotoxicity. Mol Neurodegener. 2015;10:61.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Tang L-L, Y-B W, Fang C-Q, Qu P, Gao Z-L. NDRG2 promoted secreted miR-375 in microvesicles shed from M1 microglia, which induced neuron damage. Biochem Biophys Res Commun. 2015; Tang L-L, Y-B W, Fang C-Q, Qu P, Gao Z-L. NDRG2 promoted secreted miR-375 in microvesicles shed from M1 microglia, which induced neuron damage. Biochem Biophys Res Commun. 2015;
50.
Zurück zum Zitat Verderio C, Muzio L, Turola E, Bergami A, Novellino L, Ruffini F, Riganti L, Corradini I, Francolini M, Garzetti L, et al. Myeloid microvesicles are a marker and therapeutic target for neuroinflammation. Ann Neurol. 2012;72:610–24.CrossRefPubMed Verderio C, Muzio L, Turola E, Bergami A, Novellino L, Ruffini F, Riganti L, Corradini I, Francolini M, Garzetti L, et al. Myeloid microvesicles are a marker and therapeutic target for neuroinflammation. Ann Neurol. 2012;72:610–24.CrossRefPubMed
51.
Zurück zum Zitat Garzetti L, Menon R, Finardi A, Bergami A, Sica A, Martino G, Comi G, Verderio C, Farina C, Furlan R. Activated macrophages release microvesicles containing polarized M1 or M2 mRNAs. J Leukoc Biol. 2013; Garzetti L, Menon R, Finardi A, Bergami A, Sica A, Martino G, Comi G, Verderio C, Farina C, Furlan R. Activated macrophages release microvesicles containing polarized M1 or M2 mRNAs. J Leukoc Biol. 2013;
52.
Zurück zum Zitat Hornik TC, Neniskyte U, Brown GC. Inflammation induces multinucleation of microglia via PKC inhibition of cytokinesis, generating highly phagocytic multinucleated giant cells. J Neurochem. 2014;128:650–61.CrossRefPubMed Hornik TC, Neniskyte U, Brown GC. Inflammation induces multinucleation of microglia via PKC inhibition of cytokinesis, generating highly phagocytic multinucleated giant cells. J Neurochem. 2014;128:650–61.CrossRefPubMed
53.
Zurück zum Zitat Kawahara K, Mori M, Nakayama H. NO-induced apoptosis and ER stress in microglia. Nihon Yakurigaku Zasshi. 2004;124:399–406.CrossRefPubMed Kawahara K, Mori M, Nakayama H. NO-induced apoptosis and ER stress in microglia. Nihon Yakurigaku Zasshi. 2004;124:399–406.CrossRefPubMed
54.
Zurück zum Zitat Frakes AE, Ferraiuolo L, Haidet-Phillips AM, Schmelzer L, Braun L, Miranda CJ, Ladner KJ, Bevan AK, Foust KD, Godbout JP, et al. Microglia induce motor neuron death via the classical NF-κB pathway in amyotrophic lateral sclerosis. Neuron. 2014;81:1009–23.CrossRefPubMedPubMedCentral Frakes AE, Ferraiuolo L, Haidet-Phillips AM, Schmelzer L, Braun L, Miranda CJ, Ladner KJ, Bevan AK, Foust KD, Godbout JP, et al. Microglia induce motor neuron death via the classical NF-κB pathway in amyotrophic lateral sclerosis. Neuron. 2014;81:1009–23.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Swarup V, Phaneuf D, Dupré N, Petri S, Strong M, Kriz J, Julien J-P. Deregulation of TDP-43 in amyotrophic lateral sclerosis triggers nuclear factor κB-mediated pathogenic pathways. J Exp Med. 2011;208:2429–47.CrossRefPubMedPubMedCentral Swarup V, Phaneuf D, Dupré N, Petri S, Strong M, Kriz J, Julien J-P. Deregulation of TDP-43 in amyotrophic lateral sclerosis triggers nuclear factor κB-mediated pathogenic pathways. J Exp Med. 2011;208:2429–47.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Zhao W, Beers DR, Bell S, Wang J, Wen S, Baloh RH, Appel SH. TDP-43 activates microglia through NF-kappaB and NLRP3 inflammasome. Exp Neurol. 2015;273:24–35.CrossRefPubMed Zhao W, Beers DR, Bell S, Wang J, Wen S, Baloh RH, Appel SH. TDP-43 activates microglia through NF-kappaB and NLRP3 inflammasome. Exp Neurol. 2015;273:24–35.CrossRefPubMed
57.
Zurück zum Zitat Lall D, Baloh RH. Microglia and C9orf72 in neuroinflammation and ALS and frontotemporal dementia. J Clin Invest. 2017;127:3250–8.CrossRefPubMed Lall D, Baloh RH. Microglia and C9orf72 in neuroinflammation and ALS and frontotemporal dementia. J Clin Invest. 2017;127:3250–8.CrossRefPubMed
58.
Zurück zum Zitat Lewis C-A, Manning J, Rossi F, Krieger C. The neuroinflammatory response in ALS: the roles of microglia and T cells. Neurol Res Int. 2012;2012:803701.CrossRefPubMedPubMedCentral Lewis C-A, Manning J, Rossi F, Krieger C. The neuroinflammatory response in ALS: the roles of microglia and T cells. Neurol Res Int. 2012;2012:803701.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Takaki J, Fujimori K, Miura M, Suzuki T, Sekino Y, Sato K. L-Glutamate released from activated microglia downregulates astrocytic L-glutamate transporter expression in neuroinflammation: the ‘collusion’ hypothesis for increased extracellular L-glutamate concentration in neuroinflammation. J Neuroinflammation. 2012;9:275.CrossRefPubMedPubMedCentral Takaki J, Fujimori K, Miura M, Suzuki T, Sekino Y, Sato K. L-Glutamate released from activated microglia downregulates astrocytic L-glutamate transporter expression in neuroinflammation: the ‘collusion’ hypothesis for increased extracellular L-glutamate concentration in neuroinflammation. J Neuroinflammation. 2012;9:275.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Shobha K, Vijayalakshmi K, Alladi PA, Nalini A, Sathyaprabha TN, Raju TR. Altered in-vitro and in-vivo expression of glial glutamate transporter-1 following exposure to cerebrospinal fluid of amyotrophic lateral sclerosis patients. J Neurol Sci. 2007;254:9–16.CrossRefPubMed Shobha K, Vijayalakshmi K, Alladi PA, Nalini A, Sathyaprabha TN, Raju TR. Altered in-vitro and in-vivo expression of glial glutamate transporter-1 following exposure to cerebrospinal fluid of amyotrophic lateral sclerosis patients. J Neurol Sci. 2007;254:9–16.CrossRefPubMed
61.
Zurück zum Zitat Agulhon C, Sun MY, Murphy T, Myers T, Lauderdale K, Fiacco TA. Calcium signaling and gliotransmission in normal vs. reactive astrocytes. Front Pharmacol. 2012;3 Agulhon C, Sun MY, Murphy T, Myers T, Lauderdale K, Fiacco TA. Calcium signaling and gliotransmission in normal vs. reactive astrocytes. Front Pharmacol. 2012;3
62.
Zurück zum Zitat Socodato R, Portugal CC, Canedo T, Domith I, Oliveira NA, Paes-de-Carvalho R, Relvas JB, Cossenza M. c-Src deactivation by the polyphenol 3-O-caffeoylquinic acid abrogates reactive oxygen species-mediated glutamate release from microglia and neuronal excitotoxicity. Free Radic Biol Med. 2015;79C:45–55.CrossRef Socodato R, Portugal CC, Canedo T, Domith I, Oliveira NA, Paes-de-Carvalho R, Relvas JB, Cossenza M. c-Src deactivation by the polyphenol 3-O-caffeoylquinic acid abrogates reactive oxygen species-mediated glutamate release from microglia and neuronal excitotoxicity. Free Radic Biol Med. 2015;79C:45–55.CrossRef
63.
Zurück zum Zitat Vesce S, Rossi D, Brambilla L, Volterra A. Glutamate release from astrocytes in physiological conditions and in neurodegenerative disorders characterized by neuroinflammation. Int Rev Neurobiol. 2007;82:57–71.CrossRefPubMed Vesce S, Rossi D, Brambilla L, Volterra A. Glutamate release from astrocytes in physiological conditions and in neurodegenerative disorders characterized by neuroinflammation. Int Rev Neurobiol. 2007;82:57–71.CrossRefPubMed
64.
Zurück zum Zitat Bal-Price A, Brown GC. Inflammatory neurodegeneration mediated by nitric oxide from activated glia-inhibiting neuronal respiration, causing glutamate release and excitotoxicity. J Neurosci. 2001;21:6480–91.PubMed Bal-Price A, Brown GC. Inflammatory neurodegeneration mediated by nitric oxide from activated glia-inhibiting neuronal respiration, causing glutamate release and excitotoxicity. J Neurosci. 2001;21:6480–91.PubMed
65.
Zurück zum Zitat Hensley K, Floyd RA, Gordon B, Mou S, Pye QN, Stewart C, West M, Williamson K. Temporal patterns of cytokine and apoptosis-related gene expression in spinal cords of the G93A-SOD1 mouse model of amyotrophic lateral sclerosis. J Neurochem. 2002;82:365–74.CrossRefPubMed Hensley K, Floyd RA, Gordon B, Mou S, Pye QN, Stewart C, West M, Williamson K. Temporal patterns of cytokine and apoptosis-related gene expression in spinal cords of the G93A-SOD1 mouse model of amyotrophic lateral sclerosis. J Neurochem. 2002;82:365–74.CrossRefPubMed
66.
Zurück zum Zitat Yoshihara T, Ishigaki S, Yamamoto M, Liang Y, Niwa J, Takeuchi H, Doyu M, Sobue G. Differential expression of inflammation- and apoptosis-related genes in spinal cords of a mutant SOD1 transgenic mouse model of familial amyotrophic lateral sclerosis. J Neurochem. 2002;80:158–67.CrossRefPubMed Yoshihara T, Ishigaki S, Yamamoto M, Liang Y, Niwa J, Takeuchi H, Doyu M, Sobue G. Differential expression of inflammation- and apoptosis-related genes in spinal cords of a mutant SOD1 transgenic mouse model of familial amyotrophic lateral sclerosis. J Neurochem. 2002;80:158–67.CrossRefPubMed
67.
Zurück zum Zitat Ding X, Yan Y, Li X, Li K, Ciric B, Yang J, Zhang Y, Wu S, Xu H, Chen W, et al. Silencing IFN-γ binding/signaling in astrocytes versus microglia leads to opposite effects on central nervous system autoimmunity. J Immunol. 2015;194:4251–64.CrossRefPubMedPubMedCentral Ding X, Yan Y, Li X, Li K, Ciric B, Yang J, Zhang Y, Wu S, Xu H, Chen W, et al. Silencing IFN-γ binding/signaling in astrocytes versus microglia leads to opposite effects on central nervous system autoimmunity. J Immunol. 2015;194:4251–64.CrossRefPubMedPubMedCentral
68.
Zurück zum Zitat Baruch K, Deczkowska A, Rosenzweig N, Tsitsou-Kampeli A, Sharif AM, Matcovitch-Natan O, Kertser A, David E, Amit I, Schwartz M. PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer’s disease. Nat Med. 2016;22:135–7.CrossRefPubMed Baruch K, Deczkowska A, Rosenzweig N, Tsitsou-Kampeli A, Sharif AM, Matcovitch-Natan O, Kertser A, David E, Amit I, Schwartz M. PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer’s disease. Nat Med. 2016;22:135–7.CrossRefPubMed
69.
Zurück zum Zitat Mărgăritescu O, Pirici D, Mărgăritescu C. VEGF expression in human brain tissue after acute ischemic stroke. Romanian J Morphol Embryol. 2011;52:1283–92. Mărgăritescu O, Pirici D, Mărgăritescu C. VEGF expression in human brain tissue after acute ischemic stroke. Romanian J Morphol Embryol. 2011;52:1283–92.
70.
Zurück zum Zitat Matsushita Y, Nakajima K, Tohyama Y, Kurihara T, Kohsaka S. Activation of microglia by endotoxin suppresses the secretion of glial cell line-derived neurotrophic factor (GDNF) through the action of protein kinase C alpha (PKCalpha) and mitogen-activated protein kinases (MAPKS). J Neurosci Res. 2008;86:1959–71.CrossRefPubMed Matsushita Y, Nakajima K, Tohyama Y, Kurihara T, Kohsaka S. Activation of microglia by endotoxin suppresses the secretion of glial cell line-derived neurotrophic factor (GDNF) through the action of protein kinase C alpha (PKCalpha) and mitogen-activated protein kinases (MAPKS). J Neurosci Res. 2008;86:1959–71.CrossRefPubMed
71.
Zurück zum Zitat Cudkowicz ME, Shefner JM, Schoenfeld DA, Zhang H, Andreasson KI, Rothstein JD, Drachman DB. Trial of celecoxib in amyotrophic lateral sclerosis. Ann Neurol. 2006;60:22–31.CrossRefPubMed Cudkowicz ME, Shefner JM, Schoenfeld DA, Zhang H, Andreasson KI, Rothstein JD, Drachman DB. Trial of celecoxib in amyotrophic lateral sclerosis. Ann Neurol. 2006;60:22–31.CrossRefPubMed
72.
Zurück zum Zitat Gordon PH, Moore DH, Miller RG, Florence JM, Verheijde JL, Doorish C, Hilton JF, Spitalny GM, MacArthur RB, Mitsumoto H, et al. Efficacy of minocycline in patients with amyotrophic lateral sclerosis: a phase III randomised trial. Lancet Neurol. 6:1045–53. Gordon PH, Moore DH, Miller RG, Florence JM, Verheijde JL, Doorish C, Hilton JF, Spitalny GM, MacArthur RB, Mitsumoto H, et al. Efficacy of minocycline in patients with amyotrophic lateral sclerosis: a phase III randomised trial. Lancet Neurol. 6:1045–53.
73.
Zurück zum Zitat Van Den Bosch L, Tilkin P, Lemmens G, Robberecht W. Minocycline delays disease onset and mortality in a transgenic model of ALS. Neuroreport. 2002;13:1067–70.CrossRefPubMed Van Den Bosch L, Tilkin P, Lemmens G, Robberecht W. Minocycline delays disease onset and mortality in a transgenic model of ALS. Neuroreport. 2002;13:1067–70.CrossRefPubMed
74.
Zurück zum Zitat Keller AF, Gravel M, Kriz J. Treatment with minocycline after disease onset alters astrocyte reactivity and increases microgliosis in SOD1 mutant mice. Exp Neurol. 2011;228:69–79.CrossRefPubMed Keller AF, Gravel M, Kriz J. Treatment with minocycline after disease onset alters astrocyte reactivity and increases microgliosis in SOD1 mutant mice. Exp Neurol. 2011;228:69–79.CrossRefPubMed
75.
Zurück zum Zitat Ramanathan R, Kohli A, Ingaramo Mí C, Jain A, Leng SX, Punjabi NM, Walston JD, Fedarko NS. Serum chitotriosidase, a putative marker of chronically activated macrophages, increases with normal aging. J Gerontol A Biol Sci Med Sci. 2013;68:1303–9.CrossRefPubMedPubMedCentral Ramanathan R, Kohli A, Ingaramo Mí C, Jain A, Leng SX, Punjabi NM, Walston JD, Fedarko NS. Serum chitotriosidase, a putative marker of chronically activated macrophages, increases with normal aging. J Gerontol A Biol Sci Med Sci. 2013;68:1303–9.CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat Di Rosa M, Malaguarnera G, De Gregorio C, D’Amico F, Mazzarino MC, Malaguarnera L. Modulation of chitotriosidase during macrophage differentiation. Cell Biochem Biophys. 2013;66:239–47.CrossRefPubMed Di Rosa M, Malaguarnera G, De Gregorio C, D’Amico F, Mazzarino MC, Malaguarnera L. Modulation of chitotriosidase during macrophage differentiation. Cell Biochem Biophys. 2013;66:239–47.CrossRefPubMed
77.
Zurück zum Zitat Sotgiu S, Musumeci S, Marconi S, Gini B, Bonetti B. Different content of chitin-like polysaccharides in multiple sclerosis and Alzheimer’s disease brains. J Neuroimmunol. 2008;197:70–3.CrossRefPubMed Sotgiu S, Musumeci S, Marconi S, Gini B, Bonetti B. Different content of chitin-like polysaccharides in multiple sclerosis and Alzheimer’s disease brains. J Neuroimmunol. 2008;197:70–3.CrossRefPubMed
Metadaten
Titel
Etiogenic factors present in the cerebrospinal fluid from amyotrophic lateral sclerosis patients induce predominantly pro-inflammatory responses in microglia
verfasst von
Pooja-Shree Mishra
K. Vijayalakshmi
A. Nalini
T. N. Sathyaprabha
B. W. Kramer
Phalguni Anand Alladi
T. R. Raju
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2017
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-017-1028-x

Weitere Artikel der Ausgabe 1/2017

Journal of Neuroinflammation 1/2017 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.