Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Glia ; 71(3): 616-632, 2023 03.
Article in English | MEDLINE | ID: mdl-36394300

ABSTRACT

In the central nervous system (CNS), insulin-like growth factor 1 (IGF-1) regulates myelination by oligodendrocyte (ODC) precursor cells and shows anti-apoptotic properties in neuronal cells in different in vitro and in vivo systems. Previous work also suggests that IGF-1 protects ODCs from cell death and enhances remyelination in models of toxin-induced and autoimmune demyelination. However, since evidence remains controversial, the therapeutic potential of IGF-1 in demyelinating CNS conditions is unclear. To finally shed light on the function of IGF1-signaling for ODCs, we deleted insulin-like growth factor 1 receptor (IGF1R) specifically in mature ODCs of the mouse. We found that ODC survival and myelin status were unaffected by the absence of IGF1R until 15 months of age, indicating that IGF-1 signaling does not play a major role in post-mitotic ODCs during homeostasis. Notably, the absence of IGF1R did neither affect ODC survival nor myelin status upon cuprizone intoxication or induction of experimental autoimmune encephalomyelitis (EAE), models for toxic and autoimmune demyelination, respectively. Surprisingly, however, the absence of IGF1R from ODCs protected against clinical neuroinflammation in the EAE model. Together, our data indicate that IGF-1 signaling is not required for the function and survival of mature ODCs in steady-state and disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Insulin-Like Growth Factor I , Receptor, IGF Type 1 , Animals , Mice , Cuprizone , Encephalomyelitis, Autoimmune, Experimental/metabolism , Insulin-Like Growth Factor I/metabolism , Mice, Inbred C57BL , Myelin Sheath/metabolism , Neuroinflammatory Diseases , Oligodendroglia/metabolism , Receptor, IGF Type 1/metabolism
2.
Nature ; 585(7823): 107-112, 2020 09.
Article in English | MEDLINE | ID: mdl-32728218

ABSTRACT

Treating patients who have cancer with vaccines that stimulate a targeted immune response is conceptually appealing, but cancer vaccine trials have not been successful in late-stage patients with treatment-refractory tumours1,2. We are testing melanoma FixVac (BNT111)-an intravenously administered liposomal RNA (RNA-LPX) vaccine, which targets four non-mutated, tumour-associated antigens that are prevalent in melanoma-in an ongoing, first-in-human, dose-escalation phase I trial in patients with advanced melanoma (Lipo-MERIT trial, ClinicalTrials.gov identifier NCT02410733). We report here data from an exploratory interim analysis that show that melanoma FixVac, alone or in combination with blockade of the checkpoint inhibitor PD1, mediates durable objective responses in checkpoint-inhibitor (CPI)-experienced patients with unresectable melanoma. Clinical responses are accompanied by the induction of strong CD4+ and CD8+ T cell immunity against the vaccine antigens. The antigen-specific cytotoxic T-cell responses in some responders reach magnitudes typically reported for adoptive T-cell therapy, and are durable. Our findings indicate that RNA-LPX vaccination is a potent immunotherapy in patients with CPI-experienced melanoma, and suggest the general utility of non-mutant shared tumour antigens as targets for cancer vaccination.


Subject(s)
Antineoplastic Agents/therapeutic use , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Melanoma/immunology , Melanoma/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , RNA, Neoplasm/genetics , T-Lymphocytes/immunology , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Combined Modality Therapy , Humans , Melanoma/drug therapy , Melanoma/pathology , Neoplasm Staging , T-Lymphocytes/cytology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Vaccination
3.
PLoS One ; 13(8): e0202954, 2018.
Article in English | MEDLINE | ID: mdl-30157279

ABSTRACT

BACKGROUND: Abnormal aggregation of proteins induces neuronal cell loss in neurodegenerative disorders such as Alzheimer's Disease, Creutzfeldt-Jakob Disease and Parkinson's Disease. Specific stimuli initialize conformational changes in physiological proteins, causing intra- or extracellular protein aggregation. We and other groups have identified naturally occurring autoantibodies (nAbs) as part of the human antibody pool that are able to prevent peptide fibrillation. These nAbs show a rescue effect following exposure of toxic aggregates on neurons, and they support microglial uptake of aggregated peptides. OBJECTIVE: Identification of a putative common epitope among the relevant proteins ß-Amyloid, α-Synuclein and Prion Protein for the respective nAbs. MATERIAL AND METHODS: Binding affinity between the aforementioned proteins and nAbs was tested by Dot Blot, ELISA and SPR-technology. Furthermore, the functionality of the protein-nAbs-complexes was studied in Thioflavin-T assays and microglial uptake experiments to study dependent inhibition of protein aggregation and enhancement of Fcγ mediated uptake by microglial cells. RESULTS: ß-Amyloid and Prion Protein fragment showed considerable binding affinity and functional efficacy for all applied nAbs. Thereby, no significant difference within the different nAbs was detected. In contrast, α-Synuclein was bound exclusively by nAbs-α-Synuclein, which was reproduced in all binding studies. Surprisingly, functional assays with α-Synuclein revealed no significant effect of nAbs in comparison to IVIg treatment. However, all applied nAbs as well as IVIg show a minimal functionality on the microglial uptake of α-Synuclein. CONCLUSION: nAbs-Aß, nAbs-PrP possibly display comparable affinity to the same structural epitope within Aß and PrP106-126 A117V whereas the epitope recognized by nAbs-α-Syn is only present in α-Syn. The structural similarity of Aß and PrP fragment promotes the outline for an efficient antibody for the treatment of several neurodegenerative disorders and extend the functional characteristics of the investigated nAbs.


Subject(s)
Amyloid beta-Peptides/immunology , Autoantibodies/immunology , Peptide Fragments/immunology , Prion Proteins/immunology , alpha-Synuclein/immunology , Animals , Autoantibodies/chemistry , Cell Line , Epitopes/chemistry , Epitopes/immunology , Humans , Mice
4.
Bioorg Med Chem ; 25(2): 603-608, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27887962

ABSTRACT

The idea of privileged scaffolds - that there seem to be more bioactive compounds found around some structures than others - is well established for small drug molecules, but has little significance for standalone peptide secondary structures whose adaptable shapes escape the definition of a 3D motif in the absence of a protein scaffold. Here, we joined two independent biological functions in a single highly restricted peptide to support the hypothesis that the ß-hairpin shape is the common basis of two otherwise unrelated biological recognition processes. To achieve this, the hydrophobic cluster HWX4LV from the decapeptide cyclic hairpin model peptide C1-C10cyclo-CHWEGNKLVC was included in the bicyclic peptide 2. The designed ß-hairpin peptide C4-C17, C8-C13bicyclo-KHQCHWECTZGRCRLVCGRSGS (2, Z=citrulline), serves, on the one hand, as a specific epitope for rheumatoid autoantibodies and, on the other hand, shows a not negligible antibiotic effect against the bacterial strain E. coli AS19.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Peptides/chemical synthesis , Peptides/pharmacology , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Peptides/chemistry , Protein Conformation , Structure-Activity Relationship
5.
J Neurol Sci ; 359(1-2): 356-62, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26671142

ABSTRACT

Excessive glutamate secretion leads to excitotoxicity, which has been shown to underlie neurodegenerative disorders. Excitotoxicity is in part exerted by overactivation of calpains, which promote neuronal cell death via induction of limited proteolysis of the cellular proteins p35, regulatory subunit of cyclin-dependent kinase 5, and αII-spectrin. We used primary murine neuronal cells in a model of glutamate toxicity. The protease inhibitor α1-antitrypsin was able to prevent glutamate toxicity as determined by MTT assay and immunofluorescence. Calpain and caspase 3 activity were reduced following α1-antitrypsin treatment, as assessed by calpain and caspase 3 activity assays. In addition we could observe a modulation of cleavage of the calpain/caspase substrates αII-spectrin and p35 in Western blots. In summary, α1-antitrypsin shows inhibitory effects on excitotoxicity of primary neurons involving the inhibition of calpain activity. The advantage of using α1-antitrypsin is that the substance is already in clinical use for the treatment of patients with hereditary α1-antitrypsin deficiency. Further experiments are required in animal models of neurodegenerative disorders to assess the suitability of this substance in patients suffering from Alzheimer's disease or Parkinson's disease.


Subject(s)
Calpain/metabolism , Excitatory Amino Acids/toxicity , Glutamic Acid/toxicity , Neurons/drug effects , Analysis of Variance , Animals , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Mice , Phosphotransferases/metabolism , Time Factors , alpha 1-Antitrypsin/pharmacology
6.
Semin Immunopathol ; 37(6): 607-11, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26251237

ABSTRACT

There is extensive evidence that accumulation of mononuclear phagocytes including microglial cells, monocytes, and macrophages at sites of ß-amyloid (Aß) deposition in the brain is an important pathological feature of Alzheimer's disease (AD) and related animal models, and the concentration of these cells clustered around Aß deposits is several folds higher than in neighboring areas of the brain [1-5]. Microglial cells phagocytose and clear debris, pathogens, and toxins, but they can also be activated to produce inflammatory cytokines, chemokines, and neurotoxins [6]. Over the past decade, the roles of microglial cells in AD have begun to be clarified, and we proposed that these cells play a dichotomous role in the pathogenesis of AD [4, 6-11]. Microglial cells are able to clear soluble and fibrillar Aß, but continued interactions of these cells with Aß can lead to an inflammatory response resulting in neurotoxicity. Inflammasomes are inducible high molecular weight protein complexes that are involved in many inflammatory pathological processes. Recently, Aß was found to activate the NLRP3 inflammasome in microglial cells in vitro and in vivo thereby defining a novel pathway that could lead to progression of AD [12-14]. In this manuscript, we review possible steps leading to Aß-induced inflammasome activation and discuss how this could contribute to the pathogenesis of AD.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/metabolism , Brain/immunology , Carrier Proteins/immunology , Alzheimer Disease/pathology , Brain/cytology , Brain/pathology , Cytokines/metabolism , Humans , Inflammasomes , Macrophages/immunology , Microglia/immunology , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis
7.
PLoS One ; 10(7): e0132227, 2015.
Article in English | MEDLINE | ID: mdl-26168044

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is diagnosed based upon medical history, neuropsychiatric examination, cerebrospinal fluid analysis, extensive laboratory analyses and cerebral imaging. Diagnosis is time consuming and labour intensive. Parkinson's disease (PD) is mainly diagnosed on clinical grounds. OBJECTIVE: The primary aim of this study was to differentiate patients suffering from AD, PD and healthy controls by investigating exhaled air with the electronic nose technique. After demonstrating a difference between the three groups the secondary aim was the identification of specific substances responsible for the difference(s) using ion mobility spectroscopy. Thirdly we analysed whether amyloid beta (Aß) in exhaled breath was causative for the observed differences between patients suffering from AD and healthy controls. METHODS: We employed novel pulmonary diagnostic tools (electronic nose device/ion-mobility spectrometry) for the identification of patients with neurodegenerative diseases. Specifically, we analysed breath pattern differences in exhaled air of patients with AD, those with PD and healthy controls using the electronic nose device (eNose). Using ion mobility spectrometry (IMS), we identified the compounds responsible for the observed differences in breath patterns. We applied ELISA technique to measure Aß in exhaled breath condensates. RESULTS: The eNose was able to differentiate between AD, PD and HC correctly. Using IMS, we identified markers that could be used to differentiate healthy controls from patients with AD and PD with an accuracy of 94%. In addition, patients suffering from PD were identified with sensitivity and specificity of 100%. Altogether, 3 AD patients out of 53 participants were misclassified. Although we found Aß in exhaled breath condensate from both AD and healthy controls, no significant differences between groups were detected. CONCLUSION: These data may open a new field in the diagnosis of neurodegenerative disease such as Alzheimer's disease and Parkinson's disease. Further research is required to evaluate the significance of these pulmonary findings with respect to the pathophysiology of neurodegenerative disorders.


Subject(s)
Alzheimer Disease/diagnosis , Breath Tests , Parkinson Disease/diagnosis , Aged , Amyloid beta-Peptides/analysis , Animals , Biomarkers/analysis , Blotting, Western , Breath Tests/methods , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lung/chemistry , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Peptide Fragments/analysis , Reproducibility of Results , Sensitivity and Specificity , Spectrum Analysis/methods
8.
PLoS One ; 9(12): e114566, 2014.
Article in English | MEDLINE | ID: mdl-25470145

ABSTRACT

Alpha-synuclein (α-Syn) plays a pivotal role in the pathophysiology of Parkinson's disease (PD), which can partly be modulated by innate and adaptive immune functions, and vice versa. Here, naturally occurring α-Syn autoantibodies (α-Syn-nAbs) may be effective against α-Syn pathoetiology and may serve as a PD biomarker. However, serum and cerebrospinal fluid α-Syn-nAbs levels still lack consistent evidence as required for a reliable PD biomarker. Serum and cerebrospinal fluid α-Syn-nAbs levels of 66 PD patients and 69 healthy controls were assessed using a validated ELISA assay. Moreover, potential sources of error variance including unspecific ELISA background signals, free serum hemoglobin concentrations, α-Syn plate coating procedures, and differences in α-Syn-nAbs standards, were investigated. PD patients and controls did not differ in serum (p = .49) nor cerebrospinal fluid (p = .29) α-Syn-nAbs levels. Interestingly, free serum hemoglobin concentrations were negatively correlated with α-Syn-nAbs levels in controls (Spearman ρ = -.41, p<.001), but not in PD patients (ρ = .16, p = .21). ELISA α-Syn plate coating procedures impacted inter-assay variability (same day coating: 8-16%; coating on different days: 16-58%). α-Syn-nAbs standards from different purification batches differed regarding optical density measured in ELISAs suggesting differences in α-Syn affinity. While α-Syn-nAbs levels may represent a potential PD biomarker, several methodological issues have to be considered to increase reproducibility of α-Syn-nAbs findings. Further studies using standardized protocols minimizing sources of error variance may be necessary to establish a reliable PD α-Syn-nAbs biomarker.


Subject(s)
Autoantibodies/blood , Parkinson Disease/immunology , alpha-Synuclein/immunology , Autoantibodies/cerebrospinal fluid , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Case-Control Studies , Diagnostic Errors , Female , Humans , Male , Parkinson Disease/blood , Parkinson Disease/cerebrospinal fluid , Parkinson Disease/diagnosis
9.
J Neuroinflammation ; 11: 165, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25245568

ABSTRACT

BACKGROUND: One hallmark of Alzheimer disease is microglial activation. Therapeutic approaches for this neurodegenerative disease include the modulation of microglial cells. α1-antitrypsin (A1AT) has been shown to exert anti-inflammatory effects on macrophages and lung epithelial cells and an inhibition of calpain activity in neutrophil granulocytes. Nothing is known about the effect of A1AT on microglial-mediated neuroinflammation. Our aim was to investigate the effect of A1AT on amyloid-ß (Aß)- and LPS-treated microglial cells in vitro with respect to cytokine production, stress pathways, cell viability, phagocytotic abilities and the underlying mechanisms. METHODS: Primary microglial cells were isolated from Swiss Webster mouse embryos on embryonic day 13.5. Cytokines in the supernatants of treated primary microglial cells were analyzed with ELISAs, and accumulated nitrite was detected with Griess reagents. Intracellular stress pathways were investigated in cell lysates using western blotting. Intracellular calcium levels were detected in BV-2 microglial cells loaded with the Ca2+-sensitive (fluorescent) dye Fluo-4. Calpain activity in primary microglial cells was assessed by using a calpain activity assay. Cell viability of Aß-treated microglial cells was analyzed using MTT assay. Phagocytosis of Aß was evaluated with western blot analysis. RESULTS: Upon co-administration, A1AT reduced pro-inflammatory mediators induced by LPS or Aß. Interestingly, we detected a reduction in calpain activity and in the concentration of intracellular calcium that might mediate the anti-inflammatory effects of A1AT. Inhibition of the classic activation pathways, such as phosphorylation of mitogen-activated protein kinases or activation of protein kinase A were excluded as a mechanism of A1AT-mediated effects. In addition, A1AT increased the viability of Aß-treated microglial cells and reduced Aß phagocytosis. CONCLUSIONS: We provide evidence on the mechanism of action of A1AT on microglial-mediated neuroinflammation in vitro. Our in vitro data indicate that A1AT treatment modulates microglial cells in inflammatory conditions and that this modulation is due to an inhibition of calpain activity and intracellular calcium levels. The underlying mechanisms of the effects observed here are promising for future therapeutic strategies and should thus be further pursued in transgenic mouse models of Alzheimer disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Inflammation/metabolism , Microglia/metabolism , alpha 1-Antitrypsin/metabolism , Animals , Blotting, Western , Cells, Cultured , Mice , Microglia/drug effects , alpha 1-Antitrypsin/pharmacology
10.
J Neurol Sci ; 344(1-2): 94-9, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25064444

ABSTRACT

Amyloid-ß has been shown to interact with the α7 nicotinic acetylcholine receptor on neuronal cells. Not much is known on the effect on microglial cells and whether this effect can be modulated by the endogenous α7 nicotinic acetylcholine receptor antagonist kynurenic acid. Our aim was to investigate the effect of kynurenic acid on amyloid-ß-treated BV-2 microglial cells with respect to α7 nicotinic acetylcholine receptor expression, cell viability, cytokine production and phagocytotic abilities. Therefore BV-2 cells were treated with oligomeric or fibrillar forms of amyloid-ß(1-40) and co-treated with kynurenic acid. α7 nicotinic acetylcholine receptor quantity was investigated using Western blotting. Cell viability was assessed by staining cells with fluorescein diacetate and propidium iodide. Pro-inflammatory cytokines were measured in cell culture supernatants of treated cells with ELISAs; NO with Griess reagents and amyloid-ß uptake were investigated with fluorescence-activated cell sorting and verified by Western blotting. Amyloid-ß nor kynurenic acid did have an effect on the protein level of the α7 nicotinic acetylcholine receptor. Amyloid-Beta induced cell mortality was unchanged after addition of kynurenic acid. However, kynurenic acid co-treatment reduced the pro-inflammatory cytokines tumour necrosis factor-α and IL-6 and amyloid-ß phagocytosis. We provide evidence for an immunomodulating effect of the endogenous α7 nicotinic acetylcholine receptor antagonist kynurenic acid. Our findings indicate a role for kynurenic acid in amyloid-ß associated neuroinflammation in Alzheimer disease.


Subject(s)
Kynurenic Acid/pharmacology , Microglia/drug effects , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Cell Line, Transformed , Cell Survival , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Excitatory Amino Acid Antagonists , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Peptide Fragments/pharmacology , Phagocytosis/drug effects , Tumor Necrosis Factor-alpha/metabolism , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors
11.
J Neuroinflammation ; 11: 86, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24886419

ABSTRACT

BACKGROUND: Increasing evidence suggests that inflammation associated with microglial cell activation in the substantia nigra (SN) of patients with Parkinson disease (PD) is not only a consequence of neuronal degeneration, but may actively sustain dopaminergic (DA) cell loss over time. We aimed to study whether the intracellular chaperone heat shock protein 60 (Hsp60) could serve as a signal of CNS injury for activation of microglial cells. METHODS: Hsp60 mRNA expression in the mesencephalon and the striatum of C57/BL6 mice treated with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and the Hsp60/TH mRNA ratios in the SN of PD patients and aged-matched subjects were measured. To further investigate a possible link between the neuronal Hsp60 response and PD-related cellular stress, Hsp60 immunoblot analysis and quantification in cell lysates from SH-SY5Y after treatment with 100 µM MPP+ (1-methyl-4-phenylpyridinium) at different time points (6, 12, 24 and 48 hours) compared to control cells were performed. Additional MTT and LDH assay were used. We next addressed the question as to whether Hsp60 influences the survival of TH+ neurons in mesencephalic neuron-glia cultures treated either with MPP+ (1 µM), hHsp60 (10 µg/ml) or a combination of both. Finally, we measured IL-1ß, IL-6, TNF-α and NO-release by ELISA in primary microglial cell cultures following treatment with different hHsp60 preparations. Control cultures were exposed to LPS. RESULTS: In the mesencephalon and striatum of mice treated with MPTP and also in the SN of PD patients, we found that Hsp60 mRNA was up-regulated. MPP+, the active metabolite of MPTP, also caused an increased expression and release of Hsp60 in the human dopaminergic cell line SH-SY5Y. Interestingly, in addition to being toxic to DA neurons in primary mesencephalic cultures, exogenous Hsp60 aggravated the effects of MPP+. Yet, although we demonstrated that Hsp60 specifically binds to microglial cells, it failed to stimulate the production of pro-inflammatory cytokines or NO by these cells. CONCLUSIONS: Overall, our data suggest that Hsp60 is likely to participate in DA cell death in PD but via a mechanism unrelated to cytokine release.


Subject(s)
Chaperonin 60/metabolism , Corpus Striatum/pathology , Dopaminergic Neurons/metabolism , MPTP Poisoning/pathology , Mesencephalon/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Chaperonin 60/genetics , Disease Models, Animal , Dopamine Agents/pharmacology , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Nitric Oxide/metabolism , Protein Binding/drug effects , RNA, Messenger/metabolism , Tyrosine 3-Monooxygenase/metabolism
12.
J Neuroinflammation ; 10: 5, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23317003

ABSTRACT

BACKGROUND: Naturally occurring autoantibodies against amyloid-ß (nAbs-Aß) have been shown to exert beneficial effects on transgenic Alzheimer's disease (AD) animals in vivo and on primary neurons in vitro. Not much is known about their effect on microglial cells. Our aim was to investigate the effect of nAbs-Aß on amyloid-ß (Aß)-treated microglial cells in vitro with respect to cell viability, stress pathways, cytokine production and phagocytotic abilities and whether these effects can be conveyed to neurons. METHODS: Primary microglial cells isolated from Swiss Webster mouse mesencephalons on embryonic day 13.5 were pretreated with nAbs-Aß and then treated with Aß oligomers. After 3 hours, phagocytosis as well as western blot analysis were evaluated to measure the amount of phagocytized Aß. Cell viability was analyzed using an MTT assay 24 hours after treatment. Pro-inflammatory cytokines in the supernatants were analyzed with ELISAs and then we treated primary neuronal cells with these conditioned microglia supernatants. Twenty-four hours later we did a MTT assay of the treated neurons. We further investigated the effect of a single nAbs-Aß administration on Tg2576 mice in vivo. RESULTS: Upon co-administration of Aß and nAbs-Aß no change in microglia viability was observed. However, there was an increase in phosphorylated p38 protein level, an increase in the pro-inflammatory cytokines TNF-α and IL-6 and an increase in Aß uptake by microglial cells. Treatment of primary neurons with conditioned microglia medium led to a 10% improvement in cell viability when nAbs-Aß were co-administered compared to Aß-treated cells alone. We were unable to detect changes in cytokine production in brain lysates of Tg2576 mice. CONCLUSIONS: We provide evidence on the mechanism of action of nAbs-Aß on microglia in vitro. Interestingly, our in vivo data indicate that nAbs-Aß administration should be considered as a therapeutic strategy in AD, since there is no inflammatory reaction.


Subject(s)
Amyloid beta-Peptides/immunology , Autoantibodies/physiology , Microglia/immunology , Amyloid beta-Peptides/genetics , Animals , Autoantibodies/administration & dosage , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Cricetinae , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
Glia ; 60(12): 2050-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23002008

ABSTRACT

In neurons, small-conductance calcium-activated potassium (KCNN/SK/K(Ca)2) channels maintain calcium homeostasis after N-methyl-D-aspartate (NMDA) receptor activation, thereby preventing excitotoxic neuronal death. So far, little is known about the function of KCNN/SK/K(Ca)2 channels in non-neuronal cells, such as microglial cells. In this study, we addressed the question whether KCNN/SK/K(Ca)2 channels activation affected inflammatory responses of primary mouse microglial cells upon lipopolysaccharide (LPS) stimulation. We found that N-cyclohexyl-N-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-4-pyrimidinamine (CyPPA), a positive pharmacological activator of KCNN/SK/K(Ca)2 channels, significantly reduced LPS-stimulated activation of microglia in a concentration-dependent manner. The general KCNN/SK/K(Ca)2 channel blocker apamin reverted these effects of CyPPA on microglial proliferation. Since calcium plays a central role in microglial activation, we further addressed whether KCNN/SK/K(Ca)2 channel activation affected the changes of intracellular calcium levels, [Ca(2+)](i), in microglial cells. Our data show that LPS-induced elevation of [Ca(2+)](i) was attenuated following activation of KCNN2/3/K(Ca)2.2/K(Ca)2.3 channels by CyPPA. Furthermore, CyPPA reduced downstream events including tumor necrosis factor alpha and interleukin 6 cytokine production and nitric oxide release in activated microglia. Further, we applied specific peptide inhibitors of the KCNN/SK/K(Ca)2 channel subtypes to identify which particular channel subtype mediated the observed anti-inflammatory effects. Only inhibitory peptides targeting KCNN3/SK3/K(Ca)2.3 channels, but not KCNN2/SK2/K(Ca)2.2 channel inhibition, reversed the CyPPA-effects on LPS-induced microglial proliferation. These findings revealed that KCNN3/SK3/K(Ca)2.3 channels can modulate the LPS-induced inflammatory responses in microglial cells. Thus, KCNN3/SK3/K(Ca)2.3 channels may serve as a therapeutic target for reducing microglial activity and related inflammatory responses in the central nervous system.


Subject(s)
Calcium Signaling/drug effects , Calcium Signaling/physiology , Cytokines/biosynthesis , Inflammation Mediators/physiology , Microglia/metabolism , Small-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Animals, Newborn , Apamin/pharmacology , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/physiology , Down-Regulation/drug effects , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/toxicity , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Microglia/drug effects , Pyrazoles/antagonists & inhibitors , Pyrazoles/toxicity , Pyrimidines/antagonists & inhibitors , Pyrimidines/toxicity , Small-Conductance Calcium-Activated Potassium Channels/metabolism
14.
Immunol Rev ; 248(1): 68-86, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22725955

ABSTRACT

In this article, we review the current knowledge on pathological and physiological autoantibodies directed toward structures in the central nervous system (CNS) with an emphasis on their regulation and origin. Pathological autoantibodies in the CNS that are associated with autoimmunity often lead to severe neurological deficits via inflammatory processes such as encephalitis. In some instances, however, autoantibodies function as a marker for diagnostic purposes without contributing to the pathological process and/or disease progression. The existence of naturally occurring physiological autoantibodies has been known for a long time, and their role in maintaining homeostasis is well established. Within the brain, naturally occurring autoantibodies targeting aggregated proteins have been detected and might be promising candidates for new therapeutic approaches for neurodegenerative disorders. Further evidence has demonstrated the existence of naturally occurring antibodies targeting antigens on neurons and oligodendrocytes that promote axonal outgrowth and remyelination. The numerous actions of physiological autoantibodies as well as their regulation and origin are summarized in this review.


Subject(s)
Autoantibodies/immunology , Central Nervous System/immunology , Animals , Autoantibodies/metabolism , Autoantigens/immunology , Autoantigens/metabolism , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/metabolism , Central Nervous System/metabolism , Humans , Myelin Sheath/immunology , Myelin Sheath/metabolism , Neurons/immunology , Neurons/metabolism , Paraneoplastic Syndromes/immunology , Paraneoplastic Syndromes/metabolism , Synaptic Transmission/immunology
15.
Clin Cancer Res ; 16(5): 1402-15, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20160064

ABSTRACT

PURPOSE: In small cell lung cancer cells (SCLC), various autocrine stimuli lead to the parallel activation of G(q/11) and G(12/13) proteins. Although the contribution of the G(q/11)-phospholipase C-beta cascade to mitogenic effects in SCLC cells is well established, the relevance of G(12/13) signaling is still elusive. In other tumor entities, G(12/13) activation promotes invasiveness without affecting cellular proliferation. Here, we investigate the role of G(12/13)-dependent signaling in SCLC. EXPERIMENTAL DESIGN: We used small hairpin RNA-mediated targeting of G(alpha)(12), G(alpha)(13), or both in H69 and H209 cells and analyzed the effects of G(alpha)(12) and/or G(alpha)(13) knockdown on tumor cells in vitro, tumor growth in vivo, and mitogen-activated protein kinase (MAPK) activation. RESULTS: Lentiviral expression of small hairpin RNAs resulted in robust and specific G(alpha)(12) and G(alpha)(13) knockdown as well as markedly inhibited proliferation, colony formation, and bradykinin-promoted stimulation of cell growth. Analyzing the activation status of all three major MAPK families revealed nonredundant functions of G(alpha)(12) and G(alpha)(13) in SCLC and a marked p42/p44 activation upon G(alpha)(12)/G(alpha)(13) knockdown. In a s.c. tumor xenograft mouse model, G(alpha)(12) or G(alpha)(13) downregulation led to decreased tumor growth due to reduced tumor cell proliferation. More importantly, G(alpha)(12)/G(alpha)(13) double knockdown completely abolished H69 tumorigenicity in mice. CONCLUSIONS: G(alpha)(12) and G(alpha)13) exert a complex pattern of nonredundant effects in SCLC, and in contrast to other tumor types, SCLC cell proliferation in vitro and tumorigenicity in vivo critically depend on G(12/13) signaling. Due to the complete abolishment of tumorgenicity in our study, RNAi-mediated double knockdown may provide a promising new avenue in SCLC treatment.


Subject(s)
Cell Proliferation , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Lung Neoplasms/metabolism , Small Cell Lung Carcinoma/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Mice , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Small Cell Lung Carcinoma/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...