Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Neuroendocrinology ; 103(5): 460-75, 2016.
Article in English | MEDLINE | ID: mdl-26337121

ABSTRACT

CNS ischemia results in locally confined and rapid tissue damage accompanied by a loss of neurons and their circuits. Early and time-delayed inflammatory responses are critical variables determining the extent of neural disintegration and regeneration. Inflammasomes are vital effectors in innate immunity. Their activation in brain-intrinsic immune cells contributes to ischemia-related brain damage. The steroids 17ß-estradiol (E2) and progesterone (P) are neuroprotective and anti-inflammatory. Using a transient focal rat ischemic model, we evaluated the time response of different inflammasomes in the peri-infarct zone from the early to late phases after poststroke ischemia. We show that the different inflammasome complexes reveal a specific time-oriented sequential expression pattern with a maximum at approximately 24 h after the infarct. Within the limits of antibody availability, immunofluorescence labeling demonstrated that microglia and neurons are major sources of the locally activated inflammasomes NOD-like receptor protein-3 (NLRP3) and associated speck-like protein (ASC), respectively. E2 and P given for 24 h immediately after ischemia onset reduced hypoxia-induced mRNA expression of the inflammasomes NLRC4, AIM2 and ASC, and decreased the protein levels of ASC and NLRP3. In addition, mRNA protein levels of the cytokines interleukin-1ß (IL1ß), IL18 and TNFα were reduced by the steroids. The findings provide for the first time a detailed flow chart of hypoxia-driven inflammasome regulation in the peri-infarct cerebral cortex. Further, we demonstrate that E2 and P alleviate the expression of certain inflammasome components, sometimes in a hormone-specific way. Besides directly regulating other cellular neuroprotective pathways, the control of inflammasomes by these steroids might contribute to its neuroprotective potency.


Subject(s)
Brain Infarction/etiology , Encephalitis/drug therapy , Encephalitis/etiology , Estradiol/therapeutic use , Ischemic Attack, Transient/complications , Progesterone/therapeutic use , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Brain Infarction/prevention & control , CARD Signaling Adaptor Proteins , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caspases/genetics , Caspases/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Functional Laterality , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Ischemic Attack, Transient/pathology , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats , Rats, Wistar , Reperfusion , Time Factors
2.
Glia ; 63(12): 2260-73, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26200799

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is characterized by the degeneration of motoneurons in the cerebral cortex, brainstem and spinal cord. Neuroinflammation plays an important role in the pathogenesis of ALS and involves the activation of microglia and astrocytes. Intracellular inflammasome complexes are part of the innate immunity as they sense and execute host inflammatory responses. The best characterized component is the NLRP3 inflammasome comprised of the NLR protein NLRP3, the adaptor ASC and pro-caspase 1. The NLRP3 inflammasome is critical for the activation of caspase 1 and the processing and release of IL1ß and IL18. In this study, we investigated the expression, activation and co-localization of the NLRP3 inflammasome in the spinal cord of male SOD1(G93A) mice carrying a mutant human superoxide dismutase 1 (SOD1) variant and regarded as an animal model for ALS as well as in post-mortem tissue of ALS patients. NLRP3 and its molecular components as well as IL1ß were already detectable in SOD1 mice at a pre-symptomatic stage after 9 weeks and further increased in 14 week old animals. Spinal cord astrocytes were identified as the major cell type expressing NLRP3 components. In human ALS tissue, we also found increased NLRP3, ASC, IL18 and active caspase 1 levels compared to control patients. Our findings suggest that astroglial NLRP3 inflammasome complexes are critically involved in neuroinflammation in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Carrier Proteins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/pathology , Caspase 1/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Male , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , RNA, Messenger/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
3.
Neuropathol Appl Neurobiol ; 41(3): 304-18, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24957221

ABSTRACT

AIM: Upon denervation, skeletal muscle fibres initiate complex changes in gene expression. Many of these genes are involved in muscle fibre remodelling and atrophy. Amyotrophic lateral sclerosis (ALS) leads to progressive neurodegeneration and neurogenic muscular atrophy (NMA). Disturbed calcium homeostasis and misfolded protein aggregation both in motor neurones and muscle fibres are key elements of ALS pathogenesis that are mutually interdependent. Therefore, we hypothesized that the calcium sensor STIM1 might be abnormally modified and involved in muscle fibre degeneration in ALS and other types of NMA. METHODS: We examined ALS and NMA patient biopsy and autopsy tissue and tissue from G93A SOD1 mice by immunohistochemistry and immunoblotting. RESULTS: In normal human and mouse muscle STIM1 was found to be differentially expressed in muscle fibres of different types and to concentrate at neuromuscular junctions, compatible with its known role in calcium sensing. Denervated muscle fibres of sALS and NMA cases and SOD1 mice showed diffusely increased STIM1 immunoreactivity along with ubiquitinated material. In addition, distinct focal accumulations of STIM1 were observed in target structures within denervated fibres of sALS and other NMA as well as SOD1 mouse muscles. Large STIM1-immunoreactive structures were found in ALS-8 patient muscle harbouring the P56S mutation in the ER protein VAPB. CONCLUSION: These findings suggest that STIM1 is involved in several ways in the reaction of muscle fibres to denervation, probably reflecting alterations in calcium homeostasis in denervated muscle fibres.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Membrane Proteins/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Neoplasm Proteins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Blotting, Western , Disease Models, Animal , Humans , Immunohistochemistry , Mice , Microscopy, Electron, Transmission , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Phenotype , Stromal Interaction Molecule 1
4.
Cells ; 12(14)2023 07 12.
Article in English | MEDLINE | ID: mdl-37508501

ABSTRACT

In mammals, the circadian system controls various physiological processes to maintain metabolism, behavior, and immune function during a daily 24 h cycle. Although driven by a cell-autonomous core clock in the hypothalamus, rhythmic activities are entrained to external cues, such as environmental lighting conditions. Exposure to artificial light at night (ALAN) can cause circadian disruption and thus is linked to an increased occurrence of civilization diseases in modern society. Moreover, alterations of circadian rhythms and dysregulation of immune responses, including inflammasome activation, are common attributes of neurodegenerative diseases, including Alzheimer', Parkinson's, and Huntington's disease. Although there is evidence that the inflammasome in the hippocampus is activated by stress, the direct effect of circadian disruption on inflammasome activation remains poorly understood. In the present study, we aimed to analyze whether exposure to constant light (LL) affects inflammasome activation in the mouse hippocampus. In addition to decreased circadian power and reduced locomotor activity, we found cleaved caspase 1 significantly elevated in the hippocampus of mice exposed to LL. However, we did not find hallmarks of inflammasome priming or cleavage of pro-interleukins. These findings suggest that acute circadian disruption leads to an assembled "ready to start" inflammasome, which may turn the brain more vulnerable to additional aversive stimuli.


Subject(s)
Inflammasomes , Light , Mice , Animals , Caspase 1 , Circadian Rhythm/physiology , Hippocampus , Mammals
5.
Neuroscience ; 472: 35-50, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34364953

ABSTRACT

G-protein-coupled-estrogen-receptor 1 (GPER1) is a membrane-bound receptor that mediates estrogen signaling via intracellular signaling cascades. We recently showed that GPER1 promotes the distal dendritic enrichment of hyperpolarization activated and cyclic nucleotide-gated (HCN)1 channels in CA1 stratum lacunosum-moleculare (SLM), suggesting a role of GPER1-mediated signaling in neuronal plasticity. Here we studied whether this role involves processes of structural plasticity, such as the regulation of spine and synapse density in SLM. In organotypic entorhino-hippocampal cultures from mice expressing eGFP, we analyzed spine densities in SLM after treatment with GPER1 agonist G1 (20 nM). G1 significantly increased the density of "non-stubby" spines (maturing spines with a spine head and a neck), but did so only in cultures from female mice. In support of this finding, the expression of synaptic proteins was sex-specifically altered in the cultures: G1 increased the protein (but not mRNA) expression of PSD95 and reduced the p-/n-cofilin ratio only in cultures from females. Application of E2 (2 nM) reproduced the sex-specific effect on spine density in SLM, but only partially on the expression of synaptic proteins. Spine synapse density was, however, not altered after G1-treatment, suggesting that the increased spine density did not translate into an increased spine synapse density in the culture model. Taken together, our results support a role of GPER1 in mediating structural plasticity in CA1 SLM, but suggest that in developing hippocampus, this role is sex-specific.


Subject(s)
Hippocampus , Receptors, Estrogen , Animals , Dendritic Spines/metabolism , Female , GTP-Binding Proteins , Hippocampus/metabolism , Male , Mice , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled , Synapses/metabolism
6.
Neuroscience ; 463: 288-302, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33781799

ABSTRACT

Inflammasomes are key components of the innate immune system and activation of these multiprotein platforms is a crucial event in the etiopathology of amyotrophic lateral sclerosis (ALS). Inflammasomes consist of a pattern recognition receptor (PRR), the adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) and caspase 1. Exogenous or endogenous "danger signals" can trigger inflammasome assembly and promote maturation and release of pro-inflammatory cytokines, including interleukin 1ß. Previous studies have demonstrated presence and activation of NLRP3 in spinal cord tissue from SOD1(G93A) mice and human sporadic ALS (sALS) patients. However, regulation and cell type-specific localization of other well-known PRRs has not yet been analysed in ALS. Here, we explored gene expression, protein concentration and cell type-specific localization of the NLRP1, NLRC4 and AIM2 inflammasomes in spinal cord samples from SOD1(G93A) mice and sALS patients. Transcription levels of NLRP1 and NLRC4, but not AIM2, were elevated in symptomatic SOD1(G93A) animals. Immunoblotting revealed elevated protein levels of NLRC4, which were significantly increased in sALS vs. control patients. Immunofluorescence studies revealed neuronal labelling of all investigated PRRs. Staining of AIM2 was detected in all types of glia, whereas glial type-specific labelling was observed for NLRP1 and NLRC4. Our findings revealed pathology-related and cell type-specific differences in the expression of subsets of PRRs. Besides NLRP3, NLRC4 appears to be linked more closely to ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis , Inflammasomes , Amyotrophic Lateral Sclerosis/genetics , Animals , Caspase 1 , Humans , Mice , Spinal Cord , Superoxide Dismutase-1/genetics
7.
J Neurochem ; 114(5): 1511-26, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20557428

ABSTRACT

The production of chemokines by astrocytes constitutes an important component of neuroinflammatory processes in the brain. As the transcriptional activator retinoic acid (RA), used for chemotherapy and dermatological applications, exerts anti-inflammatory effects on monocytes and lymphocytes, we have tested whether the physiologically occurring isomer, all-trans RA, affects chemokine expression by astrocytes. Under control conditions, primary cultures of murine cortical astrocytes expressed no or very low levels of CCL and CXCL chemokines. After treatment with bacterial lipopolysaccharides to simulate inflammation in vitro, we detected a strong increase in the release of CCL2 (to > 4 ng/mL in cell culture supernatant), CCL3 (> 20 ng/mL), CCL5 (> 25 ng/mL), CXCL1 (> 30 ng/mL) and CXCL2 (> 20 ng/mL). Although simultaneous exposure to RA did not significantly affect this response, 12 h pre-treatment with 0.1 microM all-trans RA strongly suppressed mRNA expression and protein release of all chemokines. The anti-inflammatory activity of RA engaged RA and retinoid X receptors and correlated with a decreased expression of the lipopolysaccharides co-receptor CD14. A minor reduction of nuclear NF-kappaB was observed but not significant, activation of Jun amino-terminal kinase, p38 and signal transducer and activator of transcription 3 were not altered by RA. The results suggest that retinoids should be further investigated as candidates for the treatment of neuroinflammation.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Chemokines/antagonists & inhibitors , Chemokines/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/pharmacology , Tretinoin/pharmacology , Animals , Cells, Cultured , Inflammation Mediators/metabolism , Mice , Mice, Inbred BALB C , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
8.
Glia ; 57(8): 807-14, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19031445

ABSTRACT

Sex hormones, for example, estrogen and progesterone, are thought to affect and delay progression of multiple sclerosis (MS) in pregnant women. Although both steroid hormones are neuroprotective in the brain and elevated during pregnancy, only estrogen was tested in clinical trials. To evaluate the role of 17beta-estradiol (E) and progesterone (P) in prevention demyelination, young adult male mice were fed with cuprizone for a defined time interval and simultaneously treated with steroids by repeated injections into the neck region. The status of myelination was analyzed by magnetic resonance imaging and conventional histological staining. The individual application of E and P resulted only in a moderate prevention of demyelination in the corpus callosum (CC). The combined treatment with both steroid hormones counteracted the process of demyelination. Expression of the mature (PLP and MBP) and premature (PDGF-alpha-R) oligodendrocyte markers were significantly increased after hormone application in the affected CC. In addition, both hormones stimulated astrogliosis and the expression of IGF-1. Microglial invasion in demyelinated CC was pronounced and additionally localized in the midline of CC after hormone treatment. These data show that sex steroids can protect the brain from demyelination and stimulate remyelination. It appears that only the administration of both hormones is fully effective. The beneficial steroid effect requires interactions with oligodendrocytes possibly by preventing their degeneration or recruitment from precursor cells which are stimulated to remyelinated fibers. The positive hormonal influence on myelination in the CNS may be a future therapeutically strategy for the treatment of MS.


Subject(s)
Corpus Callosum , Cuprizone , Demyelinating Diseases , Estradiol/pharmacology , Estrogens/pharmacology , Progesterone/pharmacology , Analysis of Variance , Animals , Corpus Callosum/drug effects , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Cytoskeleton/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Demyelinating Diseases/prevention & control , Disease Models, Animal , Drug Administration Schedule , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Myelin Proteins/genetics , Myelin Proteins/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism
9.
J Neurosci Res ; 87(6): 1343-55, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19021291

ABSTRACT

Memory impairment is outstanding within the spectrum of cognitive deficits in multiple sclerosis (MS) patients. Demyelination has been reported in the hippocampus formation of MS patients. The degree of hippocampus lesions in MS strongly correlates with progression of cognitive dysfunction. Because no appropriate animal model for the study of hippocampus demyelination has been established, we used the cuprizone mouse model to investigated demyelination in young adult and aged mice. The myelin status was analyzed by classical histological staining, immunocytochemistry for proteolipoprotein, and electron microscopy. Oligodendrocyte, astroglial, and microglia markers were studied. Cuprizone intoxication induced an almost complete demyelination of distinct hippocampus subregions to a similar extent in young adult and aged male mice. Demyelination was pronounced in a subset of white and gray matter areas, i.e., the stratum lacunosum moleculare containing the perforant path, medial alveus, stratum pyramidale in the cornu ammonis 2/3 region, and hilus region. Besides demyelination, affected areas displayed hypertrophic and hyperplastic astrocytosis. No significant effect on microglia invasion was detected at any investigated time point (0, 3, 5, and 7 weeks). We conclude that cuprizone-induced demyelination provides an adequate animal model to investigate appropriate therapy strategies for the prevention of hippocampus demyelination.


Subject(s)
Cuprizone/pharmacology , Demyelinating Diseases/pathology , Disease Models, Animal , Hippocampus/pathology , Aging , Animals , Astrocytes/pathology , Demyelinating Diseases/chemically induced , Gliosis/chemically induced , Gliosis/pathology , Hippocampus/ultrastructure , Immunohistochemistry , Indoles , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Microscopy, Electron , Multiple Sclerosis , Myelin Sheath/ultrastructure , Oligodendroglia/pathology , Reverse Transcriptase Polymerase Chain Reaction
10.
J Neurochem ; 106(1): 320-32, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18394023

ABSTRACT

Prostanoids are important mediators of inflammation and pain signaling. Although it is now well accepted that astrocytes participate in inflammatory reactions in the CNS, the molecular regulation of this activity is still largely unknown. Specifically, the regulation of prostanoid synthesis by this type of glia remains to be resolved. Recent evidence suggests that the transcriptional regulator retinoic acid (RA) is involved in regulation of the immune response. We have investigated the expression pattern of the enzymes that catalyze prostanoid and leukotriene synthesis in cultured cortical astrocytes, their stimulation by lipopolysaccharides (LPS) and their regulation by RA. The data indicate that astrocytes are an important source of prostaglandins (PGs) and that RA reduces their inflammatory biosynthesis. LPS treatment induced the expression of enzymes for the production of arachidonic acid and PGs but caused down-regulation of a PG degrading enzyme and of leukotriene synthesizing enzymes that compete with PG synthesis. Consequently, the secretion of the PGE(2) was highly increased after LPS exposure. RA counteracted the inflammatory regulation of cyclooxygenase (COX)-2 mRNA and protein in astrocytes and thereby reduced the synthesis of PGE(2) by approximately 60%. In the absence of LPS, RA enhanced the expression of COX-1 mRNA. In conclusion, RA might be effective in suppressing inflammatory processes in the brain by inhibiting PG synthesis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Astrocytes/drug effects , Encephalitis/drug therapy , Gliosis/drug therapy , Prostaglandins/biosynthesis , Tretinoin/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Dinoprostone/biosynthesis , Dinoprostone/metabolism , Encephalitis/metabolism , Encephalitis/physiopathology , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzymes/drug effects , Enzymes/metabolism , Gliosis/physiopathology , Gliosis/prevention & control , Inflammation Mediators/pharmacology , Leukotrienes/biosynthesis , Leukotrienes/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
11.
J Mol Neurosci ; 34(2): 177-85, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18172772

ABSTRACT

Neuroinflammatory processes are a common epiphenomenon for a number of neurological and neurodegenerative diseases. Besides microglia, astrocytes are implicated in brain inflammation in response to harmful stimuli and pathological processes. Bacterial endotoxins can induce the synthesis and release of proinflammatory mediators, i.e., cytokines and chemokines, by astroglia. In this study, we have investigated the effect of lipopolysaccharide (LPS) treatment on the expression of enzymes of prostanoid synthesis and degradation in cultured mouse cortical astrocytes using an Affymetrix Gene Chip array, quantitative reverse transcriptase polymerase chain reaction (RT-PCR), and an enzyme-immunosorbent assay. LPS treatment induced an upregulation of enzymes responsible for prostaglandin E2 synthesis, a downregulation of enzymes that catalyzes prostaglandin E2 (PGE2) degradation and production of proinflammatory leukotrienes. Changes in enzyme expression were accompanied by a highly significant increase in extracellular PGE2. Our data demonstrate that astrocytes are directly involved in the complex regulation of proinflammatory prostanoids in the CNS under pathological processes, thus being of potential interest as targets for therapeutical interventions. Further studies are required to unravel the different roles and interactions between astroglia and other cells of the brain-intrinsic innate immune system during inflammation.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex , Gene Expression Regulation, Enzymologic , Inflammation/metabolism , Lipopolysaccharides/immunology , Prostaglandin-Endoperoxide Synthases , Prostaglandins/metabolism , Animals , Astrocytes/cytology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Dinoprostone/genetics , Dinoprostone/metabolism , Gene Expression Profiling , Mice , Mice, Inbred BALB C , Microarray Analysis , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism
12.
J Mol Neurosci ; 35(2): 235-43, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18373222

ABSTRACT

Astroglia is well-known to be integrated in the complex regulation of neuroinflammation in the central nervous system. Astrocytes become activated and synthesize cytokines, chemokines, and prostanoids during degenerative and vulnerable processes and interact with other immune-competent cells. Degenerative disorders often occur in a brain-region-specific fashion suggesting differences in the activity and reactivity of innate immune cells. We have investigated the potency of lipopolysaccharides (LPS) to differently stimulate astrocytes from the cortex and midbrain. Astroglial cultures were prepared from Bagg albino/c mice and exposed to LPS. Astrocytes from both brain areas already differed in their capacity and profile of cytokine expression under basal unstimulated conditions. In response to LPS, we observed both a region-specific pattern of up-regulation of distinct cytokines and differences in the extent and time-course of activation. Our data demonstrate that astrocytes reveal a region-specific basal profile of cytokine expression and a selective area-specific regulation of cytokines upon LPS-induced inflammation. This makes astrocytes likely candidates to be responsible for region-specific incidence rates of neurological and neurodegenerative disorders.


Subject(s)
Astrocytes/drug effects , Astrocytes/immunology , Cerebral Cortex/cytology , Encephalitis/immunology , Lipopolysaccharides/pharmacology , Mesencephalon/cytology , Animals , Cells, Cultured , Cerebral Cortex/immunology , Cerebral Cortex/physiopathology , Cytokines/genetics , Cytokines/metabolism , Encephalitis/metabolism , Encephalitis/physiopathology , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Gene Expression/immunology , Mesencephalon/immunology , Mesencephalon/physiopathology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis
13.
Brain Pathol ; 28(1): 14-27, 2018 Jan.
Article in English | MEDLINE | ID: mdl-27880990

ABSTRACT

Nowadays, amyotrophic lateral sclerosis (ALS) is considered as a multisystem disorder, characterized by a primary degeneration of motor neurons as well as neuropathological changes in non-motor regions. Neurodegeneration in subcortical areas, such as the thalamus, are believed to contribute to cognitive and behavioral abnormalities in ALS patients. In the present study, we investigated neurodegenerative changes including neuronal loss and glia pathology in the anterodorsal thalamic nucleus (AD) of SOD1(G93A) mice, a widely used animal model for ALS. We detected massive dendrite swelling and neuronal loss in SOD1(G93A) animals, which was accompanied by a mild gliosis. Furthermore, misfolded SOD1 protein and autophagy markers were accumulating in the AD. Since innate immunity and activation inflammasomes seem to play a crucial role in ALS, we examined protein expression of Nod-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase-1 recruitment domain (ASC) and the cytokine interleukin 1 beta (IL1ß) in AD glial cells and neurons. NLRP3 and ASC were significantly up-regulated in the AD of SOD1(G93A) mice. Finally, co-localization studies revealed expression of NLRP3, ASC and IL1ß in neurons. Our study yielded two main findings: (i) neurodegenerative changes already occur at an early symptomatic stage in the AD and (ii) increased inflammasome expression may contribute to neuronal cell death. In conclusion, neurodegeneration in the anterior thalamus may critically account for cognitive changes in ALS pathology.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Anterior Thalamic Nuclei/pathology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nerve Degeneration/pathology , Neurons/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Anterior Thalamic Nuclei/physiopathology , CARD Signaling Adaptor Proteins/metabolism , Cell Death/physiology , Disease Models, Animal , Disease Progression , Female , Humans , Interleukin-1beta/metabolism , Male , Mice, Transgenic , Nerve Degeneration/physiopathology , Neuroglia/pathology , Neuroglia/physiology , Neurons/physiology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
14.
Mol Neurobiol ; 55(5): 4240-4252, 2018 May.
Article in English | MEDLINE | ID: mdl-28616718

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by progressive loss of midbrain dopaminergic neurons, resulting in motor and non-motor symptoms. The underlying pathology of non-motor symptoms is poorly understood. Discussed are pathological changes of extrastriatal brain structures. In this study, we characterized histopathological alterations of extrastriatal brain structures in the 6-hydroxydopamine (6-OHDA) PD animal model. Lesions were induced by unilateral stereotactic injections of 6-OHDA into the striatum or medial forebrain bundle of adult male mice. Loss of tyrosine hydroxylase positive (TH+) fibers as well as glia activation was quantified following stereological principles. Loss of dopaminergic innervation was further investigated by western-blotting. As expected, 6-OHDA injection into the nigrostriatal route induced retrograde degeneration of dopaminergic neurons within the substantia nigra pars compacta (SNpc), less so within the ventral tegmental area. Furthermore, we observed a region-specific drop of TH+ projection fiber density in distinct cortical regions. This pathology was most pronounced in the cingulate- and motor cortex, whereas the piriform cortex was just modestly affected. Loss of cortical TH+ fibers was not paralleled by microglia or astrocyte activation. Our results demonstrate that the loss of dopaminergic neurons within the substantia nigra pars compacta is paralleled by a cortical dopaminergic denervation in the 6-OHDA model. This model serves as a valuable tool to investigate mechanisms operant during cortical pathology in PD patients. Further studies are needed to understand why cortical dopaminergic innervation is lost in this model, and what functional consequence is associated with the observed denervation.


Subject(s)
Corpus Striatum/pathology , Animals , Astrocytes/pathology , Disease Models, Animal , Dopaminergic Neurons/pathology , Injections , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Nerve Degeneration/pathology , Oxidopamine , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/pathology
15.
World J Biol Psychiatry ; 19(3): 225-235, 2018 04.
Article in English | MEDLINE | ID: mdl-28132573

ABSTRACT

OBJECTIVES: Severe grey and white matter volume reductions were found in patients with anorexia nervosa (AN) that were linked to neuropsychological deficits while their underlying pathophysiology remains unclear. For the first time, we analysed the cellular basis of brain volume changes in an animal model (activity-based anorexia, ABA). METHODS: Female rats had 24 h/day running wheel access and received reduced food intake until a 25% weight reduction was reached and maintained for 2 weeks. RESULTS: In ABA rats, the volumes of the cerebral cortex and corpus callosum were significantly reduced compared to controls by 6% and 9%, respectively. The number of GFAP-positive astrocytes in these regions decreased by 39% and 23%, total astrocyte-covered area by 83% and 63%. In neurons no changes were observed. The findings were complemented by a 60% and 49% reduction in astrocyte (GFAP) mRNA expression. CONCLUSIONS: Volumetric brain changes in ABA animals mirror those in human AN patients. These alterations are associated with a reduction of GFAP-positive astrocytes as well as GFAP expression. Reduced astrocyte functioning could help explain neuronal dysfunctions leading to symptoms of rigidity and impaired learning. Astrocyte loss could constitute a new research target for understanding and treating semi-starvation and AN.


Subject(s)
Anorexia/pathology , Astrocytes/physiology , Cerebral Cortex/pathology , Corpus Callosum/pathology , Glial Fibrillary Acidic Protein/metabolism , Animals , Disease Models, Animal , Female , Rats , Rats, Wistar
16.
Curr Pharm Des ; 23(33): 5022-5036, 2017.
Article in English | MEDLINE | ID: mdl-28619000

ABSTRACT

The mechanisms underlying neurodegeneration in amyotrophic lateral sclerosis (ALS) are multifactorial and include genetic and environmental factors. Nowadays, it is well accepted that neuronal loss is driven by non-cell autonomous toxicity. Non-neuronal cells, such as astrocytes, have been described to significantly contribute to motoneuron cell death and disease progression in cell culture experiments and animal models of ALS. Astrocytes are essential for neuronal survival and function by regulating neurotransmitter and ion homeostasis, immune response, blood flow and glucose uptake, antioxidant defence and growth factor release. Based on their significant functions in "housekeeping" the central nervous system (CNS), they are no longer thought to be passive bystanders but rather contributors to ALS pathogenesis. Findings from animal models have broadened our knowledge about different pathomechanisms in ALS, but therapeutic approaches to impede disease progression failed. So far, there is no cure for ALS and effective medication to slow down disease progression is limited. Targeting only a single aspect of this multifactorial disease may exhibit therapeutic limitations. Hence, novel cellular targets must be defined and new pharmaceutical strategies, such as combinatorial drug therapies are urgently needed. The present review discusses the physiological role of astrocytes and current hypotheses of astrocyte pathology in ALS. Furthermore, recent investigation of potential drug candidates in astrocyte cell culture systems and animal models, as well as data obtained from clinical trials, will be addressed. The central role of astrocytes in ALS pathogenesis makes them a promising target for pharmaceutical interventions.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Astrocytes/pathology , Drug Delivery Systems/trends , Amyotrophic Lateral Sclerosis/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Drug Delivery Systems/methods , Glutamic Acid/metabolism , Humans , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Neuroprotective Agents/administration & dosage , Oxidative Stress/drug effects , Oxidative Stress/physiology
17.
Mol Neurobiol ; 54(10): 8429-8443, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27957680

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease manifested by the progressive loss of upper and lower motoneurons. The pathomechanism of ALS is complex and not yet fully understood. Neuroinflammation is believed to significantly contribute to disease progression. Inflammasome activation was recently shown in the spinal cord of human sporadic ALS patients and in the SOD1(G93A) mouse model for ALS. In the present study, we investigated the neuroprotective and anti-inflammatory effects of 17ß-estradiol (E2) treatment in pre-symptomatic and symptomatic male SOD1(G93A) mice. Symptomatic mice with E2 substitution exhibited improved motor performance correlating with an increased survival of motoneurons in the lumbar spinal cord. Expression of NLRP3 inflammasome proteins and levels of activated caspase 1 and mature interleukin 1 beta were significantly reduced in SOD1(G93A) mice supplemented with E2.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Down-Regulation/physiology , Estradiol/pharmacology , Inflammasomes/metabolism , Motor Neurons/metabolism , Superoxide Dismutase-1/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Cell Survival/drug effects , Cell Survival/physiology , Down-Regulation/drug effects , Estradiol/therapeutic use , Female , Humans , Inflammasomes/antagonists & inhibitors , Inflammasomes/genetics , Male , Mice , Mice, Transgenic , Motor Neurons/drug effects , Superoxide Dismutase-1/genetics
18.
Cell Death Differ ; 24(10): 1655-1671, 2017 10.
Article in English | MEDLINE | ID: mdl-28622300

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is characterized by the selective degeneration of motor neurons (MNs) and their target muscles. Misfolded proteins which often form intracellular aggregates are a pathological hallmark of ALS. Disruption of the functional interplay between protein degradation (ubiquitin proteasome system and autophagy) and RNA-binding protein homeostasis has recently been suggested as an integrated model that merges several ALS-associated proteins into a common pathophysiological pathway. The E102Q mutation in one such candidate gene, the endoplasmic reticulum (ER) chaperone Sigma receptor-1 (SigR1), has been reported to cause juvenile ALS. Although loss of SigR1 protein contributes to neurodegeneration in several ways, the molecular mechanisms underlying E102Q-SigR1-mediated neurodegeneration are still unclear. In the present study, we showed that the E102Q-SigR1 protein rapidly aggregates and accumulates in the ER and associated compartments in transfected cells, leading to structural alterations of the ER, nuclear envelope and mitochondria and to subsequent defects in proteasomal degradation and calcium homeostasis. ER defects and proteotoxic stress generated by E102Q-SigR1 aggregates further induce autophagy impairment, accumulation of stress granules and cytoplasmic aggregation of the ALS-linked RNA-binding proteins (RBPs) matrin-3, FUS, and TDP-43. Similar ultrastructural abnormalities as well as altered protein degradation and misregulated RBP homeostasis were observed in primary lymphoblastoid cells (PLCs) derived from E102Q-SigR1 fALS patients. Consistent with these findings, lumbar α-MNs of both sALS as well as fALS patients showed cytoplasmic matrin-3 aggregates which were not co-localized with pTDP-43 aggregates. Taken together, our results support the notion that E102Q-SigR1-mediated ALS pathogenesis comprises a synergistic mechanism of both toxic gain and loss of function involving a vicious circle of altered ER function, impaired protein homeostasis and defective RBPs.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Endoplasmic Reticulum Stress/genetics , Homeostasis/genetics , Mutation/genetics , RNA-Binding Proteins/metabolism , Receptors, sigma/genetics , Animals , Endoplasmic Reticulum/metabolism , Humans , Mice , Motor Neurons/metabolism , RNA/metabolism , Sigma-1 Receptor
19.
Mol Neurobiol ; 53(5): 3063-3075, 2016 07.
Article in English | MEDLINE | ID: mdl-25972240

ABSTRACT

Stromal cell-derived factor-1 alpha (SDF-1a) or CXCL12 is an important cytokine with multiple functions in the brain during development and in adulthood. The inflammatory response initiated by spinal cord injury (SCI) involves the processing of interleukin-1beta (IL-1ß) and IL-18 mediated by caspase-1 which is under the control of an intracellular multiprotein complex termed inflammasome. Using an SCI rat model, we found improved functional long-term recovery which is paralleled by a reduction of apoptosis after intrathecal treatment with SDF-1a. An intriguing aspect is that SDF-1a changed the number of neuroinflammatory cells in the damaged area. We further examined the cellular localization and sequential expression of several inflammasomes during SCI at 6 h, 24 h, 3 days, and 7 days as well as the role of SDF-1a as a regulatory factor for inflammasomes. Using 14-week old male Wistar rats, spinal cord contusion was applied at the thoracic segment 9, and animals were subsequently treated with SDF-1a via intrathecal application through an osmotic pump. SCI temporally increased the expression of the inflammasomes NLRP3, ASC, the inflammatory marker tumor necrosis factor-a (TNF-a), interleukin-1ß (IL-1ß) and IL-18. SDF-1a significantly reduced the levels of IL-18, IL-1b, TNF-a, NLRP3, ASC, and caspase-1. Immunofluorescence double-labeling demonstrated that microglia and neurons are major sources of the ASC and NLRP3 respectivley. Our data provide clear evidence that SCI stimulates a complex scenario of inflammasome activation at the injured site and that SDF-1a-mediated neuroprotection presumably depends on the attenuation of the inflammasome complex.


Subject(s)
Chemokine CXCL12/therapeutic use , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Animals , CARD Signaling Adaptor Proteins/metabolism , Cell Count , Chemokine CXCL12/pharmacology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Gliosis/complications , Gliosis/pathology , Injections, Spinal , Locomotion/drug effects , Male , Microglia/drug effects , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neurons/drug effects , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Time Factors
20.
World J Biol Psychiatry ; 17(4): 274-84, 2016 06.
Article in English | MEDLINE | ID: mdl-27160428

ABSTRACT

OBJECTIVES: Patients with anorexia nervosa (AN) suffer from neuropsychological deficits including memory impairments. Memory partially depends on 17ß-oestradiol (E2), which is reduced in patients with AN. We assessed whether memory functions correlate with E2 plasma levels in the activity-based anorexia (ABA) rat model. METHODS: Nine 4-week-old female Wistar rats were sacrificed directly after weight loss of 20-25% (acute starvation), whereas 17 animals had additional 2-week weight-holding (chronic starvation). E2 serum levels and novel object recognition tasks were tested before and after starvation and compared with 21 normally fed controls. RESULTS: Starvation disrupted menstrual cycle and impaired memory function, which became statistically significant in the chronic state (oestrous cycle (P < 0.001), E2 levels (P = 0.011) and object recognition memory (P = 0.042) compared to controls). E2 reduction also correlated with the loss of memory in the chronic condition (r = 0.633, P = 0.020). CONCLUSIONS: Our results demonstrate that starvation reduces the E2 levels which are associated with memory deficits in ABA rats. These effects might explain reduced memory capacity in patients with AN as a consequence of E2 deficiency and the potentially limited effectiveness of psychotherapeutic interventions in the starved state. Future studies should examine whether E2 substitution could prevent cognitive deficits and aid in earlier readiness for therapy.


Subject(s)
Anorexia Nervosa/physiopathology , Estradiol/blood , Estrous Cycle/blood , Leptin/blood , Memory Disorders/blood , Animals , Body Weight , Disease Models, Animal , Female , Humans , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL