Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Biomolecules ; 13(10)2023 09 22.
Article in English | MEDLINE | ID: mdl-37892117

ABSTRACT

Parkinson's disease (PD) is a devastating disease associated with accumulation of α-synuclein (α-Syn) within dopaminergic neurons, leading to neuronal death. PD is characterized by both motor and non-motor clinical symptoms. Several studies indicate that autophagy, an important intracellular degradation pathway, may be involved in different neurodegenerative diseases including PD. The autophagic process mediates the degradation of protein aggregates, damaged and unneeded proteins, and organelles, allowing their clearance, and thereby maintaining cell homeostasis. Impaired autophagy may cause the accumulation of abnormal proteins. Incomplete or impaired autophagy may explain the neurotoxic accumulation of protein aggregates in several neurodegenerative diseases including PD. Indeed, studies have suggested the contribution of impaired autophagy to α-Syn accumulation, the death of dopaminergic neurons, and neuroinflammation. In this review, we summarize the recent literature on the involvement of autophagy in PD pathogenesis.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/metabolism , Protein Aggregates , alpha-Synuclein/metabolism , Autophagy/physiology , Dopaminergic Neurons/metabolism
3.
J Neuroinflammation ; 20(1): 174, 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37496076

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the leading cause of dementia in the world. The pathology of AD is affiliated with the elevation of both tau (τ) and ß-amyloid (Aß) pathologies. Yet, the direct link between natural τ expression on glia cell activity and Aß remains unclear. While experiments in mouse models suggest that an increase in Aß exacerbates τ pathology when expressed under a neuronal promoter, brain pathology from AD patients suggests an appearance of τ pathology in regions without Aß. METHODS: Here, we aimed to assess the link between τ and Aß using a new mouse model that was generated by crossing a mouse model that expresses two human mutations of the human MAPT under a mouse Tau natural promoter with 5xFAD mice that express human mutated APP and PS1 in neurons. RESULTS: The new mouse model, called 5xFAD TAU, shows accelerated cognitive impairment at 2 months of age, increased number of Aß depositions at 4 months and neuritic plaques at 6 months of age. An expression of human mutated TAU in astrocytes leads to a dystrophic appearance and reduces their ability to engulf Aß, which leads to an increased brain Aß load. Astrocytes expressing mutated human TAU showed an impairment in the expression of vascular endothelial growth factor (VEGF) that has previously been suggested to play an important role in supporting neurons. CONCLUSIONS: Our results suggest the role of τ in exacerbating Aß pathology in addition to pointing out the potential role of astrocytes in disease progression. Further research of the crosstalk between τ and Aß in astrocytes may increase our understanding of the role glia cells have in the pathology of AD with the aim of identifying novel therapeutic interventions to an otherwise currently incurable disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Animals , Humans , Infant , Mice , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Brain/metabolism , Disease Models, Animal , Mice, Transgenic , tau Proteins/genetics , tau Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
Proc Natl Acad Sci U S A ; 120(21): e2220684120, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37186836

ABSTRACT

Brain insulin signaling controls peripheral energy metabolism and plays a key role in the regulation of mood and cognition. Epidemiological studies have indicated a strong connection between type 2 diabetes (T2D) and neurodegenerative disorders, especially Alzheimer's disease (AD), linked via dysregulation of insulin signaling, i.e., insulin resistance. While most studies have focused on neurons, here, we aim to understand the role of insulin signaling in astrocytes, a glial cell type highly implicated in AD pathology and AD progression. To this end, we created a mouse model by crossing 5xFAD transgenic mice, a well-recognized AD mouse model that expresses five familial AD mutations, with mice carrying a selective, inducible insulin receptor (IR) knockout in astrocytes (iGIRKO). We show that by age 6 mo, iGIRKO/5xFAD mice exhibited greater alterations in nesting, Y-maze performance, and fear response than those of mice with the 5xFAD transgenes alone. This was associated with increased Tau (T231) phosphorylation, increased Aß plaque size, and increased association of astrocytes with plaques in the cerebral cortex as assessed using tissue CLARITY of the brain in the iGIRKO/5xFAD mice. Mechanistically, in vitro knockout of IR in primary astrocytes resulted in loss of insulin signaling, reduced ATP production and glycolic capacity, and impaired Aß uptake both in the basal and insulin-stimulated states. Thus, insulin signaling in astrocytes plays an important role in the control of Aß uptake, thereby contributing to AD pathology, and highlighting the potential importance of targeting insulin signaling in astrocytes as a site for therapeutics for patients with T2D and AD.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Mice , Animals , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Insulin/metabolism , Diabetes Mellitus, Type 2/metabolism , Mice, Transgenic , Phenotype , Disease Models, Animal
5.
Int J Mol Sci ; 23(9)2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35563336

ABSTRACT

Stressful unpredictable life events have been implicated in numerous diseases. It is now becoming clear that some life periods are more vulnerable than others. As adolescence is a sensitive period in brain development, the long-term effects of stress during this period could be significant. We investigated the long-term effects of exposure to unpredictable chronic mild stress in adolescent mice on alternative splicing of Sirtuin 1. One-month-old mice were exposed to 4 weeks of UCMS and examined for anxiety and cognition at the age of 2, 4 and 6 months. We found a rise in anxious behavior immediately after the exposure to stress. Notably, there was a long-term impairment of performance in cognitive tasks and an imbalance in Sirtuin 1 and TrkB receptor alternative splicing in the stress-exposed mice compared with controls. To conclude, our results show that exposure to unpredictable chronic mild stress during adolescence affects cognition in adulthood. Understanding pathways affiliated with stress may help minimize the long-term emotional effects of an unpredictable, stressful event.


Subject(s)
Alternative Splicing , Sirtuin 1 , Stress, Psychological , Alternative Splicing/genetics , Alternative Splicing/physiology , Animals , Anxiety/genetics , Anxiety/metabolism , Cognition/physiology , Female , Mice , Sirtuin 1/genetics , Sirtuin 1/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism
6.
Front Immunol ; 13: 835774, 2022.
Article in English | MEDLINE | ID: mdl-35350789

ABSTRACT

Diabetes is a metabolic disease that may lead to different life-threatening complications. While insulin constitutes a beneficial treatment, its use may be limited due to increased degradation and an increase in side effects such as weight gain and hypoglycemia. Small molecule inhibitors to insulin-degrading enzyme (IDE) have been previously suggested as a potential treatment for diabetes through their ability to reduce insulin degradation and thus increase insulin activity. Nevertheless, their tendency to bind to the zinc ion in the catalytic site of IDE may affect other important metalloproteases and limit their clinical use. Here, we describe the isolation of an IDE-specific antibody that specifically inhibits insulin degradation by IDE. Using phage display, we generated a human IDE-specific antibody that binds human and mouse IDE with high affinity and specificity and can differentiate between active IDE to a mutated IDE with reduced catalytic activity in the range of 30 nM. We further assessed the ability of that IDE-inhibiting antibody to improve insulin activity in vivo in an STZ-induced diabetes mouse model. Since human antibodies may stimulate the mouse immune response to generate anti-human antibodies, we reformatted our inhibitory antibody to a "reverse chimeric" antibody that maintained the ability to inhibit IDE in vitro, but consisted of mouse constant regions, for reduced immunogenicity. We discovered that one intraperitoneal (IP) administration of the IDE-specific antibody in STZ-induced diabetic mice improved insulin activity in an insulin tolerance test (ITT) assay and reduced blood glucose levels. Our results suggest that antibody-mediated inhibition of IDE may be beneficial on improving insulin activity in a diabetic environment.


Subject(s)
Diabetes Mellitus, Experimental , Insulysin , Animals , Antibodies , Catalytic Domain , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Insulin/metabolism , Insulysin/metabolism , Mice
7.
Proc Natl Acad Sci U S A ; 118(24)2021 06 15.
Article in English | MEDLINE | ID: mdl-34099562

ABSTRACT

High levels of homocysteine are reported as a risk factor for Alzheimer's disease (AD). Correspondingly, inborn hyperhomocysteinemia is associated with an increased predisposition to the development of dementia in later stages of life. Yet, the mechanistic link between homocysteine accumulation and the pathological neurodegenerative processes is still elusive. Furthermore, despite the clear association between protein aggregation and AD, attempts to develop therapy that specifically targets this process have not been successful. It is envisioned that the failure in the development of efficacious therapeutic intervention may lie in the metabolomic state of affected individuals. We recently demonstrated the ability of metabolites to self-assemble and cross-seed the aggregation of pathological proteins, suggesting a role for metabolite structures in the initiation of neurodegenerative diseases. Here, we provide a report of homocysteine crystal structure and self-assembly into amyloid-like toxic fibrils, their inhibition by polyphenols, and their ability to seed the aggregation of the AD-associated ß-amyloid polypeptide. A yeast model of hyperhomocysteinemia indicates a toxic effect, correlated with increased intracellular amyloid staining that could be rescued by polyphenol treatment. Analysis of AD mouse model brain sections indicates the presence of homocysteine assemblies and the interplay between ß-amyloid and homocysteine. This work implies a molecular basis for the association between homocysteine accumulation and AD pathology, potentially leading to a paradigm shift in the understanding of AD initial pathological processes.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Homocysteine/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/ultrastructure , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/pathology , Cell Death/drug effects , Cell Line, Tumor , Disease Models, Animal , Homocysteine/chemistry , Humans , Ion Mobility Spectrometry , Kinetics , Mice, Transgenic , Models, Biological , Polyphenols/pharmacology , Saccharomyces cerevisiae/metabolism
8.
FASEB J ; 35(5): e21374, 2021 05.
Article in English | MEDLINE | ID: mdl-33835493

ABSTRACT

Inhibition of insulin-degrading enzyme (IDE) is a possible target for treating diabetes. However, it has not yet evolved into a medical intervention, mainly because most developed inhibitors target the zinc in IDE's catalytic site, potentially causing toxicity to other essential metalloproteases. Since IDE is a cellular receptor for the varicella-zoster virus (VZV), we constructed a VZV-based inhibitor. We computationally characterized its interaction site with IDE showing that the peptide specifically binds inside IDE's central cavity, however, not in close proximity to the zinc ion. We confirmed the peptide's effective inhibition on IDE activity in vitro and showed its efficacy in ameliorating insulin-related defects in types 1 and 2 diabetes mouse models. In addition, we suggest that inhibition of IDE may ameliorate the pro-inflammatory profile of CD4+ T-cells toward insulin. Together, we propose a potential role of a designed VZV-derived peptide to serve as a selectively-targeted and as an efficient diabetes therapy.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/therapy , Insulin/metabolism , Insulysin/antagonists & inhibitors , Peptide Fragments/administration & dosage , Viral Envelope Proteins/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/pathology , Enzyme Inhibitors/administration & dosage , Female , Herpesvirus 3, Human/physiology , Insulysin/genetics , Insulysin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout
9.
J Neurochem ; 158(6): 1412-1424, 2021 09.
Article in English | MEDLINE | ID: mdl-33314073

ABSTRACT

Microglia play a vital role in maintaining brain homeostasis. Their continuous sensing of surrounding micro-environments is crucial for their activity. Cross talk between specific neurons and microglia might occur through specific neurotransmitter receptors on microglia. Impairment with this interaction might result in pathological activity of microglia against potential insults. The reason for this activity in many neurodegenerative diseases such as Alzheimer's disease (AD) is not known. However, several papers report of the effects of different neurotransmitter agonists on microglial cells function that relate to their activity in AD. This review aims to summarize those works and to raise potential fundamental questions for future research.


Subject(s)
Alzheimer Disease/metabolism , Microglia/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Alzheimer Disease/pathology , Animals , Humans , Microglia/pathology , Neurons/pathology , Receptors, Cholinergic/metabolism
10.
Int J Mol Sci ; 21(9)2020 May 05.
Article in English | MEDLINE | ID: mdl-32380752

ABSTRACT

The two major proteins involved in Alzheimer's disease (AD) are the amyloid precursor protein (APP) and Tau. Here, we demonstrate that these two proteins can bind to each other. Four possible peptides APP1 (390-412), APP2 (713-730), Tau1 (19-34) and Tau2 (331-348), were predicted to be involved in this interaction, with actual binding confirmed for APP1 and Tau1. In vivo studies were performed in an Alzheimer Disease animal model-APP double transgenic (Tg) 5xFAD-as well as in 5xFAD crossed with Tau transgenic 5xFADXTau (FT), which exhibit declined cognitive reduction at four months of age. Nasal administration of APP1 and Tau1 mixture, three times a week for four or five months, reduced amyloid plaque burden as well as the level of soluble Aß 1-42 in the brain. The treatment prevented the deterioration of cognitive functions when initiated at the age of three months, before cognitive deficiency was evident, and also at the age of six months, when such deficiencies are already observed, leading to a full regain of cognitive function.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Peptide Fragments/metabolism , tau Proteins/chemistry , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Biomarkers , Cognition/drug effects , Disease Models, Animal , Fluorescent Antibody Technique , Humans , Maze Learning/drug effects , Mice , Mice, Transgenic , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Plaque, Amyloid/drug therapy , Plaque, Amyloid/etiology , Plaque, Amyloid/pathology , Protein Binding
12.
J Neural Transm (Vienna) ; 127(2): 149-158, 2020 02.
Article in English | MEDLINE | ID: mdl-32016606

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease associated with motor deficiency and rigidity. The genetic risks of the disease is reported to be between 5 and 10% depending on the background of the population. While PD is not considered an immune-mediated disease, amounting evidence in recent years suggests a major role of inflammation in the progression of PD. Markers of inflammation can be found around the regions of risk and adjacent to the appearance of Lewy bodies within the basal ganglia and the substantia nigra (SN) that are associated with PD pathology. Microglia, an important type of brain cell, has been reported to play a major role in mediating neuroinflammation and in PD disease pathology. This review aims to point out the potential role of microglia in disease progression and suggest that the interaction of microglia with the dopaminergic neurons may also facilitate the specificity of the disease in brain regions affected by PD.


Subject(s)
Disease Progression , Dopaminergic Neurons , Inflammation , Microglia , Parkinson Disease , Animals , Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Microglia/immunology , Microglia/metabolism , Parkinson Disease/genetics , Parkinson Disease/immunology , Parkinson Disease/metabolism
13.
J Neurochem ; 152(1): 61-71, 2020 01.
Article in English | MEDLINE | ID: mdl-31520492

ABSTRACT

It has been suggested that extracellular alpha synuclein (αSyn) can mediate neuroinflammation in Parkinson's disease, and that αSyn affects B-cell maturation. However, the function of αSyn in T cells is poorly understood. We hypothesized that αSyn can affect CD4+ T-cell proliferation and activity. We found that αSyn deficiency exacerbates disease progression in 8 weeks old C57BL6/J EAE-induced mice, and that αSyn-deficient CD4+ T cells have increased pro-inflammatory response to myelin antigen relative to wild-type cells, as measured by cytokine secretion of interleukin IL-17 and interferon gamma. Furthermore, expression of αSyn on a background of αSyn knockout mitigates the inflammatory responses in CD4+ T cells. We discovered that elevated levels of Nurr1, a transcription factor belonging to the orphan nuclear receptor family, are associated with the pro-inflammatory profile of αSyn-deficient CD4+ T cells. In addition, we demonstrated that silencing of Nurr1 expression using an siRNA reduces IL-17 levels and increases the levels of IL-10, an anti-inflammatory cytokine. Study of αSyn-mediated cellular pathways in CD4+ T cells may provide useful insights into the development of pro-inflammatory responses in immunity, providing future avenues for therapeutic intervention.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphocyte Activation/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , alpha-Synuclein/deficiency , Animals , Cell Proliferation , Female , Gene Expression Regulation , Gene Silencing , Inflammation/immunology , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/immunology , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Th1 Cells/immunology , alpha-Synuclein/genetics , alpha-Synuclein/physiology
14.
J Neurosci Methods ; 331: 108527, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31775012

ABSTRACT

BACKGROUND: Numerous disorders affecting the optic nerve require histological examination of whole length optic nerves and chiasm. Most methods employed to study the histopathology of the optic nerves in animal models of human diseases involve resection of a short retrobulbar section after eye globe exenteration, commonly obtained in mice. This approach might affect the morphology of the optic nerve, thus limiting accurate identification of pathological changes in the tissue. Some histological studies were performed on longer or more posterior parts of the anterior visual pathway included the chiasm. However, an accurate replicable protocol for such whole length (eye globe to chiasm) dissection is currently unavailable in published literature. NEW METHOD: Here we describe a protocol for dissecting the whole length of the optic nerves and chiasm through a craniotomy incision. RESULTS: We describe in detail the stages necessary for exposing the optic nerves, the chiasm and the optic tracts, and for detaching them with minimal traction. COMPARISON WITH EXISTING METHOD: The existing replicable method provide only a sample of the retrobulbar optic nerve and the sample might be affected by traction. Our protocol provides a whole length specimen of the optic nerve and chiasm without concern of traction artifacts. CONCLUSIONS: We present a simple and straightforward approach to isolate the complete anterior visual pathway in the mouse for histopathological evaluation.


Subject(s)
Optic Nerve Diseases , Optic Nerve , Animals , Dissection , Mice , Models, Animal , Optic Nerve/surgery , Orbit
15.
Neurobiol Aging ; 62: 105-119, 2018 02.
Article in English | MEDLINE | ID: mdl-29141186

ABSTRACT

There is a real need for new interventions for Alzheimer's disease (AD). Hyperbaric oxygen therapy (HBOT), the medical administration of 100% oxygen at conditions greater than 1 atmosphere absolute, has been used successfully to treat several neurological conditions, but its effects on AD pathology have never been thoroughly examined. Therefore, we exposed old triple-transgenic (3xTg) and non-transgenic mice to HBOT followed by behavioral, histological, and biochemical analyses. HBOT attenuated neuroinflammatory processes by reducing astrogliosis, microgliosis, and the secretion of proinflammatory cytokines (IL-1ß and TNFα) and increasing expression of scavenger receptor A, arginase1, and antiinflammatory cytokines (IL-4 and IL-10). Moreover, HBOT reduced hypoxia, amyloid burden, and tau phosphorylation in 3xTg mice and ameliorated their behavioral deficits. Therefore, we suggest that HBOT has multifaceted effects that reduce AD pathologies, even in old mice. Given that HBOT is used in the clinic to treat various indications, including neurological conditions, these results suggest HBOT as a novel therapeutic intervention for AD.


Subject(s)
Alzheimer Disease/therapy , Hyperbaric Oxygenation/methods , Inflammation/therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Arginase/metabolism , Astrocytes/pathology , Behavior, Animal , Cytokines/metabolism , Disease Models, Animal , Hypoxia/therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Phosphorylation , Scavenger Receptors, Class A/metabolism , tau Proteins/metabolism
16.
J Neurochem ; 143(5): 584-594, 2017 12.
Article in English | MEDLINE | ID: mdl-28921554

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder, of which 1% of the hereditary cases are linked to mutations in DJ-1, an oxidative stress sensor. The pathological hallmark of PD is intercellular inclusions termed Lewy Bodies, composed mainly of α-Synuclein (α-Syn) protein. Recent findings have shown that α-Syn can be transmitted from cell to cell, suggesting an important role of microglia, as the main scavenger cells of the brain, in clearing α-Syn. We previously reported that the knock down (KD) of DJ-1 in microglia increased cells' neurotoxicity to dopaminergic neurons. Here, we discovered that α-Syn significantly induced elevated secretion of the proinflammatory cytokines IL-6 and IL-1ß and a significant dose-dependent elevation in the production of nitric oxide in DJ-1 KD microglia, compared to control microglia. We further investigated the ability of DJ-1 KD microglia to uptake and degrade soluble α-Syn, and discovered that DJ-1 KD reduces cell-surface lipid raft expression in microglia and impairs their ability to uptake soluble α-Syn. Autophagy is an important mechanism for degradation of intracellular proteins and organelles. We discovered that DJ-1 KD microglia exhibit an impaired autophagy-dependent degradation of p62 and LC3 proteins, and that manipulation of autophagy had less effect on α-Syn uptake and clearance in DJ-1 KD microglia, compared to control microglia. Further studies of the link between DJ-1, α-Syn uptake and autophagy may provide useful insights into the role of microglia in the etiology of the PD.


Subject(s)
Autophagy/drug effects , Dopaminergic Neurons/drug effects , Microglia/drug effects , Phagocytosis/drug effects , Protein Deglycase DJ-1/metabolism , alpha-Synuclein/pharmacology , Animals , Cells, Cultured , Cytokines/metabolism , Humans , Mice, Inbred BALB C , Oxidative Stress/drug effects , Protein Deglycase DJ-1/deficiency , alpha-Synuclein/metabolism
17.
Neuropharmacology ; 113(Pt A): 445-456, 2017 02.
Article in English | MEDLINE | ID: mdl-27984078

ABSTRACT

Previous study demonstrated that the novel multitarget compound, MT-031 preserved in one molecule entity the beneficial properties of its parent drugs, rasagiline and rivastigmine, and exerted high dual potencies of monoamine oxidase-A (MAO-A) and cholinesterase (ChE) inhibition in acute-treated mice and neuroprotective effects against H2O2-induced neurotoxicity in human neuroblastoma SH-SY5Y cells. The present study aimed to further investigate the anti-inflammatory and protective effects of MT-031 in scopolamine mouse model and inflammatory cell cultures. Our findings demonstrated that once daily chronic administration of MT-031 (5-10 mg/kg) to mice antagonized scopolamine-induced memory and cognitive impairments, displayed brain selective MAO-A and AChE/BuChE inhibition, increased the levels of striatal dopamine (DA), serotonin (5-HT) and norepinephrine and prevented the metabolism of DA and 5-HT. In addition, MT-031 upregulated mRNA expression levels of Bcl-2, the neurotrophic factors, (e.g., brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF) and nerve growth factor (NGF)), the antioxidant enzyme catalase and the anti-inflammatory cytokine, neurotrophic tyrosine kinase receptor (Ntrk), and down-regulated the mRNA expression levels of the pro-inflammatory interleukin (IL)-6 in scopolamine-induced mice. In accordance, MT-031 was shown to reduce reactive oxygen species accumulation, increase the levels of anti-inflammatory cytokines, IL-10 and decrease the levels of the pro-inflammatory cytokines, IL-1ß, IL-6, IL-17 and interferon-gamma (IFN-γ) in activated mouse splenocytes and microglial cells. Taken together, these pharmacological properties of MT-031 can be of clinical importance for developing this novel multitarget compound as a novel drug candidate for the treatment of Alzheimer's disease.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Butyrylcholinesterase , Cholinesterase Inhibitors/administration & dosage , Monoamine Oxidase Inhibitors/administration & dosage , Neuroprotective Agents/administration & dosage , Scopolamine/toxicity , Acetylcholinesterase/metabolism , Animals , Brain/drug effects , Brain/metabolism , Butyrylcholinesterase/metabolism , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Monoamine Oxidase/metabolism
18.
Neurobiol Dis ; 96: 84-94, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27544484

ABSTRACT

In Alzheimer's disease (AD), astrocytes undergo morphological changes ranging from atrophy to hypertrophy, but the effect of such changes at the functional level is still largely unknown. Here, we aimed to investigate whether alterations in astrocyte activity in AD are transient and depend on their microenvironment, or whether they are irreversible. We established and characterized a new protocol for the isolation of adult astrocytes and discovered that astrocytes isolated from old 5xFAD mice have higher GFAP expression than astrocytes derived from WT mice, as observed in vivo. We found high C1q levels in brain sections from old 5xFAD mice in close vicinity to amyloid plaques and astrocyte processes. Interestingly, while old 5xFAD astrocytes are impaired in uptake of soluble Aß42, this effect was reversed upon an addition of exogenous C1q, suggesting a potential role for C1q in astrocyte-mediated Aß clearance. Our results suggest that scavenger receptor B1 plays a role in C1q-facilitated Aß uptake by astrocytes and that expression of scavenger receptor B1 is reduced in adult old 5xFAD astrocytes. Furthermore, old 5xFAD astrocytes show impairment in support of neuronal growth in co-culture and neurotoxicity concomitant with an elevation in IL-6 expression. Further understanding of the impact of astrocyte impairment on AD pathology may provide insights into the etiology of AD.


Subject(s)
Aging , Alzheimer Disease , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Brain/pathology , Gene Expression Regulation/genetics , Neuroprotective Agents/therapeutic use , Peptide Fragments/metabolism , Aging/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , CD11b Antigen/metabolism , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Interleukin-6/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Receptors, Complement/metabolism
19.
J Neurosci ; 36(19): 5185-92, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27170117

ABSTRACT

UNLABELLED: Multiple EGF-like domains 10 (Megf10) is a class F scavenger receptor (SR-F3) expressed on astrocytes and myosatellite cells, and recessive mutations in humans result in early-onset myopathy, areflexia, respiratory distress, and dysphagia (EMARDD). Here we report that Megf10-deficient mice have increased apoptotic cells in the developing cerebellum and have impaired phagocytosis of apoptotic cells by astrocytes ex vivo We also report that cells transfected with Megf10 gain the ability to phagocytose apoptotic neurons and that Megf10 binds with high affinity to C1q, an eat-me signal for apoptotic cells. In contrast, cells expressing Megf10 with EMARDD mutations have impaired apoptotic cell clearance and impaired binding to C1q. Our studies reveal that Megf10 is a receptor for C1q and identify a novel role for Megf10 in clearance of apoptotic cells in the mammalian developing brain with potential relevance to EMARDD patients and other CNS disorders. SIGNIFICANCE STATEMENT: Apoptosis is a universal homeostatic process and occurs in many disease conditions. Multiple EGF-like domains 10 (Megf10) is emerging as an essential receptor for synaptic pruning, clearance of neuronal debris, and for muscle differentiation. Here we define a novel Megf10-dependent pathway for apoptotic cell clearance and show that Megf10 is a receptor for C1q, an eat-me signal, that binds phosphatidylserine expressed on the surface of apoptotic cells. Understanding the pathways by which apoptotic cells are cleared in the CNS is relevant to many physiological and pathological conditions of the CNS.


Subject(s)
Apoptosis , Astrocytes/metabolism , Complement C1q/metabolism , Membrane Proteins/metabolism , Animals , Cells, Cultured , Cerebellum/cytology , Cerebellum/growth & development , Cerebellum/metabolism , Distal Myopathies/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mutation , Phagocytosis , Protein Binding
20.
J Alzheimers Dis ; 51(3): 915-27, 2016.
Article in English | MEDLINE | ID: mdl-26923027

ABSTRACT

Alzheimer's disease (AD) is the most prevalent form of dementia in elderly. Genetic studies revealed allelic segregation of the apolipoprotein E (ApoE) gene in sporadic AD and in families with higher risk of AD. The mechanisms underlying the pathological effects of ApoE4 are not yet entirely clear. Several studies indicate that autophagy, which plays an important role in degradation pathways of proteins, organelles and protein aggregates, may be impaired in AD. In the present study, we investigated the effects of ApoE4 versus the ApoE3 isoform on the process of autophagy in mouse-derived astrocytes. The results obtained reveal that under several autophagy-inducing conditions, astrocytes expressing ApoE4 exhibit lower autophagic flux compared to astrocytes expressing ApoE3. Using an in situ model, we examined the role of autophagy and the effects thereon of ApoE4 in the elimination of Aß plaques from isolated brain sections of transgenic 5xFAD mice. This revealed that ApoE4 astrocytes eliminate Aß plaques less effectively than the corresponding ApoE3 astrocytes. Additional experiments showed that the autophagy inducer, rapamycin, enhances Aß plaque degradation by ApoE4 astrocytes whereas the autophagy inhibitor, chloroquine, blocks Aß plaque degradation by ApoE3 astrocytes. Taken together, these findings show that ApoE4 impairs autophagy in astrocyte cultures and that this effect is associated with reduced capacity to clear Aß plaques. This suggests that impaired autophagy may play a role in mediating the pathological effects of ApoE4 in AD.


Subject(s)
Apolipoprotein E4/metabolism , Astrocytes/physiology , Autophagy/physiology , Amyloid beta-Peptides/metabolism , Animals , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/genetics , Astrocytes/drug effects , Astrocytes/pathology , Autophagy/drug effects , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cells, Cultured , Central Nervous System Agents/pharmacology , Chloroquine/pharmacology , Disease Models, Animal , Humans , Mice, Transgenic , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology , Sirolimus/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...