Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Alzheimers Res Ther ; 15(1): 137, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37596686

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is an age-related disease characterized by altered cognition, neuroinflammation, and neurodegeneration against which there is presently no effective cure. Brain-derived neurotrophic factor (BDNF) is a key neurotrophin involved in the learning and memory process, with a crucial role in synaptic plasticity and neuronal survival. Several findings support that a reduced BDNF expression in the human brain is associated with AD pathogenesis. BDNF has been proposed as a potential therapy for AD, but BDNF has low brain penetration. In this study, we used an innovative encapsulated cell biodelivery (ECB) device, containing genetically modified cells capable of releasing BDNF and characterized its feasibility and therapeutic effects in the novel App knock-in AD mouse model (AppNL-G-F). METHODS: ECB's containing human ARPE-19 cells genetically modified to release BDNF (ECB-BDNF devices) were stereotactically implanted bilaterally into hippocampus of 3-month-old AppNL-G-F mice. The stability of BDNF release and its effect on AD pathology were evaluated after 1, 2-, and 4-months post-implantation by immunohistochemical and biochemical analyses. Exploratory and memory performance using elevated plus maze (EPM) and Y-maze test were performed in the 4-months treatment group. Immunological reaction towards ECB-BDNF devices were studied under ex vivo and in vivo settings. RESULTS: The surgery and the ECB-BDNF implants were well tolerated without any signs of unwanted side effects or weight loss. ECB-BDNF devices did not induce host-mediated immune response under ex vivo set-up but showed reduced immune cell attachment when explanted 4-months post-implantation. Elevated BDNF staining around ECB-BDNF device proximity was detected after 1, 2, and 4 months treatment, but the retrieved devices showed variable BDNF release. A reduction of amyloid-ß (Aß) plaque deposition was observed around ECB-BDNF device proximity after 2-months of BDNF delivery. CONCLUSIONS: The result of this study supports the use of ECB device as a promising drug-delivery approach to locally administer BBB-impermeable factors for treating neurodegenerative conditions like AD. Optimization of the mouse-sized devices to reduce variability of BDNF release is needed to employ the ECB platform in future pre-clinical research and therapy development studies.


Subject(s)
Alzheimer Disease , Brain-Derived Neurotrophic Factor , Drug Delivery Systems , Animals , Mice , Alzheimer Disease/therapy , Amyloid beta-Peptides , Brain-Derived Neurotrophic Factor/therapeutic use , Feasibility Studies , Drug Delivery Systems/methods
2.
Acta Neuropathol Commun ; 11(1): 76, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37158955

ABSTRACT

Cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is caused by NOTCH3 mutations. Typical CADASIL is characterised by subcortical ischemic strokes due to severe arteriopathy and fibrotic thickening of small arteries. Arteriolar vascular smooth muscle cells (VSMCs) are the key target in CADASIL, but the potential mechanisms involved in their degeneration are still unclear. Focusing on cerebral microvessels in the frontal and anterior temporal lobes and the basal ganglia, we used advanced proteomic and immunohistochemical methods to explore the extent of inflammatory and immune responses in CADASIL subjects compared to similar age normal and other disease controls. There was variable loss of VSMC in medial layers of arteries in white matter as well as the cortex, that could not be distinguished whether NOTCH3 mutations were in the epidermal growth factor (EGFr) domains 1-6 or EGFr7-34. Proteomics of isolated cerebral microvessels showed alterations in several proteins, many associated with endoplasmic reticulum (ER) stress including heat shock proteins. Cerebral vessels with sparsely populated VSMCs also attracted robust accrual of perivascular microglia/macrophages in order CD45+ > CD163+ > CD68+cells, with > 60% of vessel walls exhibiting intercellular adhesion molecule-1 (ICAM-1) immunoreactivity. Functional VSMC cultures bearing the NOTCH3 Arg133Cys mutation showed increased gene expression of the pro-inflammatory cytokine interleukin 6 and ICAM-1 by 16- and 50-fold, respectively. We further found evidence for activation of the alternative pathway of complement. Immunolocalisation of complement Factor B, C3d and C5-9 terminal complex but not C1q was apparent in ~ 70% of cerebral vessels. Increased complement expression was corroborated in > 70% of cultured VSMCs bearing the Arg133Cys mutation independent of N3ECD immunoreactivity. Our observations suggest that ER stress and other cellular features associated with arteriolar VSMC damage instigate robust localized inflammatory and immune responses in CADASIL. Our study has important implications for immunomodulation approaches to counter the characteristic arteriopathy of CADASIL.


Subject(s)
CADASIL , Humans , CADASIL/genetics , Intercellular Adhesion Molecule-1 , Proteomics , Complement System Proteins , Cerebral Infarction
3.
Int J Mol Sci ; 23(16)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36012296

ABSTRACT

There is no cure yet available for Alzheimer's disease (AD). We recently optimized encapsulated cell biodelivery (ECB) devices releasing human mature nerve growth factor (hmNGF), termed ECB-NGF, to the basal forebrain of AD patients. The ECB-NGF delivery resulted in increased CSF cholinergic markers, improved glucose metabolism, and positive effects on cognition in AD patients. However, some ECB-NGF implants showed altered hmNGF release post-explantation. To optimize the ECB-NGF platform for future therapeutic purposes, we initiated in-vitro optimization studies by exposing ECB-NGF devices to physiological factors present within the AD brain. We report here that microglia cells can impair hmNGF release from ECB-NGF devices in-vitro, which can be reversed by transferring the devices to fresh culture medium. Further, we exposed the hmNGF secreting human ARPE-19 cell line (NGC0211) to microglia (HMC3) conditioned medium (MCM; untreated or treated with IL-1ß/IFNγ/Aß40/Aß42), and evaluated biochemical stress markers (ROS, GSH, ΔΨm, and Alamar Blue assay), cell death indicators (Annexin-V/PI), cell proliferation (CFSE retention and Ki67) and senescence markers (SA-ß-gal) in NGC0211 cells. MCMs from activated microglia reduced cell proliferation and induced cell senescence in NGC0211 cells, which otherwise resist biochemical alterations and cell death. These data indicate a critical but reversible impact of activated microglia on NGC0211 cells.


Subject(s)
Alzheimer Disease , Basal Forebrain , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Basal Forebrain/metabolism , Biomarkers , Cell Proliferation , Humans , Microglia/metabolism , Nerve Growth Factor/metabolism
4.
Cells ; 10(11)2021 10 21.
Article in English | MEDLINE | ID: mdl-34831056

ABSTRACT

Alzheimer's disease (AD) treatment is constrained due to the inability of peripherally administered therapeutic molecules to cross the blood-brain barrier. Encapsulated cell biodelivery (ECB) devices, a tissue-targeted approach for local drug release, was previously optimized for human mature nerve growth factor (hmNGF) delivery in AD patients but was found to have reduced hmNGF release over time. To understand the reason behind reduced ECB efficacy, we exposed hmNGF-releasing cells (NGC0211) in vitro to human cerebrospinal fluid (CSF) obtained from Subjective Cognitive Impairment (SCI), Lewy Body Dementia (LBD), and AD patients. Subsequently, we exposed NGC0211 cells directly to AD-related factors like amyloid-ß peptides (Aß40/42) or activated astrocyte-conditioned medium (Aß40/42/IL-1ß/TNFα-treated) and evaluated biochemical stress markers, cell death indicators, cell proliferation marker (Ki67), and hmNGF release. We found that all patients' CSF significantly reduced hmNGF release from NGC0211 cells in vitro. Aß40/42, inflammatory molecules, and activated astrocytes significantly affected NGC0211 cell proliferation without altering hmNGF release or other parameters important for essential functions of the NGC0211 cells. Long-term constant cell proliferation within the ECB device is critically important to maintain a steady cell population needed for stable mNGF release. These data show hampered proliferation of NGC0211 cells, which may lead to a decline of the NGC0211 cell population in ECBs, thereby reducing hmNGF release. Our study highlights the need for future studies to strengthen ECB-mediated long-term drug delivery approaches.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Cells, Immobilized/cytology , Nerve Growth Factor/metabolism , Alzheimer Disease/cerebrospinal fluid , Cell Line , Cell Proliferation , Cognitive Dysfunction/cerebrospinal fluid , Culture Media, Conditioned/pharmacology , Humans , Lewy Body Disease/cerebrospinal fluid , Peptides/metabolism , Stress, Physiological
5.
Adv Exp Med Biol ; 1331: 167-191, 2021.
Article in English | MEDLINE | ID: mdl-34453298

ABSTRACT

Age-dependent progressive neurodegeneration and associated cognitive dysfunction represent a serious concern worldwide. Currently, dementia accounts for the fifth highest cause of death, among which Alzheimer's disease (AD) represents more than 60% of the cases. AD is associated with progressive cognitive dysfunction which affects daily life of the affected individual and associated family. The cognitive dysfunctions are at least partially due to the degeneration of a specific set of neurons (cholinergic neurons) whose cell bodies are situated in the basal forebrain region (basal forebrain cholinergic neurons, BFCNs) but innervate wide areas of the brain. It has been explicitly shown that the delivery of the neurotrophic protein nerve growth factor (NGF) can rescue BFCNs and restore cognitive dysfunction, making NGF interesting as a potential therapeutic substance for AD. Unfortunately, NGF cannot pass through the blood-brain barrier (BBB) and thus peripheral administration of NGF protein is not viable therapeutically. NGF must be delivered in a way which will allow its brain penetration and availability to the BFCNs to modulate BFCN activity and viability. Over the past few decades, various methodologies have been developed to deliver NGF to the brain tissue. In this chapter, NGF delivery methods are discussed in the context of AD.


Subject(s)
Alzheimer Disease , Basal Forebrain , Alzheimer Disease/drug therapy , Humans , Nerve Growth Factor/metabolism , Neurons/metabolism
6.
Front Genet ; 11: 1022, 2020.
Article in English | MEDLINE | ID: mdl-33101365

ABSTRACT

Typical cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is caused by mutations in the human NOTCH3 gene. Cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy is characterized by subcortical ischemic strokes due to severe arteriopathy and fibrotic thickening of small vessels. Blood regulating vascular smooth muscle cells (VSMCs) appear as the key target in CADASIL but the pathogenic mechanisms remain unclear. With the hypothesis that brain glucose metabolism is disrupted in VSMCs in CADASIL, we investigated post-mortem tissues and VSMCs derived from CADASIL patients to explore gene expression and protein immunoreactivity of glucose transporters (GLUTs), particularly GLUT4 and GLUT2 using quantitative RT-PCR and immunohistochemical techniques. In vitro cell model analysis indicated that both GLUT4 and -2 gene expression levels were down-regulated in VSMCs derived from CADASIL patients, compared to controls. In vitro studies further indicated that the down regulation of GLUT4 coincided with impaired glucose uptake in VSMCs, which could be partially rescued by insulin treatment. Our observations on reduction in GLUTs in VSMCs are consistent with previous findings of decreased cerebral blood flow and glucose uptake in CADASIL patients. That impaired ability of glucose uptake is rescued by insulin is also consistent with previously reported lower proliferation rates of VSMCs derived from CADASIL subjects. Overall, these observations are consistent with the development of severe cerebral arteriopathy in CADASIL, in which VSMCs are replaced by widespread fibrosis.

7.
Curr Alzheimer Res ; 16(11): 1050-1054, 2019.
Article in English | MEDLINE | ID: mdl-31660827

ABSTRACT

INTRODUCTION: We recently showed that Amyloid Beta (Aß)40 accumulates in erythrocytes and possibly causes cell damage as evidenced by an increased number of assumed injured low-density (kg/L) erythrocytes. Furthermore, we have suggested a separation technique to isolate and concentrate such damaged red blood cells for subsequent analysis. OBJECTIVES: We isolated high- and low-density erythrocytes and investigated the accumulation patterns of the Aß peptides (Aß40, Aß42, and Aß43) in Alzheimer (AD), mild cognitive impairment (MCI), and Subjective Cognitive Impairment (SCI). METHODS: Whole blood was fractionated through a density gradient, resulting in two concentrated highand presumed injured low-density erythrocyte fractions. After cell lysis, intracellular Aß40, Aß42, and Aß43 were quantified by ELISA. RESULTS: In both high- and low-density erythrocytes, Aß40 displayed the lowest concentration in MCI, while it was equal and higher in AD and SCI. Aß40 was detected at a 10-fold higher level than Aß42, and in injured low-density erythrocytes, the lowest quantity of Aß42 was found in AD and MCI. Aß40 exhibited a 100-fold greater amount than Aß43, and lighter erythrocytes of MCI subjects displayed less intracellular Aß43 than SCI. CONCLUSION: Red blood cell accumulation patterns of Aß40, Aß42, and Aß43 differ significantly between AD, MCI, and SCI. The data must be verified through larger clinical trials. It is, however, tenable that Aß peptide distributions in erythrocyte subpopulations have the potential to be used for diagnostic purposes.


Subject(s)
Alzheimer Disease/blood , Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/blood , Erythrocytes/metabolism , Erythrocytes/pathology , Aged , Biomarkers/blood , Female , Humans , Male , Protein Isoforms/blood
8.
Curr Alzheimer Res ; 16(9): 834-835, 2019.
Article in English | MEDLINE | ID: mdl-31453787

ABSTRACT

BACKGROUND: Alzheimer's Disease (AD) features the accumulation of ß-amyloid in erythrocytes. The subsequent red cell damage may well affect their oxygen-carrying capabilities. 2,3- diphosphoglycerate (2,3-DPG) binds to the hemoglobin thereby promoting oxygen release. It is theorized that 2,3-DPG is reduced in AD and that the resulting hypoxia triggers erythropoietin (EPO) release. METHODS & OBJECTIVE: To explore this theory, we analyzed red cell 2,3-DPG content and EPO in AD, mild cognitive impairment, and the control group, subjective cognitive impairment. RESULTS: We studied (i) 2,3-DPG in red cells, and (ii) circulating EPO in AD, and both markers were unaffected by dementia. Disturbances of these oxygen-regulatory pathways do not appear to participate in brain hypoxia in AD.


Subject(s)
2,3-Diphosphoglycerate/blood , Alzheimer Disease/blood , Cognitive Dysfunction/blood , Erythrocytes/metabolism , Erythropoietin/blood , Aged , Biomarkers/blood , Cohort Studies , Diagnostic Self Evaluation , Female , Humans , Male , Middle Aged
9.
Front Neurosci ; 13: 38, 2019.
Article in English | MEDLINE | ID: mdl-30804738

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder associated with abnormal protein modification, inflammation and memory impairment. Aggregated amyloid beta (Aß) and phosphorylated tau proteins are medical diagnostic features. Loss of memory in AD has been associated with central cholinergic dysfunction in basal forebrain, from where the cholinergic circuitry projects to cerebral cortex and hippocampus. Various reports link AD progression with declining activity of cholinergic neurons in basal forebrain. The neurotrophic molecule, nerve growth factor (NGF), plays a major role in the maintenance of cholinergic neurons integrity and function, both during development and adulthood. Numerous studies have also shown that NGF contributes to the survival and regeneration of neurons during aging and in age-related diseases such as AD. Changes in neurotrophic signaling pathways are involved in the aging process and contribute to cholinergic and cognitive decline as observed in AD. Further, gradual dysregulation of neurotrophic factors like NGF and brain derived neurotrophic factor (BDNF) have been reported during AD development thus intensifying further research in targeting these factors as disease modifying therapies against AD. Today, there is no cure available for AD and the effects of the symptomatic treatment like cholinesterase inhibitors (ChEIs) and memantine are transient and moderate. Although many AD treatment studies are being carried out, there has not been any breakthrough and new therapies are thus highly needed. Long-term effective therapy for alleviating cognitive impairment is a major unmet need. Discussion and summarizing the new advancements of using NGF as a potential therapeutic implication in AD are important. In summary, the intent of this review is describing available experimental and clinical data related to AD therapy, priming to gain additional facts associated with the importance of NGF for AD treatment, and encapsulated cell biodelivery (ECB) as an efficient tool for NGF delivery.

10.
Mol Neurobiol ; 56(7): 4601-4619, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30361890

ABSTRACT

OMI/HTRA2 (high-temperature requirement serine protease A2) is a mitochondrial serine protease involved in several cellular processes, including autophagy, chaperone activity, and apoptosis. Few studies on the role of OMI/HTRA2 in Alzheimer's disease (AD) are available, but none on its relationship with the cholinergic system and neurotrophic factors as well as other AD-related proteins. In this study, immunohistochemical analyses revealed that AD patients had a higher cytosolic distribution of OMI/HTRA2 protein compared to controls. Quantitative analyses on brain extracts indicated a significant increase in the active form of OMI/HTRA2 in the AD brain. Activated OMI/HTRA2 protein positively correlated with stress-associated read-through acetylcholinesterase activity. In addition, α7 nicotinic acetylcholine receptor gene expression, a receptor also known to be localized on the outer membrane of mitochondria, showed a strong correlation with OMI/HTRA2 gene expression in three different brain regions. Interestingly, the activated OMI/HTRA2 levels also correlated with the activity of the acetylcholine-biosynthesizing enzyme, choline acetyltransferase (ChAT); with levels of the neurotrophic factors, NGF and BDNF; with levels of the soluble fragments of amyloid precursor protein (APP); and with gene expression of the microtubule-associated protein tau in the examined brain regions. Overall, the results demonstrate increased levels of the mitochondrial serine protease OMI/HTRA2, and a coherent pattern of association between the activated form of OMI/HTRA2 and several key proteins involved in AD pathology. In this paper, we propose a new hypothetical model to highlight the importance and needs of further investigation on the role of OMI/HTRA2 in the mitochondrial function and AD.


Subject(s)
Acetylcholine/metabolism , Alzheimer Disease/enzymology , Brain/enzymology , High-Temperature Requirement A Serine Peptidase 2/metabolism , Acetylcholinesterase/genetics , Acetylcholinesterase/metabolism , Aged , Aged, 80 and over , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Biomarkers/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Butyrylcholinesterase/metabolism , Female , Gene Expression Regulation , High-Temperature Requirement A Serine Peptidase 2/genetics , Humans , Male , Middle Aged , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , tau Proteins/genetics , tau Proteins/metabolism
11.
Eur J Cell Biol ; 97(8): 557-567, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30392756

ABSTRACT

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a familial progressive degenerative disorder and is caused by mutations in NOTCH3 gene. Previous study reported that mutant NOTCH3 is more prone to form aggregates than wild-type NOTCH3 and the mutant aggregates are resistant to degradation. We hypothesized that aggregation or accumulation of NOTCH3 could be due to impaired lysosomal-autophagy machinery in VSMC. Here, we investigated the possible cause of accumulation/aggregation of NOTCH3 in CADASIL using cerebral VSMCs derived from control and CADASIL patients carrying NOTCH3R133C mutation. Thioflavin-S-staining confirmed the increased accumulation of aggregated NOTCH3 in VSMCR133C compared to VSMCWT. Increased levels of the lysosomal marker, Lamp2, were detected in VSMCR133C, which also showed co-localization with NOTCH3 using double-immunohistochemistry. Increased level of LC3-II/LC3-I ratio was observed in VSMCR133C suggesting an accumulation of autophagosomes. This was coupled with the decreased co-localization of NOTCH3 with LC3, and Lamp2 and, further, increase of p62/SQSTM1 levels in VSMCR133C compared to the VSMCWT. In addition, Western blot analysis indicated phosphorylation of p-ERK, p-S6RP, and p-P70 S6K. Altogether, these results suggested a dysfunction in the autophagy-lysosomal pathway in VSMCR133C. The present study provides an interesting avenue of the research investigating the molecular mechanism of CADASIL.


Subject(s)
Autophagy , CADASIL/pathology , Lysosomes/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Chloroquine/pharmacology , Humans , Lysosomes/drug effects , MAP Kinase Signaling System/drug effects , Microtubule-Associated Proteins/metabolism , Models, Biological , Myocytes, Smooth Muscle/drug effects , Phosphorylation/drug effects , Proteolysis/drug effects , Receptor, Notch3/metabolism , Sequestosome-1 Protein/metabolism
12.
Exp Cell Res ; 371(1): 175-184, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30092220

ABSTRACT

Alzheimer's disease (AD) is characterized by early degeneration of cholinergic neurons and decreased levels of nerve growth factor (NGF). Thus, increasing the NGF levels by for instance encapsulated cell bio-delivery (ECB) is a potential treatment strategy. The results from our previous first-in-human studies on ECB of NGF to the basal forebrain cholinergic neurons were promising, but indicated some variability of long-term viability of the encapsulated cells and associated reduced NGF-release. Here we studied the effect of amyloid beta-peptides (Aß), interleukin 1-beta (IL-1ß), and CSF from AD, Lewy body dementia (LBD) or subjective cognitive impairment (SCI) patients on the NGF overproducing cell line NGC-0295. At physiological concentrations, neither Aß40 nor Aß42 had any major impact on cell viability or NGF-production. In contrast, IL-1ß dose-dependently affected NGF-production over time. Exposure of NGF-producing cells to CSF from AD patients showed significantly reduced NGF-release as compared to CSF from LBD or SCI patients. By mass spectrometry we found 3 proteins involved in inflammatory pathways to have an altered expression in AD CSF compared to LBD and SCI. Cell survival and NGF-release were not affected by Aß. NGF-release was affected by IL-1ß, suggesting that inflammation has a negative effect on ECB cells.


Subject(s)
Alzheimer Disease/genetics , Cerebrospinal Fluid/chemistry , Gene Expression Regulation/drug effects , Interleukin-1beta/pharmacology , Nerve Growth Factor/genetics , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Cell Line, Transformed , Cell Survival/drug effects , Cognitive Dysfunction/cerebrospinal fluid , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lewy Body Disease/cerebrospinal fluid , Lewy Body Disease/genetics , Lewy Body Disease/pathology , Nerve Growth Factor/metabolism , Peptide Fragments/pharmacology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Staurosporine/pharmacology
13.
J Cell Mol Med ; 22(6): 3016-3024, 2018 06.
Article in English | MEDLINE | ID: mdl-29536621

ABSTRACT

Cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a familial fatal progressive degenerative disorder. One of the pathological hallmarks of CADASIL is a dramatic reduction of vascular smooth muscle cells (VSMCs) in cerebral arteries. Using VSMCs from the vasculature of the human umbilical cord, placenta and cerebrum of CADASIL patients, we found that CADASIL VSMCs had a lower proliferation rate compared to control VSMCs. Exposure of control VSMCs and endothelial cells (ECs) to media derived from CADASIL VSMCs lowered the proliferation rate of all cells examined. By quantitative RT-PCR analysis, we observed increased Transforming growth factor-ß (TGFß) gene expression in CADASIL VSMCs. Adding TGFß-neutralizing antibody restored the proliferation rate of CADASIL VSMCs. We assessed proliferation differences in the presence or absence of TGFß-neutralizing antibody in ECs co-cultured with VSMCs. ECs co-cultured with CADASIL VSMCs exhibited a lower proliferation rate than those co-cultured with control VSMCs, and neutralization of TGFß normalized the proliferation rate of ECs co-cultured with CADASIL VSMCs. We suggest that increased TGFß expression in CADASIL VSMCs is involved in the reduced VSMC proliferation in CADASIL and may play a role in situ in altered proliferation of neighbouring cells in the vasculature.


Subject(s)
CADASIL/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Transforming Growth Factor beta/genetics , Antibodies, Neutralizing/pharmacology , CADASIL/metabolism , CADASIL/pathology , Cell Proliferation/genetics , Coculture Techniques , Endothelial Cells/metabolism , Gene Expression Regulation/genetics , Humans , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Transforming Growth Factor beta/antagonists & inhibitors
14.
Mol Neurobiol ; 54(2): 874-887, 2017 03.
Article in English | MEDLINE | ID: mdl-26780457

ABSTRACT

Studies on the mechanisms of neuronal amyloid-ß (Aß) internalisation are crucial for understanding the neuropathological progression of Alzheimer's disease (AD). We here investigated how extracellular Aß peptides are internalised and focused on three different pathways: (i) via endocytic mechanisms, (ii) via the receptor for advanced glycation end products (RAGE) and (iii) via the pore-forming protein perforin. Both Aß40 and Aß42 were internalised in retinoic acid differentiated neuroblastoma (RA-SH-SY5Y) cells. A higher concentration was required for Aß40 (250 nM) compared with Aß42 (100 nM). The internalised Aß40 showed a dot-like pattern of distribution whereas Aß42 accumulated in larger and distinct formations. By confocal microscopy, we showed that Aß40 and Aß42 co-localised with mitochondria, endoplasmic reticulum (ER) and lysosomes. Aß treatment of human primary cortical neurons (hPCN) confirmed our findings in RA-SH-SY5Y cells, but hPCN were less sensitive to Aß; therefore, a 20 (Aß40) and 50 (Aß42) times higher concentration was needed for inducing uptake. The blocking of endocytosis completely inhibited the internalisation of Aß peptides in RA-SH-SY5Y cells and hPCN, indicating that this is a major pathway by which Aß enters the cells. In addition, the internalisation of Aß42, but not Aß40, was reduced by 55 % by blocking RAGE. Finally, for the first time we showed that pore formation in cell membranes by perforin led to Aß internalisation in hPCN. Understanding how Aß is internalised sheds light on the pathological role of Aß and provides further ideas of inhibitory strategies for preventing Aß internalisation and the spreading of neurodegeneration in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Endocytosis/physiology , Neurons/metabolism , Peptide Fragments/metabolism , Perforin/metabolism , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Endocytosis/drug effects , Humans , Neurons/drug effects , Perforin/pharmacology
15.
J Alzheimers Dis ; 53(2): 621-30, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27163811

ABSTRACT

INTRODUCTION: Serotonin (5-HT) is involved in the pathology of Alzheimer's disease (AD). OBJECTIVE: We aimed to measure 5-HT level in platelets in AD and explore its association with cerebrospinal fluid (CSF), AD biomarkers (amyloid-ß 1-42 (Aß42), total tau (t-tau), and phosphorylated tau (p-tau)), and clinical symptoms. METHODS: 15 patients with AD and 20 patients with subjective cognitive impairment (SCI) were included. 5-HT metabolites were measured, in a specific fraction, using high performance liquid chromatography with electrochemical detection (HPLC-ECD). RESULTS: Significantly lower 5-HT concentrations were observed in AD patients compared to SCI patients both after normalization against total protein (p = 0.008) or platelet count (p = 0.019). SCI patients with lower 5-HT level have higher AD CSF biomarkers, total tau (p = 0.026) and tau/Aß42 ratio (p = 0.001), compared to those with high 5-HT levels. CONCLUSION: AD patients have reduced platelet 5-HT levels. In SCI, lower 5-HT content was associated with a higher AD-CSF biomarker burden.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Blood Platelets/metabolism , Peptide Fragments/cerebrospinal fluid , Serotonin/blood , tau Proteins/cerebrospinal fluid , Chromatography, High Pressure Liquid , Cognitive Dysfunction/blood , Cognitive Dysfunction/cerebrospinal fluid , Electrochemical Techniques , Female , Humans , Hydroxyindoleacetic Acid/blood , Male , P-Selectin/metabolism , Psychiatric Status Rating Scales , Statistics, Nonparametric
16.
J Neurochem ; 136(3): 497-502, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26500157

ABSTRACT

Accumulation of amyloid ß-peptide (Aß) in the brain is an important event in the pathogenesis of Alzheimer disease. We have used a transgenic mouse model expressing human amyloid precursor protein (APP) with the Arctic mutation to investigate whether Aß deposition is correlated with mitochondrial functions in these animals. We found evidence of mitochondrial dysfunction (i.e., decreased mitochondrial membrane potential, increased production of reactive oxygen species and oxidative DNA damage) at 6 months of age, when the mice showed very mild Aß deposition. More pronounced mitochondrial abnormalities were present in 24-month-old TgAPParc mice with more extensive Aß pathology. This study demonstrates for the first time mitochondrial dysfunction in transgenic mice with a mutation within the Aß peptide (the Arctic APP mutation), and confirms previous studies suggesting that mitochondrial dysfunction and oxidative stress is an early event in the pathogenesis of Alzheimer disease. This study demonstrates mitochondrial dysfunction in transgenic mice with a mutation within the amyloid beta (Aß) peptide (the Arctic amyloid precursor protein (APP) mutation). We found evidence of mitochondrial dysfunction (i.e. decreased mitochondrial membrane potential (MMP), increased production of reactive oxygen species (ROS) and oxidative DNA damage) at 6 months of age, when very mild Aß deposition is present in the mice. Also, the cytochrome c (COX) activity was significantly decreased in mitochondria from transgenic mice at 24 months of age.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Mitochondrial Diseases/genetics , Mutation/genetics , 8-Hydroxy-2'-Deoxyguanosine , Age Factors , Amyloid beta-Peptides/metabolism , Analysis of Variance , Animals , Brain/metabolism , Brain/pathology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Electron Transport Complex IV/metabolism , Gene Expression Regulation/genetics , Humans , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Reactive Oxygen Species/metabolism
17.
Mol Neurobiol ; 52(3): 1077-1092, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25288153

ABSTRACT

Dysfunctional Omi/HtrA2, a mitochondrial serine protease, has been implicated in various neurodegenerative disorders. Despite the wealth of evidence on the roles of Omi/HtrA2 in apoptosis, little is known about its cytosolic targets, the cleavage of which could account for the observed morphological changes such as cytoskeletal reorganizations in axons. By proteomic analysis, vimentin was identified as a substrate for Omi/HtrA2 and we have reported increased Omi/HtrA2 protease activity in Alzheimer disease (AD) brain. Here, we investigated a possible link between Omi/HtrA2 and vimentin cleavage, and consequence of this cleavage on mitochondrial distribution in neurons. In vitro protease assays showed vimentin to be cleaved by Omi/HtrA2 protease, and proximity ligation assay demonstrated an increased interaction between Omi/HtrA2 and vimentin in human primary neurons upon stress stimuli. Using differentiated neuroblastoma SH-SY5Y cells, we showed that Omi/HtrA2 under several different stress conditions induces cleavage of vimentin in wild-type as well as SH-SY5Y cells transfected with amyloid precursor protein with the Alzheimer disease-associated Swedish mutation. After stress treatment, inhibition of Omi/HtrA2 protease activity by the Omi/HtrA2 specific inhibitor, Ucf-101, reduced the cleavage of vimentin in wild-type cells. Following altered vimentin filaments integrity by stress stimuli, mitochondria was redistributed in differentiated SH-SY5Y cells and human primary neurons. In summary, the findings outlined in this paper suggest a role of Omi/HtrA2 in modulation of vimentin filamentous structure in neurons. Our results provide important findings for understanding the biological role of Omi/HtrA2 activity during stress conditions, and give knowledge of interplay between Omi/HtrA2 and vimentin which might affect mitochondrial distribution in neurons.


Subject(s)
Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Serine Endopeptidases/metabolism , Stress, Physiological/physiology , Vimentin/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Axons/ultrastructure , Cell Line, Tumor , Culture Media, Serum-Free/pharmacology , Cytosol/enzymology , High-Temperature Requirement A Serine Peptidase 2 , Humans , Interleukin-1beta/toxicity , Intermediate Filaments/ultrastructure , Isothiocyanates/toxicity , Mitochondria/physiology , Mitochondrial Proteins/antagonists & inhibitors , Mutation, Missense , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Neurogenesis/drug effects , Neurons/drug effects , Neurons/ultrastructure , Point Mutation , Pyrimidinones/pharmacology , Serine Proteinase Inhibitors/pharmacology , Sulfoxides , Thiones/pharmacology , Transfection , Tretinoin/pharmacology , Tunicamycin/toxicity
18.
PLoS One ; 8(6): e65936, 2013.
Article in English | MEDLINE | ID: mdl-23840379

ABSTRACT

Acetylcholine (ACh), the classical neurotransmitter, also affects a variety of nonexcitable cells, such as endothelia, microglia, astrocytes and lymphocytes in both the nervous system and secondary lymphoid organs. Most of these cells are very distant from cholinergic synapses. The action of ACh on these distant cells is unlikely to occur through diffusion, given that ACh is very short-lived in the presence of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), two extremely efficient ACh-degrading enzymes abundantly present in extracellular fluids. In this study, we show compelling evidence for presence of a high concentration and activity of the ACh-synthesizing enzyme, choline-acetyltransferase (ChAT) in human cerebrospinal fluid (CSF) and plasma. We show that ChAT levels are physiologically balanced to the levels of its counteracting enzymes, AChE and BuChE in the human plasma and CSF. Equilibrium analyses show that soluble ChAT maintains a steady-state ACh level in the presence of physiological levels of fully active ACh-degrading enzymes. We show that ChAT is secreted by cultured human-brain astrocytes, and that activated spleen lymphocytes release ChAT itself rather than ACh. We further report differential CSF levels of ChAT in relation to Alzheimer's disease risk genotypes, as well as in patients with multiple sclerosis, a chronic neuroinflammatory disease, compared to controls. Interestingly, soluble CSF ChAT levels show strong correlation with soluble complement factor levels, supporting a role in inflammatory regulation. This study provides a plausible explanation for the long-distance action of ACh through continuous renewal of ACh in extracellular fluids by the soluble ChAT and thereby maintenance of steady-state equilibrium between hydrolysis and synthesis of this ubiquitous cholinergic signal substance in the brain and peripheral compartments. These findings may have important implications for the role of cholinergic signaling in states of inflammation in general and in neurodegenerative disease, such as Alzheimer's disease and multiple sclerosis in particular.


Subject(s)
Alzheimer Disease/enzymology , Astrocytes/enzymology , Choline O-Acetyltransferase/blood , Choline O-Acetyltransferase/cerebrospinal fluid , Multiple Sclerosis/enzymology , Acetylcholine/metabolism , Acetylcholinesterase/blood , Acetylcholinesterase/cerebrospinal fluid , Alzheimer Disease/genetics , Animals , Astrocytes/cytology , Astrocytes/metabolism , Butyrylcholinesterase/blood , Butyrylcholinesterase/cerebrospinal fluid , Cells, Cultured , Choline O-Acetyltransferase/genetics , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , Genotype , Humans , Lymphocytes/cytology , Lymphocytes/enzymology , Mice , Multiple Sclerosis/genetics
19.
Proc Natl Acad Sci U S A ; 110(19): 7916-21, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23620518

ABSTRACT

It is well-established that subcompartments of endoplasmic reticulum (ER) are in physical contact with the mitochondria. These lipid raft-like regions of ER are referred to as mitochondria-associated ER membranes (MAMs), and they play an important role in, for example, lipid synthesis, calcium homeostasis, and apoptotic signaling. Perturbation of MAM function has previously been suggested in Alzheimer's disease (AD) as shown in fibroblasts from AD patients and a neuroblastoma cell line containing familial presenilin-2 AD mutation. The effect of AD pathogenesis on the ER-mitochondria interplay in the brain has so far remained unknown. Here, we studied ER-mitochondria contacts in human AD brain and related AD mouse and neuronal cell models. We found uniform distribution of MAM in neurons. Phosphofurin acidic cluster sorting protein-2 and σ1 receptor, two MAM-associated proteins, were shown to be essential for neuronal survival, because siRNA knockdown resulted in degeneration. Up-regulated MAM-associated proteins were found in the AD brain and amyloid precursor protein (APP)Swe/Lon mouse model, in which up-regulation was observed before the appearance of plaques. By studying an ER-mitochondria bridging complex, inositol-1,4,5-triphosphate receptor-voltage-dependent anion channel, we revealed that nanomolar concentrations of amyloid ß-peptide increased inositol-1,4,5-triphosphate receptor and voltage-dependent anion channel protein expression and elevated the number of ER-mitochondria contact points and mitochondrial calcium concentrations. Our data suggest an important role of ER-mitochondria contacts and cross-talk in AD pathology.


Subject(s)
Alzheimer Disease/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Receptor Cross-Talk , Amyloid/metabolism , Animals , Brain/metabolism , CHO Cells , Calcium/metabolism , Cell Line, Tumor , Cricetinae , Disease Models, Animal , Gene Knockdown Techniques , Hippocampus/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors , Membrane Microdomains/metabolism , Mice , Mutation , Neurons/metabolism , RNA, Small Interfering/metabolism , Receptors, sigma/metabolism , Subcellular Fractions/metabolism , Vesicular Transport Proteins/metabolism , Sigma-1 Receptor
20.
Exp Cell Res ; 319(3): 134-43, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23036509

ABSTRACT

Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is a familiar fatal progressive degenerative disorder characterized by cognitive decline, and recurrent stroke in young adults. Pathological features include a dramatic reduction of brain vascular smooth muscle cells and severe arteriopathy with the presence of granular osmophilic material in the arterial walls. Here we have investigated the cellular and mitochondrial function in vascular smooth muscle cell lines (VSMCs) established from CADASIL mutation carriers (R133C) and healthy controls. We found significantly lower proliferation rates in CADASIL VSMC as compared to VSMC from controls. Cultured CADASIL VSMCs were not more vulnerable than control cells to a number of toxic substances. Morphological studies showed reduced mitochondrial connectivity and increased number of mitochondria in CADASIL VSMCs. Transmission electron microscopy analysis demonstrated increased irregular and abnormal mitochondria in CADASIL VSMCs. Measurements of mitochondrial membrane potential (Δψ(m)) showed a lower percentage of fully functional mitochondria in CADASIL VSMCs. For a number of genes previously reported to be changed in CADASIL VSMCs, immunoblotting analysis demonstrated a significantly reduced SOD1 expression. These findings suggest that alteration of proliferation and mitochondrial function in CADASIL VSMCs might have an effect on vital cellular functions important for CADASIL pathology.


Subject(s)
CADASIL/physiopathology , Mitochondria, Muscle/physiology , Muscle, Smooth, Vascular/ultrastructure , CADASIL/genetics , CADASIL/pathology , Case-Control Studies , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Female , Humans , Infant, Newborn , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/physiology , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Models, Theoretical , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Mutation, Missense/physiology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Myocytes, Smooth Muscle/ultrastructure , Receptor, Notch3 , Receptors, Notch/genetics , Receptors, Notch/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...