Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
J Biol Chem ; 300(5): 107231, 2024 May.
Article in English | MEDLINE | ID: mdl-38537700

ABSTRACT

Aggregation of leukocyte cell-derived chemotaxin 2 (LECT2) causes ALECT2, a systemic amyloidosis that affects the kidney and liver. Previous studies established that LECT2 fibrillogenesis is accelerated by the loss of its bound zinc ion and stirring/shaking. These forms of agitation create heterogeneous shear conditions, including air-liquid interfaces that denature proteins, that are not present in the body. Here, we determined the extent to which a more physiological form of mechanical stress-shear generated by fluid flow through a network of narrow channels-drives LECT2 fibrillogenesis. To mimic blood flow through the kidney, where LECT2 and other proteins form amyloid deposits, we developed a microfluidic device consisting of progressively branched channels narrowing from 5 mm to 20 µm in width. Shear was particularly pronounced at the branch points and in the smallest capillaries. Aggregation was induced within 24 h by shear levels that were in the physiological range and well below those required to unfold globular proteins such as LECT2. EM images suggested the resulting fibril ultrastructures were different when generated by laminar flow shear versus shaking/stirring. Importantly, results from the microfluidic device showed the first evidence that the I40V mutation accelerated fibril formation and increased both the size and the density of the aggregates. These findings suggest that kidney-like flow shear, in combination with zinc loss, acts in combination with the I40V mutation to trigger LECT2 amyloidogenesis. These microfluidic devices may be of general use for uncovering mechanisms by which blood flow induces misfolding and amyloidosis of circulating proteins.


Subject(s)
Amyloid Neuropathies , Intercellular Signaling Peptides and Proteins , Kidney , Renal Plasma Flow , Humans , Amyloid/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Kidney/blood supply , Kidney/physiopathology , Stress, Mechanical , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/physiopathology , Shear Strength , Protein Aggregates
2.
FEBS J ; 289(2): 494-506, 2022 01.
Article in English | MEDLINE | ID: mdl-34482629

ABSTRACT

Light-chain (AL) amyloidosis is characterized by deposition of immunoglobulin light chains (LC) as fibrils in target organs. Alongside the full-length protein, abundant LC fragments are always present in AL deposits. Herein, by combining gel-based and mass spectrometry analyses, we identified and compared the fragmentation sites of amyloid LCs from multiple organs of an AL λ amyloidosis patient (AL-55). The positions pinpointed here in kidney and subcutaneous fat, alongside those previously detected in heart of the same patient, were aligned and mapped on the LC's dimeric and fibrillar states. All tissues contain fragmented LCs along with the full-length protein; the fragment pattern is coincident across organs, although microheterogeneity exists. Multiple cleavage positions were detected; some are shared, whereas some are organ-specific, likely due to a complex of proteases. Cleavage sites are concentrated in 'proteolysis-prone' regions, common to all tissues. Several proteolytic sites are not accessible on native dimers, while they are compatible with fibrils. Overall, data suggest that the heterogeneous ensemble of LC fragments originates in tissues and is consistent with digestion of preformed fibrils, or with the hypothesis that initial proteolytic cleavage of the constant domain triggers the amyloidogenic potential of LCs, followed by subsequent proteolytic degradation. This work provides a unique set of molecular data on proteolysis from ex vivo amyloid, which allows discussing hypotheses on role and timing of proteolytic events occurring along amyloid formation and accumulation in AL patients.


Subject(s)
Amyloid Neuropathies/genetics , Amyloid/genetics , Amyloidogenic Proteins/genetics , Amyloidosis/genetics , Immunoglobulin Light Chains/genetics , Amyloid/metabolism , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/pathology , Amyloidosis/metabolism , Amyloidosis/pathology , Endopeptidases/genetics , Humans , Immunoglobulin Light Chains/metabolism , Kinetics , Peptide Hydrolases/genetics , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Proteolysis , Thermodynamics
3.
Nat Rev Neurosci ; 23(1): 53-66, 2022 01.
Article in English | MEDLINE | ID: mdl-34815562

ABSTRACT

The current conceptualization of Alzheimer disease (AD) is driven by the amyloid hypothesis, in which a deterministic chain of events leads from amyloid deposition and then tau deposition to neurodegeneration and progressive cognitive impairment. This model fits autosomal dominant AD but is less applicable to sporadic AD. Owing to emerging information regarding the complex biology of AD and the challenges of developing amyloid-targeting drugs, the amyloid hypothesis needs to be reconsidered. Here we propose a probabilistic model of AD in which three variants of AD (autosomal dominant AD, APOE ε4-related sporadic AD and APOE ε4-unrelated sporadic AD) feature decreasing penetrance and decreasing weight of the amyloid pathophysiological cascade, and increasing weight of stochastic factors (environmental exposures and lower-risk genes). Together, these variants account for a large share of the neuropathological and clinical variability observed in people with AD. The implementation of this model in research might lead to a better understanding of disease pathophysiology, a revision of the current clinical taxonomy and accelerated development of strategies to prevent and treat AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Models, Statistical , Alzheimer Disease/psychology , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/pathology , Amyloid beta-Peptides , Animals , Humans , tau Proteins/metabolism
4.
Int J Mol Sci ; 22(8)2021 Apr 11.
Article in English | MEDLINE | ID: mdl-33920464

ABSTRACT

Salmon calcitonin is a good model for studying amyloid behavior and neurotoxicity. Its slow aggregation rate allows the purification of low molecular weight prefibrillar oligomers, which are the most toxic species. It has been proposed that these species may cause amyloid pore formation in neuronal membranes through contact with negatively charged sialic acid residues of the ganglioside GM1. In particular, it has been proposed that an electrostatic interaction may be responsible for the initial contact between prefibrillar oligomers and GM1 contained in lipid rafts. Based on this evidence, the aim of our work was to investigate whether the neurotoxic action induced by calcitonin prefibrillar oligomers could be counteracted by treatment with neuraminidase, an enzyme that removes sialic acid residues from gangliosides. Therefore, we studied cell viability in HT22 cell lines and evaluated the effects on synaptic transmission and long-term potentiation by in vitro extracellular recordings in mouse hippocampal slices. Our results showed that treatment with neuraminidase alters the surface charges of lipid rafts, preventing interaction between the calcitonin prefibrillar oligomers and GM1, and suggesting that the enzyme, depending on the concentration used, may have a partial or total protective action in terms of cell survival and modulation of synaptic transmission.


Subject(s)
Amyloid Neuropathies , Calcitonin/toxicity , Fish Proteins/toxicity , Neuraminidase/pharmacology , Salmon , Amyloid Neuropathies/chemically induced , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/pathology , Amyloid Neuropathies/prevention & control , Animals , G(M1) Ganglioside/metabolism , Male , Membrane Microdomains/metabolism , Membrane Microdomains/pathology , Mice , Mice, Inbred BALB C , Static Electricity
5.
J Peripher Nerv Syst ; 25(2): 85-101, 2020 06.
Article in English | MEDLINE | ID: mdl-32378274

ABSTRACT

Until recently, systemic amyloidoses were regarded as ineluctably disabling and life-threatening diseases. However, this field has witnessed major advances in the last decade, with significant improvements in therapeutic options and in the availability of accurate and non-invasive diagnostic tools. Outstanding progress includes unprecedented hematological response rates provided by risk-adapted regimens in light chain (AL) amyloidosis and the approval of innovative pharmacological agents for both hereditary and wild-type transthyretin amyloidosis (ATTR). Moreover, the incidence of secondary (AA) amyloidosis has continuously reduced, reflecting advances in therapeutics and overall management of several chronic inflammatory diseases. The identification and validation of novel therapeutic targets has grounded on a better knowledge of key molecular events underlying protein misfolding and aggregation and on the increasing availability of diagnostic, prognostic and predictive markers of organ damage and response to treatment. In this review, we focus on these recent advancements and discuss how they are translating into improved outcomes. Neurological involvement dominates the clinical picture in transthyretin and gelsolin inherited amyloidosis and has a significant impact on disease course and management in all patients. Neurologists, therefore, play a major role in improving patients' journey to diagnosis and in providing early access to treatment in order to prevent significant disability and extend survival.


Subject(s)
Amyloid Neuropathies , Amyloid Neuropathies/classification , Amyloid Neuropathies/diagnosis , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/therapy , Amyloid Neuropathies, Familial/diagnosis , Amyloid Neuropathies, Familial/metabolism , Amyloid Neuropathies, Familial/therapy , Humans
6.
J Alzheimers Dis ; 74(4): 1167-1187, 2020.
Article in English | MEDLINE | ID: mdl-32144981

ABSTRACT

Early changes in inhibitory synapse connectivities are thought to contribute to the excitation/inhibition imbalance preceding neurodegeneration in Alzheimer's disease (AD). Recently, we reported a robust increase in the level of different key-proteins of inhibitory synapses in hippocampal subregions of pre-symptomatic APPswe-PS1 mice, a model of cerebral amyloidosis. Besides increased inhibitory synaptic clusters on parvalbumin-positive projections in CA1 and CA3, we observed impaired communication between these two hippocampal areas of young APP-PS1 mice. Interestingly, the phosphorylation of gephyrin, a major organizer of inhibitory synapses, was also increased. Here, we demonstrate that the protein levels of CDK5, a kinase involved in the phosphorylation of gephyrin, and its regulatory protein p35 are also significantly increased in hippocampal subregions of young APP-PS1 mice. Consistently, the expression of hAPP-swe in cultured hippocampal neurons resulted in higher p35-protein levels, indicating a possible molecular link between increased Aß-production and the elevated p35/CDK5 levels seen in vivo. Further, a shRNA mediated downregulation of p35-expression in hippocampal neurons correlated with a decrease in gephyrin phosphorylation and in a reduced density of synaptic γ2-GABAA-receptor clusters. These findings, together with the detection of gephyrin colocalization with CDK5 and p35 by immunostaining and proximity ligation experiments in vivo and in vitro, are supporting our hypothesis that Aß has a profound impact on inhibitory network properties, likely mediated at least in part by p35/CDK5 signaling. This further underscores the impact of altered inhibitory synaptic transmission in AD.


Subject(s)
Amyloid Neuropathies/metabolism , Amyloid beta-Peptides/metabolism , Cyclin-Dependent Kinase 5/metabolism , Phosphotransferases/metabolism , Signal Transduction , Synapses/physiology , Amyloid Neuropathies/physiopathology , Animals , Brain/metabolism , Brain/physiopathology , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Synapses/metabolism
8.
Cell Mol Biol Lett ; 24: 69, 2019.
Article in English | MEDLINE | ID: mdl-31867046

ABSTRACT

With the first RNA interference (RNAi) drug (ONPATTRO (patisiran)) on the market, we witness the RNAi therapy field reaching a critical turning point, when further improvements in drug candidate design and delivery pipelines should enable fast delivery of novel life changing treatments to patients. Nevertheless, ignoring parallel development of RNAi dedicated in vitro pharmacological profiling aiming to identify undesirable off-target activity may slow down or halt progress in the RNAi field. Since academic research is currently fueling the RNAi development pipeline with new therapeutic options, the objective of this article is to briefly summarize the basics of RNAi therapy, as well as to discuss how to translate basic research into better understanding of related drug candidate safety profiles early in the process.


Subject(s)
Amyloid Neuropathies/therapy , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/therapeutic use , RNAi Therapeutics/methods , Amyloid Neuropathies/genetics , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/pathology , Animals , Gene Transfer Techniques , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Targeted Therapy/methods , RNA Stability , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Untranslated/antagonists & inhibitors , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism
9.
Clin Chim Acta ; 499: 70-74, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31479652

ABSTRACT

BACKGROUND: Commercial solutions of human serum albumin (HSA) are administered to critically ill patients for the treatment of shock, restoration of blood volume, and the acute management of burns. Previously, conflicting results on the effects of HSA administration have been reported varying from a favorable increase in total plasma antioxidant capacity to a higher mortality rate in traumatic brain injury (TBI) patients. These results could be partially explained due to the known heterogeneity of HSA solutions. We report the discovery of S-sulfonated human transthyretin (hTTR) in HSA solutions. METHODS: Proteomics was performed on commercially available solutions of 5% HSA by LC-MS analysis. The MS charge envelope for hTTR was deconvolved to the uncharged native hTTR parent mass (13,762 Da). The parent mass was then integrated, and relative proportions of the 2 major species of hTTR, native and S-sulfonated hTTR (13,842 Da), were calculated. RESULTS: The majority of hTTR found in 5% commercial HSA solutions is in the S-sulfonated form regardless of the age of the HSA solution. S-sulfonation of hTTR at the free cysteine residue in position 10 appears to be the result of a mixed disulfide exchange possibly with S-cysteinylated hTTR or S-cysteinylated HSA. hTTR is a tetramer composed of four identical monomers each containing a reduced cysteine residue in position 10. S-sulfonation of hTTR at this cysteine residue can destabilize the hTTR tetramer, an important step in the formation of TTR-related amyloid fibrils. CONCLUSIONS: Administration of a commercial HSA solution that already contains S-sulfonated hTTR could potentially contribute to the development of amyloid-related/polyneuropathy in the critically ill.


Subject(s)
Amyloid Neuropathies/metabolism , Prealbumin/analysis , Serum Albumin, Human/chemistry , Solutions/chemistry , Solutions/economics , Amyloid Neuropathies/pathology , Chromatography, Liquid , Cysteine/chemistry , Cysteine/metabolism , Humans , Mass Spectrometry , Oxidation-Reduction , Prealbumin/metabolism , Proteomics , Serum Albumin, Human/metabolism
10.
Amyloid ; 26(2): 55-65, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30907141

ABSTRACT

There have now been randomized controlled trials of four different therapeutics for hereditary amyloid polyneuropathy related to transthyretin (TTR) deposition and one for amyloidotic cardiomyopathy of both genetic and sporadic origin. It is likely that in the next few months those not already approved by either the US Food and Drug Administration (FDA) and/or the European Medicines Authority (EMA) will receive similar approvals for treatment for all or particular groups of patients. This is a far cry from circumstances less than 10 years ago when the only available therapy was gene replacement by liver transplant. The randomized controlled trials have shown that all the treatments (tafamidis, diflunisal, patisiran, and inotersen) are effective in the context of a clinical trial. However, we have very little idea of whether individual patients will respond in an equally positive way to all the drugs or whether there will be some who respond better to one or another or not respond at all, nor do we know whether combinations will be additive or synergistic. We lack validated markers of clinical response. While the small molecule TTR stabilizers increase serum TTR levels, the RNA-based drugs lower serum TTR. In the latter case, it is not clear that the reduction in serum TTR is related to the clinical response in a 1:1 fashion. Pharmaceutical companies have made substantial investments in the development of these agents and will clearly attempt to recoup those investments quickly. It is incumbent upon those of us who care for these patients to develop ways to assess the effects of therapy in the shortest possible time at the lowest possible cost. The better we are able to accomplish this the more likely it is that we will be able to treat the most patients in the most clinically efficient fashion regardless of their economic status. We now have the drugs we just have to figure out who should get them and when.


Subject(s)
Amyloid Neuropathies, Familial/drug therapy , Prealbumin , Precision Medicine , Randomized Controlled Trials as Topic , Amyloid Neuropathies/drug therapy , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/therapy , Amyloid Neuropathies, Familial/therapy , Benzoxazoles/therapeutic use , Diflunisal/therapeutic use , Female , Humans , Male , Oligonucleotides/therapeutic use , RNA, Small Interfering/therapeutic use , Treatment Outcome
12.
Orphanet J Rare Dis ; 13(1): 225, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30558645

ABSTRACT

BACKGROUND: Emerging evidence suggests that several factors can impact disease progression in transthyretin amyloid polyneuropathy (ATTR-PN). The present analysis used longitudinal data from Val30Met patients participating in the tafamidis (selective TTR stabilizer) clinical development program to evaluate the impact of baseline neurologic severity on disease progression in ATTR-PN. METHODS: A linear mixed-effects model for repeated measures (MMRM) was constructed using tafamidis and placebo data from the intent-to-treat Val30Met population of the original registration study as well as tafamidis data from the two consecutive open-label extension studies. The second extension study is ongoing, but a prospectively-planned interim analysis involving a cleaned and locked database was conducted (cut-off: December 31, 2014). Val30Met patients are presented by treatment groups as those who received tafamidis during the registration and open-label studies (T-T group), or who received placebo during the registration study and were switched to tafamidis in the open-label studies (P-T group). Neurologic functioning was assessed at baseline and subsequent visits using the Neuropathy Impairment Score-Lower Limbs (NIS-LL). The analysis focused on the disease trajectory over the first 18 months of treatment. RESULTS: The T-T (n = 64) and P-T (n = 61) cohorts were predominantly Caucasian and presented with early-stage neurologic disease (mean [standard deviation] baseline NIS-LL values were 8.4 [11.4] and 11.4 [13.5], respectively). The MMRM analysis demonstrated that baseline severity is an independent significant predictor of disease progression in addition to the treatment effect: patients with a lower baseline NIS-LL showed less progression than those with a higher baseline NIS-LL (p < 0.0001). Neurologic progression in the T-T group was less than in the P-T group across all levels of baseline NIS-LL (p = 0.0088), and the degree of separation increased over the 18-month period. Similar results were seen with the NIS-LL muscle weakness subscale. CONCLUSIONS: This analysis of patients with Val30Met ATTR-PN demonstrates that neurologic disease progression strongly depends on baseline neurologic severity and illustrates the disease-modifying effect of tafamidis relative to placebo across a range of baseline levels of neurologic severity and treatment durations. These data also underscore the benefit of early diagnosis and treatment with tafamidis in delaying disease progression in ATTR-PN. TRIAL REGISTRATION: NCT00409175 , NCT00791492 and NCT00925002 registered 08 December 2006, 14 November 2008 (retrospectively registered), and 19 June 2009, respectively.


Subject(s)
Amyloid Neuropathies/drug therapy , Amyloid Neuropathies/physiopathology , Benzoxazoles/therapeutic use , Adult , Amyloid Neuropathies/metabolism , Amyloidosis/drug therapy , Amyloidosis/metabolism , Disease Progression , Double-Blind Method , Female , Humans , Male , Middle Aged , Prealbumin/metabolism , Prospective Studies
13.
Proc Natl Acad Sci U S A ; 115(33): E7710-E7719, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061394

ABSTRACT

Cell-autonomous and cell-nonautonomous mechanisms of neurodegeneration appear to occur in the proteinopathies, including Alzheimer's and Parkinson's diseases. However, how neuronal toxicity is generated from misfolding-prone proteins secreted by nonneuronal tissues and whether modulating protein aggregate levels at distal locales affects the degeneration of postmitotic neurons remains unknown. We generated and characterized animal models of the transthyretin (TTR) amyloidoses that faithfully recapitulate cell-nonautonomous neuronal proteotoxicity by expressing human TTR in the Caenorhabditis elegans muscle. We identified sensory neurons with affected morphological and behavioral nociception-sensing impairments. Nonnative TTR oligomer load and neurotoxicity increased following inhibition of TTR degradation in distal macrophage-like nonaffected cells. Moreover, reducing TTR levels by RNAi or by kinetically stabilizing natively folded TTR pharmacologically decreased TTR aggregate load and attenuated neuronal dysfunction. These findings reveal a critical role for in trans modulation of aggregation-prone degradation that directly affects postmitotic tissue degeneration observed in the proteinopathies.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Prealbumin/metabolism , Protein Aggregates , Amyloid Neuropathies/genetics , Amyloid Neuropathies/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Humans , Prealbumin/genetics , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism
14.
Ann Neurol ; 80(3): 401-11, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27422051

ABSTRACT

OBJECTIVE: To systematically compare transthyretin with primary amyloid neuropathy to define their natural history and the underlying mechanisms for differences in phenotype and natural history. METHODS: All patients with defined amyloid subtype and peripheral neuropathy who completed autonomic testing and electromyography at Mayo Clinic Rochester between 1993 and 2013 were included. Medical records were reviewed for time of onset of defined clinical features. The degree of autonomic impairment was quantified using the composite autonomic severity scale. Comparisons were made between acquired and inherited forms of amyloidosis. RESULTS: One hundred one cases of amyloidosis with peripheral neuropathy were identified, 60 primary and 41 transthyretin. Twenty transthyretin cases were found to have Val30Met mutations; 21 had other mutations. Compared to primary cases, transthyretin cases had longer survival, longer time to diagnosis, higher composite autonomic severity scale scores, greater reduction of upper limb nerve conduction study amplitudes, more frequent occurrence of weakness, and later non-neuronal systemic involvement. Four systemic markers (cardiac involvement by echocardiogram, weight loss > 10 pounds, orthostatic intolerance, fatigue) in combination were highly predictive of poor survival in both groups. INTERPRETATION: These findings suggest that transthyretin has earlier and greater predilection for neural involvement and more delayed systemic involvement. The degree and rate of systemic involvement is most closely related to prognosis. Ann Neurol 2016;80:401-411.


Subject(s)
Amyloid Neuropathies/metabolism , Amyloid Neuropathies/physiopathology , Amyloid/metabolism , Prealbumin/metabolism , Aged , Female , Humans , Male , Middle Aged , Mutation , Phenotype , Prealbumin/genetics , Prognosis , Retrospective Studies
15.
Neurology ; 86(9): 821-8, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26843562

ABSTRACT

OBJECTIVE: To image ß-amyloid (Aß) plaque burden in long-term survivors of traumatic brain injury (TBI), test whether traumatic axonal injury and Aß are correlated, and compare the spatial distribution of Aß to Alzheimer disease (AD). METHODS: Patients 11 months to 17 years after moderate-severe TBI underwent (11)C-Pittsburgh compound B ((11)C-PiB)-PET, structural and diffusion MRI, and neuropsychological examination. Healthy aged controls and patients with AD underwent PET and structural MRI. Binding potential (BPND) images of (11)C-PiB, which index Aß plaque density, were computed using an automatic reference region extraction procedure. Voxelwise and regional differences in BPND were assessed. In TBI, a measure of white matter integrity, fractional anisotropy, was estimated and correlated with (11)C-PiB BPND. RESULTS: Twenty-eight participants (9 with TBI, 9 controls, 10 with AD) were assessed. Increased (11)C-PiB BPND was found in TBI vs controls in the posterior cingulate cortex and cerebellum. Binding in the posterior cingulate cortex increased with decreasing fractional anisotropy of associated white matter tracts and increased with time since injury. Compared to AD, binding after TBI was lower in neocortical regions but increased in the cerebellum. CONCLUSIONS: Increased Aß burden was observed in TBI. The distribution overlaps with, but is distinct from, that of AD. This suggests a mechanistic link between TBI and the development of neuropathologic features of dementia, which may relate to axonal damage produced by the injury.


Subject(s)
Amyloid Neuropathies/diagnosis , Amyloid Neuropathies/etiology , Amyloid beta-Peptides/metabolism , Axons/pathology , Brain Injuries/complications , Brain Injuries/diagnosis , Adult , Amyloid Neuropathies/metabolism , Biomarkers/metabolism , Brain Injuries/metabolism , Diffusion Tensor Imaging/methods , Female , Humans , Longitudinal Studies , Male , Middle Aged , Multimodal Imaging/methods , Positron-Emission Tomography/methods , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
16.
Biochem Soc Trans ; 42(5): 1286-90, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25233405

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease characterized by the aggregation of amyloid ß-peptide (Aß) into ß-sheet-rich fibrils. Although plaques containing Aß fibrils have been viewed as the conventional hallmark of AD, recent research implicates small oligomeric species formed during the aggregation of Aß in the neuronal toxicity and cognitive deficits associated with AD. We have demonstrated that oligomers, but not monomers, of Aß40 and Aß42 were found to induce calcium signalling in astrocytes but not in neurons. This cell specificity was dependent on the higher cholesterol level in the membrane of astrocytes compared with neurons. The Aß-induced calcium signal stimulated NADPH oxidase and induced increased reactive oxygen species (ROS) production. These events are detectable at physiologically relevant concentrations of Aß. Excessive ROS production and Ca²âº overload induced mitochondrial depolarization through activation of the DNA repairing enzyme poly(ADP-ribose) polymerase-1 (PARP-1) and opening mitochondrial permeability transition pore (mPTP). Aß significantly reduced the level of GSH in both astrocytes and neurons, an effect which is dependent on external calcium. Thus Aß induces a [Ca²âº]c signal in astrocytes which could regulate the GSH level in co-cultures that in the area of excessive ROS production could be a trigger for neurotoxicity. The pineal hormone melatonin, the glycoprotein clusterin and regulation of the membrane cholesterol can modify Aß-induced calcium signals, ROS production and mitochondrial depolarization, which eventually lead to neuroprotection.


Subject(s)
Amyloid Neuropathies/metabolism , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Cell Communication , Models, Biological , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Neuropathies/pathology , Animals , Astrocytes/pathology , Calcium Signaling , Glutathione/antagonists & inhibitors , Glutathione/metabolism , Humans , Membrane Potential, Mitochondrial , Neurons/pathology , Reactive Oxygen Species/metabolism
18.
J Alzheimers Dis ; 36(2): 335-47, 2013.
Article in English | MEDLINE | ID: mdl-23603398

ABSTRACT

BACKGROUND: Apolipoprotein E (ApoE) is the major apolipoprotein present in the high-density lipoprotein-like particles in the central nervous system (CNS). ApoE is involved in various protective functions in CNS including cholesterol transport, anti-inflammatory, and antioxidant effects. An ApoE peptide would be expected to exert protective effects on neuroinflammation. OBJECTIVE: To determine the effects of an ApoE mimetic peptide Ac-hE18A-NH2 on amyloid-ß pathology. METHOD: Using human APP/PS1ΔE9 transgenic mice and in vitro studies, we have evaluated the effect of an ApoE mimetic peptide, Ac-hE18A-NH2, on amyloid plaque deposition and inflammation. RESULTS: Administration of Ac-hE18A-NH2 to APP/PS1ΔE9 mice for 6 weeks (50 µg/mouse, 3 times a week) significantly improved cognition with a concomitant decrease in amyloid plaque deposition and reduced activated microglia and astrocytes, and increased brain ApoE levels. Oligomeric Aß42 (oAß42) and oxidized PAPC (ox-PAPC) inhibited secretion of ApoE in U251 cells, a human astrocyte cell line, and this effect was ameliorated in the presence of peptide Ac-hE18A-NH2. The peptide also increased Aß42 uptake in a cell line of human macrophages. CONCLUSIONS: Peptide Ac-hE18A-NH2 attenuates the effects of oxidative stress on ApoE secretion, inhibits amyloid plaque deposition, and thus could be beneficial in the treatment of Alzheimer's disease.


Subject(s)
Amyloid Neuropathies/drug therapy , Antipsychotic Agents/therapeutic use , Brain/metabolism , Lipoproteins/therapeutic use , Peptide Fragments/therapeutic use , Amyloid Neuropathies/complications , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Apolipoproteins E/metabolism , Brain/drug effects , Cell Line, Transformed , Cholesterol/blood , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Mutation/genetics , Peptide Fragments/metabolism , Plaque, Amyloid/drug therapy , Plaque, Amyloid/etiology , Plaque, Amyloid/genetics , Presenilin-1/genetics , Transfection
19.
Nat Rev Drug Discov ; 11(3): 185-6, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22378262

ABSTRACT

In November 2011, tafamidis (Vyndaqel; Pfizer), a small molecule that inhibits the dissociation of transthyretin tetramers, was granted marketing authorization by the European Commission for the treatment of transthyretin amyloidosis in adult patients with stage 1 symptomatic polyneuropathy to delay peripheral neurological impairment.


Subject(s)
Amyloid Neuropathies/drug therapy , Benzoxazoles/therapeutic use , Prealbumin/antagonists & inhibitors , Amyloid Neuropathies/metabolism , Animals , Benzoxazoles/chemistry , Benzoxazoles/metabolism , Clinical Trials as Topic/trends , Humans , Prealbumin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...