Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
2.
Clin Biochem ; 68: 50-54, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30991036

ABSTRACT

OBJECTIVES: Fluoroquinolone antibiotics are commonly used in the treatment of infections and have previously been confirmed to cross-react with previous generations of opiates immunoassays. In this work we evaluated the cross-reactivity of the three fluoroquinolones in use at our institution with a panel of 10 urine drug screens. DESIGN AND METHODS: Drug preparations of levofloxacin, ciprofloxacin, and moxifloxacin that were designed for intravenous delivery were added to drug-free urine at varying concentrations. Spiked urine samples were screened for illicit and therapeutic drugs on an Abbott Architect c16000 automated chemistry analyzer. Percent cross-reactivity was calculated. RESULTS: Levofloxacin displayed clinically relevant cross-reactivity with the Abbott MULTIGENT opiates and Thermo CEDIA® buprenorphine immunoassays but did not cross-react with the Abbott MULTIGENT oxycodone or methadone immunoassays. Moxifloxacin displayed clinically relevant cross-reactivity only with the Abbott MULTIGENT amphetamine/methamphetamine assay. Ciprofloxacin did not cross-react with any of the 10 immunoassays. CONCLUSIONS: This study demonstrates that levofloxacin cross-reacts with modern immunoassays for two related opioids (buprenorphine and morphine) and moxifloxacin cross-reacts with the amphetamine/methamphetamine assay. Urine concentrations of these fluoroquinolones that are consistent with therapeutic use produced results above commonly used-cutoffs for positivity. This underscores the necessity of confirmatory testing of presumptively positive urine drug screens.


Subject(s)
Buprenorphine/chemistry , Cross Reactions , Fluoroquinolones/chemistry , Immunoassay/methods , Opiate Alkaloids/chemistry , Amphetamine/chemistry , Analgesics, Opioid/chemistry , Humans , Levofloxacin/chemistry , Substance Abuse Detection
3.
Neurobiol Dis ; 120: 98-106, 2018 12.
Article in English | MEDLINE | ID: mdl-30195075

ABSTRACT

Mechanisms underlying α-synuclein (αSyn) mediated neurodegeneration are poorly understood. Intramuscular (IM) injection of αSyn fibrils in human A53T transgenic M83+/- mice produce a rapid model of α-synucleinopathy with highly predictable onset of motor impairment. Using varying doses of αSyn seeds, we show that αSyn-induced phenotype is largely dose-independent. We utilized the synchrony of this IM model to explore the temporal sequence of αSyn pathology, neurodegeneration and neuroinflammation. Longitudinal tracking showed that while motor neuron death and αSyn pathology occur within 2 months post IM, astrogliosis appears at a later timepoint, implying neuroinflammation is a consequence, rather than a trigger, in this prionoid model of synucleinopathy. Initiating at 3 months post IM, immune activation dominates the pathologic landscape in terminal IM-seeded M83+/- mice, as revealed by unbiased transcriptomic analyses. Our findings provide insights into the role of neuroinflammation in αSyn mediated proteostasis and neurodegeneration, which will be key in designing potential therapies.


Subject(s)
Motor Neurons/metabolism , Nerve Degeneration/metabolism , alpha-Synuclein/biosynthesis , Animals , Brain/immunology , Brain/metabolism , Brain/pathology , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Transgenic , Motor Neurons/immunology , Motor Neurons/pathology , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , alpha-Synuclein/immunology
7.
Neurosci Lett ; 661: 114-120, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-28964772

ABSTRACT

Neurofilaments are a major component of the axonal cytoskeleton in neurons and have been implicated in a number of neurodegenerative diseases due to their presence within characteristic pathological inclusions. Their contributions to these diseases are not yet fully understood, but previous studies investigated the effects of ablating the obligate subunit of neurofilaments, low molecular mass neurofilament subunit (NFL), on disease phenotypes in transgenic mouse models of Alzheimer's disease and tauopathy. Here, we tested the effects of ablating NFL in α-synuclein M83 transgenic mice expressing the human pathogenic A53T mutation, by breeding them onto an NFL null background. The induction and spread of α-synuclein inclusion pathology was triggered by the injection of preformed α-synuclein fibrils into the gastrocnemius muscle or hippocampus in M83 versus M83/NFL null mice. We observed no difference in the post-injection time to motor-impairment and paralysis endpoint or amount and distribution of α-synuclein inclusion pathology in the muscle injected M83 and M83/NFL null mice. Hippocampal injected M83/NFL null mice displayed subtle region-specific differences in the amount of α-synuclein inclusions however, pathology was observed in the same regions as the M83 mice. Overall, we observed only minor differences in the induction and transmission of α-synuclein pathology in these induced models of synucleinopathy in the presence or absence of NFL. This suggests that NFL and neurofilaments do not play a major role in influencing the induction and transmission of α-synuclein aggregation.


Subject(s)
Intermediate Filaments/metabolism , Neurons/metabolism , Prions/metabolism , alpha-Synuclein/metabolism , Animals , Brain/metabolism , Inclusion Bodies/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurodegenerative Diseases/pathology
8.
Hum Mol Genet ; 26(24): 4906-4915, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29036344

ABSTRACT

Parkinson's disease (PD) is one of many neurodegenerative diseases termed synucleinopathies, neuropathologically defined by inclusions containing aggregated α-synuclein (αS). αS gene (SNCA) mutations can directly cause autosomal dominant PD. In vitro studies demonstrated that SNCA missense mutations may either enhance or diminish αS aggregation but cross-seeding of mutant and wild-type αS proteins appear to reduce aggregation efficiency. Here, we extended these studies by assessing the effects of seeded αS aggregation in αS transgenic mice through intracerebral or peripheral injection of various mutant αS fibrils. We observed modestly decreased time to paralysis in mice transgenic for human A53T αS (line M83) intramuscularly injected with H50Q, G51D or A53E αS fibrils relative to wild-type αS fibrils. Conversely, E46K αS fibril seeding was significantly delayed and less efficient in the same experimental paradigm. However, the amount and distribution of αS inclusions in the central nervous system were similar for all αS fibril muscle injected mice that developed paralysis. Mice transgenic for human αS (line M20) injected in the hippocampus with wild-type, H50Q, G51D or A53E αS fibrils displayed induction of αS inclusion pathology that increased and spread over time. By comparison, induction of αS aggregation following the intrahippocampal injection of E46K αS fibrils in M20 mice was much less efficient. These findings show that H50Q, G51D or A53E can efficiently cross-seed and induce αS pathology in vivo. In contrast, E46K αS fibrils are intrinsically inefficient at seeding αS inclusion pathology. Consistent with previous in vitro studies, E46K αS polymers are likely distinct aggregated conformers that may represent a unique prion-like strain of αS.


Subject(s)
Parkinson Disease/genetics , alpha-Synuclein/metabolism , Animals , Central Nervous System/metabolism , Gene Expression/genetics , Gene Expression Regulation/genetics , Humans , Inclusion Bodies/metabolism , Mice , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/physiology
9.
Mol Neurodegener ; 12(1): 40, 2017 05 29.
Article in English | MEDLINE | ID: mdl-28552073

ABSTRACT

BACKGROUND: Prionoid transmission of α-synuclein (αSyn) aggregates along neuroanatomically connected projections is posited to underlie disease progression in α-synucleinopathies. Here, we specifically wanted to study whether this prionoid progression occurs via direct inter-neuronal transfer and, if so, would intrastriatal injection of αSyn aggregates lead to nigral degeneration. METHODS: To test prionoid transmission of αSyn aggregates along the nigro-striatal pathway, we injected amyloidogenic αSyn aggregates into two different regions of the striatum of adult human wild type αSyn transgenic mice (Line M20) or non-transgenic (NTG) mice and aged for 4 months. RESULTS: M20 mice injected in internal capsule (IC) or caudate putamen (CPu) regions of the striatum showed florid αSyn inclusion pathology distributed throughout the neuraxis, irrespective of anatomic connectivity. These αSyn inclusions were found in different cell types including neurons, astrocytes and even ependymal cells. On the other hand, intra-striatal injection of αSyn fibrils into NTG mice resulted in sparse αSyn pathology, mostly localized in the striatum and entorhinal cortex. Interestingly, NTG mice injected with preformed human αSyn fibrils showed no induction of αSyn inclusion pathology, suggesting the presence of a species barrier for αSyn fibrillar seeds. Modest levels of nigral dopaminergic (DA) neuronal loss was observed exclusively in substantia nigra (SN) of M20 cohorts injected in the IC, even in the absence of frank αSyn inclusions in DA neurons. None of the NTG mice or CPu-injected M20 mice showed DA neurodegeneration. Interestingly, the pattern and distribution of induced αSyn pathology corresponded with neuroinflammation especially in the SN of M20 cohorts. Hypermorphic reactive astrocytes laden with αSyn inclusions were abundantly present in the brains of M20 mice. CONCLUSIONS: Overall, our findings show that the pattern and extent of dissemination of αSyn pathology does not necessarily follow expected neuroanatomic connectivity. Further, the presence of intra-astrocytic αSyn pathology implies that glial cells participate in αSyn transmission and possibly have a role in non-cell autonomous disease modification.


Subject(s)
Brain/pathology , Neurodegenerative Diseases/pathology , alpha-Synuclein/toxicity , Animals , Humans , Mice , Mice, Transgenic
10.
J Virol ; 91(2)2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27852849

ABSTRACT

Misfolded α-synuclein (αS) is hypothesized to spread throughout the central nervous system (CNS) by neuronal connectivity leading to widespread pathology. Increasing evidence indicates that it also has the potential to invade the CNS via peripheral nerves in a prion-like manner. On the basis of the effectiveness following peripheral routes of prion administration, we extend our previous studies of CNS neuroinvasion in M83 αS transgenic mice following hind limb muscle (intramuscular [i.m.]) injection of αS fibrils by comparing various peripheral sites of inoculations with different αS protein preparations. Following intravenous injection in the tail veins of homozygous M83 transgenic (M83+/+) mice, robust αS pathology was observed in the CNS without the development of motor impairments within the time frame examined. Intraperitoneal (i.p.) injections of αS fibrils in hemizygous M83 transgenic (M83+/-) mice resulted in CNS αS pathology associated with paralysis. Interestingly, injection with soluble, nonaggregated αS resulted in paralysis and pathology in only a subset of mice, whereas soluble Δ71-82 αS, human ßS, and keyhole limpet hemocyanin (KLH) control proteins induced no symptoms or pathology. Intraperitoneal injection of αS fibrils also induced CNS αS pathology in another αS transgenic mouse line (M20), albeit less robustly in these mice. In comparison, i.m. injection of αS fibrils was more efficient in inducing CNS αS pathology in M83 mice than i.p. or tail vein injections. Furthermore, i.m. injection of soluble, nonaggregated αS in M83+/- mice also induced paralysis and CNS αS pathology, although less efficiently. These results further demonstrate the prion-like characteristics of αS and reveal its efficiency to invade the CNS via multiple routes of peripheral administration. IMPORTANCE: The misfolding and accumulation of α-synuclein (αS) inclusions are found in a number of neurodegenerative disorders and is a hallmark feature of Parkinson's disease (PD) and PD-related diseases. Similar characteristics have been observed between the infectious prion protein and αS, including its ability to spread from the peripheral nervous system and along neuroanatomical tracts within the central nervous system. In this study, we extend our previous results and investigate the efficiency of intravenous (i.v.), intraperitoneal (i.p.), and intramuscular (i.m.) routes of injection of αS fibrils and other protein controls. Our data reveal that injection of αS fibrils via these peripheral routes in αS-overexpressing mice are capable of inducing a robust αS pathology and in some cases cause paralysis. Furthermore, soluble, nonaggregated αS also induced αS pathology, albeit with much less efficiency. These findings further support and extend the idea of αS neuroinvasion from peripheral exposures.


Subject(s)
Central Nervous System Diseases/genetics , Central Nervous System Diseases/pathology , alpha-Synuclein/administration & dosage , Animals , Brain/metabolism , Brain/pathology , Central Nervous System Diseases/mortality , Central Nervous System Diseases/physiopathology , Disease Models, Animal , Inclusion Bodies/metabolism , Mice , Mice, Transgenic , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Phenotype , Protein Aggregates , Protein Aggregation, Pathological , Spinal Cord/metabolism , Spinal Cord/pathology , alpha-Synuclein/metabolism
11.
Acta Neuropathol Commun ; 4(1): 80, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27503460

ABSTRACT

Pathological inclusions containing aggregated, highly phosphorylated (at serine129) α-synuclein (αS pSer129) are characteristic of a group of neurodegenerative diseases termed synucleinopathies. Antibodies to the pSer129 epitope can be highly sensitive in detecting αS inclusions in human tissue and experimental models of synucleinopathies. However, the generation of extensively specific pSer129 antibodies has been problematic, in some cases leading to the misinterpretation of αS inclusion pathology. One common issue is cross-reactivity to the low molecular mass neurofilament subunit (NFL) phosphorylated at Ser473. Here, we generated a series of monoclonal antibodies to the pSer129 αS and pSer473 NFL epitopes. We determined the relative abilities of the known αS kinases, polo-like kinases (PLK) 1, 2 and 3 and casein kinase (CK) II in phosphorylating NFL and αS, while using this information to characterize the specificity of the new antibodies. NFL can be phosphorylated by PLK1, 2 and 3 at Ser473; however CKII shows the highest phosphorylation efficiency and specificity for this site. Conversely, PLK3 is the most efficient kinase at phosphorylating αS at Ser129, but there is overlay in the ability of these kinases to phosphorylate both epitopes. Antibody 4F8, generated to the pSer473 NFL epitope, was relatively specific for phosphorylated NFL, however it could uniquely cross-react with pSer129 αS when highly phosphorylated, further showing the structural similarity between these phospho-epitopes. All of the new pSer129 antibodies detected pathological αS inclusions in human brains and mouse and cultured cell experimental models of induced synucleinopathies. Several of these pSer129 αS antibodies reacted with the pSer473 NFL epitope, but 2 clones (LS3-2C2 and LS4-2G12) did not. However, LS3-2C2 demonstrated cross-reactivity with other proteins. Our findings further demonstrate the difficulties in generating specific pSer129 αS antibodies, but highlights that the use of multiple antibodies, such as those generated here, can provide a sensitive and accurate assessment of αS pathology.


Subject(s)
Antibodies/immunology , Epitopes/chemistry , Neurofilament Proteins/genetics , Neurofilament Proteins/immunology , alpha-Synuclein/chemistry , alpha-Synuclein/immunology , Animals , Brain/metabolism , Brain/pathology , Casein Kinase II/metabolism , Cells, Cultured , Epitopes/immunology , Escherichia coli , Female , Humans , Hybridomas , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/immunology , Sequence Homology, Amino Acid , Serine/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , alpha-Synuclein/deficiency , alpha-Synuclein/genetics
12.
Mol Neurodegener ; 10: 32, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26223783

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder that is pathologically characterized by loss of dopaminergic neurons from the substantia nigra, the presence of aggregated α-synuclein (αS) and evidence of neuroinflammation. Experimental studies have shown that the cerebral injection of recombinant fibrillar αS, especially in αS transgenic mouse models, can induce the formation and spread of αS inclusion pathology. However, studies reporting this phenomenon did not consider the presence of lipopolysaccharide (LPS) in the injected αS, produced in E. coli, as a potential confound. The objectives of this study are to develop a method to remove the LPS contamination and investigate the differences in pathologies induced by αS containing LPS or αS highly purified of LPS. RESULTS AND CONCLUSIONS: We were able to remove >99.5% of the LPS contamination from the αS preparations through the addition of a cation exchange step during purification. The αS pathology induced by injection of fibrils produced from αS containing LPS or purified of LPS, showed a similar distribution pattern; however, there was less spread into the cortex of the mice injected with αS containing higher levels of LPS. As previously reported, injection of αS fibrils could induce astrogliosis, and αS inclusions were present within astrocytes in mice injected with fibrils comprised of αS with or without cation exchange purification. Furthermore, we identified the presence of αS pathology in ependymal cells in both groups of mice, which suggests the involvement of a novel mechanism for spread in this model of αS pathology.


Subject(s)
Endotoxins/pharmacology , Parkinsonian Disorders/chemically induced , alpha-Synuclein/toxicity , Animals , Astrocytes/pathology , Cell Count , Cells, Cultured , Chromatography, Ion Exchange , Disease Progression , Drug Contamination , Endotoxins/isolation & purification , Entorhinal Cortex/drug effects , Entorhinal Cortex/pathology , Escherichia coli/chemistry , Escherichia coli/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Humans , Hydrophobic and Hydrophilic Interactions , Inclusion Bodies/chemistry , Inflammation , Injections , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Parkinsonian Disorders/pathology , Plaque, Amyloid/chemistry , Protein Conformation , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity , alpha-Synuclein/administration & dosage , alpha-Synuclein/isolation & purification
13.
Acta Neuropathol ; 130(1): 145-57, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25917047

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a degenerative disorder affecting upper and lower motor neurons, but it is increasingly recognized to affect other systems, with cognitive impairment resembling frontotemporal dementia (FTD) in some patients. We report clinical and pathologic findings of a family with ALS due to a truncating mutation, p.Gly141X, in copper/zinc superoxide dismutase (SOD1). The proband presented clinically with FTD and later showed progressive motor neuron disease, while all other family members had early-onset and rapidly progressive ALS without significant cognitive deficits. Pathologic examination of both the proband and her daughter revealed degeneration of corticospinal tracts and motor neurons in brain and spinal cord compatible with ALS. On the other hand, the proband also had neocortical and limbic system degeneration with pleomorphic neuronal cytoplasmic inclusions. Extramotor pathology in her daughter was relatively restricted to the hypothalamus and extrapyramidal system, but not the neocortex. The inclusions in the proband and her daughter were immunoreactive for SOD1, but negative for TAR DNA-binding protein of 43 kDa (TDP-43). In the proband, a number of the neocortical inclusions were immunopositive for α-internexin, initially suggesting a diagnosis of atypical FTLD, but there was no evidence of fused in sarcoma (FUS) immunoreactivity, which is often detected in atypical FTLD. Analogous to atypical FTLD, neuronal inclusions had variable co-localization of SOD1 and α-internexin. The current classification of FTLD is based on the major constituent protein: FTLD-tau, FTLD-TDP-43, and FTLD-FUS. The proband in this family indicates that SOD1, while rare, can also be the substrate of FTLD, in addition to the more common presentation of ALS. The explanation for clinical and pathologic heterogeneity of SOD1 mutations, including the p.Gly141X mutation, remains unresolved.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Brain/pathology , Frontotemporal Lobar Degeneration/genetics , Mutation , Superoxide Dismutase/genetics , Adult , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Brain/metabolism , Family , Fatal Outcome , Female , Frontotemporal Lobar Degeneration/pathology , Frontotemporal Lobar Degeneration/physiopathology , Humans , Male , Middle Aged , Pedigree , Superoxide Dismutase-1
14.
Neurosci Lett ; 597: 43-8, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25892596

ABSTRACT

Mutations in the gene that encodes α-synuclein (αS) are a known cause of Parkinson's disease. αS is also the major component of pathological inclusions that characterize this disorder and a spectrum of other neurodegenerative diseases termed synucleinopathies. The effects of the most recently identified αS mutation, A53E, on αS aggregation were studied in vitro and in cell culture models. The A53E mutation in αS impedes the formation of aggregated, amyloid protein in vitro compared to wild-type αS. Under certain conditions, A53E αS can still form elongated amyloid fibrils with similar morphology, but with thinner width compared to wild-type αS. Using amyloid seeding of αS in cell culture studies, we demonstrate that significantly less A53E αS could be induced to aggregate compared to wild-type αS, although the mutant protein was still able to form mature inclusions within some cells. Furthermore, expression of A53E αS enhanced toxicity in cells experiencing mitochondrial stress. These findings indicate that the A53E mutation in αS reduces the propensity of αS to aggregate both in vitro and in the cellular environment, and may lead to cellular toxicity through other mechanisms.


Subject(s)
Amyloid/chemistry , Amyloid/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Animals , Cell Line, Tumor , Cell Survival , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mutation , Piperidines/toxicity , Protein Aggregates , Pyrazoles/toxicity , alpha-Synuclein/genetics
15.
Acta Neuropathol ; 129(1): 53-64, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25367383

ABSTRACT

Hippocampal sclerosis (HpScl) is frequent in frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP), but it also occurs in dementia of the elderly with or without accompanying Alzheimer type pathology. HpScl has been hypothesized to be a neurodegenerative process given its association with TDP-43 pathology, but this is still controversial. TDP-43 pathology is found in Lewy body disease (LBD), but no study has focused on the pathologic and genetic characteristics of HpScl in LBD. We found HpScl in 5.2% of 669 LBD cases (289 transitional and 380 diffuse). Older age, higher Braak neurofibrillary tangle (NFT) stage, and presence of TDP-43 pathology were associated with HpScl. There was no difference in the frequency of HpScl between transitional and diffuse LBD, suggesting that Lewy-related pathology appears to have no direct association with HpScl. All HpScl cases had TDP-43 pathology consistent with Type A pattern. HpScl cases harbored genetic variation in TMEM106B that has been previously associated with FTLD-TDP. Interestingly, the severity of TDP-43-positive fine neurites in CA1 sector, a possible pathologic precursor of HpScl, was associated with the TMEM106B variant. These results demonstrate HpScl in LBD is a TDP-43 proteinopathy and is similar to FTLD-TDP Type A. Furthermore, a subset of LBD cases without HpScl ("pre-HpScl") had similar pathologic and genetic characteristics to typical HpScl, suggesting that the spectrum of HpScl pathology may be wider than previously thought. Some cases with many extracellular NFTs also had a similar profile. We suggest that HpScl is "masked" in these cases.


Subject(s)
Frontotemporal Lobar Degeneration/pathology , Hippocampus/pathology , Lewy Body Disease/pathology , TDP-43 Proteinopathies/pathology , Aged , Aged, 80 and over , Apolipoprotein E4/genetics , DNA-Binding Proteins/metabolism , Female , Frontotemporal Lobar Degeneration/epidemiology , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Hippocampus/metabolism , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Lewy Body Disease/epidemiology , Lewy Body Disease/genetics , Lewy Body Disease/metabolism , Male , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neurites/metabolism , Neurites/pathology , Progranulins , Sclerosis/epidemiology , Sclerosis/genetics , Sclerosis/metabolism , TDP-43 Proteinopathies/epidemiology , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism
16.
J Neurochem ; 131(6): 859-67, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24984882

ABSTRACT

The discoveries of mutations in SNCA were seminal findings that resulted in the knowledge that α-synuclein (αS) is the major component of Parkinson's disease-associated Lewy bodies. Since the pathologic roles of these protein inclusions and SNCA mutations are not completely established, we characterized the aggregation properties of the recently identified SNCA mutations, H50Q and G51D, to provide novel insights. The properties of recombinant H50Q, G51D, and wild-type αS to polymerize and aggregate into amyloid were studied using (trans,trans)-1-bromo-2,5-bis-(4-hydroxy)styrylbenzene fluorometry, sedimentation analyses, electron microscopy, and atomic force microscopy. These studies showed that the H50Q mutation increases the rate of αS aggregation, whereas the G51D mutation has the opposite effect. However, H50Q and G51D αS could still be similarly induced to form intracellular aggregates from the exposure to exogenous amyloidogenic seeds under conditions that promote their cellular entry. Both mutant αS proteins, but especially G51D, promoted cellular toxicity under cellular stress conditions. These findings reveal that the novel pathogenic SNCA mutations, H50Q and G51D, have divergent effects on aggregation properties relative to the wild-type protein, with G51D αS demonstrating reduced aggregation despite presenting with earlier disease onset, suggesting that these mutants promote different mechanisms of αS pathogenesis. The α-synuclein (SNCA) mutations, H50Q and G51D, cause Parkinson's disease. We found that H50Q increases and G51D decreases the rate of α-synuclein aggregation in vitro, and cells over-expressing the mutant proteins show decreased viability when stressed, compared to wild type. G51D is the first SNCA mutation to show decreased α-synuclein aggregation, suggesting a distinct disease mechanism to other SNCA mutations.


Subject(s)
Mutation/genetics , Protein Aggregates/genetics , alpha-Synuclein/genetics , Amyloid/metabolism , Cells, Cultured , Humans , Parkinson Disease/genetics , Parkinson Disease/metabolism , alpha-Synuclein/metabolism
17.
Acta Neuropathol ; 128(3): 411-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24899141

ABSTRACT

Hippocampal sclerosis of the elderly (HpScl) and Alzheimer's disease (AD), especially the limbic-predominant subtype (LP-AD), are amnestic syndromes that can be difficult to distinguish. To complicate matters, a subset has concomitant HpScl and AD (HpScl-AD). We examined a large cohort of autopsy-confirmed cases of HpScl, HpScl-AD, LP-AD, and typical AD to identify distinct clinical, genetic, and pathologic characteristics. HpScl cases were significantly older at death and had a substantially slower rate of cognitive decline than the AD subtypes. Genetic analysis revealed that the AD groups (AD, LP-AD, and HpScl-AD) were more likely to be APOE ε4 carriers. In contrast, the HpScl groups (HpScl and HpScl-AD) were more likely to exhibit genetic variants in GRN and TMEM106B that are associated with frontotemporal lobar degeneration. The HpScl groups had a high frequency of TDP-43 pathology that was most often Type A morphology and distribution, while typical AD and LP-AD had a significantly lower frequency of TDP-43 pathology that was most often Type B. These results suggest that HpScl and AD are pathologically and genetically distinct and non-synergistic neurodegenerative processes that present with amnestic dementia. Pure HpScl and HpScl with concomitant AD occur most often in elderly individuals.


Subject(s)
Alzheimer Disease , Apolipoproteins E/genetics , DNA-Binding Proteins/genetics , Hippocampus/pathology , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Aged , Aged, 80 and over , Aging/genetics , Aging/pathology , Alzheimer Disease/classification , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cognition Disorders/etiology , Cognition Disorders/genetics , DNA Mutational Analysis , Female , Humans , Longitudinal Studies , Male , Neurofibrillary Tangles/pathology , Neuropsychological Tests , Plaque, Amyloid/pathology , Progranulins , Sclerosis/pathology , Statistics, Nonparametric
18.
Neurology ; 82(21): 1871-8, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24771538

ABSTRACT

OBJECTIVE: We aimed to investigate the relationship between plasma and CSF progranulin (PGRN) levels. METHODS: Plasma and CSF PGRN were measured in a cohort of 345 subjects from the Mayo Clinic Study of Aging by ELISA. Single nucleotide polymorphism genotyping was performed using TaqMan assays. Associations between PGRN and sex, age at sample collection, diagnosis, single nucleotide polymorphism genotypes (GRN, SORT1, and APOE), and Pittsburgh compound B score were explored separately in CSF and plasma using single variable linear regression models. Pearson partial correlation coefficient was used to estimate the correlation of PGRN in CSF and plasma. RESULTS: Plasma (p = 0.0031) and CSF (p = 0.0044) PGRN significantly increased with age, whereas plasma PGRN levels were 7% lower (p = 0.0025) and CSF PGRN levels 5% higher (p = 0.0024) in male compared with female participants. Correcting for age and sex, higher plasma PGRN was associated with higher CSF PGRN (partial r = 0.17, p = 0.004). In plasma, both rs5848 (GRN; p = 0.002) and rs646776 (SORT1; p = 3.56E-7) were associated with PGRN, while only rs5848 showed highly significant association in CSF (p = 5.59E-14). Age, sex, rs5848 genotype, and plasma PGRN together accounted for only 18% of the variability observed in CSF PGRN. CONCLUSIONS: While some correlation exists between plasma and CSF PGRN, age, sex, and genetic factors differently affect PGRN levels. Therefore, caution should be taken when using plasma PGRN to predict PGRN changes in the brain. These findings further highlight that plasma PGRN levels may not accurately predict clinical features or response to future frontotemporal lobar degeneration therapies.


Subject(s)
Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/genetics , Polymorphism, Single Nucleotide , Age Factors , Aged , Aged, 80 and over , Female , Genetic Association Studies , Humans , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/cerebrospinal fluid , Male , Progranulins , Sex Factors
19.
Acta Neuropathol ; 127(5): 645-65, 2014 May.
Article in English | MEDLINE | ID: mdl-24659240

ABSTRACT

In order to further evaluate the parameters whereby intracerebral administration of recombinant α-synuclein (αS) induces pathological phenotypes in mice, we conducted a series of studies where αS fibrils were injected into the brains of M83 (A53T) and M47 (E46K) αS transgenic (Tg) mice, and non-transgenic (nTg) mice. Using multiple markers to assess αS inclusion formation, we find that injected fibrillar human αS induced widespread cerebral αS inclusion formation in the M83 Tg mice, but in both nTg and M47 Tg mice, induced αS inclusion pathology is largely restricted to the site of injection. Furthermore, mouse αS fibrils injected into nTg mice brains also resulted in inclusion pathology restricted to the site of injection with no evidence for spread. We find no compelling evidence for extensive spread of αS pathology within white matter tracts, and we attribute previous reports of white matter tract spreading to cross-reactivity of the αS pSer129/81A antibody with phosphorylated neurofilament subunit L. These studies suggest that, with the exception of the M83 Tg mice which appear to be uniquely susceptible to induction of inclusion pathology by exogenous forms of αS, there are significant barriers in mice to widespread induction of αS pathology following intracerebral administration of amyloidogenic αS.


Subject(s)
Amyloidosis/metabolism , Amyloidosis/pathology , Brain/metabolism , Brain/pathology , alpha-Synuclein/metabolism , Animals , Cells, Cultured , Escherichia coli , Gene Transfer Techniques , Humans , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Neural Pathways/metabolism , Neural Pathways/pathology , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Recombinant Proteins/metabolism , Species Specificity , alpha-Synuclein/genetics
20.
Neurology ; 81(15): 1332-41, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24027057

ABSTRACT

OBJECTIVE: To identify potential genetic modifiers contributing to the phenotypic variability that is detected in patients with repeat expansions in chromosome 9 open reading frame 72 (C9ORF72), we investigated the frequency of these expansions in a cohort of 334 subjects previously found to carry mutations in genes known to be associated with a spectrum of neurodegenerative diseases. METHODS: A 2-step protocol, with a fluorescent PCR and a repeat-primed PCR, was used to determine the presence of hexanucleotide expansions in C9ORF72. For one double mutant, we performed Southern blots to assess expansion sizes, and immunohistochemistry to characterize neuropathology. RESULTS: We detected C9ORF72 repeat expansions in 4 of 334 subjects (1.2% [or 1.8% of 217 families]). All these subjects had behavioral phenotypes and also harbored well-known pathogenic mutations in either progranulin (GRN: p.C466LfsX46, p.R493X, p.C31LfsX35) or microtubule-associated protein tau (MAPT: p.P301L). Southern blotting of one double mutant with a p.C466LfsX46 GRN mutation demonstrated a long repeat expansion in brain (>3,000 repeats), and immunohistochemistry showed mixed neuropathology with characteristics of both C9ORF72 expansions and GRN mutations. CONCLUSIONS: Our findings indicate that co-occurrence of 2 evidently pathogenic mutations could contribute to the pleiotropy that is detected in patients with C9ORF72 repeat expansions. These findings suggest that patients with known mutations should not be excluded from further studies, and that genetic counselors should be aware of this phenomenon when advising patients and their family members.


Subject(s)
DNA Repeat Expansion/genetics , Genetic Predisposition to Disease/genetics , Neurodegenerative Diseases/genetics , Proteins/genetics , Aged , Aged, 80 and over , Autopsy , C9orf72 Protein , Cohort Studies , Female , Follow-Up Studies , Genetic Testing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Microtubule-Associated Proteins/genetics , Middle Aged , Phenotype , Progranulins , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...