Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Acta Neuropathol Commun ; 12(1): 70, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38698465

RESUMEN

The majority of patients with Parkinson disease (PD) experience a loss in their sense of smell and accumulate insoluble α-synuclein aggregates in their olfactory bulbs (OB). Subjects affected by a SARS-CoV-2-linked illness (COVID-19) also frequently experience hyposmia. We previously postulated that microglial activation as well as α-synuclein and tau misprocessing can occur during host responses following microbial encounters. Using semiquantitative measurements of immunohistochemical signals, we examined OB and olfactory tract specimens collected serially at autopsies between 2020 and 2023. Deceased subjects comprised 50 adults, which included COVID19 + patients (n = 22), individuals with Lewy body disease (e.g., PD; dementia with Lewy bodies (n = 6)), Alzheimer disease (AD; n = 3), and other neurodegenerative disorders (e.g., progressive supranuclear palsy (n = 2); multisystem atrophy (n = 1)). Further, we included neurologically healthy controls (n = 9), and added subjects with an inflammation-rich brain disorder as neurological controls (NCO; n = 7). When probing for microglial and histiocytic reactivity in the anterior olfactory nuclei (AON) by anti-CD68 immunostaining, scores were consistently elevated in NCO and AD cases. In contrast, microglial signals on average were not significantly altered in COVID19 + patients relative to healthy controls, although anti-CD68 reactivity in their OB and tracts declined with progression in age. Mild-to-moderate increases in phospho-α-synuclein and phospho-tau signals were detected in the AON of tauopathy- and synucleinopathy-afflicted brains, respectively, consistent with mixed pathology, as described by others. Lastly, when both sides were available for comparison in our case series, we saw no asymmetry in the degree of pathology of the left versus right OB and tracts. We concluded from our autopsy series that after a fatal course of COVID-19, microscopic changes in the rostral, intracranial portion of the olfactory circuitry -when present- reflected neurodegenerative processes seen elsewhere in the brain. In general, microglial reactivity correlated best with the degree of Alzheimer's-linked tauopathy and declined with progression of age in COVID19 + patients.


Asunto(s)
COVID-19 , Microglía , Bulbo Olfatorio , Humanos , COVID-19/patología , COVID-19/complicaciones , Bulbo Olfatorio/patología , Bulbo Olfatorio/metabolismo , Anciano , Masculino , Femenino , Anciano de 80 o más Años , Persona de Mediana Edad , Microglía/patología , Microglía/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , SARS-CoV-2 , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/metabolismo
2.
Cell Death Dis ; 15(4): 246, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38575601

RESUMEN

Parkinson's disease (PD) is a debilitating neurodegenerative disease characterized by the loss of midbrain dopaminergic neurons (DaNs) and the abnormal accumulation of α-Synuclein (α-Syn) protein. Currently, no treatment can slow nor halt the progression of PD. Multiplications and mutations of the α-Syn gene (SNCA) cause PD-associated syndromes and animal models that overexpress α-Syn replicate several features of PD. Decreasing total α-Syn levels, therefore, is an attractive approach to slow down neurodegeneration in patients with synucleinopathy. We previously performed a genetic screen for modifiers of α-Syn levels and identified CDK14, a kinase of largely unknown function as a regulator of α-Syn. To test the potential therapeutic effects of CDK14 reduction in PD, we ablated Cdk14 in the α-Syn preformed fibrils (PFF)-induced PD mouse model. We found that loss of Cdk14 mitigates the grip strength deficit of PFF-treated mice and ameliorates PFF-induced cortical α-Syn pathology, indicated by reduced numbers of pS129 α-Syn-containing cells. In primary neurons, we found that Cdk14 depletion protects against the propagation of toxic α-Syn species. We further validated these findings on pS129 α-Syn levels in PD patient neurons. Finally, we leveraged the recent discovery of a covalent inhibitor of CDK14 to determine whether this target is pharmacologically tractable in vitro and in vivo. We found that CDK14 inhibition decreases total and pathologically aggregated α-Syn in human neurons, in PFF-challenged rat neurons and in the brains of α-Syn-humanized mice. In summary, we suggest that CDK14 represents a novel therapeutic target for PD-associated synucleinopathy.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Sinucleinopatías , Animales , Humanos , Ratones , Ratas , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Sinucleinopatías/metabolismo , Sinucleinopatías/patología
3.
Mater Today Bio ; 25: 100992, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38371467

RESUMEN

Modern in vitro technologies for preclinical research, including organ-on-a-chip, organoids- and assembloid-based systems, have rapidly emerged as pivotal tools for elucidating disease mechanisms and assessing the efficacy of putative therapeutics. In this context, advanced in vitro models of Parkinson's Disease (PD) offer the potential to accelerate drug discovery by enabling effective platforms that recapitulate both physiological and pathological attributes of the in vivo environment. Although these systems often aim at replicating the PD-associated loss of dopaminergic (DA) neurons, only a few have modelled the degradation of dopaminergic pathways as a way to mimic the disruption of downstream regulation mechanisms that define the characteristic motor symptoms of the disease. To this end, assembloids have been successfully employed to recapitulate neuronal pathways between distinct brain regions. However, the investigation and characterization of these connections through neural tracing and electrophysiological analysis remain a technically challenging and time-consuming process. Here, we present a novel bioengineered platform consisting of surface-grown midbrain and striatal organoids at opposite sides of a self-assembled DA pathway. In particular, dopaminergic neurons and striatal GABAergic neurons spontaneously form DA connections across a microelectrode array (MEA), specifically integrated for the real-time monitoring of electrophysiological development and stimuli response. Calcium imaging data showed spiking synchronicity of the two organoids forming the inter-organoid pathways (IOPs) demonstrating that they are functionally connected. MEA recordings confirm a more robust response to the DA neurotoxin 6-OHDA compared to midbrain organoids alone, thereby validating the potential of this technology to generate highly tractable, easily extractable real-time functional readouts to investigate the dysfunctional dopaminergic network of PD patients.

4.
Mov Disord ; 38(7): 1127-1142, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37156737

RESUMEN

BACKGROUND: More than 200 years after James Parkinsondescribed a clinical syndrome based on his astute observations, Parkinson's disease (PD) has evolved into a complex entity, akin to the heterogeneity of other complex human syndromes of the central nervous system such as dementia, motor neuron disease, multiple sclerosis, and epilepsy. Clinicians, pathologists, and basic science researchers evolved arrange of concepts andcriteria for the clinical, genetic, mechanistic, and neuropathological characterization of what, in their best judgment, constitutes PD. However, these specialists have generated and used criteria that are not necessarily aligned between their different operational definitions, which may hinder progress in solving the riddle of the distinct forms of PD and ultimately how to treat them. OBJECTIVE: This task force has identified current in consistencies between the definitions of PD and its diverse variants in different domains: clinical criteria, neuropathological classification, genetic subtyping, biomarker signatures, and mechanisms of disease. This initial effort for "defining the riddle" will lay the foundation for future attempts to better define the range of PD and its variants, as has been done and implemented for other heterogeneous neurological syndromes, such as stroke and peripheral neuropathy. We strongly advocate for a more systematic and evidence-based integration of our diverse disciplines by looking at well-defined variants of the syndrome of PD. CONCLUSION: Accuracy in defining endophenotypes of "typical PD" across these different but interrelated disciplines will enable better definition of variants and their stratification in therapeutic trials, a prerequisite for breakthroughs in the era of precision medicine. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/genética , Síndrome , Biomarcadores , Predicción , Sistema Nervioso Central/patología
6.
Mov Disord ; 38(3): 378-384, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36645106

RESUMEN

Two recent, high-profile manuscripts reported negative results with two parallel approaches of passive immunization targeting α-synuclein in a population of patients with early Parkinson's disease (PD). These phase II studies failed to show a bona fide disease-modifying neuroprotective effect on PD progression, despite the evidence that these antibodies effectively bind native α-synuclein in human serum. Here, we discuss the possible reasons that could help explain the lack of clinical efficacy. In particular, we highlight (1) the wealth of evidence supporting the notion of α-synuclein as a valid therapeutic target; (2) the lack of evidence of target engagement in the aforementioned studies, especially of the elusive oligomeric species, the likely culprits in disease pathogenesis and/or its propagation; (3) the limitations, especially in terms of timing passive immunization, of preclinical models, where the same α-synuclein antibodies succeeded in mitigating disease manifestations; (4) the consideration of possibly intervening at an even earlier stage of disease in future trials; and (5) the multitude of strategies beyond passive immunization that could be used to combat α-synuclein-mediated neurodegeneration, if in the end the current approach is not fruitful. Overall, our perception is that converging developments in the field, among them novel bioassays and biomarkers, improved cellular and animal models and objective measurements of motor activities integrated into clinical trials, if further optimized, will gradually move the momentum of the field forward. This, to better test the concept of whether α-synuclein-targeting therapies can indeed deliver the "holy grail" of neuroprotection to the benefit of the PD community. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Animales , Humanos , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/inmunología , Anticuerpos/inmunología , Anticuerpos/uso terapéutico , Biomarcadores , Enfermedad de Parkinson/tratamiento farmacológico , Resultado del Tratamiento
7.
Acta Neuropathol Commun ; 11(1): 19, 2023 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-36691076

RESUMEN

We recently discovered that the expression of PRKN, a young-onset Parkinson disease-linked gene, confers redox homeostasis. To further examine the protective effects of parkin in an oxidative stress model, we first combined the loss of prkn with Sod2 haploinsufficiency in mice. Although adult prkn-/-//Sod2± animals did not develop dopamine cell loss in the S. nigra, they had more reactive oxidative species and a higher concentration of carbonylated proteins in the brain; bi-genic mice also showed a trend for more nitrotyrosinated proteins. Because these redox changes were seen in the cytosol rather than mitochondria, we next explored the thiol network in the context of PRKN expression. We detected a parkin deficiency-associated increase in the ratio of reduced glutathione (GSH) to oxidized glutathione (GSSG) in murine brain, PRKN-linked human cortex and several cell models. This shift resulted from enhanced recycling of GSSG back to GSH via upregulated glutathione reductase activity; it also correlated with altered activities of redox-sensitive enzymes in mitochondria isolated from mouse brain (e.g., aconitase-2; creatine kinase). Intriguingly, human parkin itself showed glutathione-recycling activity in vitro and in cells: For each GSSG dipeptide encountered, parkin regenerated one GSH molecule and was S-glutathionylated by the other (GSSG + P-SH [Formula: see text] GSH + P-S-SG), including at cysteines 59, 95 and 377. Moreover, parkin's S-glutathionylation was reversible by glutaredoxin activity. In summary, we found that PRKN gene expression contributes to the network of available thiols in the cell, including by parkin's participation in glutathione recycling, which involves a reversible, posttranslational modification at select cysteines. Further, parkin's impact on redox homeostasis in the cytosol can affect enzyme activities elsewhere, such as in mitochondria. We posit that antioxidant functions of parkin may explain many of its previously described, protective effects in vertebrates and invertebrates that are unrelated to E3 ligase activity.


Asunto(s)
Glutatión , Proteínas , Adulto , Ratones , Humanos , Animales , Disulfuro de Glutatión/metabolismo , Glutatión/metabolismo , Proteínas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Ubiquitina-Proteína Ligasas/genética , Antioxidantes , Cisteína/metabolismo , Encéfalo/metabolismo , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/metabolismo , Mamíferos/metabolismo
8.
NPJ Parkinsons Dis ; 8(1): 94, 2022 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-35906250

RESUMEN

Several recent publications described algorithms to identify subjects with Parkinson's disease (PD). In creating the "PREDIGT Score", we previously developed a hypothesis-driven, simple-to-use formula to potentially calculate the incidence of PD. Here, we tested its performance in the 'De Novo Parkinson Study' (DeNoPa) and 'Parkinson's Progression Marker Initiative' (PPMI); the latter included participants from the 'FOllow Up persons with Neurologic Disease' (FOUND) cohort. Baseline data from 563 newly diagnosed PD patients and 306 healthy control subjects were evaluated. Based on 13 variables, the original PREDIGT Score identified recently diagnosed PD patients in the DeNoPa, PPMI + FOUND and the pooled cohorts with area-under-the-curve (AUC) values of 0.88 (95% CI 0.83-0.92), 0.79 (95% CI 0.72-0.85), and 0.84 (95% CI 0.8-0.88), respectively. A simplified version (8 variables) generated AUC values of 0.92 (95% CI 0.89-0.95), 0.84 (95% CI 0.81-0.87), and 0.87 (0.84-0.89) in the DeNoPa, PPMI, and the pooled cohorts, respectively. In a two-step, screening-type approach, self-reported answers to a questionnaire (step 1) distinguished PD patients from controls with an AUC of 0.81 (95% CI 0.75-0.86). Adding a single, objective test (Step 2) further improved classification. Among seven biological markers explored, hyposmia was the most informative. The composite AUC value measured 0.9 (95% CI 0.88-0.91) in DeNoPa and 0.89 (95% CI 0.84-0.94) in PPMI. These results reveal a robust performance of the original PREDIGT Score to distinguish newly diagnosed PD patients from controls in two established cohorts. We also demonstrate the formula's potential applicability to enriching for PD subjects in a population screening-type approach.

9.
Bioinformatics ; 38(6): 1593-1599, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-34951624

RESUMEN

MOTIVATION: Bioinformatic tools capable of annotating, rapidly and reproducibly, large, targeted lipidomic datasets are limited. Specifically, few programs enable high-throughput peak assessment of liquid chromatography-electrospray ionization tandem mass spectrometry data acquired in either selected or multiple reaction monitoring modes. RESULTS: We present here Bayesian Annotations for Targeted Lipidomics, a Gaussian naïve Bayes classifier for targeted lipidomics that annotates peak identities according to eight features related to retention time, intensity, and peak shape. Lipid identification is achieved by modeling distributions of these eight input features across biological conditions and maximizing the joint posterior probabilities of all peak identities at a given transition. When applied to sphingolipid and glycerophosphocholine selected reaction monitoring datasets, we demonstrate over 95% of all peaks are rapidly and correctly identified. AVAILABILITY AND IMPLEMENTATION: BATL software is freely accessible online at https://complimet.ca/batl/ and is compatible with Safari, Firefox, Chrome and Edge. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Lipidómica , Programas Informáticos , Teorema de Bayes , Espectrometría de Masas , Cromatografía Liquida/métodos
10.
Arch Biochem Biophys ; 704: 108869, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33819447

RESUMEN

Parkinson's disease (PD) is one of the fastest-growing neurodegenerative disorders of increasing global prevalence. It represents the second most common movement disorder after tremor and the second most common neurodegenerative disorder after Alzheimer's disease. The incidence rate of idiopathic PD increases steadily with age, however, some variants of autosomal recessive inheritance are present with an early age-at-onset (ARPD). Approximately 50 percent of ARPD cases have been linked to bi-allelic mutations in genes encoding Parkin, DJ-1, and PINK1. Each protein has been implicated in maintaining proper mitochondrial function, which is particularly important for neuronal health. Aberrant post-translational modifications of these proteins may disrupt their cellular functions and thus contributing to the development of idiopathic PD. Some post-translational modifictions can be attributed to the dysregulation of potentially harmful reactive oxygen and nitrogen species inside the cell, which promote oxidative and nitrosative stress, respectively. Unlike oxidative modifications, the covalent modification by Nitric Oxide under nitrosative stress, leading to S-nitrosylation of Parkin, DJ-1; and PINK1, is less studied. Here, we review the available literature on S-nitrosylation of these three proteins, their implications in the pathogenesis of PD, and provide an overview of currently known, denitrosylating systems in eukaryotic cells.


Asunto(s)
Óxido Nítrico/metabolismo , Estrés Nitrosativo , Enfermedad de Parkinson/metabolismo , Proteína Desglicasa DJ-1/metabolismo , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Ubiquitina-Proteína Ligasas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Humanos , Mutación , Óxido Nítrico/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Proteína Desglicasa DJ-1/genética , Proteínas Quinasas/genética , Ubiquitina-Proteína Ligasas/genética
11.
Acta Neuropathol ; 141(5): 725-754, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33694021

RESUMEN

The mechanisms by which parkin protects the adult human brain from Parkinson disease remain incompletely understood. We hypothesized that parkin cysteines participate in redox reactions and that these are reflected in its posttranslational modifications. We found that in post mortem human brain, including in the Substantia nigra, parkin is largely insoluble after age 40 years; this transition is linked to its oxidation, such as at residues Cys95 and Cys253. In mice, oxidative stress induces posttranslational modifications of parkin cysteines that lower its solubility in vivo. Similarly, oxidation of recombinant parkin by hydrogen peroxide (H2O2) promotes its insolubility and aggregate formation, and in exchange leads to the reduction of H2O2. This thiol-based redox activity is diminished by parkin point mutants, e.g., p.C431F and p.G328E. In prkn-null mice, H2O2 levels are increased under oxidative stress conditions, such as acutely by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxin exposure or chronically due to a second, genetic hit; H2O2 levels are also significantly increased in parkin-deficient human brain. In dopamine toxicity studies, wild-type parkin, but not disease-linked mutants, protects human dopaminergic cells, in part through lowering H2O2. Parkin also neutralizes reactive, electrophilic dopamine metabolites via adduct formation, which occurs foremost at the primate-specific residue Cys95. Further, wild-type but not p.C95A-mutant parkin augments melanin formation in vitro. By probing sections of adult, human midbrain from control individuals with epitope-mapped, monoclonal antibodies, we found specific and robust parkin reactivity that co-localizes with neuromelanin pigment, frequently within LAMP-3/CD63+ lysosomes. We conclude that oxidative modifications of parkin cysteines are associated with protective outcomes, which include the reduction of H2O2, conjugation of reactive dopamine metabolites, sequestration of radicals within insoluble aggregates, and increased melanin formation. The loss of these complementary redox effects may augment oxidative stress during ageing in dopamine-producing cells of mutant PRKN allele carriers, thereby enhancing the risk of Parkinson's-linked neurodegeneration.


Asunto(s)
Envejecimiento/metabolismo , Dopamina/metabolismo , Mesencéfalo/metabolismo , Degeneración Nerviosa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/patología , Animales , Niño , Preescolar , Femenino , Humanos , Masculino , Mesencéfalo/patología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Degeneración Nerviosa/patología , Oxidación-Reducción , Adulto Joven
12.
Sci Signal ; 13(662)2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33323410

RESUMEN

The prevalence, presentation, and progression of Alzheimer's disease (AD) differ between men and women, although ß-amyloid (Aß) deposition is a pathological hallmark of AD in both sexes. Aß-induced activation of the neuronal glutamate receptor mGluR5 is linked to AD progression. However, we found that mGluR5 exhibits distinct sex-dependent profiles. Specifically, mGluR5 isolated from male mouse cortical and hippocampal tissues bound with high affinity to Aß oligomers, whereas mGluR5 from female mice exhibited no such affinity. This sex-selective Aß-mGluR5 interaction did not appear to depend on estrogen, but rather Aß interaction with cellular prion protein (PrPC), which was detected only in male mouse brain homogenates. The ternary complex between mGluR5, Aß oligomers, and PrPC was essential to elicit mGluR5-dependent pathological suppression of autophagy in primary neuronal cultures. Pharmacological inhibition of mGluR5 reactivated autophagy, mitigated Aß pathology, and reversed cognitive decline in male APPswe/PS1ΔE9 mice, but not in their female counterparts. Aß oligomers also bound with high affinity to human mGluR5 isolated from postmortem donor male cortical brain tissue, but not that from female samples, suggesting that this mechanism may be relevant to patients. Our findings indicate that mGluR5 does not contribute to Aß pathology in females, highlighting the complexity of mGluR5 pharmacology and Aß signaling that supports the need for sex-specific stratification in clinical trials assessing AD therapeutics.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Corteza Cerebral/metabolismo , Multimerización de Proteína , Receptor del Glutamato Metabotropico 5/metabolismo , Caracteres Sexuales , Transducción de Señal , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Receptor del Glutamato Metabotropico 5/genética
13.
Mov Disord ; 35(11): 1922-1932, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33053225

RESUMEN

On May 26, 2020, Dr. Oleh Hornykiewicz died at the age of 93 years. His twin discoveries in the early 1960s of dopamine deficiency in the brains of subjects with Parkinson's disease and the amelioration of patients' symptoms by levodopa therapy represent milestone events in the history of medicine. These breakthroughs enabled much-needed relief for millions of patients suffering from neurological disorders every year and have given rise to the field of dopamine signaling in the regulation of complex behaviors in primates. What did Dr. Hornykiewicz, who was actively engaged in research until shortly before his 91st birthday, wish to pass on to younger scientists? What were his thoughts regarding the elusive cause of Parkinson's disease? How did he wish to be remembered? Here, the authors, one a former student and the other an admired colleague, recall messages conveyed by Dr. Hornykiewicz in public lectures; they also share the content of conversations and letters exchanged with him since 2004, as he began to reflect on his legacy. Through Dr. Hornykiewicz's own words and writings, the picture emerges of an extraordinarily committed scientist, who was exemplary in his professional integrity, who knew how to deploy a gallous sense of humor, who displayed little patience for physicians offering advice, and who kept any sense of pride over his monumental contributions private. When asked at the age of 91 years about the secrets of his long and fulfilled career in neuroscience, he identified himself as "a mad scientist.…I am someone who continuously fantasizes. I am someone who chases fantastical ideas and who keeps on dreaming…", and as a man who was supported by the loving companionship of his wife, Christine. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Neurociencias , Enfermedad de Parkinson , Médicos , Dopamina , Historia del Siglo XX , Humanos , Levodopa , Masculino , Enfermedad de Parkinson/tratamiento farmacológico
14.
Clin Invest Med ; 43(2): E1-13, 2020 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-32593271

RESUMEN

PURPOSE: Integrated MD/PhD programs are relatively new in Canada and represent a platform to train the next generation of clinician-scientists. However, MD/PhD programs vary substantially by structure, funding and mentorship opportunities, and there exists a paucity of data on the overall students' successes and challenges. The purpose of this study is to assess objective and subjective metrics of the MD/PhD Program at the University of Ottawa. METHODS: Students in all years of the program were invited to complete a 58- question survey, and the resulting data were analyzed by descriptive statistics. RESULTS: Our survey had an 88.5% (23/26) participation rate. The program has been gaining interest and the number of applications increased by 178% between 2013 and 2018. Tuition support was considered an essential element in accepting the admission offer, as 47.8% of students would have declined admission without full tuition coverage. The MD/PhD students were heavily engaged in scholarly activities, with an average of 8.3 presentations/ publications per respondent. Respondents indicated low satisfaction with formal career planning advice (28.6% satisfied/very satisfied) and program transition guidance (22.2%). When delivered informally by peers, both career planning advice and program transition guidance were experienced as more satisfying (65.2% and 63.6%, respectively). Only 34.8% of survey respondents identified as female, highlighting the challenge of achieving diversity in clinician-scientist training programs. CONCLUSION: Our report contributes to the body of knowledge on concrete obstacles experienced by students within MD/PhD programs and key areas that can be improved upon-locally, provincially and nationally-to further advance student success.


Asunto(s)
Investigación Biomédica , Médicos , Canadá , Niño , Femenino , Humanos , Mentores , Encuestas y Cuestionarios
15.
Microbes Infect ; 22(1): 40-45, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31430539

RESUMEN

Receptor interacting protein kinase 3 (Ripk3) is a signal relay protein involved in initiation of programmed cell death (necroptosis) and modulation of inflammasome activation. While caspase 1 and 11 are pro-inflammatory caspases responsible for unleashing inflammation and cell death by enzymatic activation of the executioners of inflammation and cell death (pyroptosis). Upon Salmonella infection, the host mounts a pro-inflammatory response which require Ripk3 and Caspase1/11. Here we show that bone marrow derived macrophages with combined deficiency of Ripk3 and Casp1/11 are highly resistant to Salmonella induced cell death, and that these macrophages show an anti-inflammatory cytokine profile. We confirm what was previously known that mice deficient in Casp1/11 have impaired ability to control Salmonella burden, and that the absence of Ripk3 alone does not influence the innate immune responses to Salmonella infection. However, we describe a synergistic role of Ripk3 and Casp1/11 in regulating Salmonella in vivo burden and that Ripk3-dependent host protection in the absence of Casp1/11 is evident during infection by sifA-expressing Salmonella. In summary, we show that the Ripk3 protection to Salmonella infection is obscured by presence of Caspase 1/11 and that the Ripk3-dependent protection requires a phagosome-bound Salmonella.


Asunto(s)
Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Inflamasomas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Carga Bacteriana , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Caspasa 1/deficiencia , Caspasas Iniciadoras/deficiencia , Muerte Celular , Citocinas/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Salmonella/fisiología , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología
16.
Sci Transl Med ; 11(511)2019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31554740

RESUMEN

Variants in the leucine-rich repeat kinase-2 (LRRK2) gene are associated with Parkinson's disease, leprosy, and Crohn's disease, three disorders with inflammation as an important component. Because of its high expression in granulocytes and CD68-positive cells, LRRK2 may have a function in innate immunity. We tested this hypothesis in two ways. First, adult mice were intravenously inoculated with Salmonella typhimurium, resulting in sepsis. Second, newborn mouse pups were intranasally infected with reovirus (serotype 3 Dearing), which induced encephalitis. In both mouse models, wild-type Lrrk2 expression was protective and showed a sex effect, with female Lrrk2-deficient animals not controlling infection as well as males. Mice expressing Lrrk2 carrying the Parkinson's disease-linked p.G2019S mutation controlled infection better, with reduced bacterial growth and longer animal survival during sepsis. This gain-of-function effect conferred by the p.G2019S mutation was mediated by myeloid cells and was abolished in animals expressing a kinase-dead Lrrk2 variant, p.D1994S. Mouse pups with reovirus-induced encephalitis that expressed the p.G2019S Lrrk2 mutation showed increased mortality despite lower viral titers. The p.G2019S mutant Lrrk2 augmented immune cell chemotaxis and generated more reactive oxygen species during virulent infection. Reovirus-infected brains from mice expressing the p.G2019S mutant Lrrk2 contained higher concentrations of α-synuclein. Animals expressing one or two p.D1994S Lrrk2 alleles showed lower mortality from reovirus-induced encephalitis. Thus, Lrrk2 alleles may alter the course of microbial infections by modulating inflammation, and this may be dependent on the sex and genotype of the host as well as the type of pathogen.


Asunto(s)
Alelos , Infecciones/enzimología , Infecciones/genética , Inflamación/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Caracteres Sexuales , Animales , Encéfalo/patología , Encéfalo/virología , Quimiotaxis , Encefalitis/virología , Femenino , Humanos , Infecciones/inmunología , Infecciones/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/deficiencia , Leucocitos/enzimología , Masculino , Ratones Endogámicos C57BL , Mutación/genética , Especies Reactivas de Oxígeno/metabolismo , Reoviridae/fisiología , Salmonella typhimurium/crecimiento & desarrollo , Sepsis/microbiología , Análisis de Supervivencia , alfa-Sinucleína/metabolismo
17.
J Lipid Res ; 60(1): 200-211, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30413651

RESUMEN

Cerebrosides, including glucosylceramides (GlcCers) and galactosylceramides (GalCers), are important membrane components of animal cells with deficiencies resulting in devastating lysosomal storage disorders. Their quantification is essential for disease diagnosis and a better understanding of disease mechanisms. The simultaneous quantification of GlcCer and GalCer isomers is, however, particularly challenging due to their virtually identical structures. To address this challenge, we developed a new LC/MS-based method using differential ion mobility spectrometry (DMS) capable of rapidly and reproducibly separating and quantifying isomeric cerebrosides in a single run. We show that this LC/ESI/DMS/MS/MS method exhibits robust quantitative performance within an analyte concentration range of 2.8-355 nM. We further report the simultaneous quantification of nine GlcCers (16:0, 18:0, 20:0, 22:0, 23:0, 24:1, 24:0, 25:0, and 26:0) and five GalCers (16:0, 22:0, 23:0, 24:1, and 24:0) molecular species in human plasma, as well as six GalCers (18:0, 22:0, 23:0, 24:1, 24:0 and 25:0) and two GlcCers (24:1 and 24:0) in human cerebrospinal fluid. Our method expands the potential of DMS technology in the field of glycosphingolipid analysis for both biomarker discovery and drug screening by enabling the unambiguous assignment and quantification of cerebroside lipid species in biological samples.


Asunto(s)
Cerebrósidos/química , Cerebrósidos/aislamiento & purificación , Cromatografía Liquida/métodos , Espectrometría de Movilidad Iónica/métodos , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masas en Tándem/métodos , Cerebrósidos/sangre , Cerebrósidos/líquido cefalorraquídeo , Cromatografía Liquida/normas , Femenino , Humanos , Espectrometría de Movilidad Iónica/normas , Isomerismo , Persona de Mediana Edad , Estándares de Referencia , Espectrometría de Masa por Ionización de Electrospray/normas , Espectrometría de Masas en Tándem/normas , Factores de Tiempo
18.
Proc Natl Acad Sci U S A ; 115(22): E5164-E5173, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29760073

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) has been implicated in both familial and sporadic Parkinson's disease (PD), yet its pathogenic role remains unclear. A previous screen in Drosophila identified Scar/WAVE (Wiskott-Aldrich syndrome protein-family verproline) proteins as potential genetic interactors of LRRK2 Here, we provide evidence that LRRK2 modulates the phagocytic response of myeloid cells via specific modulation of the actin-cytoskeletal regulator, WAVE2. We demonstrate that macrophages and microglia from LRRK2-G2019S PD patients and mice display a WAVE2-mediated increase in phagocytic response, respectively. Lrrk2 loss results in the opposite effect. LRRK2 binds and phosphorylates Wave2 at Thr470, stabilizing and preventing its proteasomal degradation. Finally, we show that Wave2 also mediates Lrrk2-G2019S-induced dopaminergic neuronal death in both macrophage-midbrain cocultures and in vivo. Taken together, a LRRK2-WAVE2 pathway, which modulates the phagocytic response in mice and human leukocytes, may define an important role for altered immune function in PD.


Asunto(s)
Citofagocitosis/fisiología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Células Mieloides/citología , Enfermedad de Parkinson/fisiopatología , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Línea Celular , Drosophila , Humanos , Ratones , Microglía , Células Mieloides/fisiología , Transducción de Señal/fisiología
19.
J Neuroinflammation ; 14(1): 164, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28821274

RESUMEN

BACKGROUND: Efforts to identify fluid biomarkers of Parkinson's disease (PD) have intensified in the last decade. As the role of inflammation in PD pathophysiology becomes increasingly recognized, investigators aim to define inflammatory signatures to help elucidate underlying mechanisms of disease pathogenesis and aid in identification of patients with inflammatory endophenotypes that could benefit from immunomodulatory interventions. However, discordant results in the literature and a lack of information regarding the stability of inflammatory factors over a 24-h period have hampered progress. METHODS: Here, we measured inflammatory proteins in serum and CSF of a small cohort of PD (n = 12) and age-matched healthy control (HC) subjects (n = 6) at 11 time points across 24 h to (1) identify potential diurnal variation, (2) reveal differences in PD vs HC, and (3) to correlate with CSF levels of amyloid ß (Aß) and α-synuclein in an effort to generate data-driven hypotheses regarding candidate biomarkers of PD. RESULTS: Despite significant variability in other factors, a repeated measures two-way analysis of variance by time and disease state for each analyte revealed that serum IFNγ, TNF, and neutrophil gelatinase-associated lipocalin (NGAL) were stable across 24 h and different between HC and PD. Regression analysis revealed that C-reactive protein (CRP) was the only factor with a strong linear relationship between CSF and serum. PD and HC subjects showed significantly different relationships between CSF Aß proteins and α-synuclein and specific inflammatory factors, and CSF IFNγ and serum IL-8 positively correlated with clinical measures of PD. Finally, linear discriminant analysis revealed that serum TNF and CSF α-synuclein discriminated between PD and HC with a minimum of 82% sensitivity and 83% specificity. CONCLUSIONS: Our findings identify a panel of inflammatory factors in serum and CSF that can be reliably measured, distinguish between PD and HC, and monitor inflammation as disease progresses or in response to interventional therapies. This panel may aid in generating hypotheses and feasible experimental designs towards identifying biomarkers of neurodegenerative disease by focusing on analytes that remain stable regardless of time of sample collection.


Asunto(s)
Mediadores de Inflamación/sangre , Mediadores de Inflamación/líquido cefalorraquídeo , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/líquido cefalorraquídeo , alfa-Sinucleína/sangre , alfa-Sinucleína/líquido cefalorraquídeo , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Femenino , Humanos , Masculino , Enfermedad de Parkinson/diagnóstico , Índice de Severidad de la Enfermedad
20.
J Neural Transm (Vienna) ; 124(6): 721-738, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28477284

RESUMEN

Braak and Del Tredici have proposed that typical Parkinson disease (PD) has its origins in the olfactory bulb and gastrointestinal tract. However, the role of the olfactory system has insufficiently been explored in the pathogeneses of PD and Alzheimer disease (AD) in laboratory models. Here, we demonstrate applications of a new method to process mouse heads for microscopy by sectioning, mounting, and staining whole skulls ('holocranohistochemistry'). This technique permits the visualization of the olfactory system from the nasal cavity to mitral cells and dopamine-producing interneurons of glomeruli in the olfactory bulb. We applied this method to two specific goals: first, to visualize PD- and AD-linked gene expression in the olfactory system, where we detected abundant, endogenous α-synuclein and tau expression in the olfactory epithelium. Furthermore, we observed amyloid-ß plaques and proteinase-K-resistant α-synuclein species, respectively, in cranial nerve-I of APP- and human SNCA-over-expressing mice. The second application of the technique was to the modeling of gene-environment interactions in the nasal cavity of mice. We tracked the infection of a neurotropic respiratory-enteric-orphan virus from the nose pad into cranial nerves-I (and -V) and monitored the ensuing brain infection. Given its abundance in the olfactory epithelia, we questioned whether α-synuclein played a role in innate host defenses to modify the outcome of infections. Indeed, Snca-null mice were more likely to succumb to viral encephalitis versus their wild-type littermates. Moreover, using a bacterial sepsis model, Snca-null mice were less able to control infection after intravenous inoculation with Salmonella typhimurium. Together, holocranohistochemistry enabled new discoveries related to α-synuclein expression and its function in mice. Future studies will address: the role of Mapt and mutant SNCA alleles in infection paradigms; the contribution of xenobiotics in the initiation of idiopathic PD; and the safety to the host when systemically targeting α-synuclein by immunotherapy.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/virología , Encefalitis Viral/virología , Mucosa Olfatoria/anatomía & histología , Mucosa Olfatoria/metabolismo , Infecciones por Reoviridae/virología , alfa-Sinucleína/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Modelos Animales de Enfermedad , Encefalitis Viral/inmunología , Encefalitis Viral/mortalidad , Encefalitis Viral/patología , Femenino , Cabeza , Humanos , Inmunohistoquímica , Masculino , Orthoreovirus Mamífero 3 , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/anatomía & histología , Vías Nerviosas/diagnóstico por imagen , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Mucosa Olfatoria/patología , Neuronas Receptoras Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/virología , Infecciones por Reoviridae/inmunología , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/patología , Salmonella typhimurium , Conservación de Tejido/métodos , alfa-Sinucleína/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA