Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 90
Filtrer
1.
Neural Regen Res ; 20(2): 574-586, 2025 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-38819068

RÉSUMÉ

JOURNAL/nrgr/04.03/01300535-202502000-00033/figure1/v/2024-05-28T214302Z/r/image-tiff There is a need to develop interventions to slow or reverse the degeneration of dopamine neurons in Parkinson's disease after diagnosis. Given that preclinical and clinical studies suggest benefits of dietary n-3 polyunsaturated fatty acids, such as docosahexaenoic acid, and exercise in Parkinson's disease, we investigated whether both could synergistically interact to induce recovery of the dopaminergic pathway. First, mice received a unilateral stereotactic injection of 6-hydroxydopamine into the striatum to establish an animal model of nigrostriatal denervation. Four weeks after lesion, animals were fed a docosahexaenoic acid-enriched or a control diet for the next 8 weeks. During this period, the animals had access to a running wheel, which they could use or not. Docosahexaenoic acid treatment, voluntary exercise, or the combination of both had no effect on (i) distance traveled in the open field test, (ii) the percentage of contraversive rotations in the apomorphine-induction test or (iii) the number of tyrosine-hydroxylase-positive cells in the substantia nigra pars compacta. However, the docosahexaenoic acid diet increased the number of tyrosine-hydroxylase-positive terminals and induced a rise in dopamine concentrations in the lesioned striatum. Compared to docosahexaenoic acid treatment or exercise alone, the combination of docosahexaenoic acid and exercise (i) improved forelimb balance in the stepping test, (ii) decreased the striatal DOPAC/dopamine ratio and (iii) led to increased dopamine transporter levels in the lesioned striatum. The present results suggest that the combination of exercise and docosahexaenoic acid may act synergistically in the striatum of mice with a unilateral lesion of the dopaminergic system and provide support for clinical trials combining nutrition and physical exercise in the treatment of Parkinson's disease.

2.
Neurobiol Dis ; 198: 106542, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38810948

RÉSUMÉ

A number of post-mortem studies conducted in transplanted Huntington's disease (HD) patients from various trials have reported the presence of pathological and misfolded proteins, in particular mutant huntingtin (mHtt) and phosphorylated tau neuropil threads, in the healthy grafted tissue. Here, we extended these observations with histological analysis of post-mortem tissue from three additional HD patients who had received similar striatal allografts from the fetal tissue transplantation trial conducted in Los Angeles in 1998. Immunohistochemical staining was performed using anti-mHtt antibodies, EM48 and MW7, as well as anti-hyperphosphorylated tau antibodies, AT8 and CP13. Immunofluorescence was used to assess the colocalization of EM48+ mHtt aggregates with the neuronal marker MAP2 and/or the extracellular matrix protein phosphacan in both the host and grafts. We confirmed the presence of mHtt aggregates within grafts of all three cases as well as tau neuropil threads in the grafts of two of the three transplanted HD patients. Phosphorylated tau was also variably expressed in the host cerebral cortex of all three subjects. While mHtt inclusions were present within neurons (immunofluorescence co-localization of MAP2 and EM48) as well as within the extracellular matrix of the host (immunofluorescence co-localization of phosphacan and EM48), their localization was limited to the extracellular matrix in the grafted tissue. This study corroborates previous findings that both mHtt and tau pathology can be found in the host and grafts of HD patients years post-grafting.


Sujet(s)
Protéine huntingtine , Maladie de Huntington , Neurones , Protéines tau , Humains , Maladie de Huntington/anatomopathologie , Maladie de Huntington/métabolisme , Maladie de Huntington/génétique , Protéines tau/métabolisme , Protéines tau/génétique , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Mâle , Adulte d'âge moyen , Femelle , Neurones/métabolisme , Neurones/anatomopathologie , Adulte , Transplantation de tissu foetal/méthodes , Sujet âgé , Transplantation de tissu cérébral/méthodes
3.
J Neurol ; 271(6): 3682-3689, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38607433

RÉSUMÉ

Our limited understanding of complex neurodegenerative disorders has held us back on the development of efficient therapies. While several approaches are currently being considered, it is still unclear what will be most successful. Among the latest and more novel ideas, the concept of blood or plasma transfusion from young healthy donors to diseased patients is gaining momentum and attracting attention beyond the scientific arena. While young or healthy blood is enriched with protective and restorative components, blood from older subjects may accumulate neurotoxic agents or be impoverished of beneficial factors. In this commentary, we present an overview of the compelling evidence collected in various animal models of brain diseases (e.g., Alzheimer, Parkinson, Huntington) to the actual clinical trials that have been conducted to test the validity of blood-related treatments in neurodegenerative diseases and argue in favor of such approach.


Sujet(s)
Maladies neurodégénératives , Humains , Maladies neurodégénératives/thérapie , Animaux , Encéphale/imagerie diagnostique , Transfusion sanguine/méthodes
6.
Neurobiol Dis ; 190: 106376, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38092268

RÉSUMÉ

In Huntington disease (HD), the mutant huntingtin (mtHTT) protein is the principal cause of pathological changes that initiate primarily along the cortico-striatal axis. mtHTT is ubiquitously expressed and there is, accordingly, growing recognition that HD is a systemic disorder with functional interplay between the brain and the periphery. We have developed a monoclonal antibody, C6-17, targeting an exposed region of HTT near the aa586 Caspase 6 cleavage site. As recently published, mAB C6-17 can block cell-to-cell propagation of mtHTT in vitro. In order to reduce the burden of the mutant protein in vivo, we queried whether extracellular mtHTT could be therapeutically targeted in YAC128 HD mice. In a series of proof of concept experiments, we found that systemic mAB C6-17 treatment resulted in the distribution of the mAB C6-17 to peripheral and CNS tissues and led to the reduction of HTT protein levels. Compared to CTRL mAB or vehicle treated mice, the mAB C6-17 treated YAC128 animals showed improved body weight and motor behaviors, a delayed progression in motor deficits and reduced striatal EM48 immunoreactivity. These results provide the first proof of concept for the feasibility and therapeutic efficacy of an antibody-based anti-HTT passive immunization approach and suggest this modality as a potential new HD treatment strategy.


Sujet(s)
Maladie de Huntington , Souris , Animaux , Maladie de Huntington/métabolisme , Souris transgéniques , Protéines de tissu nerveux/métabolisme , Protéines nucléaires/métabolisme , Protéine huntingtine/génétique , Immunothérapie , Modèles animaux de maladie humaine , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Évolution de la maladie
7.
Mol Psychiatry ; 28(10): 4070-4083, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37749233

RÉSUMÉ

Developing effective treatments for patients with Huntington's disease (HD)-a neurodegenerative disorder characterized by severe cognitive, motor and psychiatric impairments-is proving extremely challenging. While the monogenic nature of this condition enables to identify individuals at risk, robust biomarkers would still be extremely valuable to help diagnose disease onset and progression, and especially to confirm treatment efficacy. If measurements of cerebrospinal fluid neurofilament levels, for example, have demonstrated use in recent clinical trials, other proteins may prove equal, if not greater, relevance as biomarkers. In fact, proteins such as tau could specifically be used to detect/predict cognitive affectations. We have herein reviewed the literature pertaining to the association between tau levels and cognitive states, zooming in on Alzheimer's disease, Parkinson's disease and traumatic brain injury in which imaging, cerebrospinal fluid, and blood samples have been interrogated or used to unveil a strong association between tau and cognition. Collectively, these areas of research have accrued compelling evidence to suggest tau-related measurements as both diagnostic and prognostic tools for clinical practice. The abundance of information retrieved in this niche of study has laid the groundwork for further understanding whether tau-related biomarkers may be applied to HD and guide future investigations to better understand and treat this disease.


Sujet(s)
Maladie d'Alzheimer , Maladie de Huntington , Humains , Maladie de Huntington/diagnostic , Maladie de Huntington/métabolisme , Protéines tau/métabolisme , Maladie d'Alzheimer/diagnostic , Marqueurs biologiques/liquide cérébrospinal , Cognition , Peptides bêta-amyloïdes
9.
Neurobiol Dis ; 180: 106091, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36967065

RÉSUMÉ

In a previous study, we have shown that parabiotic coupling of a knock-in mouse model (zQ175) of Huntington's disease (HD) to wild-type (WT) littermates resulted in a worsening of the normal phenotype as seen by detection of mutant huntingtin protein (mHTT) aggregates within peripheral organs and the cerebral cortex as well as vascular abnormalities in WT mice. In contrast, parabiosis improved disease features in the zQ175 mice such as reduction of mHTT aggregate number in the liver and cortex, decrease in blood-brain barrier (BBB) permeability and attenuation of mitochondrial impairments. While the shared circulation mediated these effects, no specific factor was identified. To better understand which blood elements were involved in the aforementioned changes, WT and zQ175 mice underwent parabiotic surgery prior to exposing one of the paired animals to irradiation. The irradiation procedure successfully eliminated the hematopoietic niche followed by repopulation with cells originating from the non-irradiated parabiont, as measured by the quantification of mHTT levels in peripheral blood mononuclear cells. Although irradiation of the WT parabiont, causing the loss of healthy hematopoietic cells, did lead to a few alterations in mitochondrial function in the muscle (TOM40 levels), and increased neuroinflammation in the striatum (GFAP levels), most of the changes observed were likely attributable to the irradiation procedure itself (e.g. mHTT aggregates in cortex and liver; cellular stress in peripheral organs). However, factors such as mHTT aggregation in the brain and periphery, and BBB leakage, which were improved in zQ175 mice when paired to WT littermates in the previous parabiosis experiment, were unaffected by perturbation of the hematopoietic niche. It would therefore appear that cells of the hematopoietic stem cell niche are largely uninvolved in the beneficial effects of parabiosis.


Sujet(s)
Maladie de Huntington , Souris , Animaux , Souris transgéniques , Maladie de Huntington/génétique , Agranulocytes/métabolisme , Modèles animaux de maladie humaine , Phénotype , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme
10.
J Huntingtons Dis ; 12(1): 15-29, 2023.
Article de Anglais | MEDLINE | ID: mdl-36806513

RÉSUMÉ

There is increasing evidence for the presence of pathological forms of tau in tissues of both Huntington's disease (HD) patients and animal models of this condition. While cumulative studies of the past decade have led to the proposition that this disorder could also be considered a tauopathy, the implications of tau in cellular toxicity and consequent behavioral impairments are largely unknown. In fact, recent animal work has challenged the contributory role of tau in HD pathogenesis/pathophysiology. This review presents the supporting and opposing arguments for the involvement of tau in HD, highlighting the discrepancies that have emerged. Reflecting on what is known in other tauopathies, the putative mechanisms through which tau could initiate and/or contribute to pathology are discussed, shedding light on the future research directions that could be considered to confirm, or rule out, the clinical relevance of tau in HD.


Sujet(s)
Maladie de Huntington , Tauopathies , Animaux , Protéines tau , Maladie de Huntington/anatomopathologie , Tauopathies/anatomopathologie , Modèles animaux de maladie humaine
11.
Cell Mol Life Sci ; 80(2): 45, 2023 Jan 18.
Article de Anglais | MEDLINE | ID: mdl-36651994

RÉSUMÉ

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder resulting from a CAG expansion in the huntingtin (HTT) gene, which leads to the production and accumulation of mutant huntingtin (mHTT). While primarily considered a disorder of the central nervous system, multiple changes have been described to occur throughout the body, including activation of the immune system. In other neurodegenerative disorders, activation of the immune system has been shown to include the production of antibodies against disease-associated pathological proteins. However, the existence of mHTT-targeted antibodies has never been reported. In this study, we assessed the presence and titer of antibodies recognizing HTT/mHTT in patients with HD (n = 66) and age- and gender-matched healthy controls (n = 66) using a combination of Western blotting and ELISA. Together, these analyses revealed that antibodies capable of recognizing HTT/mHTT were detectable in the plasma samples of all participants, including healthy controls. When antibody levels were monitored at different disease stages, it was observed that antibodies against full-length mHTT were highest in patients with severe disease while antibodies against HTTExon1 were elevated in patients with mild disease. Combined, these results suggest that antibodies detecting different forms of mHTT peak at different disease stages.


Sujet(s)
Maladie de Huntington , Maladies neurodégénératives , Humains , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Maladie de Huntington/métabolisme , Anticorps
12.
Mol Neurodegener ; 17(1): 38, 2022 05 31.
Article de Anglais | MEDLINE | ID: mdl-35637520

RÉSUMÉ

Drug-refractory forms of neurological diseases could find their next breakthrough therapy in non-pharmacological approaches to brain repair. Lentini et al. present the potential of in situ brain regeneration to address neurodegeneration in the epileptic brain.


Sujet(s)
Lésions encéphaliques , Encéphale , Humains
13.
PLoS Biol ; 20(3): e3001578, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35263320

RÉSUMÉ

Neurodegenerative disorders refer to a group of diseases commonly associated with abnormal protein accumulation and aggregation in the central nervous system. However, the exact role of protein aggregation in the pathophysiology of these disorders remains unclear. This gap in knowledge is due to the lack of experimental models that allow for the spatiotemporal control of protein aggregation, and the investigation of early dynamic events associated with inclusion formation. Here, we report on the development of a light-inducible protein aggregation (LIPA) system that enables spatiotemporal control of α-synuclein (α-syn) aggregation into insoluble deposits called Lewy bodies (LBs), the pathological hallmark of Parkinson disease (PD) and other proteinopathies. We demonstrate that LIPA-α-syn inclusions mimic key biochemical, biophysical, and ultrastructural features of authentic LBs observed in PD-diseased brains. In vivo, LIPA-α-syn aggregates compromise nigrostriatal transmission, induce neurodegeneration and PD-like motor impairments. Collectively, our findings provide a new tool for the generation, visualization, and dissection of the role of α-syn aggregation in PD.


Sujet(s)
Maladie de Parkinson , alpha-Synucléine , Analyse de regroupements , Humains , Corps de Lewy/métabolisme , Corps de Lewy/anatomopathologie , Maladie de Parkinson/métabolisme , Agrégats de protéines , alpha-Synucléine/métabolisme
14.
Mol Ther ; 30(4): 1500-1522, 2022 04 06.
Article de Anglais | MEDLINE | ID: mdl-35051614

RÉSUMÉ

Huntington's disease is classically described as a neurodegenerative disorder of monogenic aetiology. The disease is characterized by an abnormal polyglutamine expansion in the huntingtin gene, which drives the toxicity of the mutated form of the protein. However, accumulation of the microtubule-associated protein tau, which is involved in a number of neurological disorders, has also been observed in patients with Huntington's disease. In order to unravel the contribution of tau hyperphosphorylation to hallmark features of Huntington's disease, we administered weekly intraperitoneal injections of the anti-tau pS202 CP13 monoclonal antibody to zQ175 mice and characterized the resulting behavioral and biochemical changes. After 12 weeks of treatment, motor impairments, cognitive performance and general health were improved in zQ175 mice along with a significant reduction in hippocampal pS202 tau levels. Despite the lack of effect of CP13 on neuronal markers associated with Huntington's disease pathology, tau-targeting enzymes and gliosis, CP13 was shown to directly impact mutant huntingtin aggregation such that brain levels of amyloid fibrils and huntingtin oligomers were decreased, while larger huntingtin protein aggregates were increased. Investigation of CP13 treatment of Huntington's disease patient-derived induced pluripotent stem cells (iPSCs) revealed a reduction in pS202 levels in differentiated cortical neurons and a rescue of neurite length. Collectively, these findings suggest that attenuating tau pathology could mitigate behavioral and molecular hallmarks associated with Huntington's disease.


Sujet(s)
Maladie de Huntington , Cellules souches pluripotentes induites , Animaux , Encéphale/métabolisme , Modèles animaux de maladie humaine , Humains , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/métabolisme , Maladie de Huntington/thérapie , Immunisation passive , Cellules souches pluripotentes induites/métabolisme , Souris , Neurones/métabolisme
15.
Mol Psychiatry ; 27(1): 269-280, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34711942

RÉSUMÉ

If theories postulating that pathological proteins associated with neurodegenerative disorders behave similarly to prions were initially viewed with reluctance, it is now well-accepted that this occurs in several disease contexts. Notably, it has been reported that protein misfolding and subsequent prion-like properties can actively participate in neurodegenerative disorders. While this has been demonstrated in multiple cellular and animal model systems related to Alzheimer's and Parkinson's diseases, the prion-like properties of the mutant huntingtin protein (mHTT), associated with Huntington's disease (HD), have only recently been considered to play a role in this pathology, a concept our research group has contributed to extensively. In this review, we summarize the last few years of in vivo research in the field and speculate on the relationship between prion-like events and human HD. By interpreting observations primarily collected in in vivo models, our discussion will aim to discriminate which experimental factors contribute to the most efficient types of prion-like activities of mHTT and which routes of propagation may be more relevant to the human condition. A look back at nearly a decade of experimentation will inform future research and whether therapeutic strategies may emerge from this new knowledge.


Sujet(s)
Maladie de Huntington , Maladies neurodégénératives , Prions , Animaux , Modèles animaux de maladie humaine , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/métabolisme , Maladies neurodégénératives/métabolisme , Prions/génétique , Prions/métabolisme
16.
Cells ; 10(11)2021 10 30.
Article de Anglais | MEDLINE | ID: mdl-34831178

RÉSUMÉ

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in childhood. Recently, we demonstrated the overexpression of both DNA methyltransferase 3A (DNMT3A) and 3B (DNMT3B) in RMS tumour biopsies and cell lines compared to normal skeletal muscle. Radiotherapy may often fail due to the abnormal expression of some molecules able to drive resistance mechanisms. The aim of this study was to analyse the involvement of DNMT3A and DNMT3B in radioresistance in RMS. RNA interference experiments against DNMT3A/3B were performed in embryonal RMS cells, upon ionizing radiation (IR) exposure and the effects of the combined treatment on RMS cells were analysed. DNMT3A and DNMT3B knocking down increased the sensitivity of RMS cells to IR, as indicated by the drastic decrease of colony formation ability. Interestingly, DNMT3A/3B act in two different ways: DNMT3A silencing triggers the cellular senescence program by up-regulating p16 and p21, whilst DNMT3B depletion induces significant DNA damage and impairs the DNA repair machinery (ATM, DNA-PKcs and Rad51 reduction). Our findings demonstrate for the first time that DNMT3A and DNMT3B overexpression may contribute to radiotherapy failure, and their inhibition might be a promising radiosensitizing strategy, mainly in the treatment of patients with metastatic or recurrent RMS tumours.


Sujet(s)
DNA (cytosine-5-)-methyltransferase/métabolisme , DNA methyltransferase 3A/métabolisme , Radiotolérance , Rhabdomyosarcome embryonnaire/radiothérapie , Cycle cellulaire/effets des radiations , Différenciation cellulaire/effets des radiations , Lignée cellulaire tumorale , Prolifération cellulaire/effets des radiations , Vieillissement de la cellule/effets des radiations , Clones cellulaires , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , DNA (cytosine-5-)-methyltransferase/génétique , Altération de l'ADN , DNA methyltransferase 3A/génétique , Activation enzymatique/effets des radiations , Régulation de l'expression des gènes tumoraux , Extinction de l'expression des gènes/effets des radiations , Histone/métabolisme , Humains , Développement musculaire/effets des radiations , Radiotolérance/génétique , Rayonnement ionisant , Rhabdomyosarcome embryonnaire/génétique , Régulation positive/génétique , p38 Mitogen-Activated Protein Kinases/métabolisme ,
17.
Cell Rep ; 37(6): 109975, 2021 11 09.
Article de Anglais | MEDLINE | ID: mdl-34758317

RÉSUMÉ

Dopamine (DA) neurons in the ventral tier of the substantia nigra pars compacta (SNc) degenerate prominently in Parkinson's disease, while those in the dorsal tier are relatively spared. Defining the molecular, functional, and developmental characteristics of each SNc tier is crucial to understand their distinct susceptibility. We demonstrate that Sox6 expression distinguishes ventrally and dorsally biased DA neuron populations in the SNc. The Sox6+ population in the ventral SNc includes an Aldh1a1+ subset and is enriched in gene pathways that underpin vulnerability. Sox6+ neurons project to the dorsal striatum and show activity correlated with acceleration. Sox6- neurons project to the medial, ventral, and caudal striatum and respond to rewards. Moreover, we show that this adult division is encoded early in development. Overall, our work demonstrates a dual origin of the SNc that results in DA neuron cohorts with distinct molecular profiles, projections, and functions.


Sujet(s)
Corps strié/anatomopathologie , Neurones dopaminergiques/anatomopathologie , Régulation de l'expression des gènes au cours du développement , Maladie de Parkinson/anatomopathologie , Facteurs de transcription SOX-D/métabolisme , Facteurs de transcription SOX-D/physiologie , Substantia nigra/anatomopathologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Études cas-témoins , Corps strié/métabolisme , Dopamine/métabolisme , Neurones dopaminergiques/métabolisme , Femelle , Humains , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Adulte d'âge moyen , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Facteurs de transcription SOX-D/génétique , Substantia nigra/métabolisme , Aire tegmentale ventrale/métabolisme , Aire tegmentale ventrale/anatomopathologie
18.
Int J Mol Sci ; 22(19)2021 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-34639012

RÉSUMÉ

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood. About 25% of RMS expresses fusion oncoproteins such as PAX3/PAX7-FOXO1 (fusion-positive, FP) while fusion-negative (FN)-RMS harbors RAS mutations. Radiotherapy (RT) plays a crucial role in local control but metastatic RMS is often radio-resistant. HDAC inhibitors (HDACi) radio-sensitize different cancer cells types. Thus, we evaluated MS-275 (Entinostat), a Class I and IV HDACi, in combination with RT on RMS cells in vitro and in vivo. MS-275 reversibly hampered cell survival in vitro in FN-RMS RD (RASmut) and irreversibly in FP-RMS RH30 cell lines down-regulating cyclin A, B, and D1, up-regulating p21 and p27 and reducing ERKs activity, and c-Myc expression in RD and PI3K/Akt/mTOR activity and N-Myc expression in RH30 cells. Further, MS-275 and RT combination reduced colony formation ability of RH30 cells. In both cell lines, co-treatment increased DNA damage repair inhibition and reactive oxygen species formation, down-regulated NRF2, SOD, CAT and GPx4 anti-oxidant genes and improved RT ability to induce G2 growth arrest. MS-275 inhibited in vivo growth of RH30 cells and completely prevented the growth of RT-unresponsive RH30 xenografts when combined with radiation. Thus, MS-275 could be considered as a radio-sensitizing agent for the treatment of intrinsically radio-resistant PAX3-FOXO1 RMS.


Sujet(s)
Benzamides/pharmacologie , Protéines de fusion oncogènes/génétique , Facteurs de transcription PAX/génétique , Pyridines/pharmacologie , Radiotolérance/effets des médicaments et des substances chimiques , Radiotolérance/génétique , Radiosensibilisants/pharmacologie , Rhabdomyosarcome/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des radiations , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Souris , Rhabdomyosarcome/métabolisme , Rhabdomyosarcome/radiothérapie
19.
Parkinsonism Relat Disord ; 91: 66-76, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34536727

RÉSUMÉ

BACKGROUND: The COVID-19 pandemic has necessitated the social isolation of the population and the rapid implementation of remote care for patients with neurodegenerative diseases. The objective of this study was to explore the perceived impact of confinement in patients with Parkinson's disease and document the effects of gender and living environment. METHODS: We recruited two cohorts from the Canadian provinces of Québec and Alberta, which differed in the dynamics of COVID-19 spreading at the time of the study, and administered a questionnaire on the perceived effects of confinement on daily living and disease management. RESULTS: The data reveals that approximately half of the patients experienced a change in one or more clinical symptoms, with differences observed between gender (e.g. day-to-day changes in slowness in men, aggravated headaches in women) and geographic location (e.g. increased depression in Alberta but reduced sleep quality in Québec). Furthermore, participants identifying as women or living in Alberta implemented more frequently home or online exercise. Lastly, high levels of satisfaction with phone or video consultations did not translate into a sustained interest to pursue this mode of healthcare. CONCLUSIONS: This study suggests that COVID-19-related confinement affected Parkinson's disease manifestation and management. Patients also reported varying levels of interest to continue remote care. A number of differences reported in our study were seemingly related to gender and living environment.


Sujet(s)
Attitude envers la santé , COVID-19 , Exercice physique , Maladie de Parkinson/thérapie , Isolement social , Télémédecine , Activités de la vie quotidienne , Sujet âgé , Alberta , Canada , Études de cohortes , Prise en charge de la maladie , Femelle , Humains , Mâle , Adulte d'âge moyen , Maladie de Parkinson/physiopathologie , Maladie de Parkinson/psychologie , Québec , Caractéristiques de l'habitat , SARS-CoV-2 , Facteurs sexuels
20.
Int J Radiat Biol ; 97(7): 943-957, 2021.
Article de Anglais | MEDLINE | ID: mdl-33979259

RÉSUMÉ

PURPOSE: Herein we describe the in vitro and in vivo activity of FK228 (Romidepsin), an inhibitor of class I HDACs, in counteracting and radiosensitizing embryonal (ERMS, fusion-negative) and alveolar (ARMS, fusion-positive) rhabdomyosarcoma (RMS). METHODS: RH30 (ARMS, fusion-positive) and RD (ERMS, fusion-negative) cell lines and human multipotent mesenchymal stromal cells (HMSC) were used. Flow cytometry analysis, RT-qPCR, western blotting and enzymatic assays were performed. Irradiation was delivered by using an x-6 MV photon linear accelerator. FK228 (1.2 mg/kg) in vivo activity, combined or not with radiation therapy (2 Gy), was assessed in murine xenografts. RESULTS: Compared to HMSC, RMS expressed low levels of class I HDACs. In vitro, FK228, as single agents, reversibly downregulated class I HDACs expression and activity and induced oxidative stress, DNA damage and a concomitant growth arrest associated with PARP-1-mediated transient non-apoptotic cell death. Surviving cells upregulated the expression of cyclin A, B, D1, p27, Myc and activated PI3K/Akt/mTOR and MAPK signaling, known to be differently involved in cancer chemoresistance. Interestingly, while no radiosensitizing effects were detected, in vitro or in vivo, on RD cells, FK228 markedly radiosensitized RH30 cells by impairing antioxidant and DSBs repair pathways in vitro. Further, FK228 when combined with RT in vivo significantly reduced tumor mass in mouse RH30 xenografts. CONCLUSION: FK228 did not show antitumor activity as a single agent whilst its combination with RT resulted in radiosensitization of fusion-positive RMS cells, thus representing a possible strategy for the treatment of the most aggressive RMS subtype.


Sujet(s)
Transformation cellulaire néoplasique , Depsipeptides/pharmacologie , Phénotype , Radiosensibilisants/pharmacologie , Rhabdomyosarcome/anatomopathologie , Animaux , Apoptose/effets des radiations , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des radiations , Lignée cellulaire tumorale , Humains , Souris
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE