Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Neurobiol Dis ; 180: 106086, 2023 05.
Article in English | MEDLINE | ID: mdl-36933673

ABSTRACT

The role of alpha-synuclein in Parkinson's disease has been heavily investigated since its discovery as a component of Lewy bodies. Recent rodent data demonstrate that alpha-synuclein strain structure is critical for differential propagation and toxicity. Based on these findings, we have compared, for the first time, in this pilot study, the capacity of two alpha-synuclein strains and patient-derived Lewy body extracts to model synucleinopathies after intra-putaminal injection in the non-human primate brain. Functional alterations triggered by these injections were evaluated in vivo using glucose positron emission tomography imaging. Post-mortem immunohistochemical and biochemical analyses were used to detect neuropathological alterations in the dopaminergic system and alpha-synuclein pathology propagation. In vivo results revealed a decrease in glucose metabolism more pronounced in alpha-synuclein strain-injected animals. Histology showed a decreased number of dopaminergic tyrosine hydroxylase-positive cells in the substantia nigra to different extents according to the inoculum used. Biochemistry revealed that alpha-synuclein-induced aggregation, phosphorylation, and propagation in different brain regions are strain-specific. Our findings show that distinct alpha-synuclein strains can induce specific patterns of synucleinopathy in the non-human primate, changes in the nigrostriatal pathway, and functional alterations that resemble early-stage Parkinson's disease.


Subject(s)
Parkinson Disease , Synucleinopathies , Animals , alpha-Synuclein/metabolism , Parkinson Disease/metabolism , Pilot Projects , Lewy Bodies/metabolism , Synucleinopathies/pathology , Substantia Nigra/metabolism , Dopamine/metabolism , Primates/metabolism
3.
Nat Commun ; 10(1): 4357, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31554807

ABSTRACT

Cell therapy products (CTP) derived from pluripotent stem cells (iPSCs) may constitute a renewable, specifically differentiated source of cells to potentially cure patients with neurodegenerative disorders. However, the immunogenicity of CTP remains a major issue for therapeutic approaches based on transplantation of non-autologous stem cell-derived neural grafts. Despite its considerable side-effects, long-term immunosuppression, appears indispensable to mitigate neuro-inflammation and prevent rejection of allogeneic CTP. Matching iPSC donors' and patients' HLA haplotypes has been proposed as a way to access CTP with enhanced immunological compatibility, ultimately reducing the need for immunosuppression. In the present work, we challenge this paradigm by grafting autologous, MHC-matched and mis-matched neuronal grafts in a primate model of Huntington's disease. Unlike previous reports in unlesioned hosts, we show that in the absence of immunosuppression MHC matching alone is insufficient to grant long-term survival of neuronal grafts in the lesioned brain.


Subject(s)
Graft Rejection/immunology , Huntington Disease/therapy , Induced Pluripotent Stem Cells/transplantation , Major Histocompatibility Complex/immunology , Neurons/transplantation , Animals , Cell Differentiation/immunology , Cytotoxicity, Immunologic/immunology , Disease Models, Animal , Histocompatibility Testing , Humans , Huntington Disease/immunology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Neurons/cytology , Neurons/immunology , Primates , Rats, Nude , Transplantation, Autologous
4.
Neurobiol Dis ; 130: 104484, 2019 10.
Article in English | MEDLINE | ID: mdl-31132407

ABSTRACT

As research progresses in the understanding of the molecular and cellular mechanisms underlying neurodegenerative diseases like Huntington's disease (HD) and expands towards preclinical work for the development of new therapies, highly relevant animal models are increasingly needed to test new hypotheses and to validate new therapeutic approaches. In this light, we characterized an excitotoxic lesion model of striatal dysfunction in non-human primates (NHPs) using cognitive and motor behaviour assessment as well as functional imaging and post-mortem anatomical analyses. NHPs received intra-striatal stereotaxic injections of quinolinic acid bilaterally in the caudate nucleus and unilaterally in the left sensorimotor putamen. Post-operative MRI scans showed atrophy of the caudate nucleus and a large ventricular enlargement in all 6 NHPs that correlated with post-mortem measurements. Behavioral analysis showed deficits in 2 analogues of the Wisconsin card sorting test (perseverative behavior) and in an executive task, while no deficits were observed in a visual recognition or an episodic memory task at 6 months following surgery. Spontaneous locomotor activity was decreased after lesion and the incidence of apomorphine-induced dyskinesias was significantly increased at 3 and 6 months following lesion. Positron emission tomography scans obtained at end-point showed a major deficit in glucose metabolism and D2 receptor density limited to the lesioned striatum of all NHPs compared to controls. Post-mortem analyses revealed a significant loss of medium-sized spiny neurons in the striatum, a loss of neurons and fibers in the globus pallidus, a unilateral decrease in dopaminergic neurons of the substantia nigra and a loss of neurons in the motor and dorsolateral prefrontal cortex. Overall, we show that this robust NHP model presents specific behavioral (learning, execution and retention of cognitive tests) and metabolic functional deficits that, to the best of our knowledge, are currently not mimicked in any available large animal model of striatal dysfunction. Moreover, we used non-invasive, translational techniques like behavior and imaging to quantify such deficits and found that they correlate to a significant cell loss in the striatum and its main input and output structures. This model can thus significantly contribute to the pre-clinical longitudinal evaluation of the ability of new therapeutic cell, gene or pharmacotherapy approaches in restoring the functionality of the striatal circuitry.


Subject(s)
Cognitive Dysfunction , Disease Models, Animal , Huntington Disease , Motor Disorders , Animals , Cognitive Dysfunction/chemically induced , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Huntington Disease/chemically induced , Huntington Disease/pathology , Huntington Disease/physiopathology , Longitudinal Studies , Macaca fascicularis , Male , Motor Disorders/chemically induced , Quinolinic Acid/toxicity
5.
J Cereb Blood Flow Metab ; 39(7): 1191-1203, 2019 07.
Article in English | MEDLINE | ID: mdl-29381130

ABSTRACT

Focused ultrasound (FUS) in combination with microbubbles is capable of noninvasive, site-targeted delivery of drugs through the blood-brain barrier (BBB). Although acoustic parameters are reproducible in small animals, their control remains challenging in primates due to skull heterogeneity. This study describes a 7-T magnetic resonance (MR)-guided FUS system designed for BBB disruption in non-human primates (NHP) with a robust feedback control based on passive cavitation detection (PCD). Contrast enhanced T1-weighted MR images confirmed the BBB opening in NHP sonicated during 2 min with 500-kHz frequency, pulse length of 10 ms, and pulse repetition frequency of 5 Hz. The safe acoustic pressure range from 185 ± 22 kPa to 266 ± 4 kPa in one representative case was estimated from combining data from the acoustic beam profile with the BBB opening and hemorrhage profiles obtained from MR images. A maximum amount of MR contrast agent at focus was observed at 30 min after sonication with a relative contrast enhancement of 67% ± 15% (in comparison to that found in muscles). The feedback control based on PCD using relative spectra was shown to be robust, allowing comparisons across animals and experimental sessions. Finally, we also demonstrated that PCD can test acoustic coupling conditions, which improves the efficacy and safety of ultrasound transmission into the brain.


Subject(s)
Blood-Brain Barrier/physiology , Feedback, Physiological/physiology , Macaca fascicularis , Microbubbles/therapeutic use , Ultrasonography/methods , Animals , Blood-Brain Barrier/diagnostic imaging , Brain/drug effects , Brain/physiology , Brain Diseases/drug therapy , Drug Delivery Systems , Magnetic Resonance Imaging/methods , Male , Sonication/methods
6.
Methods Mol Biol ; 1780: 267-284, 2018.
Article in English | MEDLINE | ID: mdl-29856024

ABSTRACT

Huntington's disease (HD) is a monogenic, autosomal dominant inherited fatal disease that affects 1 in 10,000 people worldwide. Given its unique genetic characteristics, HD would appear as one of the most straightforward neurodegenerative diseases to replicate in animal models. Indeed, mutations in the HTT gene have been used to generate a variety of animal models that display differential pathologies and have significantly increased our understanding of the pathological mechanisms of HD. However, decades of efforts have also shown the complexity of recapitulating the human condition in other species. Here we describe the three different types of models that have been generated in nonhuman primate species, stating their advantages and limitations and attempt to give a critical perspective of their translational value to test the efficacy of novel therapeutic strategies. Obtaining construct, phenotypic, and predictive validity has proven to be challenging in most animal models of human diseases. In HD in particular, it is hard to assess the predictive validity of a new therapeutic strategy when no effective "benchmark" treatment is available in the clinic. In this light, only phenotypic/face validity and construct validity are discussed.


Subject(s)
Behavior Observation Techniques/methods , Disease Models, Animal , Huntington Disease/pathology , Primates , Translational Research, Biomedical/methods , Animals , Animals, Genetically Modified , Atrophy/chemically induced , Behavior Observation Techniques/economics , Behavior Observation Techniques/instrumentation , Behavior, Animal , Brain/drug effects , Brain/pathology , Brain/physiopathology , Genetic Therapy/adverse effects , Genetic Therapy/methods , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/etiology , Huntington Disease/physiopathology , Huntington Disease/therapy , Mutation , Neurotoxins/administration & dosage , Neurotoxins/toxicity , Stereotaxic Techniques/instrumentation , Translational Research, Biomedical/instrumentation , Treatment Outcome
7.
Data Brief ; 16: 37-42, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29167818

ABSTRACT

Validation data for segmentation algorithms dedicated to preclinical images is fiercely lacking, especially when compared to the large number of databases of Human brain images and segmentations available to the academic community. Not only is such data essential for validating methods, it is also needed for objectively comparing concurrent algorithms and detect promising paths, as segmentation challenges have shown for clinical images. The dataset we present here is a first step in this direction. It comprises 10 T2-weighted MRIs of healthy adult macaque brains, acquired on a 7 T magnet, along with corresponding manual segmentations into 17 brain anatomic labelled regions spread over 5 hierarchical levels based on a previously published macaque atlas (Calabrese et al., 2015) [1]. By giving access to this unique dataset, we hope to provide a reference needed by the non-human primate imaging community. This dataset was used in an article presenting a new primate brain morphology analysis pipeline, Primatologist (Balbastre et al., 2017) [2]. Data is available through a NITRC repository (https://www.nitrc.org/projects/mircen_macset).

8.
Neuroimage ; 162: 306-321, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28899745

ABSTRACT

Because they bridge the genetic gap between rodents and humans, non-human primates (NHPs) play a major role in therapy development and evaluation for neurological disorders. However, translational research success from NHPs to patients requires an accurate phenotyping of the models. In patients, magnetic resonance imaging (MRI) combined with automated segmentation methods has offered the unique opportunity to assess in vivo brain morphological changes. Meanwhile, specific challenges caused by brain size and high field contrasts make existing algorithms hard to use routinely in NHPs. To tackle this issue, we propose a complete pipeline, Primatologist, for multi-region segmentation. Tissue segmentation is based on a modular statistical model that includes random field regularization, bias correction and denoising and is optimized by expectation-maximization. To deal with the broad variety of structures with different relaxing times at 7 T, images are segmented into 17 anatomical classes, including subcortical regions. Pre-processing steps insure a good initialization of the parameters and thus the robustness of the pipeline. It is validated on 10 T2-weighted MRIs of healthy macaque brains. Classification scores are compared with those of a non-linear atlas registration, and the impact of each module on classification scores is thoroughly evaluated.


Subject(s)
Algorithms , Brain/anatomy & histology , Macaca/anatomy & histology , Neuroimaging/methods , Software , Animals , Atlases as Topic , Image Processing, Computer-Assisted , Imaging, Three-Dimensional/methods , Magnetic Resonance Imaging
9.
Stroke ; 48(8): 2301-2305, 2017 08.
Article in English | MEDLINE | ID: mdl-28526764

ABSTRACT

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) is a devastating form of stroke with neurological outcomes dependent on the occurrence of delayed cerebral ischemia. It has been shown in rodents that some of the mechanisms leading to delayed cerebral ischemia are related to a decreased circulation of the cerebrospinal fluid (CSF) within the brain parenchyma. Here, we evaluated the cerebral circulation of the CSF in a nonhuman primate in physiological condition and after SAH. METHODS: We first evaluated in physiological condition the circulation of the brain CSF in Macacafacicularis, using magnetic resonance imaging of the temporal DOTA-Gd distribution after its injection into the CSF. Then, animals were subjected to a minimally invasive SAH before an MRI evaluation of the impact of SAH on the brain parenchymal CSF circulation. RESULTS: We first demonstrate that the CSF actively penetrates the brain parenchyma. Two hours after injection, almost the entire brain is labeled by DOTA-Gd. We also show that our model of SAH in nonhuman primate displays the characteristics of SAH in humans and leads to a dramatic impairment of the brain parenchymal circulation of the CSF. CONCLUSIONS: The CSF actively penetrates within the brain parenchyma in the gyrencephalic brain, as described for the glymphatic system in rodent. This parenchymal CSF circulation is severely impaired by SAH.


Subject(s)
Brain/metabolism , Cerebrovascular Circulation/physiology , Parenchymal Tissue/metabolism , Severity of Illness Index , Subarachnoid Hemorrhage/cerebrospinal fluid , Animals , Brain/diagnostic imaging , Macaca fascicularis , Male , Parenchymal Tissue/diagnostic imaging , Primates , Subarachnoid Hemorrhage/diagnostic imaging
10.
Int J Surg ; 23(Pt B): 267-272, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26403068

ABSTRACT

Neural cell transplantation has long been considered as an option for the treatment of neurodegenerative disorders. To date, several patients with Parkinson's and Huntington's diseases have been treated with human fetal-derived neurons with disparate results. However, the limited efficacy to date combined with the scarce availability of human fetal tissues and ethical concerns render this procedure inapplicable to a wide population scale. With a view to overcoming these shortcomings, transplantation of pig-derived cell precursors has been proposed and applied in preclinical and clinical trials. Recently long-term survival (more than 18 months) associated with clinical efficacy has been reported following transplantation of genetically engineered porcine neural precursors in fully immunosuppressed primate recipients. Despite the promising results obtained to date, several questions remain unanswered. In particular, the ideal xenogeneic cell-products to transplant, the extent of the immune response against the implanted xenograft and the most suitable therapeutic strategies to improve engraftment are all issues that still need to be thoroughly addressed. The present review describes the current knowledge in the pig-to-primate xenotransplantation field. In this context, recent data on human-to-nonhuman primate xenogeneic stem cell-based treatments for neurological disorders are discussed.


Subject(s)
Neurodegenerative Diseases/therapy , Neurons/transplantation , Animals , Humans , Immunosuppression Therapy/methods , Primates , Swine , Transplantation, Heterologous/methods
SELECTION OF CITATIONS
SEARCH DETAIL