Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Neurobiol Dis ; 180: 106086, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36933673

RESUMO

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.


Assuntos
Doença de Parkinson , Sinucleinopatias , Animais , alfa-Sinucleína/metabolismo , Doença de Parkinson/metabolismo , Projetos Piloto , Corpos de Lewy/metabolismo , Sinucleinopatias/patologia , Substância Negra/metabolismo , Dopamina/metabolismo , Primatas/metabolismo
3.
Nat Commun ; 10(1): 4357, 2019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31554807

RESUMO

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.


Assuntos
Rejeição de Enxerto/imunologia , Doença de Huntington/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Complexo Principal de Histocompatibilidade/imunologia , Neurônios/transplante , Animais , Diferenciação Celular/imunologia , Citotoxicidade Imunológica/imunologia , Modelos Animais de Doenças , Teste de Histocompatibilidade , Humanos , Doença de Huntington/imunologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/imunologia , Neurônios/citologia , Neurônios/imunologia , Primatas , Ratos Nus , Transplante Autólogo
4.
Neurobiol Dis ; 130: 104484, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31132407

RESUMO

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.


Assuntos
Disfunção Cognitiva , Modelos Animais de Doenças , Doença de Huntington , Transtornos Motores , Animais , Disfunção Cognitiva/induzido quimicamente , Corpo Estriado/patologia , Corpo Estriado/fisiopatologia , Doença de Huntington/induzido quimicamente , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Estudos Longitudinais , Macaca fascicularis , Masculino , Transtornos Motores/induzido quimicamente , Ácido Quinolínico/toxicidade
5.
J Cereb Blood Flow Metab ; 39(7): 1191-1203, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-29381130

RESUMO

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.


Assuntos
Barreira Hematoencefálica/fisiologia , Retroalimentação Fisiológica/fisiologia , Macaca fascicularis , Microbolhas/uso terapêutico , Ultrassonografia/métodos , Animais , Barreira Hematoencefálica/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Encefalopatias/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Imageamento por Ressonância Magnética/métodos , Masculino , Sonicação/métodos
6.
Methods Mol Biol ; 1780: 267-284, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29856024

RESUMO

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.


Assuntos
Técnicas de Observação do Comportamento/métodos , Modelos Animais de Doenças , Doença de Huntington/patologia , Primatas , Pesquisa Translacional Biomédica/métodos , Animais , Animais Geneticamente Modificados , Atrofia/induzido quimicamente , Técnicas de Observação do Comportamento/economia , Técnicas de Observação do Comportamento/instrumentação , Comportamento Animal , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/etiologia , Doença de Huntington/fisiopatologia , Doença de Huntington/terapia , Mutação , Neurotoxinas/administração & dosagem , Neurotoxinas/toxicidade , Técnicas Estereotáxicas/instrumentação , Pesquisa Translacional Biomédica/instrumentação , Resultado do Tratamento
7.
Data Brief ; 16: 37-42, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29167818

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-28899745

RESUMO

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.


Assuntos
Algoritmos , Encéfalo/anatomia & histologia , Macaca/anatomia & histologia , Neuroimagem/métodos , Software , Animais , Atlas como Assunto , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional/métodos , Imageamento por Ressonância Magnética
9.
Stroke ; 48(8): 2301-2305, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28526764

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Circulação Cerebrovascular/fisiologia , Tecido Parenquimatoso/metabolismo , Índice de Gravidade de Doença , Hemorragia Subaracnóidea/líquido cefalorraquidiano , Animais , Encéfalo/diagnóstico por imagem , Macaca fascicularis , Masculino , Tecido Parenquimatoso/diagnóstico por imagem , Primatas , Hemorragia Subaracnóidea/diagnóstico por imagem
10.
Int J Surg ; 23(Pt B): 267-272, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26403068

RESUMO

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.


Assuntos
Doenças Neurodegenerativas/terapia , Neurônios/transplante , Animais , Humanos , Terapia de Imunossupressão/métodos , Primatas , Suínos , Transplante Heterólogo/métodos
11.
Eur J Pharmacol ; 759: 118-26, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25841877

RESUMO

Animal models have been highly questioned for their ability to predict the efficacy of different therapeutic strategies for neurodegenerative diseases. The increasing number of phase I/II clinical trials that fail to proceed to further stages of drug development has discredited the pertinence of such investigations. However, critical analysis of the data has often revealed errors and partially explained the lack of efficacy, opening the way to a refinement in designing pre-clinical studies. In parallel, many promising methods of drug delivery to the brain such as gene therapy or cell therapy have considerably advanced thanks to the clinical failures in the past 10 years. As methodological advances appear and knowledge becomes available, scientists will be faced with the choice of how to test new strategies or re-test old ones. With the hardening of social views and legislation regarding animal experimentation, there is increasing pressure to find alternative methods of assessment that predict efficacy (such as computational based models), or to perform efficacy trials directly in patients and only safety assays in animals. In this review we will focus on Parkinson׳s disease and on the impact of a body of data issued from NHP studies. We will attempt to critically examine the advantages and limitations of various approaches from the perspective of the animal model used to address specific questions.


Assuntos
Modelos Animais de Doenças , Doença de Parkinson/terapia , Pesquisa Translacional Biomédica/métodos , Animais , Transplante de Células , Terapia Genética , Humanos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Primatas , Especificidade da Espécie
12.
J Neurosci ; 35(6): 2817-29, 2015 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-25673868

RESUMO

Astrocyte reactivity is a hallmark of neurodegenerative diseases (ND), but its effects on disease outcomes remain highly debated. Elucidation of the signaling cascades inducing reactivity in astrocytes during ND would help characterize the function of these cells and identify novel molecular targets to modulate disease progression. The Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway is associated with reactive astrocytes in models of acute injury, but it is unknown whether this pathway is directly responsible for astrocyte reactivity in progressive pathological conditions such as ND. In this study, we examined whether the JAK/STAT3 pathway promotes astrocyte reactivity in several animal models of ND. The JAK/STAT3 pathway was activated in reactive astrocytes in two transgenic mouse models of Alzheimer's disease and in a mouse and a nonhuman primate lentiviral vector-based model of Huntington's disease (HD). To determine whether this cascade was instrumental for astrocyte reactivity, we used a lentiviral vector that specifically targets astrocytes in vivo to overexpress the endogenous inhibitor of the JAK/STAT3 pathway [suppressor of cytokine signaling 3 (SOCS3)]. SOCS3 significantly inhibited this pathway in astrocytes, prevented astrocyte reactivity, and decreased microglial activation in models of both diseases. Inhibition of the JAK/STAT3 pathway within reactive astrocytes also increased the number of huntingtin aggregates, a neuropathological hallmark of HD, but did not influence neuronal death. Our data demonstrate that the JAK/STAT3 pathway is a common mediator of astrocyte reactivity that is highly conserved between disease states, species, and brain regions. This universal signaling cascade represents a potent target to study the role of reactive astrocytes in ND.


Assuntos
Doença de Alzheimer/fisiopatologia , Astrócitos , Doença de Huntington/fisiopatologia , Janus Quinases , Fator de Transcrição STAT3 , Transdução de Sinais , Doença de Alzheimer/patologia , Animais , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Humanos , Doença de Huntington/patologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética
13.
PLoS One ; 8(1): e52680, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23300984

RESUMO

The development of dyskinesias following chronic L-DOPA replacement therapy remains a major problem in the long-term treatment of Parkinson's disease. This study aimed at evaluating the effect of IRC-082451 (base of BN82451), a novel multitargeting hybrid molecule, on L-DOPA-induced dyskinesias (LIDs) and hypolocomotor activity in a non-human primate model of PD. IRC-082451 displays multiple properties: it inhibits neuronal excitotoxicity (sodium channel blocker), oxidative stress (antioxidant) and neuroinflammation (cyclooxygenase inhibitor) and is endowed with mitochondrial protective properties. Animals received daily MPTP injections until stably parkinsonian. A daily treatment with increasing doses of L-DOPA was administered to parkinsonian primates until the appearance of dyskinesias. Then, different treatment regimens and doses of IRC-082451 were tested and compared to the benchmark molecule amantadine. Primates were regularly filmed and videos were analyzed with specialized software. A novel approach combining the analysis of dyskinesias and locomotor activity was used to determine efficacy. This analysis yielded the quantification of the total distance travelled and the incidence of dyskinesias in 7 different body parts. A dose-dependent efficacy of IRC-082451 against dyskinesias was observed. The 5 mg/kg dose was best at attenuating the severity of fully established LIDs. Its effect was significantly different from that of amantadine since it increased spontaneous locomotor activity while reducing LIDs. This dose was effective both acutely and in a 5-day sub-chronic treatment. Moreover, positron emission tomography scans using radiolabelled dopamine demonstrated that there was no direct interference between treatment with IRC-082451 and dopamine metabolism in the brain. Finally, post-mortem analysis indicated that this reduction in dyskinesias was associated with changes in cFOS, FosB and ARC mRNA expression levels in the putamen. The data demonstrates the antidyskinetic efficacy of IRC-082451 in a primate model of PD with motor complications and opens the way to the clinical application of this treatment for the management of LIDs.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Antiparkinsonianos/farmacologia , Discinesias/metabolismo , Levodopa/metabolismo , Doença de Parkinson/tratamento farmacológico , Tiazóis/farmacologia , Amantadina/farmacologia , Animais , Antioxidantes/farmacologia , Comportamento Animal , Cromatografia Líquida de Alta Pressão , Inibidores de Ciclo-Oxigenase/farmacologia , Modelos Animais de Doenças , Dopaminérgicos/farmacologia , Imuno-Histoquímica , Macaca fascicularis , Imageamento por Ressonância Magnética , Masculino , Neurônios/efeitos dos fármacos , Estresse Oxidativo , Tomografia por Emissão de Pósitrons
14.
J Cereb Blood Flow Metab ; 30(4): 849-56, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19997117

RESUMO

Heat shock protein 27 (HSP27) has a major role in mediating survival responses to a range of central nervous system insults, functioning as a protein chaperone, an antioxidant, and through inhibition of cell death pathways. We have used transgenic mice overexpressing HSP27 (HSP27tg) to examine the role of HSP27 in cerebral ischemia, using model of permanent middle cerebral artery occlusion (MCAO). Infarct size was evaluated using multislice T(2)-weighted anatomical magnetic resonance imaging (MRI) after 24 h. A significant reduction of 30% in infarct size was detected in HSP27tg animals compared with wild-type (WT) littermates. To gain some insight into the mechanisms contributing to cell death and its attenuation by HSP27, we monitored the effect of induction of c-jun and ATF3 on tissue survival in MCAO and their effects on the expression of endogenous mouse HSP25 and HSP70. It is important that, the c-jun induction seen at 4 h tended to be localized to regions that were salvageable in HSP27tg mice but became infarcted in WT animals. Our results provide support for the powerful neuroprotective effects of HSP27 in cerebral ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Encéfalo , Proteínas de Choque Térmico HSP27/metabolismo , Fármacos Neuroprotetores/metabolismo , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Circulação Cerebrovascular/fisiologia , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Hibridização In Situ , Infarto da Artéria Cerebral Média/patologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Chaperonas Moleculares , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fluxo Sanguíneo Regional/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA