Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuropathol Appl Neurobiol ; 48(4): e12800, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35156715

RESUMO

AIMS: An obstacle to developing new treatment strategies for Alzheimer's disease (AD) has been the inadequate translation of findings in current AD transgenic rodent models to the prediction of clinical outcomes. By contrast, nonhuman primates (NHPs) share a close neurobiology with humans in virtually all aspects relevant to developing a translational AD model. The present investigation used African green monkeys (AGMs) to refine an inducible NHP model of AD based on the administration of amyloid-beta oligomers (AßOs), a key upstream initiator of AD pathology. METHODS: AßOs or vehicle were repeatedly delivered over 4 weeks to age-matched young adult AGMs by intracerebroventricular (ICV) or intrathecal (IT) injections. Induction of AD-like pathology was assessed in subregions of the medial temporal lobe (MTL) by quantitative immunohistochemistry (IHC) using the AT8 antibody to detect hyperphosphorylated tau. Hippocampal volume was measured by magnetic resonance imaging (MRI) scans prior to, and after, intrathecal injections. RESULTS: IT administration of AßOs in young adult AGMs revealed an elevation of tau phosphorylation in the MTL cortical memory circuit compared with controls. The largest increases were detected in the entorhinal cortex that persisted for at least 12 weeks after dosing. MRI scans showed a reduction in hippocampal volume following AßO injections. CONCLUSIONS: Repeated IT delivery of AßOs in young adult AGMs led to an accelerated AD-like neuropathology in MTL, similar to human AD, supporting the value of this translational model to de-risk the clinical trial of diagnostic and therapeutic strategies.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Chlorocebus aethiops , Fosforilação , Primatas/metabolismo , Lobo Temporal/patologia , Proteínas tau/metabolismo
2.
Stem Cells Transl Med ; 10(2): 278-290, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32997443

RESUMO

Nongenetic methodologies to reduce undesirable proliferation would be valuable when generating dopamine neurons from stem cells for transplantation in Parkinson's disease (PD). To this end, we modified an established method for controlled differentiation of human induced pluripotent stem cells (iPSCs) into midbrain dopamine neurons using two distinct methods: omission of FGF8 or the in-process use of the DNA cross-linker mitomycin-C (MMC). We transplanted the cells to athymic rats with unilateral 6-hydroxydopamine lesions and monitored long-term survival and function of the grafts. Transplants of cells manufactured using MMC had low proliferation while still permitting robust survival and function comparable to that seen with transplanted dopamine neurons derived using genetic drug selection. Conversely, cells manufactured without FGF8 survived transplantation but exhibited poor in vivo function. Our results suggest that MMC can be used to reduce the number of proliferative cells in stem cell-derived postmitotic neuron preparations for use in PD cell therapy.


Assuntos
Neurônios Dopaminérgicos , Células-Tronco Pluripotentes Induzidas , Mitomicina , Doença de Parkinson , Animais , Diferenciação Celular , Proliferação de Células , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Mitomicina/farmacologia , Doença de Parkinson/terapia , Ratos , Transplante de Células-Tronco
4.
Proc Natl Acad Sci U S A ; 117(49): 31177-31188, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33219123

RESUMO

A transplanted stem cell's engagement with a pathologic niche is the first step in its restoring homeostasis to that site. Inflammatory chemokines are constitutively produced in such a niche; their binding to receptors on the stem cell helps direct that cell's "pathotropism." Neural stem cells (NSCs), which express CXCR4, migrate to sites of CNS injury or degeneration in part because astrocytes and vasculature produce the inflammatory chemokine CXCL12. Binding of CXCL12 to CXCR4 (a G protein-coupled receptor, GPCR) triggers repair processes within the NSC. Although a tool directing NSCs to where needed has been long-sought, one would not inject this chemokine in vivo because undesirable inflammation also follows CXCL12-CXCR4 coupling. Alternatively, we chemically "mutated" CXCL12, creating a CXCR4 agonist that contained a strong pure binding motif linked to a signaling motif devoid of sequences responsible for synthetic functions. This synthetic dual-moity CXCR4 agonist not only elicited more extensive and persistent human NSC migration and distribution than did native CXCL 12, but induced no host inflammation (or other adverse effects); rather, there was predominantly reparative gene expression. When co-administered with transplanted human induced pluripotent stem cell-derived hNSCs in a mouse model of a prototypical neurodegenerative disease, the agonist enhanced migration, dissemination, and integration of donor-derived cells into the diseased cerebral cortex (including as electrophysiologically-active cortical neurons) where their secreted cross-corrective enzyme mediated a therapeutic impact unachieved by cells alone. Such a "designer" cytokine receptor-agonist peptide illustrates that treatments can be controlled and optimized by exploiting fundamental stem cell properties (e.g., "inflammo-attraction").


Assuntos
Quimiocina CXCL12/genética , Neurônios/metabolismo , Ligação Proteica/genética , Receptores CXCR4/genética , Astrócitos/metabolismo , Astrócitos/patologia , Movimento Celular/genética , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Humanos , Células-Tronco Pluripotentes Induzidas , Inflamação/genética , Ligantes , Mutagênese/genética , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/terapia , Neurônios/patologia
5.
Cell Rep ; 31(6): 107622, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32402283

RESUMO

To date, no stem cell therapy has been directed to specific recipients-and, conversely, withheld from others-based on a clinical or molecular profile congruent with that cell's therapeutic mechanism-of-action (MOA) for that condition. We address this challenge preclinically with a prototypical scenario: human neural stem cells (hNSCs) against perinatal/neonatal cerebral hypoxic-ischemic injury (HII). We demonstrate that a clinically translatable magnetic resonance imaging (MRI) algorithm, hierarchical region splitting, provides a rigorous, expeditious, prospective, noninvasive "biomarker" for identifying subjects with lesions bearing a molecular profile indicative of responsiveness to hNSCs' neuroprotective MOA. Implanted hNSCs improve lesional, motor, and/or cognitive outcomes only when there is an MRI-measurable penumbra that can be forestalled from evolving into necrotic core; the core never improves. Unlike the core, a penumbra is characterized by a molecular profile associated with salvageability. Hence, only lesions characterized by penumbral > core volumes should be treated with cells, making such measurements arguably a regenerative medicine selection biomarker.


Assuntos
Biomarcadores/metabolismo , Lesões Encefálicas/terapia , Medicina Regenerativa/métodos , Transplante de Células-Tronco/métodos , Animais , Modelos Animais de Doenças , Ratos , Ratos Sprague-Dawley
6.
Stem Cell Reports ; 9(1): 149-161, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28579395

RESUMO

A major challenge for clinical application of pluripotent stem cell therapy for Parkinson's disease (PD) is large-scale manufacturing and cryopreservation of neurons that can be efficiently prepared with minimal manipulation. To address this obstacle, midbrain dopamine neurons were derived from human induced pluripotent stem cells (iPSC-mDA) and cryopreserved in large production lots for biochemical and transplantation studies. Cryopreserved, post-mitotic iPSC-mDA neurons retained high viability with gene, protein, and electrophysiological signatures consistent with midbrain floor-plate lineage. To test therapeutic efficacy, cryopreserved iPSC-mDA neurons were transplanted without subculturing into the 6-OHDA-lesioned rat and MPTP-lesioned non-human-primate models of PD. Grafted neurons retained midbrain lineage with extensive fiber innervation in both rodents and monkeys. Behavioral assessment in 6-OHDA-lesioned rats demonstrated significant reversal in functional deficits up to 6 months post transplantation with reinnervation of the host striatum and no aberrant growth, supporting the translational development of pluripotent cell-based therapies in PD.


Assuntos
Criopreservação , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/transplante , Células-Tronco Pluripotentes Induzidas/citologia , Doença de Parkinson/terapia , Animais , Linhagem Celular , Corpo Estriado/citologia , Corpo Estriado/patologia , Criopreservação/métodos , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Haplorrinos , Humanos , Mesencéfalo/citologia , Mesencéfalo/patologia , Neurogênese , Doença de Parkinson/patologia , Ratos , Ratos Sprague-Dawley
9.
J Comp Neurol ; 522(12): 2689-90, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24942075
10.
Stem Cells Transl Med ; 3(6): 692-701, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24744393

RESUMO

Transplanted multipotent human fetal neural stem cells (hfNSCs) significantly improved the function of parkinsonian monkeys in a prior study primarily by neuroprotection, with only 3%-5% of cells expressing a dopamine (DA) phenotype. In this paper, we sought to determine whether further manipulation of the neural microenvironment by overexpression of a developmentally critical molecule, glial cell-derived neurotrophic factor (GDNF), in the host striatum could enhance DA differentiation of hfNSCs injected into the substantia nigra and elicit growth of their axons to the GDNF-expressing target. hfNSCs were transplanted into the midbrain of 10 green monkeys exposed to 1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine. GDNF was delivered concomitantly to the striatum via an adeno-associated virus serotype 5 vector, and the fate of grafted cells was assessed after 11 months. Donor cells remained predominantly within the midbrain at the injection site and sprouted numerous neurofilament-immunoreactive fibers that appeared to course rostrally toward the striatum in parallel with tyrosine hydroxylase-immunoreactive fibers from the host substantia nigra but did not mature into DA neurons. This work suggests that hfNSCs can generate neurons that project long fibers in the adult primate brain. However, in the absence of region-specific signals and despite GDNF overexpression, hfNSCs did not differentiate into mature DA neurons in large numbers. It is encouraging, however, that the adult primate brain appeared to retain axonal guidance cues. We believe that transplantation of stem cells, specifically instructed ex vivo to yield DA neurons, could lead to reconstruction of some portion of the nigrostriatal pathway and prove beneficial for the parkinsonian condition.


Assuntos
Terapia Genética/métodos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Intoxicação por MPTP/terapia , Mesencéfalo/cirurgia , Células-Tronco Neurais/transplante , Neuritos/transplante , Neurogênese , Medicina Regenerativa/métodos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Linhagem Celular , Linhagem da Célula , Forma Celular , Sobrevivência Celular , Chlorocebus aethiops , Dependovirus/genética , Modelos Animais de Doenças , Vetores Genéticos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Humanos , Intoxicação por MPTP/induzido quimicamente , Intoxicação por MPTP/genética , Intoxicação por MPTP/metabolismo , Intoxicação por MPTP/patologia , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neuritos/metabolismo , Neuritos/patologia , Nicho de Células-Tronco , Fatores de Tempo , Transdução Genética , Transfecção , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA