Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neuropathol Appl Neurobiol ; 48(4): e12800, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35156715

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Chlorocebus aethiops , Fosforilación , Primates/metabolismo , Lóbulo Temporal/patología , Proteínas tau/metabolismo
2.
J Neural Transm (Vienna) ; 127(5): 785-791, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32172471

RESUMEN

The substantial contributions of Dr. Gerald Stern to past and current treatments for Parkinson's disease patients are reviewed, which form the foundation for an evaluation of future options to control symptoms and halt progression of the disease. These opportunities will depend on a greater understanding of the relative contributions of the environment, genetic and epigenetic influences to disease onset, and promise to emerge as strategies for improving mitochondrial function, halting accumulation of synuclein and neuromelanin, in addition to refinement of stem cell and gene therapies. Such advances will be achieved through deployment of improved models for the disease.


Asunto(s)
Neurología/historia , Enfermedad de Parkinson/terapia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Neurología/tendencias , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo
3.
Biochem Biophys Res Commun ; 510(1): 78-84, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30660369

RESUMEN

Generating human organs inside interspecies chimeras might one day produce patient-specific organs for clinical applications, but further advances in identifying human chimera-competent pluripotent stem (PS) cells are needed. Moreover, the potential for human PS cells to contribute to the brains in human-animal chimeras raises ethical questions. The use of non-human primate (NHP) chimera-competent PS cells would allow one to test interspecies organogenesis strategies while also bypassing such ethical concerns. Here, we provide the first evidence for a putative chimera-competent pluripotent state in NHPs. Using histone deacetylase (HDAC) and selective kinase inhibition, we converted the PS cells of an Old World monkey, the African Green monkey (aGM), to an ERK-independent cellular state that can be propagated in culture conditions similar to those that sustain chimera-competency in rodent cells. The obtained stem cell lines indefinitely self-renew in MEK inhibitor-containing culture media lacking serum replacement and FGF. Compared to conventional PS cells, the novel stem cells express elevated levels of KLF4, exhibit more intense nuclear staining for TFE3, and manifest increased mitochondrial membrane depolarization. These data are preliminary but indicate that the key to deriving primate chimera-competent PS cells is to shield cells from the activation of ERK, PKC, and WNT signaling. Because of the similarity of aGMs to humans, the more ethically palatable use of NHP cells, and the more similar gestation length between aGMs and large animals such as sheep, the aGM cell lines described herein will serve as a useful tool for evaluating the efficacy and safety of interspecies organogenesis strategies. Future studies will examine chimera-competency and generalizability to human cells.


Asunto(s)
Quimera/embriología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Células Madre Pluripotentes/citología , Animales , Bioética , Células Cultivadas , Chlorocebus aethiops , Humanos , Factor 4 Similar a Kruppel , Organogénesis
4.
J Neurosci ; 36(10): 3049-63, 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26961958

RESUMEN

Calorie restriction (CR) is neuroprotective in Parkinson's disease (PD) although the mechanisms are unknown. In this study we hypothesized that elevated ghrelin, a gut hormone with neuroprotective properties, during CR prevents neurodegeneration in an 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD. CR attenuated the MPTP-induced loss of substantia nigra (SN) dopamine neurons and striatal dopamine turnover in ghrelin WT but not KO mice, demonstrating that ghrelin mediates CR's neuroprotective effect. CR elevated phosphorylated AMPK and ACC levels in the striatum of WT but not KO mice suggesting that AMPK is a target for ghrelin-induced neuroprotection. Indeed, exogenous ghrelin significantly increased pAMPK in the SN. Genetic deletion of AMPKß1 and 2 subunits only in dopamine neurons prevented ghrelin-induced AMPK phosphorylation and neuroprotection. Hence, ghrelin signaling through AMPK in SN dopamine neurons mediates CR's neuroprotective effects. We consider targeting AMPK in dopamine neurons may recapitulate neuroprotective effects of CR without requiring dietary intervention.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Restricción Calórica , Ghrelina/metabolismo , Intoxicación por MPTP/patología , Intoxicación por MPTP/prevención & control , Enfermedad de Parkinson/fisiopatología , Transducción de Señal/fisiología , Proteínas Quinasas Activadas por AMP/genética , Animales , Proteínas de Unión al Calcio/metabolismo , Recuento de Células , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Ghrelina/genética , Ghrelina/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Intoxicación por MPTP/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tirosina 3-Monooxigenasa/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(32): 11876-81, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25071172

RESUMEN

Prolyl endopeptidase (PREP) has been implicated in neuronal functions. Here we report that hypothalamic PREP is predominantly expressed in the ventromedial nucleus (VMH), where it regulates glucose-induced neuronal activation. PREP knockdown mice (Prep(gt/gt)) exhibited glucose intolerance, decreased fasting insulin, increased fasting glucagon levels, and reduced glucose-induced insulin secretion compared with wild-type controls. Consistent with this, central infusion of a specific PREP inhibitor, S17092, impaired glucose tolerance and decreased insulin levels in wild-type mice. Arguing further for a central mode of action of PREP, isolated pancreatic islets showed no difference in glucose-induced insulin release between Prep(gt/gt) and wild-type mice. Furthermore, hyperinsulinemic euglycemic clamp studies showed no difference between Prep(gt/gt) and wild-type control mice. Central PREP regulation of insulin and glucagon secretion appears to be mediated by the autonomic nervous system because Prep(gt/gt) mice have elevated sympathetic outflow and norepinephrine levels in the pancreas, and propranolol treatment reversed glucose intolerance in these mice. Finally, re-expression of PREP by bilateral VMH injection of adeno-associated virus-PREP reversed the glucose-intolerant phenotype of the Prep(gt/gt) mice. Taken together, our results unmask a previously unknown player in central regulation of glucose metabolism and pancreatic function.


Asunto(s)
Glucagón/metabolismo , Hipotálamo/enzimología , Insulina/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Glucemia/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Técnica de Clampeo de la Glucosa , Intolerancia a la Glucosa/enzimología , Intolerancia a la Glucosa/etiología , Hipotálamo/fisiología , Indoles/farmacología , Secreción de Insulina , Canales Iónicos/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas Mitocondriales/genética , Páncreas/metabolismo , Fosforilación , Prolil Oligopeptidasas , Receptor de Insulina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/genética , Inhibidores de Serina Proteinasa/farmacología , Tiazolidinas/farmacología , Proteína Desacopladora 1 , Núcleo Hipotalámico Ventromedial/enzimología , Núcleo Hipotalámico Ventromedial/fisiología
6.
J Neurochem ; 137(3): 460-71, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26872221

RESUMEN

The gut hormone ghrelin is widely beneficial in many disease states. However, ghrelin exists in two distinctive isoforms, each with its own metabolic profile. In Parkinson's Disease (PD) acylated ghrelin administration is neuroprotective, however, the role of des-acylated ghrelin remains unknown. In this study, we wanted to identify the relative contribution each isoform plays using the MPTP model of PD. Chronic administration of acylated ghrelin in mice lacking both isoforms of ghrelin (Ghrelin KO) attenuated the MPTP-induced loss on tyrosine hydroxylase (TH) neuronal number and volume and TH protein expression in the nigrostriatal pathway. Moreover, acylated ghrelin reduced the increase in glial fibrillary acidic protein and Ionized calcium binding adaptor molecule 1 microglia in the substantia nigra. However, injection of acylated ghrelin also elevated plasma des-acylated ghrelin, indicating in vivo deacetylation. Next, we chronically administered des-acylated ghrelin to Ghrelin KO mice and observed no neuroprotective effects in terms of TH cell number, TH protein expression, glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1 cell number. The lack of a protective effect was mirrored in ghrelin-O-acyltransferase KO mice, which lack the ability to acylate ghrelin and consequently these mice have chronically increased plasma des-acyl ghrelin. Plasma corticosterone was elevated in ghrelin-O-acyltransferase KO mice and with des-acylated ghrelin administration. Overall, our studies suggest that acylated ghrelin is the isoform responsible for in vivo neuroprotection and that pharmacological approaches preventing plasma conversion from acyl ghrelin to des-acyl ghrelin may have clinical efficacy to help slow or prevent the debilitating effects of PD. Ghrelin exists in the plasma as acyl and des-acyl ghrelin. We determined the form responsible for in vivo neuroprotection in a mouse model of Parkinson's disease. Although exogenous acyl ghrelin is deacylated in situ to des-acyl, only acyl ghrelin was neuroprotective by attenuating dopamine cell loss and glial activation. Acyl ghrelin is a therapeutic option to reduce Parkinson's Disease progression. Cover Image for this issue: doi: 10.1111/jnc.13316.


Asunto(s)
Ghrelina/análogos & derivados , Ghrelina/farmacología , Intoxicación por MPTP/prevención & control , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson Secundaria/prevención & control , Acilación , Aciltransferasas/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Recuento de Células , Ghrelina/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Vías Nerviosas/patología , Neuronas/patología , Fármacos Neuroprotectores/química , Tirosina 3-Monooxigenasa/metabolismo
7.
J Neurosci ; 34(43): 14443-54, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25339755

RESUMEN

For >30 years, positron emission tomography (PET) has proven to be a powerful approach for measuring aspects of dopaminergic transmission in the living human brain; this technique has revealed important relationships between dopamine D2-like receptors and dimensions of normal behavior, such as human impulsivity, and psychopathology, particularly behavioral addictions. Nevertheless, PET is an indirect estimate that lacks cellular and functional resolution and, in some cases, is not entirely pharmacologically specific. To identify the relationships between PET estimates of D2-like receptor availability and direct in vitro measures of receptor number, affinity, and function, we conducted neuroimaging and behavioral and molecular pharmacological assessments in a group of adult male vervet monkeys. Data gathered from these studies indicate that variation in D2-like receptor PET measurements is related to reversal-learning performance and sensitivity to positive feedback and is associated with in vitro estimates of the density of functional dopamine D2-like receptors. Furthermore, we report that a simple behavioral measure, eyeblink rate, reveals novel and crucial links between neuroimaging assessments and in vitro measures of dopamine D2 receptors.


Asunto(s)
Parpadeo/fisiología , Cuerpo Estriado/fisiología , Aprendizaje Discriminativo/fisiología , Retroalimentación Fisiológica/fisiología , Receptores de Dopamina D2/fisiología , Animales , Chlorocebus aethiops , Masculino , Estimulación Luminosa/métodos , Factores de Tiempo
8.
J Cell Mol Med ; 19(1): 249-56, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25283241

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disease caused by the loss of dopaminergic neurons in the substantia nigra. Cell-replacement therapies have emerged as a promising strategy to slow down or replace neuronal loss. Compared to other stem cell types, endometrium-derived stem cells (EDSCs) are an attractive source of stem cells for cellular therapies because of their ease of collection and vast differentiation potential. Here we demonstrate that endometrium-derived stem cells may be transplanted into an MPTP exposed monkey model of PD. After injection into the striatum, endometrium-derived stem cells engrafted, exhibited neuron-like morphology, expressed tyrosine hydroxylase (TH) and increased the numbers of TH positive cells on the transplanted side and dopamine metabolite concentrations in vivo. Our results suggest that endometrium-derived stem cells may provide a therapeutic benefit in the primate model of PD and may be used in stem cell based therapies.


Asunto(s)
Endometrio/citología , Enfermedad de Parkinson/terapia , Trasplante de Células Madre , Células Madre/citología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Recuento de Células , Movimiento Celular , Femenino , Ácido Homovanílico/metabolismo , Masculino , Neuronas/metabolismo , Enfermedad de Parkinson/patología , Primates , Tirosina 3-Monooxigenasa/metabolismo
9.
Proc Natl Acad Sci U S A ; 109(8): 3125-30, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22315408

RESUMEN

Schizophrenia is characterized by affective, cognitive, neuromorphological, and molecular abnormalities that may have a neurodevelopmental origin. MicroRNAs (miRNAs) are small noncoding RNA sequences critical to neurodevelopment and adult neuronal processes by coordinating the activity of multiple genes within biological networks. We examined the expression of 854 miRNAs in prefrontal cortical tissue from 100 control, schizophrenic, and bipolar subjects. The cyclic AMP-responsive element binding- and NMDA-regulated microRNA miR-132 was significantly down-regulated in both the schizophrenic discovery cohort and a second, independent set of schizophrenic subjects. Analysis of miR-132 target gene expression in schizophrenia gene-expression microarrays identified 26 genes up-regulated in schizophrenia subjects. Consistent with NMDA-mediated hypofunction observed in schizophrenic subjects, administration of an NMDA antagonist to adult mice results in miR-132 down-regulation in the prefrontal cortex. Furthermore, miR-132 expression in the murine prefrontal cortex exhibits significant developmental regulation and overlaps with critical neurodevelopmental processes during adolescence. Adult prefrontal expression of miR-132 can be down-regulated by pharmacologic inhibition of NMDA receptor signaling during a brief postnatal period. Several key genes, including DNMT3A, GATA2, and DPYSL3, are regulated by miR-132 and exhibited altered expression either during normal neurodevelopment or in tissue from adult schizophrenic subjects. Our data suggest miR-132 dysregulation and subsequent abnormal expression of miR-132 target genes contribute to the neurodevelopmental and neuromorphological pathologies present in schizophrenia.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/fisiopatología , Regulación de la Expresión Génica , MicroARNs/genética , Esquizofrenia/genética , Esquizofrenia/fisiopatología , Adulto , Animales , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Trastorno Bipolar/tratamiento farmacológico , Trastorno Bipolar/genética , Trastorno Bipolar/fisiopatología , Encéfalo/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Bases de Datos Genéticas , Demografía , Modelos Animales de Enfermedad , Factor de Transcripción GATA2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ratones , MicroARNs/metabolismo , Proteínas Musculares/metabolismo , N-Metilaspartato/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Sistemas de Lectura Abierta/genética , Reacción en Cadena de la Polimerasa , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo , Reproducibilidad de los Resultados , Esquizofrenia/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos
10.
Int J Neuropsychopharmacol ; 18(6)2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25522392

RESUMEN

BACKGROUND: Cognitive deficits are a core symptom of schizophrenia, yet they remain particularly resistant to treatment. The model provided by repeatedly exposing adult nonhuman primates to phencyclidine has generated important insights into the neurobiology of these deficits, but it remains possible that administration of this psychotomimetic agent during the pre-adult period, when the dorsolateral prefrontal cortex in human and nonhuman primates is still undergoing significant maturation, may provide a greater understanding of schizophrenia-related cognitive deficits. METHODS: The effects of repeated phencyclidine treatment on spine synapse number, dopamine turnover and BDNF expression in dorsolateral prefrontal cortex, and working memory accuracy were examined in pre-adult monkeys. RESULTS: One week following phencyclidine treatment, juvenile and adolescent male monkeys demonstrated a greater loss of spine synapses in dorsolateral prefrontal cortex than adult male monkeys. Further studies indicated that in juvenile males, a cognitive deficit existed at 4 weeks following phencyclidine treatment, and this impairment was associated with decreased dopamine turnover, decreased brain derived neurotrophic factor messenger RNA, and a loss of dendritic spine synapses in dorsolateral prefrontal cortex. In contrast, female juvenile monkeys displayed no cognitive deficit at 4 weeks after phencyclidine treatment and no alteration in dopamine turnover or brain derived neurotrophic factor messenger RNA or spine synapse number in dorsolateral prefrontal cortex. In the combined group of male and female juvenile monkeys, significant linear correlations were detected between dopamine turnover, spine synapse number, and cognitive performance. CONCLUSIONS: As the incidence of schizophrenia is greater in males than females, these findings support the validity of the juvenile primate phencyclidine model and highlight its potential usefulness in understanding the deficits in dorsolateral prefrontal cortex in schizophrenia and developing novel treatments for the cognitive deficits associated with schizophrenia.


Asunto(s)
Conducta Animal , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cognición , Dopamina/metabolismo , Fenciclidina , Corteza Prefrontal/metabolismo , Esquizofrenia/metabolismo , Psicología del Esquizofrénico , Médula Espinal/metabolismo , Sinapsis/metabolismo , Factores de Edad , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Masculino , Memoria a Corto Plazo , Corteza Prefrontal/fisiopatología , Esquizofrenia/inducido químicamente , Esquizofrenia/patología , Esquizofrenia/fisiopatología , Factores Sexuales , Médula Espinal/fisiopatología , Médula Espinal/ultraestructura , Sinapsis/ultraestructura , Factores de Tiempo
11.
Mol Ther ; 21(12): 2160-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23913185

RESUMEN

We combined viral vector delivery of human glial-derived neurotrophic factor (GDNF) with the grafting of dopamine (DA) precursor cells from fetal ventral mesencephalon (VM) to determine whether these strategies would improve the anti-Parkinson's effects in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys, an animal model for Parkinson's disease (PD). Both strategies have been reported as individually beneficial in animal models of PD, leading to clinical studies. GDNF delivery has also been reported to augment VM tissue implants, but no combined studies have been done in monkeys. Monkeys were treated with MPTP and placed into four balanced treatment groups receiving only recombinant adeno-associated virus serotype 5 (rAAV5)/hu-GDNF, only fetal DA precursor cells, both together, or a buffered saline solution (control). The combination of fetal precursors with rAAV5/hu-GDNF showed significantly higher striatal DA concentrations compared with the other treatments, but did not lead to greater functional improvement in this study. For the first time under identical conditions in primates, we show that all three treatments lead to improvement compared with control animals.


Asunto(s)
Dependovirus/genética , Dopamina/metabolismo , Trasplante de Tejido Fetal , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Intoxicación por MPTP/terapia , Mesencéfalo/trasplante , Enfermedad de Parkinson/terapia , Animales , Conducta Animal , Trasplante de Tejido Encefálico , Chlorocebus aethiops , Terapia Combinada , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Terapia Genética , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Virus de la Anemia Infecciosa Equina/genética , Intoxicación por MPTP/fisiopatología , Intoxicación por MPTP/psicología , Masculino , Mesencéfalo/citología , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/psicología
12.
Int J Neuropsychopharmacol ; 16(4): 905-12, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22947206

RESUMEN

Parkinson's disease is usually characterized as a movement disorder; however, cognitive abilities that are dependent on the prefrontal cortex decline at an early stage of the disease in most patients. The changes that underlie cognitive deficits in Parkinson's disease are not well understood. We hypothesize that reduced dopamine signalling in the prefrontal cortex in Parkinson's disease is a harbinger of detrimental synaptic changes in pyramidal neurons in the prefrontal cortex, whose function is necessary for normal cognition. Our previous data showed that monkeys exposed to the neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), but not exhibiting overt motor deficits (motor-asymptomatic), displayed cognitive deficits in prefrontal cortex-dependent tasks. The present results demonstrate that motor-asymptomatic MPTP-treated monkeys have a reduced dopamine concentration and a substantially lower number (50%) of asymmetric (excitatory) spine synapses in layer II/III, but not layer V, of the dorsolateral prefrontal cortex, compared to controls. In contrast, neither dopamine concentration nor asymmetric synapse number was altered in the entorhinal cortex of MPTP-treated monkeys. Together, these findings suggest that the number of asymmetric spine synapses on dendrites in the prefrontal cortex is dopamine-dependent and that the loss of synapses may be a morphological substrate of the cognitive deficits induced by a reduction in dopamine neurotransmission in this region.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Espinas Dendríticas/metabolismo , Dopamina/metabolismo , Trastornos Parkinsonianos/metabolismo , Corteza Prefrontal/metabolismo , Sinapsis/metabolismo , Animales , Chlorocebus aethiops , Trastornos del Conocimiento/patología , Espinas Dendríticas/patología , Masculino , Trastornos Parkinsonianos/patología , Corteza Prefrontal/patología , Sinapsis/patología
13.
Synapse ; 67(9): 580-5, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23468413

RESUMEN

Several addictive or neurotoxic drugs are dependent on the dopamine transporter (DAT) and/or vesicular monoamine transporter (VMAT2) to exert their detrimental effects on dopamine neurons. For example, methamphetamine and MPTP are substrates for both DAT and VMAT2, with the ratio of DAT to VMAT2 in striatum being a determinant of the degree of toxicity inflicted by these drugs on dopamine neurons. Thus, the susceptibility of dopamine neurons to agents whose pharmacology involves DAT and VMAT2 may vary during development if the ontogeny of DAT and VMAT2 differs, and this is relevant as exposure of dopamine neurons to toxic agents during development is hypothesized to underlie some neurological or psychiatric disorders. However, the relative expression of DAT and VMAT2 has not been studied in either primate or nonprimate fetal brain, and this was addressed in the present study by measuring the binding of specific radioligands of DAT and VMAT2 to striatal membranes from nonhuman primates at mid-gestation, late-gestation, and the postnatal and adult periods. Dopamine concentration was also determined in striatal tissue from the same brains. These data indicate that in striatum of primates, unlike rodents, there is a sharp increase in DAT and VMAT2 expression after mid-gestation, with adult levels being attained at the time of birth. In addition, this study demonstrated that there is a coordinated expression of DAT and VMAT2 from the time of mid-gestation to adulthood.


Asunto(s)
Cuerpo Estriado/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Animales , Chlorocebus aethiops , Cuerpo Estriado/embriología , Cuerpo Estriado/crecimiento & desarrollo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Ensayo de Unión Radioligante , Proteínas de Transporte Vesicular de Monoaminas/genética
14.
Biol Sex Differ ; 14(1): 65, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37770961

RESUMEN

BACKGROUND: Paraoxonase 2 (PON2) and neuronal uncoupling proteins (UCP4 and UCP5) possess antioxidant, anti-apoptotic activities and minimize accumulation of reactive oxygen species in mitochondria. While age and sex are risk factors for several disorders that are linked with oxidative stress, no study has explored the age- and sex-dependent expression of PON2 isoforms, UCP4 and UCP5 in primate brain or identified a drug to activate UCP4 and UCP5 in vivo. Preclinical studies suggest that the peroxisome proliferator-activated receptor gamma agonist, pioglitazone (PIO), can be neuroprotective, although the mechanism responsible is unclear. Our previous studies demonstrated that pioglitazone activates PON2 in primate brain and we hypothesized that pioglitazone also induces UCP4/5. This study was designed to elucidate the age- and sex-dependent expression of PON2 isoforms, UCP4 and UCP5, in addition to examining the impact of systemic PIO treatment on UCP4 and UCP5 expression in primate brain. METHODS: Western blot technique was used to determine the age- and sex-dependent expression of UCP4 and UCP5 in substantia nigra and striatum of African green monkeys. In addition, we tested the impact of daily oral pioglitazone (5 mg/kg/day) or vehicle for 1 or 3 weeks on expression of UCP4 and UCP5 in substantia nigra and striatum in adult male monkeys. PIO levels in plasma and cerebrospinal fluid (CSF) were determined using LC-MS. RESULTS: We found no sex-based difference in the expression of PON2 isoforms, UCP4 and UCP5 in striatum and substantia nigra of young monkeys. However, we discovered that adult female monkeys exhibit greater expression of PON2 isoforms than males in substantia nigra and striatum. Our data also revealed that adult male monkeys exhibit greater expression of UCP4 and UCP5 than females in substantia nigra but not in striatum. PIO increased UCP4 and UCP5 expression in substantia nigra and striatum at 1 week, but after 3 weeks of treatment this activation had subsided. CONCLUSIONS: Our findings demonstrate a sex-, age- and region-dependent profile to the expression of PON2, UCP4 and UCP5. These data establish a biochemical link between PPARγ, PON2, UCP4 and UCP5 in primate brain and demonstrate that PON2, UCP4 and UCP5 can be pharmacologically stimulated in vivo, revealing a novel mechanism for observed pioglitazone-induced neuroprotection. We anticipate that these outcomes will contribute to the development of novel neuroprotective treatments for Parkinson's disease and other CNS disorders.


Parkinson's disease (PD) is less common in women than men, which may be related to the protective effect of high levels of estrogens in women that maintain the activity of neuroprotective proteins in brain mitochondria. Our previous work suggests that paraoxonase-2 (PON2), uncoupling protein-4 (UCP4) and uncoupling protein-5 (UCP5) play vital roles in maintaining the health of brain dopamine neurons that are lost in PD. This work tested the hypothesis that female primate brains expresses higher levels of these proteins than males. In addition, this research investigated whether estrogen regulates the expression these factors and whether they can be pharmacologically activated later in life to protect dopamine neurons at a time when symptoms of PD typically emerge. The results indicate that before puberty when estrogen levels in females are relatively low, there is no difference in PON2, UCP4, UCP5 brain levels between males and females, but in adults PON2 is up to 3 × higher in females compared with males in regions relevant to PD, consistent with estrogen activation of PON2. Earlier studies have shown that pioglitazone can be neuroprotective in several adverse brain conditions, although the mechanism is not clear. The current research demonstrates that pioglitazone transiently activates by about twofold the expression of PON2, UCP4, UCP5 in vivo in primate brain, suggesting their involvement in the neuroprotective properties of the drug. Overall, the current data provides impetus for further work on activating protective factors that alter mitochondrial dynamics and function, leading to improved understanding and treatment of multiple diseases.


Asunto(s)
Encéfalo , Caracteres Sexuales , Animales , Femenino , Masculino , Chlorocebus aethiops , Pioglitazona/farmacología , Pioglitazona/metabolismo , Encéfalo/metabolismo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacología , Mitocondrias , Oxidación-Reducción
15.
Free Radic Biol Med ; 178: 215-218, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34890766

RESUMEN

Mitochondrial dysfunction and oxidative stress contribute to the neuropathology of neurodegenerative disorders such as Parkinson's disease (PD). Paraoxonase-2 (PON2) is a mitochondrial protein that mitigates oxidative stress, enhances mitochondrial function and exhibits anti-inflammatory properties. Previously, we have documented sex-based variation in PON2 with higher brain PON2 expression in female (2-fold) as compared to male African green monkeys. This aim of this study is to identify PON2 isoforms and explore the region-based variations in the protein level of PON2 in brain of African green monkeys. Male and female brain tissue samples (striatum, hippocampus, occipital cortex, dorsolateral prefrontal cortex) from African green monkeys (Chlorocebus sabaeus) were analyzed by western blotting technique for PON2 expression. We found two PON2 isoforms (39 and 41 kDa) in each examined brain region of male and female monkeys. Male monkeys showed no significant difference in the expression level of PON2 isoforms among different brain regions whereas female monkeys showed a significant difference in the expression level of PON2 isoforms in all examined regions except dorsolateral prefrontal cortex. In addition, the result revealed highest expression of PON2 protein in striatum compared to other brain regions in both male and female monkeys. This report is the first to quantify expression of PON2 isoforms in different brain regions and it also establishes the existence of sex as well as region-based variation in PON2 protein expression in primate brain. Since PON2 serves a protective role for dopaminergic neurons it should be considered as a druggable target for oxidative stress-related neurodegenerative disorders like PD. We anticipate that the outcome of this study will contribute to the development of neuroprotective strategies in PD.


Asunto(s)
Arildialquilfosfatasa , Corteza Prefontal Dorsolateral , Animales , Arildialquilfosfatasa/metabolismo , Encéfalo/metabolismo , Chlorocebus aethiops , Femenino , Masculino , Estrés Oxidativo , Isoformas de Proteínas/genética
16.
Neurochem Int ; 152: 105222, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34767873

RESUMEN

Paraoxonase-2 (PON2) enhances mitochondria function and protects against oxidative stress. Stimulating its expression has therapeutic potential for diseases where oxidative stress plays a significant role in the pathology, such as Parkinson's disease. Clinical and preclinical evidence suggest that the anti-diabetic drug pioglitazone may provide neuroprotection in Parkinson's disease, Alzheimer's disease, and stroke, but the biochemical pathway(s) responsible has not been fully elucidated. To determine the effect of pioglitazone on PON2 expression we treated male African green monkeys with oral pioglitazone (5 mg/kg/day) for 1 and 3 weeks. We found that pioglitazone increased PON2 mRNA and protein expression in brain following 1 week of treatment, however, by 3 weeks of treatment PON2 expression had returned to baseline. This transient increase was detected in substantia nigra, striatum, hippocampus, and dorsolateral prefrontal cortex The short-term impact of pioglitazone on PON2 expression in striatum may contribute to the discrepancy in the potency of the drug between short-term animal models and clinical trials for Parkinson's disease. Both PON2 and pioglitazone's receptor, peroxisome proliferator-activated receptor gamma (PPARγ), possess sex- and brain region-dependent expression, which may play a role in the short-term effect of pioglitazone and provide clues to extending the beneficial effects of PON2 activation.


Asunto(s)
Arildialquilfosfatasa/efectos de los fármacos , Encéfalo/efectos de los fármacos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Pioglitazona/farmacología , Factores Sexuales , Animales , Arildialquilfosfatasa/metabolismo , Encéfalo/metabolismo , Femenino , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Pioglitazona/metabolismo , Primates/metabolismo
17.
J Cell Mol Med ; 15(4): 747-55, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20406327

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder caused by the loss of dopaminergic neurons. Adult human endometrial derived stem cells (HEDSC), a readily obtainable type of mesenchymal stem-like cell, were used to generate dopaminergic cells and for transplantation. Cells expressing CD90, platelet derived growth factor (PDGF)-Rß and CD146 but not CD45 or CD31 were differentiated in vitro into dopaminergic neurons that exhibited axon projections, pyramidal cell bodies and dendritic projections that recapitulate synapse formation; these cells also expressed the neural marker nestin and tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis. Whole cell patch clamp recording identified G-protein coupled inwardly rectifying potassium current 2 channels characteristic of central neurons. A 1-methyl 4-phenyl 1,2,3,6-tetrahydro pyridine induced animal model of PD was used to demonstrate the ability of labelled HEDSC to engraft, migrate to the site of lesion, differentiate in vivo and significantly increase striatal dopamine and dopamine metabolite concentrations. HEDSC are a highly inducible source of allogenic stem cells that rescue dopamine concentrations in an immunocompetent PD mouse model.


Asunto(s)
Dopamina/biosíntesis , Endometrio/citología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Trasplante de Células Madre , Células Madre/citología , Adulto , Animales , Diferenciación Celular , Movimiento Celular , Modelos Animales de Enfermedad , Fenómenos Electrofisiológicos , Femenino , Citometría de Flujo , Humanos , Ratones , Neostriado/metabolismo , Neostriado/patología , Neurogénesis , Células Madre/metabolismo
18.
Neurobiol Dis ; 43(1): 152-62, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21406233

RESUMEN

Guanidinopropionic acid (GPA) increases AMPK activity, mitochondrial function and biogenesis in muscle and improves physiological function, for example during aging. Mitochondrial dysfunction is a major contributor to the pathogenesis of Parkinson's disease. Here we tested whether GPA prevents neurodegeneration of the nigrostriatal dopamine system in MPTP-treated mice. Mice were fed a diet of 1% GPA or normal chow for 4 weeks and then treated with either MPTP or saline. Indices of nigrostriatal function were examined by HPLC, immunohistochemistry, stereology, electron microscopy and mitochondrial respiration. MPTP intoxication decreased TH neurons in the SNpc of normal chow-fed mice; however GPA-fed mice remarkably exhibited no loss of TH neurons in the SNpc. MPTP caused a decrease in striatal dopamine of both normal chow- and GPA-fed mice, although this effect was significantly attenuated in GPA-fed mice. GPA-fed mice showed increased AMPK activity, mitochondrial respiration and mitochondrial number in nigrostriatal TH neurons, suggesting that the neuroprotective effects of GPA involved AMPK-dependent increases in mitochondrial function and biogenesis. MPTP treatment produced a decrease in mitochondrial number and volume in normal chow-fed mice but not GPA-fed mice. Our results show the neuroprotective properties of GPA in a mouse model of Parkinson's disease are partially mediated by AMPK and mitochondrial function. Mitochondrial dysfunction is a common problem in neurodegeneration and thus GPA may slow disease progression in other models of neurodegeneration.


Asunto(s)
Cuerpo Estriado/fisiología , Dopamina/metabolismo , Guanidinas/farmacología , Mitocondrias/fisiología , Fármacos Neuroprotectores/administración & dosificación , Trastornos Parkinsonianos/tratamiento farmacológico , Propionatos/farmacología , Sustancia Negra/fisiología , Regulación hacia Arriba/fisiología , Administración Oral , Animales , Cuerpo Estriado/efectos de los fármacos , Dopamina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Regulación hacia Arriba/efectos de los fármacos
19.
Int J Neuropsychopharmacol ; 14(10): 1411-5, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21733230

RESUMEN

Schizophrenia patients, long-term abusers of phencyclidine (PCP), and monkeys treated with PCP all exhibit enduring cognitive deficits. Evidence indicates that loss of prefrontal cortex spine synapses results in cognitive dysfunction, suggesting the presence of synaptic pathology in the monkey PCP model; however, there is no direct evidence of such changes. In this study we use the monkey PCP model of schizophrenia to investigate at the ultrastructural level whether remodelling of dorsolateral prefrontal cortex (DLPFC) asymmetric spine synapses occurs following PCP. Subchronic PCP treatment resulted in a decrease in the number of asymmetric spine synapses, which was greater in layer II/III than layer V of DLPFC, compared to vehicle-treated controls. This decrease may contribute to PCP-induced cognitive dysfunction in the non-human primate model and perhaps in schizophrenia. Thus, the synapse loss in the PCP model provides a novel target for the development of potential treatments of cognitive dysfunction in this model and in schizophrenia.


Asunto(s)
Trastornos del Conocimiento/patología , Cognición , Espinas Dendríticas/patología , Fenciclidina , Corteza Prefrontal/patología , Esquizofrenia/patología , Sinapsis/patología , Animales , Chlorocebus aethiops , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/psicología , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Masculino , Microscopía Electrónica de Transmisión , Corteza Prefrontal/ultraestructura , Esquizofrenia/inducido químicamente , Psicología del Esquizofrénico , Sinapsis/ultraestructura
20.
Mol Ther ; 18(3): 588-93, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20010918

RESUMEN

Vectors derived from adeno-associated virus (AAV) are promising candidates for neural cell transduction in vivo because they are nonpathogenic and achieve long-term transduction in the central nervous system. AAV serotype 2 (AAV2) is the most widely used AAV vector in clinical trials based largely on its ability to transduce neural cells in the rodent and primate brain. Prior work in rodents suggests that other serotypes might be more efficient; however, a systematic evaluation of vector transduction efficiency has not yet been performed in the primate brain. In this study, AAV viral vectors of serotypes 1-6 with an enhanced green-fluorescent protein (GFP) reporter gene were generated at comparable titers, and injected in equal amounts into the brains of Chlorocebus sabaeus. Vector injections were placed in the substantia nigra (SN) and the caudate nucleus (CD). One month after injection, immunohistochemistry for GFP was performed and the total number of GFP+ cells was calculated using unbiased stereology. AAV5 was the most efficient vector, not only transducing significantly more cells than any other serotype, but also transducing both NeuN+ and glial-fibrillary-acidic protein positive (GFAP+) cells. These results suggest that AAV5 is a more effective vector than AAV2 at delivering potentially therapeutic transgenes to the nigrostriatal system of the primate brain.


Asunto(s)
Cuerpo Estriado/metabolismo , Dependovirus/metabolismo , Sustancia Negra/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica/métodos , Microscopía Confocal , Neuronas/metabolismo , Primates
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA