Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Yale J Biol Med ; 91(3): 333-342, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30258320

RESUMEN

The shortage of human organs for transplantation is a devastating medical problem. One way to expand organ supply is to derive functional organs from patient-specific stem cells. Due to their capacity to grow indefinitely in the laboratory and differentiate into any cell type of the human body, patient-specific pluripotent stem (PS) cells harbor the potential to provide an inexhaustible supply of donor cells for transplantation. However, current efforts to generate functional organs from PS cells have so far been unsuccessful. An alternative and promising strategy is to generate human organs inside large animal species through a technique called interspecies blastocyst complementation. In this method, animals comprised of cells from human and animal species are generated by injecting donor human PS cells into animal host embryos. Critical genes for organ development are knocked out by genome editing, allowing donor human PS cells to populate the vacated niche. In principle, this experimental approach will produce a desired organ of human origin inside a host animal. In this mini-review, we focus on recent advances that may bring the promise of blastocyst complementation to clinical practice. While CRISPR/Cas9 has accelerated the creation of transgenic large animals such as pigs and sheep, we propose that further advances in the generation of chimera-competent human PS cells are needed to achieve interspecies blastocyst complementation. It will also be necessary to define the constituents of the species barrier, which inhibits efficient colonization of host animal embryos with human cells. Interspecies blastocyst complementation is a promising approach to help overcome the organ shortage facing the practice of clinical medicine today.


Asunto(s)
Células Madre Pluripotentes/citología , Animales , Diferenciación Celular/fisiología , Humanos , Trasplante de Órganos
3.
Stroke ; 47(4): 1109-16, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26956259

RESUMEN

BACKGROUND AND PURPOSE: Ischemic stroke is the leading cause of upper extremity motor impairments. Although several well-characterized experimental stroke models exist, modeling of upper extremity motor impairments, which are unique to primates, is not well established. Cortical representation of dexterous movements in nonhuman primates is functionally and topographically similar to that in humans. In this study, we characterize the African green monkey model of focal ischemia reperfusion with a defined syndrome, impaired dexterous movements. METHODS: Cerebral ischemia was induced by transient occlusion of the M3 segment of the left middle cerebral artery. Motor and cognitive functions after stroke were evaluated using the object retrieval task with barrier-detour. Postmortem magnetic resonance imaging and histopathology were performed to map and characterize the infarct. RESULTS: The middle cerebral artery occlusion consistently produced a necrotic infarct localized in the sensorimotor cortex in the middle cerebral artery territory. The infarction was reproducible and resulted in significant loss of fine motor function characterized by impaired dexterity. No significant cognitive impairment was detected. Magnetic resonance imaging and histopathology demonstrated consistent and significant loss of tissue on the left parietal cortex by the central sulcus covering the sensorimotor area. The results suggest that this species has less collateralization, which closely resembles humans. CONCLUSIONS: The reported nonhuman primate model produces a defined and reproducible syndrome relevant to our understanding of ischemic stroke, cortical representation, and sensorimotor integration controlling dexterous movements. This model will be useful in basic and translational research addressing loss of arm function and dexterity.


Asunto(s)
Brazo/fisiopatología , Isquemia Encefálica/fisiopatología , Cognición/fisiología , Dedos/fisiopatología , Desempeño Psicomotor/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/fisiopatología , Destreza Motora/fisiología , Corteza Sensoriomotora/fisiopatología
4.
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
5.
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
6.
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
7.
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
8.
J Med Primatol ; 41(3): 158-62, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22296111

RESUMEN

BACKGROUND: Focus on the placenta as an agent of fetal development and offspring health outcomes is growing. Primate research facilities or zoos may collect and fix placental tissue for long-term storage, but little is known about the effects of formalin fixation on the non-human primate placenta. METHODS: We obtained 48 vervet monkey placentas from the St. Kitts Biomedical Research Foundation. We investigated via correlation coefficients and ANOVAs the effects of gestational age and original fresh weight on weight change due to fixation. We also used linear regression models to determine whether fixed tissue weight was predictive of fresh weight and gestational age. RESULTS: Although the vervet monkey placenta is described as bidiscoid, 14.6% of the placentas in this sample were fused into a single mass. A decrease in weight was the most common response to formalin fixation, with the greatest degree of loss experienced by the heaviest placentas (ANOVA, F=5.99, P=0.005). Gestational age was unrelated to weight change. Those placentas that increased in weight had the lowest fresh weights. Fixed weights significantly predicted both fresh weight and gestational age (r(2) =0.78, P<0.00001; r(2) =0.76, P<0.00001, respectively). CONCLUSIONS: This paper adds to a sparse literature on the vervet monkey placenta. That fixed placentas are excellent predictors of both fresh weight and gestational age suggests that banked tissue may be a valuable resource for reconstructing aspects of individual life history, although caution must be exercised given the variability of weight change as a function of original placental size.


Asunto(s)
Chlorocebus aethiops/anatomía & histología , Placenta/anatomía & histología , Conservación de Tejido/veterinaria , Animales , Chlorocebus aethiops/fisiología , Femenino , Edad Gestacional , Tamaño de los Órganos , Embarazo
9.
Am J Primatol ; 74(5): 433-41, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22311723

RESUMEN

Ultrasound assessments of fetal growth have been used in other species of primates to estimate fetal age, but there are no published morphometrics for the St. Kitts green monkey (Chlorocebus sabaeus), a species that has been important for studies of transplantation of fetal tissue into the brain as potential treatment for degenerative disease. Previous studies with other primate species have used relatively small numbers of pregnancies, measured repeatedly, to derive regressions for predicting fetal age from ultrasound studies. The present study derives data from 967 pregnancies, collected over a 9-year period, for predicting fetal age from ultrasound measurements of crown rump length, biparietal diameter, head circumference, abdominal circumference, and femur length in the St. Kitts green monkey. Linear and polynomial regressions were determined from pregnancies dated from a 3- to 4-day breeding period and confirmed in a second, independent group of pregnant monkeys with more extended breeding times to determine their accuracy for predicting fetal age. Although similar to morphometrics reported in other monkey species, there were some significant differences. These data will improve the estimates of fetal ages in previously published studies of St. Kitts green monkeys and provide more precise estimates of fetal age in studies of fetal development, genomics, and reproductive toxicology.


Asunto(s)
Chlorocebus aethiops/fisiología , Edad Gestacional , Resultado del Embarazo/veterinaria , Animales , Antropometría , Femenino , Modelos Lineales , Modelos Estadísticos , Embarazo , San Kitts y Nevis , Ultrasonografía Prenatal/veterinaria
10.
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
11.
Stem Cells Dev ; 29(19): 1294-1307, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32715987

RESUMEN

Patient-specific stem cells derived from somatic cell nuclear transfer (SCNT) embryos or from induced pluripotent stem cells (iPSCs) could be used to treat various diseases with minimal immune rejection. Many studies using these cells have been conducted in rats and mice; however, there exist numerous dissimilarities between the rodents and humans limiting the clinical predictive power and experimental utility of rodent experiments alone. Nonhuman primates (NHPs) share greater homology to human than rodents in all respects, including genomics, physiology, biochemistry, and the immune system. Thus, experimental data obtained from monkey studies would be more predictive for designing an effective cell replacement therapy in humans. Unfortunately, there are few iPSC lines and even fewer SCNT lines that have been derived in NHPs, hampering broader studies in regenerative medicine. One promising potential therapy would be the replacement of dopamine neurons that are lost in Parkinson's disease. After dopamine depletion by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), the African green monkey (Chlorocebus sabaeus) shows the most complete model of Parkinsonism compared with other species and brain pathology and behavioral changes are almost identical to those in humans after accidental exposure to MPTP. Therefore, we have developed a SCNT procedure to generate multiple pluripotent stem cell lines in this species for studies of possible treatment of Parkinsonism and for comparing with cells derived from iPSCs. Using 24 female monkeys as egg donors and 7 somatic cell donor monkeys, we have derived 11 SCNT embryonic stem cell lines that expressed typical stemness genes and formed all three germ layer derivatives. We also derived two iPSC lines using an episome-mediated reprogramming factor delivery system. This report describes the process for deriving these cell lines and proving their pluripotency for differentiation into various potentially therapeutic cells.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Técnicas de Transferencia Nuclear , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Bandeo Cromosómico , Clonación de Organismos , Medios de Cultivo , Análisis Citogenético , ADN/genética , Neuronas Dopaminérgicas/metabolismo , Desarrollo Embrionario , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Genotipo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Mitocondrias/metabolismo , Ovario/fisiología , Tirosina 3-Monooxigenasa/metabolismo
12.
Mol Ther ; 16(7): 1252-60, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18500254

RESUMEN

We report a DNA shuffling-based approach for developing cell type-specific vectors through directed evolution. Capsid genomes of adeno-associated virus (AAV) serotypes 1-9 were randomly fragmented and reassembled using PCR to generate a chimeric capsid library. A single infectious clone (chimeric-1829) containing genome fragments from AAV1, 2, 8, and 9 was isolated from an integrin minus hamster melanoma cell line previously shown to have low permissiveness to AAV. Molecular modeling studies suggest that AAV2 contributes to surface loops at the icosahedral threefold axis of symmetry, while AAV1 and 9 contribute to two- and fivefold symmetry interactions, respectively. The C-terminal domain (AAV9) was identified as a critical structural determinant of melanoma tropism through rational mutagenesis. Chimeric-1829 utilizes heparan sulfate as a primary receptor and transduces melanoma cells more efficiently than all serotypes. Further, chimeric-1829 demonstrates altered tropism in rodent skeletal muscle, liver, and brain including nonhuman primates. We determined a unique immunological profile based on neutralizing antibody (NAb) titer and crossreactivity studies strongly supporting isolation of a synthetic laboratory-derived capsid variant. Application of this technology to alternative cell/tissue types using AAV or other viral capsid sequences is likely to yield a new class of biological nanoparticles as vectors for human gene transfer.


Asunto(s)
Barajamiento de ADN , Dependovirus/genética , Vectores Genéticos/aislamiento & purificación , Genoma Viral/genética , Nanopartículas , Animales , Anticuerpos/inmunología , Encéfalo/metabolismo , Cápside/inmunología , Cricetinae , Dependovirus/ultraestructura , Evolución Molecular Dirigida , Biblioteca de Genes , Vectores Genéticos/genética , Humanos , Hígado/metabolismo , Melanoma , Ratones , Ratones Endogámicos BALB C , Músculo Esquelético/metabolismo , Primates , Transducción Genética , Internalización del Virus
13.
Methods Mol Biol ; 2005: 221-231, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31175656

RESUMEN

The search for a better animal model to simulate human disease has been a "holy grail" of biomedical research for decades. Recent identification of different types of pluripotent stem cells (PS cells) and advances in chimera research might soon permit the generation of interspecies chimeras from closely related species, such as those between humans and other primates. Here, we suggest that the creation of human-primate chimeras-specifically, the transfer of human stem cells into (non-ape) primate hosts-could surpass the limitations of current monkey models of neurological and psychiatric disease, but would also raise important ethical considerations concerning the use of monkeys in invasive research. Questions regarding the scientific value and ethical concerns raised by the prospect of human-monkey chimeras are more urgent in light of recent advances in PS cell research and attempts to generate interspecies chimeras between humans and animals. While some jurisdictions prohibit the introduction of human PS cells into monkey preimplantation embryos, other jurisdictions may permit and even encourage such experiments. Therefore, it is useful to consider blastocyst complementation experiments more closely in light of advances that could make these chimeras possible and to consider the ethical and political issues that are raised.


Asunto(s)
Discusiones Bioéticas , Modelos Animales de Enfermedad , Ética en Investigación , Trasplante de Células Madre/ética , Quimera por Trasplante , Animales , Haplorrinos , Humanos
14.
Neuropsychopharmacology ; 33(3): 491-6, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17507917

RESUMEN

The mechanism responsible for the therapeutic effects of the prototypical atypical antipsychotic drug, clozapine, is still not understood; however, there is persuasive evidence from in vivo studies in normal rodents and primates that the ability to elevate dopamine neurotransmission preferentially in the prefrontal cortex is a key component to the beneficial effects of clozapine in schizophrenia. Theoretically, such an effect of clozapine would counteract the deficient dopaminergic innervation of the prefrontal cortex that appears to be part of the pathophysiology of schizophrenia. We have previously shown that following repeated, intermittent administrations of phencyclidine to monkeys there is lowered prefrontal cortical dopamine transmission and impairment of cognitive performance that is dependent on the prefrontal cortex; these biochemical and behavioral changes therefore model certain aspects of schizophrenia. We now investigate the effects of clozapine on the dopamine projections to prefrontal cortex, nucleus accumbens, and striatum in control monkeys and in those withdrawn from repeated phencyclidine treatment, using a dose regimen of clozapine that ameliorates the cognitive deficits described in the primate phencyclidine (PCP) model. In normal monkeys, clozapine elevated dopamine turnover in all prefrontal cortical, but not subcortical, regions analyzed. In the primate PCP model, clozapine normalized dopamine (DA) turnover in the dorsolateral prefrontal cortex, prelimbic cortex, and cingulate cortex. Thus, the present data support the hypothesis that the therapeutic effects of clozapine in this primate model and perhaps in schizophrenia may be related at least in part to the restoration of DA tone in the prefrontal cortex.


Asunto(s)
Antipsicóticos/farmacología , Clozapina/farmacología , Dopamina/fisiología , Alucinógenos/toxicidad , Fenciclidina/toxicidad , Corteza Prefrontal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Química Encefálica/efectos de los fármacos , Chlorocebus aethiops , Dopamina/metabolismo , Femenino , Masculino , Memoria/efectos de los fármacos , Corteza Prefrontal/metabolismo , Desempeño Psicomotor/efectos de los fármacos
15.
Stem Cells Dev ; 27(23): 1599-1604, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30319057

RESUMEN

The search for a better animal model to simulate human disease has been a "holy grail" of biomedical research for decades. Recent identification of different types of pluripotent stem (PS) cells and advances in chimera research might soon permit the generation of interspecies chimeras from closely related species, such as those between humans and other primates. In this study, we suggest that the creation of human-primate chimeras-specifically, the transfer of human stem cells into (non-ape) primate hosts-could not only surpass the limitations of current monkey models of neurological and psychiatric disease but would also raise important ethical considerations concerning the use of monkeys in invasive research. Questions regarding the scientific value and ethical concerns raised by the prospect of human-monkey chimeras are more urgent in light of recent advances in PS cell research and attempts to generate interspecies chimeras between humans and animals. While some jurisdictions prohibit the introduction of human PS cells into monkey preimplantation embryos, other jurisdictions may permit and even encourage such experiments. Therefore, it is useful to consider blastocyst complementation experiments more closely in light of advances that could make these chimeras possible and to consider the ethical and political issues that are raised.


Asunto(s)
Modelos Animales de Enfermedad , Haplorrinos/genética , Trastornos Mentales/genética , Enfermedades Neurodegenerativas/genética , Quimera por Trasplante/genética , Animales , Investigaciones con Embriones/ética , Haplorrinos/fisiología , Humanos , Trastornos Mentales/patología , Enfermedades Neurodegenerativas/patología , Trasplante de Células Madre/ética , Trasplante de Células Madre/métodos , Trasplante de Células Madre/normas , Quimera por Trasplante/fisiología
17.
J Comp Neurol ; 525(3): 498-512, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27418401

RESUMEN

Clinical trials testing the hypothesis that fetal dopamine grafts would provide antiparkinsonian benefit in patients who had already developed side effects from their long-term use of L-dopa revealed, in some cases, the presence of dyskinesias even in the absence of L-dopa. The form, intensity, and frequency of these dyskinesias were quite variable, but their manifestation slowed the clinical development of cell replacement therapies. Rodent models of graft-induced dyskinesias (GIDs) have been proposed, but their accuracy in modeling GIDs has been questioned because they usually require amphetamine for their presentation. The present study attempted to model GIDs in parkinsonian monkeys and, for the first time, to test the effect of grafts on previously dyskinetic monkeys. Toward this end, monkeys were rendered parkinsonian with n-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and dyskinetic with levodopa. They then received intraputamenal grafts of fetal dopaminergic cells, control cerebellar cells, or vehicle bilaterally and were studied for 18 months. Dopaminergic cells were grafted in a manner designed to produce either "hot spot" or "widespread" striatal innervation. Although levodopa-induced dyskinesias could be elicited postoperatively, GIDs were never observed in any animal at any time after grafting. Grafted monkeys were also challenged with levodopa but did not show any greater responses to these challenges than before grafting. These studies support the development of future dopamine neuron cell transplantation therapy-based approaches, indicating that in relevant primate models with appropriate cell preparation methodology, with successful graft survival and putamenal dopamine innervation, there is no evidence of graft-induced dyskinesias. J. Comp. Neurol. 525:498-512, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Cerebelo/trasplante , Neuronas Dopaminérgicas/trasplante , Discinesia Inducida por Medicamentos/fisiopatología , Trasplante de Tejido Fetal , Intoxicación por MPTP/terapia , Mesencéfalo/trasplante , Neuronas/trasplante , Animales , Antiparkinsonianos/toxicidad , Calbindinas/metabolismo , Núcleo Caudado/patología , Núcleo Caudado/fisiopatología , Cerebelo/metabolismo , Chlorocebus aethiops , Dopamina/administración & dosificación , Dopamina/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Levodopa/toxicidad , Intoxicación por MPTP/patología , Intoxicación por MPTP/fisiopatología , Masculino , Mesencéfalo/embriología , Mesencéfalo/metabolismo , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Putamen/patología , Putamen/fisiopatología , Putamen/cirugía , Tirosina 3-Monooxigenasa/metabolismo
18.
Front Neurosci ; 10: 12, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26858591

RESUMEN

Accumulation of alpha-synuclein (α-syn) in Lewy bodies and neurites of midbrain dopamine neurons is diagnostic for Parkinson's disease (PD), leading to the proposal that PD is a toxic gain-of-function synucleinopathy. Here we discuss the alternative viewpoint that α-syn displacement from synapses by misfolding and aggregation results in a toxic loss-of-function. In support of this hypothesis we provide evidence from our pilot study demonstrating that knockdown of endogenous α-syn in dopamine neurons of non-human primates reproduces the pattern of nigrostriatal degeneration characteristic of PD.

19.
Cell Transplant ; 25(11): 1945-1966, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27213850

RESUMEN

Cell therapy has attracted considerable interest as a promising therapeutic alternative for patients with Parkinson's disease (PD). Clinical studies have shown that grafted fetal neural tissue can achieve considerable biochemical and clinical improvements in PD. However, the source of fetal tissue grafts is limited and ethically controversial. Human parthenogenetic stem cells offer a good alternative because they are derived from unfertilized oocytes without destroying potentially viable human embryos and can be used to generate an unlimited supply of neural cells for transplantation. We have previously reported that human parthenogenetic stem cell-derived neural stem cells (hpNSCs) successfully engraft, survive long term, and increase brain dopamine (DA) levels in rodent and nonhuman primate models of PD. Here we report the results of a 12-month transplantation study of hpNSCs in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned African green monkeys with moderate to severe clinical parkinsonian symptoms. The hpNSCs manufactured under current good manufacturing practice (cGMP) conditions were injected bilaterally into the striatum and substantia nigra of immunosuppressed monkeys. Transplantation of hpNSCs was safe and well tolerated by the animals with no dyskinesia, tumors, ectopic tissue formation, or other test article-related serious adverse events. We observed that hpNSCs promoted behavioral recovery; increased striatal DA concentration, fiber innervation, and number of dopaminergic neurons; and induced the expression of genes and pathways downregulated in PD compared to vehicle control animals. These results provide further evidence for the clinical translation of hpNSCs and support the approval of the world's first pluripotent stem cell-based phase I/IIa study for the treatment of PD (Clinical Trial Identifier NCT02452723).


Asunto(s)
Intoxicación por MPTP/terapia , Células-Madre Neurales/trasplante , Recuperación de la Función/fisiología , Animales , Conducta Animal , Encéfalo/metabolismo , Encéfalo/patología , Diferenciación Celular , Células Cultivadas , Chlorocebus aethiops , Análisis por Conglomerados , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Femenino , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Inmunohistoquímica , Cariotipo , Intoxicación por MPTP/inducido químicamente , Intoxicación por MPTP/patología , Masculino , Células-Madre Neurales/citología , Partenogénesis
20.
Hum Gene Ther ; 16(12): 1484-503, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16390279

RESUMEN

Late infantile neuronal ceroid lipofuscinosis (LINCL), a pediatric autosomal recessive neurodegenerative lysosomal storage disorder, results from mutations in the CLN2 gene and consequent deficiency in tripeptidyl-peptidase I (TPP-I) and progressive destruction of neurons. We have previously demonstrated that CNS gene transfer of AAV2(CU)hCLN2 (an AAV2-based vector expressing the human CLN2 cDNA) in rats and nonhuman primates mediates long-term TPP-I expression in the CNS neurons [Sondhi, D., Peterson, D.A., Giannaris, E.L., Sanders, C.T., Mendez, B.S., De, B., Rostkowski, A., Blancard, B., Bjugstad, K., Sladek, J.R., Redmond, D.E., Leopold, P.L., Kaminsky, S.M., Hackett, N.R., and Crystal, R.G. (2005). Gene Ther. 12, 1618-1632]. The present study tests the hypothesis that direct CNS administration of a clinical-grade AAV2(CU)hCLN2 vector to the CNS of rats and nonhuman primates at doses scalable to humans has a long-term safety profile acceptable for initiating clinical trials. Fischer 344 rats were injected bilaterally via the striatum with 2 x 10(10) particle units (PU) of AAV2(CU)hCLN2, using saline as a control. At 13, 26, and 52 weeks, vector and phosphate-buffered salineinjected rats were killed (n = 6 per time point), and blood, brain, and distant organs were assessed. There were no biologically significant differences between control and vector groups for complete blood count, serum chemistry, and neutralizing anti-AAV2 antibody levels. CNS administration of AAV2 CUhCLN2 did not result in any pathological changes in the brain that were attributable to the vector, although microscopic changes were observed along the track consistent with needle trauma. A total dose of 3.6 x 10(10) or 3.6 x 10(11) PU of AAV2(CU)hCLN2 was administered to the CNS of African Green monkeys at 12 locations, targeting the caudate nucleus, hippocampus, and overlying cortices. Monkeys (n = 3 at each dose) were killed 1, 13, 26, or 52 weeks after injection. Controls included sham-injected, saline-injected, and AAV2(CU)Null-injected (3.6 x 10(11) PU) monkeys. There were no biologically significant differences among vector-injected and control groups in any parameter of the general assessment, complete blood count, or serum chemistry assessed at multiple time points after vector administration. Importantly, no abnormal behavior was observed in any group in videotaped neurological assessment, where behaviors were quantified before administration and at multiple time points afterward. Histopathological examination of the CNS demonstrated that 1 week after administration, AAV2(CU)hCLN2 produced transient minor white matter edema with reactive glial cells in the corona radiata of the cerebrum along the injection track and in the surrounding white matter. This abnormality was not observed at 13, 26, or 52 weeks. Together with the long-term gene expression after gene transfer, these findings supported the initiation of clinical trials to assess the safety of AAV2(CU)hCLN2 administration to individuals with LINCL.


Asunto(s)
Técnicas de Transferencia de Gen/efectos adversos , Terapia Genética , Vectores Genéticos/administración & dosificación , Lipofuscinosis Ceroideas Neuronales/terapia , Animales , Edema Encefálico/etiología , Sistema Nervioso Central , Chlorocebus aethiops , Dependovirus/genética , Expresión Génica , Terapia Genética/efectos adversos , Masculino , Lipofuscinosis Ceroideas Neuronales/genética , Plásmidos/genética , Ratas , Ratas Endogámicas F344 , Tripeptidil Peptidasa 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA