Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 290
Filter
1.
Expert Rev Clin Immunol ; 18(1): 15-29, 2022 01.
Article in English | MEDLINE | ID: mdl-34882509

ABSTRACT

INTRODUCTION: More than one million breast augmentation procedures using silicone breast implants (SBI) have been performed worldwide. Adverse events of SBI include local complications such as pain, swelling, redness, infections, capsular contracture, implant rupture, and gel-bleed. Furthermore, patients experience systemic symptoms such as chronic fatigue, arthralgias, myalgias, pyrexia, sicca, and cognitive dysfunction. These symptoms received different names such as autoimmune/autoinflammatory syndrome induced by adjuvants (ASIA) due to silicone incompatibility syndrome and breast implant illness (BII). Because of chronic immune activation, BII/ASIA, allergies, autoimmune diseases, immune deficiencies, and finally lymphomas may develop in SBI patients. AREAS COVERED: Causality for SBI-related BII/ASIA is reviewed. To address the role of silicone implants in promoting causality, we utilized the Bradford Hill criteria, with results highlighted in this article. EXPERT OPINION: We conclude that there is a causal association between SBIs and BII/ASIA. Using data derived from patients with BII/ASIA and from other medically implanted devices, there appears to be clear pathogenic relationship between SBI and BII/ASIA. Breast implants cause characteristic systemic reactions in certain women, leading to symptoms of sufficient severity to warrant device removal. The morbidity suffered is variable. SBI removal resolves the symptoms in most women, and removal is the most effective treatment.


Subject(s)
Autoimmune Diseases , Breast Implantation , Breast Implants , Immunologic Deficiency Syndromes , Autoimmune Diseases/etiology , Breast Implantation/adverse effects , Breast Implants/adverse effects , Female , Humans , Immunologic Deficiency Syndromes/etiology , Silicones/adverse effects
2.
Oper Dent ; 46(1): 100-106, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33882137

ABSTRACT

CLINICAL RELEVANCE: Removing laminate veneers on anterior teeth by using an Er,Cr:YSGG dental laser can be completed faster than previously reported while maintaining thermal safety.


Subject(s)
Lasers, Solid-State , Aluminum Silicates , Ceramics , Lasers, Solid-State/therapeutic use
3.
Stem Cells Dev ; 29(19): 1294-1307, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32715987

ABSTRACT

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.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Nuclear Transfer Techniques , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Chromosome Banding , Cloning, Organism , Culture Media , Cytogenetic Analysis , DNA/genetics , Dopaminergic Neurons/metabolism , Embryonic Development , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Genotype , Humans , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Ovary/physiology , Tyrosine 3-Monooxygenase/metabolism
4.
Comput Methods Programs Biomed ; 186: 105184, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31715280

ABSTRACT

BACKGROUND AND OBJECTIVE: Model-based lung mechanics monitoring can provide clinically useful information for guiding mechanical ventilator treatment in intensive care. However, many methods of measuring lung mechanics are not appropriate for both fully and partially sedated patients, and are unable provide lung mechanics metrics in real-time. This study proposes a novel method of using lung mechanics identified during passive expiration to estimate inspiratory lung mechanics for spontaneously breathing patients. METHODS: Relationships between inspiratory and expiratory modeled lung mechanics were identified from clinical data from 4 fully sedated patients. The validity of these relationships were assessed using data from a further 4 spontaneously breathing patients. RESULTS: For the fully sedated patients, a linear relationship was identified between inspiratory and expiratory elastance, with slope 1.04 and intercept 1.66. The r value of this correlation was 0.94. No cohort-wide relationship was determined for airway resistance. Expiratory elastance measurements in spontaneously breathing patients were able to produce reasonable estimates of inspiratory elastance after adjusting for the identified difference between them. CONCLUSIONS: This study shows that when conventional methods fail, typically ignored expiratory data may be able to provide clinicians with the information needed about patient condition to guide MV therapy.


Subject(s)
Exhalation , Inhalation , Respiration , Airway Resistance , Humans , Models, Biological , Respiration, Artificial
5.
Methods Mol Biol ; 2005: 221-231, 2019.
Article in English | MEDLINE | ID: mdl-31175656

ABSTRACT

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.


Subject(s)
Bioethical Issues , Disease Models, Animal , Ethics, Research , Stem Cell Transplantation/ethics , Transplantation Chimera , Animals , Haplorhini , Humans
6.
Biochem Biophys Res Commun ; 510(1): 78-84, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30660369

ABSTRACT

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.


Subject(s)
Chimera/embryology , Extracellular Signal-Regulated MAP Kinases/physiology , Pluripotent Stem Cells/cytology , Animals , Bioethics , Cells, Cultured , Chlorocebus aethiops , Humans , Kruppel-Like Factor 4 , Organogenesis
7.
Stem Cells Dev ; 27(23): 1599-1604, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30319057

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Haplorhini/genetics , Mental Disorders/genetics , Neurodegenerative Diseases/genetics , Transplantation Chimera/genetics , Animals , Embryo Research/ethics , Haplorhini/physiology , Humans , Mental Disorders/pathology , Neurodegenerative Diseases/pathology , Stem Cell Transplantation/ethics , Stem Cell Transplantation/methods , Stem Cell Transplantation/standards , Transplantation Chimera/physiology
8.
Yale J Biol Med ; 91(3): 333-342, 2018 09.
Article in English | MEDLINE | ID: mdl-30258320

ABSTRACT

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.


Subject(s)
Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/physiology , Humans , Organ Transplantation
9.
Math Biosci ; 284: 21-31, 2017 02.
Article in English | MEDLINE | ID: mdl-27301378

ABSTRACT

Randomised control trials have sought to seek to improve mechanical ventilation treatment. However, few trials to date have shown clinical significance. It is hypothesised that aside from effective treatment, the outcome metrics and sample sizes of the trial also affect the significance, and thus impact trial design. In this study, a Monte-Carlo simulation method was developed and used to investigate several outcome metrics of ventilation treatment, including 1) length of mechanical ventilation (LoMV); 2) Ventilator Free Days (VFD); and 3) LoMV-28, a combination of the other metrics. As these metrics have highly skewed distributions, it also investigated the impact of imposing clinically relevant exclusion criteria on study power to enable better design for significance. Data from invasively ventilated patients from a single intensive care unit were used in this analysis to demonstrate the method. Use of LoMV as an outcome metric required 160 patients/arm to reach 80% power with a clinically expected intervention difference of 25% LoMV if clinically relevant exclusion criteria were applied to the cohort, but 400 patients/arm if they were not. However, only 130 patients/arm would be required for the same statistical significance at the same intervention difference if VFD was used. A Monte-Carlo simulation approach using local cohort data combined with objective patient selection criteria can yield better design of ventilation studies to desired power and significance, with fewer patients per arm than traditional trial design methods, which in turn reduces patient risk. Outcome metrics, such as VFD, should be used when a difference in mortality is also expected between the two cohorts. Finally, the non-parametric approach taken is readily generalisable to a range of trial types where outcome data is similarly skewed.


Subject(s)
Models, Theoretical , Monte Carlo Method , Randomized Controlled Trials as Topic/statistics & numerical data , Respiration, Artificial/statistics & numerical data , Sample Size , Humans
10.
J Comp Neurol ; 525(3): 498-512, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27418401

ABSTRACT

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.


Subject(s)
Cerebellum/transplantation , Dopaminergic Neurons/transplantation , Dyskinesia, Drug-Induced/physiopathology , Fetal Tissue Transplantation , MPTP Poisoning/therapy , Mesencephalon/transplantation , Neurons/transplantation , Animals , Antiparkinson Agents/toxicity , Calbindins/metabolism , Caudate Nucleus/pathology , Caudate Nucleus/physiopathology , Cerebellum/metabolism , Chlorocebus aethiops , Dopamine/administration & dosage , Dopamine/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Levodopa/toxicity , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , Male , Mesencephalon/embryology , Mesencephalon/metabolism , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Putamen/pathology , Putamen/physiopathology , Putamen/surgery , Tyrosine 3-Monooxygenase/metabolism
11.
Cell Transplant ; 25(11): 1945-1966, 2016 11.
Article in English | MEDLINE | ID: mdl-27213850

ABSTRACT

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).


Subject(s)
MPTP Poisoning/therapy , Neural Stem Cells/transplantation , Recovery of Function/physiology , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Cell Differentiation , Cells, Cultured , Chlorocebus aethiops , Cluster Analysis , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Female , Gene Expression Regulation , Gene Regulatory Networks , Humans , Immunohistochemistry , Karyotype , MPTP Poisoning/chemically induced , MPTP Poisoning/pathology , Male , Neural Stem Cells/cytology , Parthenogenesis
12.
Stroke ; 47(4): 1109-16, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26956259

ABSTRACT

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.


Subject(s)
Arm/physiopathology , Brain Ischemia/physiopathology , Cognition/physiology , Fingers/physiopathology , Psychomotor Performance/physiology , Stroke/physiopathology , Animals , Chlorocebus aethiops , Disease Models, Animal , Infarction, Middle Cerebral Artery/physiopathology , Motor Skills/physiology , Sensorimotor Cortex/physiopathology
13.
Front Neurosci ; 10: 12, 2016.
Article in English | MEDLINE | ID: mdl-26858591

ABSTRACT

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.

14.
Leukemia ; 30(3): 594-604, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26530011

ABSTRACT

PRDM1/Blimp1, a master regulator of B-cell terminal differentiation, has been identified as a tumor suppressor gene in aggressive lymphomas, including diffuse large B-cell lymphoma (DLBCL). It has been shown in DLBCL and Hodgkin lymphoma that PRDM1 is downregulated by cellular microRNAs. In this study, we identify the Epstein-Barr virus (EBV) microRNA (miRNA), EBV-miR-BHRF1-2, as a viral miRNA regulator of PRDM1. EBV-miR-BHRF1-2 repressed luciferase reporter activity by specific interaction with the seed region within the PRDM1 3' untranslated region. EBV-miR-BHRF1-2 inhibition upregulated PRDM1 protein expression in lymphoblastoid cell lines (LCL), supporting a role of miR-BHRF1-2 in PRDM1 downregulation in vivo. Discordance of PRDM1 messenger RNA and protein expressions is associated with high EBV-miR-BHRF1-2 levels in LCLs and primary post-transplant EBV-positive DLBCL. Enforced expression of PRDM1-induced apoptosis and cell cycle arrest in LCL cells. Inhibition of EBV-miR-BHRF1-2 negatively regulates cell cycle and decreases expression of SCARNA20, a small nucleolar RNA that is also downregulated by PRDM1 overexpression. The interaction between EBV-miR-BHRF1-2 and PRDM1 may be one of the mechanisms by which EBV-miR-BHRF1-2 promotes EBV lymphomagenesis. Our results support the potential of EBV-miR-BHRF1-2 as a therapeutic target in EBV-associated lymphoma.


Subject(s)
Carcinogenesis/genetics , Epstein-Barr Virus Infections/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, Large B-Cell, Diffuse/genetics , MicroRNAs/genetics , Repressor Proteins/genetics , Viral Proteins/genetics , 3' Untranslated Regions , Base Sequence , Binding Sites , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Cycle Checkpoints , Cell Line, Tumor , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , MicroRNAs/metabolism , Molecular Sequence Data , Paraffin Embedding , Positive Regulatory Domain I-Binding Factor 1 , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Repressor Proteins/metabolism , Signal Transduction , Tissue Fixation , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism
15.
Cell Transplant ; 24(4): 673-80, 2015.
Article in English | MEDLINE | ID: mdl-25839107

ABSTRACT

The possibility of enhancing endogenous brain repair following neurological disorders, such as Parkinson's disease (PD), is of considerable recent interest. One such mechanism may exist in the striatum as an upregulated population of tyrosine hydroxylase (TH)-immunoreactive neurons that appear after 1-methyl-4-phenyl-1,2,3,6-tetra-hydropyridine (MPTP) lesions in nonhuman primates as well as in humans with PD. An intriguing possibility is that these endogenous neurons reflect a compensatory mechanism to mitigate the loss of striatal DA due to progressive destruction of the nigrostriatal pathway. The possibility of enhancing the number and function of this population is attractive; however, it is crucial to gain further information about these cells in order to comprehend more fully their possible therapeutic potential. The current research was designed to investigate the fate of this endogenous population in African green monkeys rendered parkinsonian by MPTP lesions. Specifically, we assessed changes in the numbers of striatal neurons expressing TH at differing stages of the toxin-induced behavioral disability and discovered a close relationship with symptom severity and striatal DA neuron numbers. Increased numbers of striatal TH-positive neurons were associated with MPTP treatment that produced parkinsonian symptoms compared to numbers of these neurons in MPTP-treated asymptomatic animals and untreated controls. Expression of striatal DA neurons peaked at the manifestation of symptoms in mild/moderate animals and remained stable in animals that were severely parkinsonian. Furthermore, in severely debilitated animals that improved after fetal dopaminergic grafts, we discovered a return to control levels of the endogenous population. Taken together, our results further support the concept that this population of DA neurons responds to variations in striatal DA tone and may serve as a compensatory mechanism to restore striatal DA levels in the context of significant depletion. Artificially manipulating this endogenous population could prove beneficial for PD treatment, especially for individuals in early disease stages.


Subject(s)
Dopaminergic Neurons/metabolism , MPTP Poisoning/pathology , Tyrosine 3-Monooxygenase/metabolism , Animals , Caudate Nucleus/metabolism , Chlorocebus aethiops , Disease Models, Animal , MPTP Poisoning/metabolism , Male , Severity of Illness Index
16.
Cell Transplant ; 24(4): 681-90, 2015.
Article in English | MEDLINE | ID: mdl-25839189

ABSTRACT

Recent studies indicate that human pluripotent stem cell (PSC)-based therapies hold great promise in 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 viable human embryos and can be used to generate an unlimited supply of neural stem cells for transplantation. Here we evaluate for the first time the safety and engraftment of human parthenogenetic stem cell-derived neural stem cells (hpNSCs) in two animal models: 6-hydroxydopamine (6-OHDA)-lesioned rodents and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated nonhuman primates (NHPs). In both rodents and nonhuman primates, we observed successful engraftment and higher dopamine levels in hpNSC-transplanted animals compared to vehicle control animals, without any adverse events. These results indicate that hpNSCs are safe, well tolerated, and could potentially be a source for cell-based therapies in PD.


Subject(s)
MPTP Poisoning/therapy , Neural Stem Cells/transplantation , Ovum/cytology , Parkinson Disease, Secondary/therapy , Animals , Brain/metabolism , Brain/pathology , Chlorocebus aethiops , Chromatography, High Pressure Liquid , Disease Models, Animal , Dopamine/analysis , Dopamine/metabolism , Humans , Immunohistochemistry , Microscopy, Fluorescence , Neural Stem Cells/cytology , Oxidopamine/toxicity , Parkinson Disease, Secondary/chemically induced , Rats , Rats, Sprague-Dawley , Tissue Distribution , Transplantation, Heterologous
17.
J Cell Mol Med ; 19(1): 249-56, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25283241

ABSTRACT

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.


Subject(s)
Endometrium/cytology , Parkinson Disease/therapy , Stem Cell Transplantation , Stem Cells/cytology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Cell Count , Cell Movement , Female , Homovanillic Acid/metabolism , Male , Neurons/metabolism , Parkinson Disease/pathology , Primates , Tyrosine 3-Monooxygenase/metabolism
18.
Int J Neuropsychopharmacol ; 18(6)2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25522392

ABSTRACT

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.


Subject(s)
Behavior, Animal , Brain-Derived Neurotrophic Factor/metabolism , Cognition , Dopamine/metabolism , Phencyclidine , Prefrontal Cortex/metabolism , Schizophrenia/metabolism , Schizophrenic Psychology , Spinal Cord/metabolism , Synapses/metabolism , Age Factors , Animals , Chlorocebus aethiops , Disease Models, Animal , Disease Susceptibility , Female , Male , Memory, Short-Term , Prefrontal Cortex/physiopathology , Schizophrenia/chemically induced , Schizophrenia/pathology , Schizophrenia/physiopathology , Sex Factors , Spinal Cord/physiopathology , Spinal Cord/ultrastructure , Synapses/ultrastructure , Time Factors
19.
Stem Cells Transl Med ; 3(6): 692-701, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24744393

ABSTRACT

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.


Subject(s)
Genetic Therapy/methods , Glial Cell Line-Derived Neurotrophic Factor/metabolism , MPTP Poisoning/therapy , Mesencephalon/surgery , Neural Stem Cells/transplantation , Neurites/transplantation , Neurogenesis , Regenerative Medicine/methods , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Cell Line , Cell Lineage , Cell Shape , Cell Survival , Chlorocebus aethiops , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor/genetics , Humans , MPTP Poisoning/chemically induced , MPTP Poisoning/genetics , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Mesencephalon/metabolism , Mesencephalon/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurites/metabolism , Neurites/pathology , Stem Cell Niche , Time Factors , Transduction, Genetic , Transfection , Up-Regulation
20.
J Comp Neurol ; 522(12): 2729-40, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24610674

ABSTRACT

The potential for "replacement cells" to restore function in Parkinson's disease has been widely reported over the past 3 decades, rejuvenating the central nervous system rather than just relieving symptoms. Most such experiments have used fetal or embryonic sources that may induce immunological rejection and generate ethical concerns. Autologous sources, in which the cells to be implanted are derived from recipients' own cells after reprogramming to stem cells, direct genetic modifications, or epigenetic modifications in culture, could eliminate many of these problems. In a previous study on autologous brain cell transplantation, we demonstrated that adult monkey brain cells, obtained from cortical biopsies and kept in culture for 7 weeks, exhibited potential as a method of brain repair after low doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) caused dopaminergic cell death. The present study exposed monkeys to higher MPTP doses to produce significant parkinsonism and behavioral impairments. Cerebral cortical cells were biopsied from the animals, held in culture for 7 weeks to create an autologous neural cell "ecosystem" and reimplanted bilaterally into the striatum of the same six donor monkeys. These cells expressed neuroectodermal and progenitor markers such as nestin, doublecortin, GFAP, neurofilament, and vimentin. Five to six months after reimplantation, histological analysis with the dye PKH67 and unbiased stereology showed that reimplanted cells survived, migrated bilaterally throughout the striatum, and seemed to exert a neurorestorative effect. More tyrosine hydroxylase-immunoreactive neurons and significant behavioral improvement followed reimplantation of cultured autologous neural cells as a result of unknown trophic factors released by the grafts.


Subject(s)
Brain/cytology , MPTP Poisoning/complications , MPTP Poisoning/surgery , Mental Disorders/etiology , Neurons/transplantation , Recovery of Function/physiology , Animals , Biopsy , Cell Count , Chlorocebus aethiops , Disease Models, Animal , Linear Models , Male , Mental Disorders/therapy , Necrosis/etiology , Organic Chemicals , Time Factors , Transplantation, Autologous/methods , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...