Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Stem Cells Transl Med ; 13(2): 177-190, 2024 Feb 14.
Article En | MEDLINE | ID: mdl-38016184

Stroke is a leading cause of death in the US and around the world but with limited treatment options. Survivors often present with long-term cognitive and neurological deficits. Stem cell-based therapy has emerged as a potential treatment for stroke. While stem cell transplantation in stroke has reached clinical trials, mostly safety outcomes have been reported with efficacy readouts warranting more studies. In an effort to optimize the stem cell regimen for stroke, here we conducted vis-a-vis comparison of different routes of transplantation, namely, intracerebral, intraarterial, and intranasal delivery of expanded human CD34 + stem cells, called ProtheraCytes, in the established stroke model of transient middle cerebral artery occlusion (MCAO) using adult Sprague-Dawley rats. After adjusting for the dose and subacute timing of cell delivery, animals were randomly assigned to receive either ProtheraCytes or vehicle. Motor and neurological assays from days 7 to 28 post-stroke revealed significant functional recovery across all 3 delivery routes of ProtheraCytes compared to vehicle-treated stroke rats. Additionally, ProtheraCytes-transplanted stroke rats displayed significantly reduced infarct size and cell loss in the peri-infarct area coupled with enhanced neurogenesis and angiogenesis compared to vehicle-treated stroke rats. These results highlight the safety and efficacy of transplanting ProtheraCytes, including via the minimally invasive intranasal route, in conferring robust and stable behavioral and histological positive outcomes in experimental stroke.


Brain Ischemia , Ischemic Stroke , Stroke , Rats , Humans , Animals , Rats, Sprague-Dawley , Stroke/therapy , Stroke/pathology , Infarction, Middle Cerebral Artery/therapy , Infarction, Middle Cerebral Artery/pathology , Stem Cells/pathology , Neurogenesis , Brain Ischemia/therapy , Disease Models, Animal , Recovery of Function
2.
Sci Rep ; 13(1): 19665, 2023 11 11.
Article En | MEDLINE | ID: mdl-37952030

We have previously shown that intracardiac delivery of autologous CD34+ cells after acute myocardial infarction (AMI) is safe and leads to long term improvement. We are now conducting a multicenter, randomized, controlled Phase I/IIb study in post-AMI to investigate the safety and efficacy of intramyocardial injection of expanded autologous CD34+ cells (ProtheraCytes) (NCT02669810). Here, we conducted a series of in vitro studies characterizing the growth factor secretion, exosome secretion, gene expression, cell surface markers, differentiation potential, and angiogenic potential of ProtheraCytes clinical batches to develop a potency assay. We show that ProtheraCytes secrete vascular endothelial growth factor (VEGF) and its concentration is significantly correlated with the number of CD34+ cells obtained after expansion. ProtheraCytes also secrete exosomes containing proangiogenic miRNAs (126, 130a, 378, 26a), antiapoptotic miRNAs (21 and 146a), antifibrotic miRNAs (133a, 24, 29b, 132), and miRNAs promoting myocardial regeneration (199a and 590). We also show that ProtheraCytes have in vitro angiogenic activity, express surface markers of endothelial progenitor cells, and can differentiate in vitro into endothelial cells. After the in vitro characterization of multiple ProtheraCytes clinical batches, we established that measuring the concentration of VEGF provided the most practical, reliable, and consistent potency assay.


Endothelial Progenitor Cells , MicroRNAs , Myocardial Infarction , Humans , Antigens, CD34/metabolism , Endothelial Progenitor Cells/metabolism , MicroRNAs/metabolism , Myocardial Infarction/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/metabolism
3.
Front Bioeng Biotechnol ; 11: 1150522, 2023.
Article En | MEDLINE | ID: mdl-37288358

Knee osteoarthritis (OA) is a degenerative joint disease of the knee that results from the progressive loss of articular cartilage. It is most common in the elderly and affects millions of people worldwide, leading to a continuous increase in the number of total knee replacement surgeries. These surgeries improve the patient's physical mobility, but can lead to late infection, loosening of the prosthesis, and persistent pain. We would like to investigate if cell-based therapies can avoid or delay such surgeries in patients with moderate OA by injecting expanded autologous peripheral blood derived CD34+ cells (ProtheraCytes®) into the articular joint. In this study we evaluated the survival of ProtheraCytes® when exposed to synovial fluid and their performance in vitro with a model consisting of their co-culture with human OA chondrocytes in separate layers of Transwells and in vivo with a murine model of OA. Here we show that ProtheraCytes® maintain high viability (>95%) when exposed for up to 96 hours to synovial fluid from OA patients. Additionally, when co-cultured with OA chondrocytes, ProtheraCytes® can modulate the expression of some chondrogenic (collagen II and Sox9) and inflammatory/degrading (IL1ß, TNF, and MMP-13) markers at gene or protein levels. Finally, ProtheraCytes® survive after injection into the knee of a collagenase-induced osteoarthritis mouse model, engrafting mainly in the synovial membrane, probably due to the fact that ProtheraCytes® express CD44, a receptor of hyaluronic acid, which is abundantly present in the synovial membrane. This report provides preliminary evidence of the therapeutic potential of CD34+ cells on OA chondrocytes in vitro and their survival after in vivo implantation in the knee of mice and merits further investigation in future preclinical studies in OA models.

4.
Diagn Microbiol Infect Dis ; 101(4): 115537, 2021 Dec.
Article En | MEDLINE | ID: mdl-34619569

We aim to study kinetics of anti-SARS-CoV-2 IgG antibody levels in subjects with COVID-19 for up to 11 months and the potential influential factors. The study was a prospective longitudinal study. The analyses were based on 77 serum/plasma samples with a mean of 4 samples per participant (range 1 - 18) in 20 participants with at least one positive Polymerase Chain Reaction testing result from 19 March 2020 up to 10 February 2021. Among the subjects (median age 34.5 years, 65% male), IgG level declined with the follow-up time (per month; geometric mean ratio [GMR] 0.73; 95% CI, 0.72 - 0.74). In a small sample of subjects from the general population with COVID-19, IgG levels declined non-linearly from month 2 to 11 with individual heterogeneity in quantity and changing speed and may be associated with gender, race and the loss of smell and taste.


COVID-19/blood , Immunoglobulin G/blood , SARS-CoV-2/immunology , Adult , Aged , Antibodies, Viral , COVID-19/immunology , COVID-19/virology , Female , Follow-Up Studies , Humans , Kinetics , Longitudinal Studies , Male , Middle Aged , Prospective Studies , Time Factors , Young Adult
5.
Theranostics ; 9(4): 1029-1046, 2019.
Article En | MEDLINE | ID: mdl-30867814

International Stem Cell Corporation human parthenogenetic neural stem cells (ISC-hpNSC) have potential therapeutic value for patients suffering from traumatic brain injury (TBI). Here, we demonstrate the behavioral and histological effects of transplanting ISC-hpNSC intracerebrally in an animal model of TBI. Methods: Sprague-Dawley rats underwent a moderate controlled cortical impact TBI surgery. Transplantation occurred at 72 h post-TBI with functional readouts of behavioral and histological deficits conducted during the subsequent 3-month period after TBI. We characterized locomotor, neurological, and cognitive performance at baseline (before TBI), then on days 0, 1, 7, 14, 30, 60, and 90 (locomotor and neurological), and on days 28-30, 58-60, and 88-90 (cognitive) after TBI. Following completion of behavioral testing at 3 months post-TBI, animals were euthanized by transcardial perfusion and brains harvested to histologically characterize the extent of brain damage. Neuronal survival was revealed by Nissl staining, and stem cell engraftment and host tissue repair mechanisms such as the anti-inflammatory response in peri-TBI lesion areas were examined by immunohistochemical analyses. Results: We observed that TBI groups given high and moderate doses of ISC-hpNSC had an improved swing bias on an elevated body swing test for motor function, increased scores on forelimb akinesia and paw grasp neurological tests, and committed significantly fewer errors on a radial arm water maze test for cognition. Furthermore, histological analyses indicated that high and moderate doses of stem cells increased the expression of phenotypic markers related to the neural lineage and myelination and decreased reactive gliosis and inflammation in the brain, increased neuronal survival in the peri-impact area of the cortex, and decreased inflammation in the spleen at 90 days post-TBI. Conclusion: These results provide evidence that high and moderate doses of ISC-hpNSC ameliorate TBI-associated histological alterations and motor, neurological, and cognitive deficits.


Brain Injuries, Traumatic/therapy , Brain Regeneration , Neural Stem Cells/physiology , Stem Cell Transplantation/methods , Animals , Cognition , Disease Models, Animal , Humans , Locomotion , Rats, Sprague-Dawley , Treatment Outcome
6.
Methods Mol Biol ; 1919: 43-57, 2019.
Article En | MEDLINE | ID: mdl-30656620

We have previously shown that human parthenogenetic stem cells (hpSC) can be chemically directed to differentiate into a homogeneous population of multipotent neural stem cells (hpNSC) that are scalable, cryopreservable, express all the appropriate neural markers, and can be further differentiated into functional dopaminergic neurons. Differentiation of hpSC into hpNSC provides a platform to study the molecular basis of human neural differentiation, to develop cell culture models of neural disease, and to provide neural stem cells for the treatment of neurodegenerative diseases. Additionally, the hpNSC that are generated could serve as a platform for drug discovery and the determination of pharmaceutical-induced neural toxicity. Here, we describe in detail the stepwise protocol that was developed in our laboratory that facilitates the highly efficient and reproducible differentiation of hpSC into hpNSC.


Cell Differentiation , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Cell Culture Techniques , Humans , Immunohistochemistry , Immunophenotyping , Microscopy , Neural Stem Cells/metabolism , Neurons/cytology , Pluripotent Stem Cells/metabolism , Stem Cell Transplantation
7.
Stem Cells Dev ; 27(14): 951-957, 2018 07 15.
Article En | MEDLINE | ID: mdl-29882481

In this commentary we discuss International Stem Cell Corporation's (ISCO's) approach to developing a pluripotent stem cell based treatment for Parkinson's disease (PD). In 2016, ISCO received approval to conduct the world's first clinical study of a pluripotent stem cell based therapy for PD. The Australian regulatory agency Therapeutic Goods Administration (TGA) and the Melbourne Health's Human Research Ethics Committee (HREC) independently reviewed ISCO's extensive preclinical data and granted approval for the evaluation of a novel human parthenogenetic derived neural stem cell (NSC) line, ISC-hpNSC, in a PD phase 1 clinical trial ( ClinicalTrials.gov NCT02452723). This is a single-center, open label, dose escalating 12-month study with a 5-year follow-up evaluating a number of objective and patient-reported safety and efficacy measures. A total of 6 years of safety and efficacy data will be collected from each patient. Twelve participants are recruited in this study with four participants per single dose cohort of 30, 50, and 70 million ISC-hpNSC. The grafts are placed bilaterally in the caudate nucleus, putamen, and substantia nigra by magnetic resonance imaging-guided stereotactic surgery. Participants are 30-70 years old with idiopathic PD ≤13 years duration and unified PD rating scale motor score (Part III) in the "OFF" state ≤49. This trial is fully funded by ISCO with no economic involvement from the patients. It is worth noting that ISCO underwent an exhaustive review process and successfully answered the very comprehensive, detailed, and specific questions posed by the TGA and HREC. The regulatory/ethic review process is based on applying scientific and clinical expertise to decision-making, to ensure that the benefits to consumers outweigh any risks associated with the use of medicines or novel therapies.


Neural Stem Cells/transplantation , Parkinson Disease/therapy , Stem Cell Transplantation , Stem Cells/cytology , Australia , Cell Differentiation/genetics , Clinical Trials as Topic , Humans , Magnetic Resonance Imaging , Parkinson Disease/pathology , Pluripotent Stem Cells
8.
J Tissue Eng Regen Med ; 12(5): 1261-1272, 2018 05.
Article En | MEDLINE | ID: mdl-29490116

Repair or regeneration of hyaline cartilage in knees, shoulders, intervertebral discs, and other assorted joints is a major therapeutic target. To date, therapeutic strategies utilizing chondrocytes or mesenchymal stem cells are limited by expandability or the generation of mechanically inferior cartilage. Our objective is to generate robust cartilage-specific matrix from human mesenchymal stem cells suitable for further therapeutic development. Human mesenchymal stem cells, in an alginate 3D format, were supplied with individual sugars and chains which comprise the glycan component of proteoglycans in articular cartilage (galactose, hyaluronic acid, glucuronic acid, and xylose) during chondrogenesis. After an initial evaluation for proteoglycan deposition utilizing Alcian blue, the tissue was further evaluated for viability, structural elements, and hypertrophic status. With the further addition of serum, a substantial increase was observed in viability, the amount of proteoglycan deposition, glycosaminoglycan production, and an enhancement of Hyaluronic Acid, Collagen II and Aggrecan deposition. Suppression of hypertrophic markers (COL1A1, COL10A1, MMP13, and RUNX2) was also observed. When mesenchymal stem cells were supplied with the raw building materials of proteoglycans and a limited amount of serum during chondrogenesis, it resulted in the generation of viable hyaline-like cartilage with deposition of structural components which exceeded previously reported in vitro-based cartilage.


Carbohydrates/pharmacology , Cell Differentiation , Chondrogenesis/drug effects , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/cytology , Cartilage, Articular/drug effects , Cartilage, Articular/growth & development , Cell Differentiation/drug effects , Cell Survival/drug effects , Collagen Type II/metabolism , Glycosaminoglycans/metabolism , Humans , Hyaluronic Acid/pharmacology , Mesenchymal Stem Cells/drug effects , Proteoglycans/metabolism , Serum
9.
Stem Cells ; 35(4): 872-885, 2017 04.
Article En | MEDLINE | ID: mdl-27888558

Genomic aberrations have been identified in many human pluripotent stem cell (hPSC) cultures. Commonly observed duplications in portions of chromosomes 12p and 17q have been associated with increases in genetic instability and resistance to apoptosis, respectively. However, the phenotypic consequences related to sporadic mutations have not been evaluated to date. Here, we report on the effects of a single-copy deletion of the chr17p13.1 region, a sporadic mutation that spontaneously arose independently in several subclones of a human embryonic stem cell culture. Compared to cells with two normal copies of chr17p13.1 ("wild-type"), the cells with a single-copy deletion of this region ("mutant") displayed a selective advantage when exposed to stressful conditions, and retained a higher percentage of cells expressing the pluripotency marker POU5F1/OCT4 after 2 weeks of in vitro differentiation. Knockdown of TP53, which is a gene encompassed by the deleted region, in wild-type cells mimicked the chr17p13.1 deletion phenotype. Thus, sporadic mutations in hPSCs can have phenotypic effects that may impact their utility for clinical applications. Stem Cells 2017;35:872-885.


Gene Dosage , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromosomes, Human, Pair 17/genetics , Clone Cells , DNA Damage , DNA Repair/drug effects , Etoposide/pharmacology , Gene Expression Profiling , Gene Knockdown Techniques , Human Embryonic Stem Cells/drug effects , Humans , Phenotype , RNA, Small Interfering/metabolism , Staurosporine/pharmacology
10.
Sci Rep ; 6: 34478, 2016 Sep 30.
Article En | MEDLINE | ID: mdl-27686862

Human pluripotent stem cells (PSC) have the potential to revolutionize regenerative medicine. However undifferentiated PSC can form tumors and strict quality control measures and safety studies must be conducted before clinical translation. Here we describe preclinical tumorigenicity and biodistribution safety studies that were required by the US Food and Drug Administration (FDA) and Australian Therapeutic Goods Administration (TGA) prior to conducting a Phase I clinical trial evaluating the safety and tolerability of human parthenogenetic stem cell derived neural stem cells ISC-hpNSC for treating Parkinson's disease (ClinicalTrials.gov Identifier NCT02452723). To mitigate the risk of having residual PSC in the final ISC-hpNSC population, we conducted sensitive in vitro assays using flow cytometry and qRT-PCR analyses and in vivo assays to determine acute toxicity, tumorigenicity and biodistribution. The results from these safety studies show the lack of residual undifferentiated PSC, negligible tumorigenic potential by ISC-hpNSC and provide additional assurance to their clinical application.

11.
Bioessays ; 38 Suppl 1: S86-95, 2016 07.
Article En | MEDLINE | ID: mdl-27417126

Human pluripotent stem cells (hPSCs) have the potential to fundamentally change the way that we go about treating and understanding human disease. Despite this extraordinary potential, these cells also have an innate capability to form tumors in immunocompromised individuals when they are introduced in their pluripotent state. Although current therapeutic strategies involve transplantation of only differentiated hPSC derivatives, there is still a concern that transplanted cell populations could contain a small percentage of cells that are not fully differentiated. In addition, these cells have been frequently reported to acquire genetic alterations that, in some cases, are associated with certain types of human cancers. Here, we try to separate the panic from reality and rationally evaluate the true tumorigenic potential of these cells. We also discuss a recent study examining the effect of culture conditions on the genetic integrity of hPSCs. Finally, we present a set of sensible guidelines for minimizing the tumorigenic potential of hPSC-derived cells. © 2016 The Authors. Inside the Cell published by Wiley Periodicals, Inc.


Cell Transformation, Neoplastic , Neoplasms/pathology , Pluripotent Stem Cells/pathology , Cell Culture Techniques , Genomic Instability , Humans , Neoplasms/genetics , Neoplasms/prevention & control
12.
Cell Transplant ; 25(11): 1945-1966, 2016 11.
Article En | MEDLINE | ID: mdl-27213850

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).


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
13.
Cell Transplant ; 24(4): 681-90, 2015.
Article En | MEDLINE | ID: mdl-25839189

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.


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
14.
PLoS One ; 10(2): e0118307, 2015.
Article En | MEDLINE | ID: mdl-25714340

The self-renewal and differentiation capacities of human pluripotent stem cells (hPSCs) make them a promising source of material for cell transplantation therapy, drug development, and studies of cellular differentiation and development. However, the large numbers of cells necessary for many of these applications require extensive expansion of hPSC cultures, a process that has been associated with genetic and epigenetic alterations. We have performed a combinatorial study on both hESCs and hiPSCs to compare the effects of enzymatic vs. mechanical passaging, and feeder-free vs. mouse embryonic fibroblast feeder substrate, on the genetic and epigenetic stability and the phenotypic characteristics of hPSCs. In extensive experiments involving over 100 continuous passages, we observed that both enzymatic passaging and feeder-free culture were associated with genetic instability, higher rates of cell proliferation, and persistence of OCT4/POU5F1-positive cells in teratomas, with enzymatic passaging having the stronger effect. In all combinations of culture conditions except for mechanical passaging on feeder layers, we noted recurrent deletions in the genomic region containing the tumor suppressor gene TP53, which was associated with decreased mRNA expression of TP53, as well as alterations in the expression of several downstream genes consistent with a decrease in the activity of the TP53 pathway. Among the hESC cultures, we also observed culture-associated variations in global gene expression and DNA methylation. The effects of enzymatic passaging and feeder-free conditions were also observed in hiPSC cultures. Our results highlight the need for careful assessment of the effects of culture conditions on cells intended for clinical therapies.


Epigenesis, Genetic , Genome, Human , Genomic Instability , Human Embryonic Stem Cells/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Line , Cell Self Renewal , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Chromosome Aberrations , Chromosome Deletion , Chromosome Duplication , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 20 , DNA Methylation , Gene Expression Profiling , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/pathology , Humans , Phenotype , Pluripotent Stem Cells/metabolism , Polymorphism, Single Nucleotide , Time Factors , Tumor Suppressor Protein p53/genetics
15.
Sci Rep ; 3: 1463, 2013.
Article En | MEDLINE | ID: mdl-23492920

New small molecules that regulate the step-wise differentiation of human pluripotent stem cells into dopaminergic neurons have been identified. The steroid, guggulsterone, was found to be the most effective inducer of neural stem cells into dopaminergic neurons. These neurons are extensively characterized and shown to be functional. We believe this new approach offers a practical route to creating neurons of sufficient quality to be used to treat Parkinson's disease patients.


Cell Differentiation/drug effects , Dopaminergic Neurons/cytology , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Pregnenediones/pharmacology , Cell Culture Techniques , Cell Line , Cell Transplantation/methods , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Gene Expression Profiling , Humans , Membrane Potentials/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Oligonucleotide Array Sequence Analysis , Parkinson Disease/surgery , Patch-Clamp Techniques , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , Reverse Transcriptase Polymerase Chain Reaction
16.
Cell Stem Cell ; 10(5): 620-34, 2012 May 04.
Article En | MEDLINE | ID: mdl-22560082

Human pluripotent stem cells (hPSCs) are potential sources of cells for modeling disease and development, drug discovery, and regenerative medicine. However, it is important to identify factors that may impact the utility of hPSCs for these applications. In an unbiased analysis of 205 hPSC and 130 somatic samples, we identified hPSC-specific epigenetic and transcriptional aberrations in genes subject to X chromosome inactivation (XCI) and genomic imprinting, which were not corrected during directed differentiation. We also found that specific tissue types were distinguished by unique patterns of DNA hypomethylation, which were recapitulated by DNA demethylation during in vitro directed differentiation. Our results suggest that verification of baseline epigenetic status is critical for hPSC-based disease models in which the observed phenotype depends on proper XCI or imprinting and that tissue-specific DNA methylation patterns can be accurately modeled during directed differentiation of hPSCs, even in the presence of variations in XCI or imprinting.


Genetic Variation , Pluripotent Stem Cells/physiology , Cell Differentiation , Cells, Cultured , Chromosome Aberrations , Chromosomes, Human, X , DNA Methylation , Genomic Imprinting , Humans , Organ Specificity , Recurrence , Stem Cell Niche , X Chromosome Inactivation
17.
Regen Med ; 7(1): 37-45, 2012 Jan.
Article En | MEDLINE | ID: mdl-22168496

Human parthenogenetic stem cells are derived from the inner cell mass of blastocysts obtained from unfertilized oocytes that have been stimulated to develop without any participation of male gamete. As parthenogenesis does not involve the destruction of a viable human embryo, the derivation and use of human parthenogenetic stem cells does not raise the same ethical concerns as conventional embryonic stem cells. Human parthenogenetic stem cells are similar to embryonic stem cells in their proliferation and multilineage in vitro differentiation capacity. The aim of this study is to derive multipotent neural stem cells from human parthenogenetic stem cells that are stable to passaging and cryopreservation, and have the ability to further differentiate into functional neurons. Immunocytochemistry, quantitative real-time PCR, or FACS were used to confirm that the derived neural stem cells express neural markers such as NES, SOX2 and MS1. The derived neural stem cells keep uniform morphology for at least 30 passages and can be spontaneously differentiated into cells with neuron morphology that express TUBB3 and MAP2, and fire action potentials. These results suggest that parthenogenetic stem cells are a very promising and potentially unlimited source for the derivation of multipotent neural stem cells that can be used for therapeutic applications.


Cell Differentiation , Cell Lineage , Neurons/cytology , Parthenogenesis , Stem Cells/cytology , Cell Aggregation , Cell Line , Electrophysiological Phenomena , Epithelium/metabolism , Humans , Male , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/metabolism , Phenotype
18.
J Vis Exp ; (57): e3177, 2011 Nov 07.
Article En | MEDLINE | ID: mdl-22158256

Pluripotent stem cells (PSCs) have the unique characteristic that they can differentiate into cells from all three germ layers. This makes them a potentially valuable tool for the treatment of many different diseases. With the advent of induced pluripotent stem cells (iPSCs) and continuing research with human embryonic stem cells (hESCs) there is a need for assays that can demonstrate that a particular cell line is pluripotent. Germline transmission has been the gold standard for demonstrating the pluripotence of mouse embryonic stem cell (mESC) lines(1,2,3). Using this assay, researchers can show that a mESC line can make all cell types in the embryo including germ cells(4). With the generation of human ESC lines(5,6), the appropriate assay to prove pluripotence of these cells was unclear since human ESCs cannot be tested for germline transmission. As a surrogate, the teratoma assay is currently used to demonstrate the pluripotency of human pluripotent stem cells (hPSCs)(7,8,9). Though this assay has recently come under scrutiny and new technologies are being actively explored, the teratoma assay is the current gold standard(7). In this assay, the cells in question are injected into an immune compromised mouse. If the cells are pluripotent, a teratoma will eventually develop and sections of the tumor will show tissues from all 3 germ layers(10). In the teratoma assay, hPSCs can be injected into different areas of the mouse. The most common injection sites include the testis capsule, the kidney capsule, the liver; or into the leg either subcutaneously or intramuscularly(11). Here we describe a robust protocol for the generation of teratomas from hPSCs using the testis capsule as the site for tumor growth.


Cell Transformation, Neoplastic/pathology , Neoplastic Stem Cells/pathology , Pluripotent Stem Cells/pathology , Teratoma/pathology , Testicular Neoplasms/pathology , Animals , Male , Mice , Mice, SCID
19.
Nat Methods ; 8(10): 829-31, 2011 Sep 04.
Article En | MEDLINE | ID: mdl-21892153

For some highly endangered species there are too few reproductively capable animals to maintain adequate genetic diversity, and extraordinary measures are necessary to prevent extinction. We report generation of induced pluripotent stem cells (iPSCs) from two endangered species: a primate, the drill, Mandrillus leucophaeus and the nearly extinct northern white rhinoceros, Ceratotherium simum cottoni. iPSCs may eventually facilitate reintroduction of genetic material into breeding populations.


Endangered Species , Induced Pluripotent Stem Cells/cytology , Mandrillus , Perissodactyla , Animals , Species Specificity
20.
Cell Res ; 21(11): 1551-63, 2011 Nov.
Article En | MEDLINE | ID: mdl-21894191

Rapid and dependable methods for isolating human pluripotent stem cell (hPSC) populations are urgently needed for quality control in basic research and in cell-based therapy applications. Using lectin arrays, we analyzed glycoproteins extracted from 26 hPSC samples and 22 differentiated cell samples, and identified a small group of lectins with distinctive binding signatures that were sufficient to distinguish hPSCs from a variety of non-pluripotent cell types. These specific biomarkers were shared by all the 12 human embryonic stem cell and the 14 human induced pluripotent stem cell samples examined, regardless of the laboratory of origin, the culture conditions, the somatic cell type reprogrammed, or the reprogramming method used. We demonstrated a practical application of specific lectin binding by detecting hPSCs within a differentiated cell population with lectin-mediated staining followed by fluorescence microscopy and flow cytometry, and by enriching and purging viable hPSCs from mixed cell populations using lectin-mediated cell separation. Global gene expression analysis showed pluripotency-associated differential expression of specific fucosyltransferases and sialyltransferases, which may underlie these differences in protein glycosylation and lectin binding. Taken together, our results show that protein glycosylation differs considerably between pluripotent and non-pluripotent cells, and demonstrate that lectins may be used as biomarkers to monitor pluripotency in stem cell populations and for removal of viable hPSCs from mixed cell populations.


Biomarkers/metabolism , Glycomics , Lectins/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Biotin/chemistry , Biotin/metabolism , Cell Separation , Cells, Cultured , Embryonic Stem Cells/cytology , Fucosyltransferases/metabolism , Gene Expression Profiling , Glycosylation , Humans , Induced Pluripotent Stem Cells/cytology , Lectins/chemistry , Protein Array Analysis , Protein Binding , Sialyltransferases/metabolism
...