Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
STAR Protoc ; 4(4): 102674, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37897731

RESUMO

Prospective isolation of defined cell types is critical for the functional study of stem cells, especially in primary human tissues. Here, we present a protocol for purifying 10 transcriptomically and functionally distinct neural stem and progenitor cell types from the developing human brain using fluorescence-activated cell sorting. We describe steps for tissue dissociation, staining, and cell sorting as well as downstream functional experiments for measuring clonogenicity, differentiation, and engraftment potential of purified populations. For complete details on the use and execution of this protocol, please refer to Liu et al. (2023).1.


Assuntos
Encéfalo , Células-Tronco , Humanos , Diferenciação Celular , Separação Celular , Citometria de Fluxo
2.
Cell ; 186(6): 1179-1194.e15, 2023 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-36931245

RESUMO

The human brain undergoes rapid development at mid-gestation from a pool of neural stem and progenitor cells (NSPCs) that give rise to the neurons, oligodendrocytes, and astrocytes of the mature brain. Functional study of these cell types has been hampered by a lack of precise purification methods. We describe a method for prospectively isolating ten distinct NSPC types from the developing human brain using cell-surface markers. CD24-THY1-/lo cells were enriched for radial glia, which robustly engrafted and differentiated into all three neural lineages in the mouse brain. THY1hi cells marked unipotent oligodendrocyte precursors committed to an oligodendroglial fate, and CD24+THY1-/lo cells marked committed excitatory and inhibitory neuronal lineages. Notably, we identify and functionally characterize a transcriptomically distinct THY1hiEGFRhiPDGFRA- bipotent glial progenitor cell (GPC), which is lineage-restricted to astrocytes and oligodendrocytes, but not to neurons. Our study provides a framework for the functional study of distinct cell types in human neurodevelopment.


Assuntos
Células-Tronco Neurais , Camundongos , Animais , Humanos , Células-Tronco Neurais/metabolismo , Neurônios , Diferenciação Celular/fisiologia , Neuroglia/metabolismo , Encéfalo , Astrócitos
3.
Clin Cancer Res ; 27(23): 6467-6478, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34475101

RESUMO

PURPOSE: Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, a key process of cancer metabolism. PKM2 is preferentially expressed by glioblastoma (GBM) cells with minimal expression in healthy brain. We describe the development, validation, and translation of a novel PET tracer to study PKM2 in GBM. We evaluated 1-((2-fluoro-6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA-23) in cell culture, mouse models of GBM, healthy human volunteers, and patients with GBM. EXPERIMENTAL DESIGN: [18F]DASA-23 was synthesized with a molar activity of 100.47 ± 29.58 GBq/µmol and radiochemical purity >95%. We performed initial testing of [18F]DASA-23 in GBM cell culture and human GBM xenografts implanted orthotopically into mice. Next, we produced [18F]DASA-23 under FDA oversight, and evaluated it in healthy volunteers and a pilot cohort of patients with glioma. RESULTS: In mouse imaging studies, [18F]DASA-23 clearly delineated the U87 GBM from surrounding healthy brain tissue and had a tumor-to-brain ratio of 3.6 ± 0.5. In human volunteers, [18F]DASA-23 crossed the intact blood-brain barrier and was rapidly cleared. In patients with GBM, [18F]DASA-23 successfully outlined tumors visible on contrast-enhanced MRI. The uptake of [18F]DASA-23 was markedly elevated in GBMs compared with normal brain, and it identified a metabolic nonresponder within 1 week of treatment initiation. CONCLUSIONS: We developed and translated [18F]DASA-23 as a new tracer that demonstrated the visualization of aberrantly expressed PKM2 for the first time in human subjects. These results warrant further clinical evaluation of [18F]DASA-23 to assess its utility for imaging therapy-induced normalization of aberrant cancer metabolism.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Animais , Neoplasias Encefálicas/patologia , Compostos de Diazônio , Glioblastoma/patologia , Glicólise , Humanos , Camundongos , Tomografia por Emissão de Pósitrons/métodos , Piruvato Quinase/metabolismo , Ácidos Sulfanílicos
4.
Nat Commun ; 11(1): 2713, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32483127

RESUMO

Despite their rapidly-expanding therapeutic potential, human pluripotent stem cell (hPSC)-derived cell therapies continue to have serious safety risks. Transplantation of hPSC-derived cell populations into preclinical models has generated teratomas (tumors arising from undifferentiated hPSCs), unwanted tissues, and other types of adverse events. Mitigating these risks is important to increase the safety of such therapies. Here we use genome editing to engineer a general platform to improve the safety of future hPSC-derived cell transplantation therapies. Specifically, we develop hPSC lines bearing two drug-inducible safeguards, which have distinct functionalities and address separate safety concerns. In vitro administration of one small molecule depletes undifferentiated hPSCs >106-fold, thus preventing teratoma formation in vivo. Administration of a second small molecule kills all hPSC-derived cell-types, thus providing an option to eliminate the entire hPSC-derived cell product in vivo if adverse events arise. These orthogonal safety switches address major safety concerns with pluripotent cell-derived therapies.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/genética , Edição de Genes/métodos , Células-Tronco Pluripotentes/metabolismo , Transplante de Células-Tronco/métodos , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia , Teratoma/genética , Teratoma/metabolismo , Teratoma/prevenção & controle
5.
Cell Stem Cell ; 24(5): 821-828.e5, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-31051134

RESUMO

Genome editing of human pluripotent stem cells (hPSCs) provides powerful opportunities for in vitro disease modeling, drug discovery, and personalized stem cell-based therapeutics. Currently, only small edits can be engineered with high frequency, while larger modifications suffer from low efficiency and a resultant need for selection markers. Here, we describe marker-free genome editing in hPSCs using Cas9 ribonucleoproteins (RNPs) in combination with AAV6-mediated DNA repair template delivery. We report highly efficient and bi-allelic integration frequencies across multiple loci and hPSC lines, achieving mono-allelic editing frequencies of up to 94% at the HBB locus. Using this method, we show robust bi-allelic correction of homozygous sickle cell mutations in a patient-derived induced PSC (iPSC) line. Thus, this strategy shows significant utility for generating hPSCs with large gene integrations and/or single-nucleotide changes at high frequency and without the need for introducing selection genes, enhancing the applicability of hPSC editing for research and translational uses.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Dependovirus/genética , Genótipo , Células-Tronco Pluripotentes/fisiologia , Proteína 9 Associada à CRISPR/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Reparo do DNA , Edição de Genes/métodos , Frequência do Gene , Engenharia Genética , Vetores Genéticos/genética , Recombinação Homóloga , Humanos , Patologia Molecular , Doadores de Tecidos
6.
iScience ; 15: 524-535, 2019 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-31132746

RESUMO

Human neural stem cells (NSCs) offer therapeutic potential for neurodegenerative diseases, such as inherited monogenic nervous system disorders, and neural injuries. Gene editing in NSCs (GE-NSCs) could enhance their therapeutic potential. We show that NSCs are amenable to gene targeting at multiple loci using Cas9 mRNA with synthetic chemically modified guide RNAs along with DNA donor templates. Transplantation of GE-NSC into oligodendrocyte mutant shiverer-immunodeficient mice showed that GE-NSCs migrate and differentiate into astrocytes, neurons, and myelin-producing oligodendrocytes, highlighting the fact that GE-NSCs retain their NSC characteristics of self-renewal and site-specific global migration and differentiation. To show the therapeutic potential of GE-NSCs, we generated GALC lysosomal enzyme overexpressing GE-NSCs that are able to cross-correct GALC enzyme activity through the mannose-6-phosphate receptor pathway. These GE-NSCs have the potential to be an investigational cell and gene therapy for a range of neurodegenerative disorders and injuries of the central nervous system, including lysosomal storage disorders.

7.
Nat Methods ; 15(12): 1045-1047, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30504872

RESUMO

Scarless genome editing in human pluripotent stem cells (hPSCs) represents a goal for both precise research applications and clinical translation of hPSC-derived therapies. Here we established a versatile and efficient method that combines CRISPR-Cas9-mediated homologous recombination with positive-negative selection of edited clones to generate scarless genetic changes in hPSCs.


Assuntos
Sistemas CRISPR-Cas , Células-Tronco Embrionárias/metabolismo , Edição de Genes , Genoma Humano , Recombinação Homóloga , Células-Tronco Pluripotentes/metabolismo , RNA Interferente Pequeno/genética , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes/citologia
8.
J Neurosci ; 37(38): 9269-9287, 2017 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-28847814

RESUMO

The interaction of transplanted stem cells with local cellular and molecular cues in the host CNS microenvironment may affect the potential for repair by therapeutic cell populations. In this regard, spinal cord injury (SCI), Alzheimer's disease, and other neurological injuries and diseases all exhibit dramatic and dynamic changes to the host microenvironment over time. Previously, we reported that delayed transplantation of human CNS-derived neural stem cells (hCNS-SCns) at 9 or 30 d post-SCI (dpi) resulted in extensive donor cell migration, predominantly neuronal and oligodendrocytic donor cell differentiation, and functional locomotor improvements. Here, we report that acute transplantation of hCNS-SCns at 0 dpi resulted in localized astroglial differentiation of donor cells near the lesion epicenter and failure to produce functional improvement in an all-female immunodeficient mouse model. Critically, specific immunodepletion of neutrophils (polymorphonuclear leukocytes) blocked hCNS-SCns astroglial differentiation near the lesion epicenter and rescued the capacity of these cells to restore function. These data represent novel evidence that a host immune cell population can block the potential for functional repair derived from a therapeutic donor cell population, and support targeting the inflammatory microenvironment in combination with cell transplantation after SCI.SIGNIFICANCE STATEMENT The interaction of transplanted cells with local cellular and molecular cues in the host microenvironment is a key variable that may shape the translation of neurotransplantation research to the clinical spinal cord injury (SCI) human population, and few studies have investigated these events. We show that the specific immunodepletion of polymorphonuclear leukocyte neutrophils using anti-Ly6G inhibits donor cell astrogliosis and rescues the capacity of a donor cell population to promote locomotor improvement after SCI. Critically, our data demonstrate novel evidence that a specific host immune cell population can block the potential for functional repair derived from a therapeutic donor cell population.


Assuntos
Regeneração Nervosa/imunologia , Células-Tronco Neurais/transplante , Neurogênese/imunologia , Neutrófilos/imunologia , Neutrófilos/patologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/terapia , Animais , Comunicação Celular , Diferenciação Celular/imunologia , Movimento Celular , Feminino , Camundongos , Camundongos SCID , Células-Tronco Neurais/imunologia , Recuperação de Função Fisiológica , Nicho de Células-Tronco
9.
J Immunol ; 199(3): 1069-1085, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28687659

RESUMO

Inflammatory processes play a key role in pathophysiology of many neurologic diseases/trauma, but the effect of immune cells and factors on neurotransplantation strategies remains unclear. We hypothesized that cellular and humoral components of innate immunity alter fate and migration of human neural stem cells (hNSC). In these experiments, conditioned media collected from polymorphonuclear leukocytes (PMN) selectively increased hNSC astrogliogenesis and promoted cell migration in vitro. PMN were shown to generate C1q and C3a; exposure of hNSC to PMN-synthesized concentrations of these complement proteins promoted astrogliogenesis and cell migration. Furthermore, in vitro, Abs directed against C1q and C3a reversed the fate and migration effects observed. In a proof-of-concept in vivo experiment, blockade of C1q and C3a transiently altered hNSC migration and reversed astroglial fate after spinal cord injury. Collectively, these data suggest that modulation of the innate/humoral inflammatory microenvironment may impact the potential of cell-based therapies for recovery and repair following CNS pathology.


Assuntos
Astrócitos/fisiologia , Diferenciação Celular/fisiologia , Complemento C1q/biossíntese , Complemento C3a/biossíntese , Células-Tronco Neurais/fisiologia , Neutrófilos/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Movimento Celular , Células Cultivadas , Complemento C1q/antagonistas & inibidores , Complemento C1q/genética , Complemento C1q/imunologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/genética , Complemento C3a/imunologia , Meios de Cultivo Condicionados , Humanos , Imunidade Inata , Camundongos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/imunologia , Neutrófilos/imunologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia
10.
Stem Cell Reports ; 8(6): 1534-1548, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28479305

RESUMO

Multipotent human central nervous system-derived neural stem cells transplanted at doses ranging from 10,000 (low) to 500,000 (very high) cells differentiated predominantly into the oligodendroglial lineage. However, while the number of engrafted cells increased linearly in relationship to increasing dose, the proportion of oligodendrocytic cells declined. Increasing dose resulted in a plateau of engraftment, enhanced neuronal differentiation, and increased distal migration caudal to the transplantation sites. Dose had no effect on terminal sensory recovery or open-field locomotor scores. However, total human cell number and decreased oligodendroglial proportion were correlated with hindlimb girdle coupling errors. Conversely, greater oligodendroglial proportion was correlated with increased Ab step pattern, decreased swing speed, and increased paw intensity, consistent with improved recovery. These data suggest that transplant dose, and/or target niche parameters can regulate donor cell engraftment, differentiation/maturation, and lineage-specific migration profiles.


Assuntos
Diferenciação Celular , Células-Tronco Neurais/transplante , Neurônios/citologia , Oligodendroglia/citologia , Traumatismos da Medula Espinal/terapia , Animais , Antígenos Nucleares/metabolismo , Linhagem da Célula , Movimento Celular , Células Cultivadas , Microambiente Celular , Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Recuperação de Função Fisiológica , Nicho de Células-Tronco
11.
Nature ; 539(7629): 384-389, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27820943

RESUMO

The ß-haemoglobinopathies, such as sickle cell disease and ß-thalassaemia, are caused by mutations in the ß-globin (HBB) gene and affect millions of people worldwide. Ex vivo gene correction in patient-derived haematopoietic stem cells followed by autologous transplantation could be used to cure ß-haemoglobinopathies. Here we present a CRISPR/Cas9 gene-editing system that combines Cas9 ribonucleoproteins and adeno-associated viral vector delivery of a homologous donor to achieve homologous recombination at the HBB gene in haematopoietic stem cells. Notably, we devise an enrichment model to purify a population of haematopoietic stem and progenitor cells with more than 90% targeted integration. We also show efficient correction of the Glu6Val mutation responsible for sickle cell disease by using patient-derived stem and progenitor cells that, after differentiation into erythrocytes, express adult ß-globin (HbA) messenger RNA, which confirms intact transcriptional regulation of edited HBB alleles. Collectively, these preclinical studies outline a CRISPR-based methodology for targeting haematopoietic stem cells by homologous recombination at the HBB locus to advance the development of next-generation therapies for ß-haemoglobinopathies.


Assuntos
Anemia Falciforme/genética , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Marcação de Genes , Terapia Genética/métodos , Células-Tronco Hematopoéticas/metabolismo , Globinas beta/genética , Alelos , Anemia Falciforme/patologia , Anemia Falciforme/terapia , Animais , Antígenos CD34/metabolismo , Proteínas Associadas a CRISPR/metabolismo , Diferenciação Celular , Linhagem da Célula , Separação Celular , Dependovirus/genética , Eritrócitos , Feminino , Citometria de Fluxo , Genes Reporter , Recombinação Homóloga , Humanos , Imãs , Camundongos Endogâmicos NOD , Camundongos SCID , Microesferas , Mutação , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Talassemia beta/genética , Talassemia beta/terapia
12.
Stem Cell Res ; 15(2): 341-53, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26298025

RESUMO

The effect of transplantation dose on the spatiotemporal dynamics of human neural stem cell (hNSC) engraftment has not been quantitatively evaluated in the central nervous system. We investigated changes over time in engraftment/survival, proliferation, and migration of multipotent human central nervous system-derived neural stem cells (hCNS-SCns) transplanted at doses ranging from 10,000 to 500,000 cells in spinal cord injured immunodeficient mice. Transplant dose was inversely correlated with measures of donor cell proliferation at 2 weeks post-transplant (WPT) and dose-normalized engraftment at 16 WPT. Critically, mice receiving the highest cell dose exhibited an engraftment plateau, in which the total number of engrafted human cells never exceeded the initial dose. These data suggest that donor cell expansion was inversely regulated by target niche parameters and/or transplantation density. Investigation of the response of donor cells to the host microenvironment should be a key variable in defining target cell dose in pre-clinical models of CNS disease and injury.


Assuntos
Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/terapia , Animais , Apoptose , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Humanos , Hospedeiro Imunocomprometido , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neurais/citologia , Transplante Heterólogo
13.
Stem Cell Reports ; 2(5): 620-32, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24936450

RESUMO

The microenvironment is a critical mediator of stem cell survival, proliferation, migration, and differentiation. The majority of preclinical studies involving transplantation of neural stem cells (NSCs) into the CNS have focused on injured or degenerating microenvironments, leaving a dearth of information as to how NSCs differentially respond to intact versus damaged CNS. Furthermore, single, terminal histological endpoints predominate, providing limited insight into the spatiotemporal dynamics of NSC engraftment and migration. We investigated the early and long-term engraftment dynamics of human CNS stem cells propagated as neurospheres (hCNS-SCns) following transplantation into uninjured versus subacutely injured spinal cords of immunodeficient NOD-scid mice. We stereologically quantified engraftment, survival, proliferation, migration, and differentiation at 1, 7, 14, 28, and 98 days posttransplantation, and identified injury-dependent alterations. Notably, the injured microenvironment decreased hCNS-SCns survival, delayed and altered the location of proliferation, influenced both total and fate-specific migration, and promoted oligodendrocyte maturation.


Assuntos
Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/terapia , Animais , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Proliferação de Células , Humanos , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neurais/citologia , Oligodendroglia/citologia , Traumatismos da Medula Espinal/patologia , Fatores de Tempo , Transplante Heterólogo
14.
Stem Cells Transl Med ; 2(12): 961-74, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24191264

RESUMO

The spinal cord injury (SCI) microenvironment undergoes dynamic changes over time, which could potentially affect survival or differentiation of cells in early versus delayed transplantation study designs. Accordingly, assessment of safety parameters, including cell survival, migration, fate, sensory fiber sprouting, and behavioral measures of pain sensitivity in animals receiving transplants during the chronic postinjury period is required for establishing a potential therapeutic window. The goal of the study was assessment of safety parameters for delayed transplantation of human central nervous system-derived neural stem cells (hCNS-SCns) by comparing hCNS-SCns transplantation in the subacute period, 9 days postinjury (DPI), versus the chronic period, 60 DPI, in contusion-injured athymic nude rats. Although the number of surviving human cells after chronic transplantation was lower, no changes in cell migration were detected between the 9 and 60 DPI cohorts; however, the data suggest chronic transplantation may have enhanced the generation of mature oligodendrocytes. The timing of transplantation did not induce changes in allodynia or hyperalgesia measures. Together, these data support the safety of hCNS-SCns transplantation in the chronic period post-SCI.


Assuntos
Células-Tronco Neurais/transplante , Neurogênese , Oligodendroglia/transplante , Traumatismos da Medula Espinal/cirurgia , Medula Espinal/patologia , Transplante de Células-Tronco/métodos , Animais , Biomarcadores/metabolismo , Linhagem da Célula , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Doença Crônica , Modelos Animais de Doenças , Marcha , Sobrevivência de Enxerto , Humanos , Hiperalgesia/patologia , Hiperalgesia/fisiopatologia , Atividade Motora , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Limiar da Dor , Ratos , Ratos Nus , Recuperação de Função Fisiológica , Esferoides Celulares , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Transplante de Células-Tronco/efeitos adversos , Fatores de Tempo
15.
Stem Cell Res Ther ; 4(4): 102, 2013 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-23987648

RESUMO

Human neural stem cell transplants have potential as therapeutic candidates to treat a vast number of disorders of the central nervous system (CNS). StemCells, Inc. has purified human neural stem cells and developed culture conditions for expansion and banking that preserve their unique biological properties. The biological activity of these human central nervous system stem cells (HuCNS-SC®) has been analyzed extensively in vitro and in vivo. When formulated for transplantation, the expanded and cryopreserved banked cells maintain their stem cell phenotype, self-renew and generate mature oligodendrocytes, neurons and astrocytes, cells normally found in the CNS. In this overview, the rationale and supporting data for pursuing neuroprotective strategies and clinical translation in the three components of the CNS (brain, spinal cord and eye) are described. A phase I trial for a rare myelin disorder and phase I/II trial for spinal cord injury are providing intriguing data relevant to the biological properties of neural stem cells, and the early clinical outcomes compel further development.


Assuntos
Células-Tronco Neurais , Transplante de Células-Tronco/métodos , Diferenciação Celular , Humanos
16.
Stem Cells Transl Med ; 2(10): 731-44, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23981724

RESUMO

Clinical immunosuppression protocols use calcineurin inhibitors, such as cyclosporine A (CsA) or tacrolimus (FK506), or mammalian target of rapamycin (mTOR) inhibitors, such as sirolimus (rapamycin). These compounds alter immunophilin ligand signaling pathways, which are known to interact downstream with mediators for human neural stem cell (hNSC) differentiation and proliferation, suggesting that immunosuppressants may directly alter hNSC properties. We investigated whether immunosuppressants can exert direct effects on the differentiation, proliferation, survival, and migration of human central nervous system-derived stem cells propagated as neurospheres (hCNS-SCns) in vitro and in an in vivo model of spinal cord injury. We identified unique, immunosuppressant-dependent effects on hCNS-SCns differentiation and proliferation in vitro. All immunosuppressants tested increased neuronal differentiation, and CsA and rapamycin inhibited proliferation in vitro. No immunosuppressant-mediated effects on hCNS-SCns survival or migration in vitro were detected. These data suggested that immunosuppressant administration could alter hCNS-SCns properties in vivo. We tested this hypothesis by administering immunosuppressants to constitutively immunodeficient spinal cord injured mice and assessed survival, proliferation, differentiation, and migration of hCNS-SCns after 14 weeks. In parallel, we administered immunosuppressants to immunocompetent spinal cord injury (SCI) mice and also evaluated hCNS-SCns engraftment and fate. We identified no effect of immunosuppressants on the overall hCNS-SCns fate profile in either xenotransplantation model. Despite a lower level of human cell engraftment in immunocompetent SCI mice, functional locomotor recovery was observed in animals receiving hCNS-SCns transplantation with no evidence of allodynia. These data suggest that local cues in the microenvironment could exert a stronger influence on hCNS-SCns than circulating levels of immunosuppressants; however, differences between human and rodent metabolism/pharmokinetics and xenograft versus allograft paradigms could be determining factors.


Assuntos
Imunossupressores/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Recuperação de Função Fisiológica/efeitos dos fármacos , Transplante Heterólogo
17.
Stem Cells Transl Med ; 2(3): 204-16, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23413374

RESUMO

Neural stem cell transplantation may have the potential to yield repair and recovery of function in central nervous system injury and disease, including spinal cord injury (SCI). Multiple pathological processes are initiated at the epicenter of a traumatic spinal cord injury; these are generally thought to make the epicenter a particularly hostile microenvironment. Conversely, the injury epicenter is an appealing potential site of therapeutic human central nervous system-derived neural stem cell (hCNS-SCns) transplantation because of both its surgical accessibility and the avoidance of spared spinal cord tissue. In this study, we compared hCNS-SCns transplantation into the SCI epicenter (EPI) versus intact rostral/caudal (R/C) parenchyma in contusion-injured athymic nude rats, and assessed the cell survival, differentiation, and migration. Regardless of transplantation site, hCNS-SCns survived and proliferated; however, the total number of hCNS-SCns quantified in the R/C transplant animals was twice that in the EPI animals, demonstrating increased overall engraftment. Migration and fate profile were unaffected by transplantation site. However, although transplantation site did not alter the proportion of human astrocytes, EPI transplantation shifted the localization of these cells and exhibited a correlation with calcitonin gene-related peptide fiber sprouting. Critically, no changes in mechanical allodynia or thermal hyperalgesia were observed. Taken together, these data suggest that the intact parenchyma may be a more favorable transplantation site than the injury epicenter in the subacute period post-SCI.


Assuntos
Células-Tronco Neurais/transplante , Neurônios/transplante , Traumatismos da Medula Espinal/cirurgia , Nicho de Células-Tronco , Transplante de Células-Tronco/métodos , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Comportamento Animal , Biomarcadores/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Humanos , Hiperalgesia/etiologia , Hiperalgesia/fisiopatologia , Hiperalgesia/psicologia , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Neurônios/patologia , Limiar da Dor , Ratos , Ratos Nus , Esferoides Celulares , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/psicologia , Transplante de Células-Tronco/efeitos adversos , Fatores de Tempo
18.
Sci Transl Med ; 4(155): 155ra136, 2012 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23052293

RESUMO

Shiverer-immunodeficient (Shi-id) mice demonstrate defective myelination in the central nervous system (CNS) and significant ataxia by 2 to 3 weeks of life. Expanded, banked human neural stem cells (HuCNS-SCs) were transplanted into three sites in the brains of neonatal or juvenile Shi-id mice, which were asymptomatic or showed advanced hypomyelination, respectively. In both groups of mice, HuCNS-SCs engrafted and underwent preferential differentiation into oligodendrocytes. These oligodendrocytes generated compact myelin with normalized nodal organization, ultrastructure, and axon conduction velocities. Myelination was equivalent in neonatal and juvenile mice by quantitative histopathology and high-field ex vivo magnetic resonance imaging, which, through fractional anisotropy, revealed CNS myelination 5 to 7 weeks after HuCNS-SC transplantation. Transplanted HuCNS-SCs generated functional myelin in the CNS, even in animals with severe symptomatic hypomyelination, suggesting that this strategy may be useful for treating dysmyelinating diseases.


Assuntos
Doenças Desmielinizantes/terapia , Bainha de Mielina/metabolismo , Células-Tronco Neurais/citologia , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Humanos , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Camundongos , Células-Tronco Neurais/fisiologia , Transplante de Células-Tronco
19.
Neurochem Int ; 59(3): 432-44, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21762743

RESUMO

The utilization of neural stem cells and their progeny in applications such as disease modelling, drug screening or safety assessment will require the development of robust methods for consistent, high quality uniform cell production. Previously, we described the generation of adherent, homogeneous, non-immortalized mouse and human neural stem cells derived from both brain tissue and pluripotent embryonic stem cells (Conti et al., 2005; Sun et al., 2008). In this study, we report the isolation or derivation of stable neurogenic human NS (hNS) lines from different regions of the 8-9 gestational week fetal human central nervous system (CNS) using new serum-free media formulations including animal component-free conditions. We generated more than 20 adherent hNS lines from whole brain, cortex, lobe, midbrain, hindbrain and spinal cord. We also compared the adherent hNS to some aspects of the human CNS-stem cells grown as neurospheres (hCNS-SCns), which were derived from prospectively isolated CD133(+)CD24(-/lo) cells from 16 to 20 gestational week fetal brain. We found, by RT-PCR and Taqman low-density array, that some of the regionally isolated lines maintained their regional identity along the anteroposterior axis. These NS cells exhibit the signature marker profile of neurogenic radial glia and maintain neurogenic and multipotential differentiation ability after extensive long-term expansion. Similarly, hCNS-SC can be expanded either as neurospheres or in extended adherent monolayer with a morphology and marker expression profile consistent with radial glia NS cells. We demonstrate that these lines can be efficiently genetically modified with standard nucleofection protocols for both protein overexpression and siRNA knockdown of exogenously expressed and endogenous genes exemplified with GFP and Nestin. To investigate the functional maturation of neuronal progeny derived from hNS we (a) performed Agilent whole genome microarray gene expression analysis from cultures undergoing neuronal differentiation for up to 32 days and found increased expression over time for a number of drugable target genes including neurotransmitter receptors and ion channels and (b) conducted a neuropharmacology study utilizing Fura-2 Ca(2+) imaging which revealed a clear shift from an initial glial reaction to carbachol to mature neuron-specific responses to glutamate and potassium after prolonged neuronal differentiation. Fully automated culture and scale-up of select hNS was achieved; cells supplied by the robot maintained the molecular profile of multipotent NS cells and performed faithfully in neuronal differentiation experiments. Here, we present validation and utility of a human neural lineage-restricted stem cell-based assay platform, including scale-up and automation, genetic engineering and functional characterization of differentiated progeny.


Assuntos
Neurônios/citologia , Células-Tronco/citologia , Animais , Adesão Celular , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Reação em Cadeia da Polimerase/métodos , Transplante de Células-Tronco
20.
PLoS One ; 5(8): e12272, 2010 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-20806064

RESUMO

BACKGROUND: Traumatic spinal cord injury (SCI) results in partial or complete paralysis and is characterized by a loss of neurons and oligodendrocytes, axonal injury, and demyelination/dysmyelination of spared axons. Approximately 1,250,000 individuals have chronic SCI in the U.S.; therefore treatment in the chronic stages is highly clinically relevant. Human neural stem cells (hCNS-SCns) were prospectively isolated based on fluorescence-activated cell sorting for a CD133(+) and CD24(-/lo) population from fetal brain, grown as neurospheres, and lineage restricted to generate neurons, oligodendrocytes and astrocytes. hCNS-SCns have recently been transplanted sub-acutely following spinal cord injury and found to promote improved locomotor recovery. We tested the ability of hCNS-SCns transplanted 30 days post SCI to survive, differentiate, migrate, and promote improved locomotor recovery. METHODS AND FINDINGS: hCNS-SCns were transplanted into immunodeficient NOD-scid mice 30 days post spinal cord contusion injury. hCNS-SCns transplanted mice demonstrated significantly improved locomotor recovery compared to vehicle controls using open field locomotor testing and CatWalk gait analysis. Transplanted hCNS-SCns exhibited long-term engraftment, migration, limited proliferation, and differentiation predominantly to oligodendrocytes and neurons. Astrocytic differentiation was rare and mice did not exhibit mechanical allodynia. Furthermore, differentiated hCNS-SCns integrated with the host as demonstrated by co-localization of human cytoplasm with discrete staining for the paranodal marker contactin-associated protein. CONCLUSIONS: The results suggest that hCNS-SCns are capable of surviving, differentiating, and promoting improved locomotor recovery when transplanted into an early chronic injury microenvironment. These data suggest that hCNS-SCns transplantation has efficacy in an early chronic SCI setting and thus expands the "window of opportunity" for intervention.


Assuntos
Encéfalo/citologia , Diferenciação Celular , Locomoção/fisiologia , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/cirurgia , Células-Tronco/citologia , Animais , Movimento Celular , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Traumatismos da Medula Espinal/patologia , Transplante de Células-Tronco , Células-Tronco/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...