Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Stem Cell Reports ; 17(4): 711-714, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35334219

RESUMO

The manipulation of human leukocyte antigens (HLAs) and immune modulatory factors in "universal" human pluripotent stem cells (PSCs) holds promise for immunological tolerance without HLA matching. This paradigm raises concerns should "universal" grafts become virally infected. Furthermore, immunological manipulation might functionally impair certain progeny, such as hematopoietic stem cells. We discuss the risks and benefits of hypoimmunogenic PSCs, and the need to further advance HLA matching and autologous strategies.


Assuntos
Pandemias , Células-Tronco Pluripotentes , Antígenos HLA , Humanos , Transplante de Células-Tronco/efeitos adversos
2.
Nat Commun ; 11(1): 517, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980631

RESUMO

Posttranslational modification (PTM) of proteins represents an important cellular mechanism for controlling diverse functions such as signalling, localisation or protein-protein interactions. AMPylation (also termed adenylylation) has recently been discovered as a prevalent PTM for regulating protein activity. In human cells AMPylation has been exclusively studied with the FICD protein. Here we investigate the role of AMPylation in human neurogenesis by introducing a cell-permeable propargyl adenosine pronucleotide probe to infiltrate cellular AMPylation pathways and report distinct modifications in intact cancer cell lines, human-derived stem cells, neural progenitor cells (NPCs), neurons and cerebral organoids (COs) via LC-MS/MS as well as imaging methods. A total of 162 AMP modified proteins were identified. FICD-dependent AMPylation remodelling accelerates differentiation of neural progenitor cells into mature neurons in COs, demonstrating a so far unknown trigger of human neurogenesis.


Assuntos
Monofosfato de Adenosina/metabolismo , Proteínas de Membrana/metabolismo , Neurogênese , Nucleotidiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Adenosina/metabolismo , Sequência de Aminoácidos , Catepsina B/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Regulação para Baixo , Humanos , Proteínas de Membrana/química , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Nucleotidiltransferases/química , Organoides/metabolismo
3.
Genome Biol ; 20(1): 155, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31387612

RESUMO

We describe a highly sensitive, quantitative, and inexpensive technique for targeted sequencing of transcript cohorts or genomic regions from thousands of bulk samples or single cells in parallel. Multiplexing is based on a simple method that produces extensive matrices of diverse DNA barcodes attached to invariant primer sets, which are all pre-selected and optimized in silico. By applying the matrices in a novel workflow named Barcode Assembly foR Targeted Sequencing (BART-Seq), we analyze developmental states of thousands of single human pluripotent stem cells, either in different maintenance media or upon Wnt/ß-catenin pathway activation, which identifies the mechanisms of differentiation induction. Moreover, we apply BART-Seq to the genetic screening of breast cancer patients and identify BRCA mutations with very high precision. The processing of thousands of samples and dynamic range measurements that outperform global transcriptomics techniques makes BART-Seq first targeted sequencing technique suitable for numerous research applications.


Assuntos
Perfilação da Expressão Gênica/métodos , Genômica/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Análise de Sequência de RNA/métodos , Neoplasias da Mama/genética , Análise Custo-Benefício , Células-Tronco Embrionárias/metabolismo , Feminino , Perfilação da Expressão Gênica/economia , Genômica/economia , Sequenciamento de Nucleotídeos em Larga Escala/economia , Humanos , Células-Tronco Pluripotentes/metabolismo , Análise de Sequência de RNA/economia , Análise de Célula Única/economia , Análise de Célula Única/métodos , Via de Sinalização Wnt , Fluxo de Trabalho
4.
Stem Cell Reports ; 12(5): 861-868, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31006630

RESUMO

The neural crest (NC) gives rise to a multitude of fetal tissues, and its misregulation is implicated in congenital malformations. Here, we investigated molecular mechanisms pertaining to NC-related symptoms in Bohring-Opitz syndrome (BOS), a developmental disorder linked to mutations in the Polycomb group factor Additional sex combs-like 1 (ASXL1). Genetically edited human pluripotent stem cell lines that were differentiated to NC progenitors and then xenotransplanted into chicken embryos demonstrated an impairment of NC delamination and emigration. Molecular analysis showed that ASXL1 mutations correlated with reduced activation of the transcription factor ZIC1 and the NC gene regulatory network. These findings were supported by differentiation experiments using BOS patient-derived induced pluripotent stem cell lines. Expression of truncated ASXL1 isoforms (amino acids 1-900) recapitulated the NC phenotypes in vitro and in ovo, raising the possibility that truncated ASXL1 variants contribute to BOS pathology. Collectively, we expand the understanding of truncated ASXL1 in BOS and in the human NC.


Assuntos
Diferenciação Celular/genética , Craniossinostoses/genética , Perfilação da Expressão Gênica/métodos , Deficiência Intelectual/genética , Mutação , Crista Neural/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteínas Repressoras/genética , Animais , Linhagem Celular , Células Cultivadas , Embrião de Galinha , Craniossinostoses/metabolismo , Craniossinostoses/patologia , Redes Reguladoras de Genes , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Crista Neural/citologia , Células-Tronco Pluripotentes/citologia , Proteínas Repressoras/metabolismo , Transplante Heterólogo
5.
Nat Med ; 25(4): 561-568, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30858616

RESUMO

Malformations of the human cortex represent a major cause of disability1. Mouse models with mutations in known causal genes only partially recapitulate the phenotypes and are therefore not unlimitedly suited for understanding the molecular and cellular mechanisms responsible for these conditions2. Here we study periventricular heterotopia (PH) by analyzing cerebral organoids derived from induced pluripotent stem cells (iPSCs) of patients with mutations in the cadherin receptor-ligand pair DCHS1 and FAT4 or from isogenic knockout (KO) lines1,3. Our results show that human cerebral organoids reproduce the cortical heterotopia associated with PH. Mutations in DCHS1 and FAT4 or knockdown of their expression causes changes in the morphology of neural progenitor cells and result in defective neuronal migration dynamics only in a subset of neurons. Single-cell RNA-sequencing (scRNA-seq) data reveal a subpopulation of mutant neurons with dysregulated genes involved in axon guidance, neuronal migration and patterning. We suggest that defective neural progenitor cell (NPC) morphology and an altered navigation system in a subset of neurons underlie this form of PH.


Assuntos
Movimento Celular , Cérebro/patologia , Neurônios/patologia , Organoides/patologia , Heterotopia Nodular Periventricular/patologia , Proteínas Relacionadas a Caderinas , Caderinas/genética , Linhagem Celular , Humanos , Recém-Nascido , Mutação/genética , Análise de Sequência de RNA , Análise de Célula Única , Imagem com Lapso de Tempo , Proteínas Supressoras de Tumor/genética
6.
J Cell Sci ; 132(5)2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30745340

RESUMO

The GGGGCC (G4C2) repeat expansion mutation in the C9ORF72 gene is the most common genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Transcription of the repeat and formation of nuclear RNA foci, which sequester specific RNA-binding proteins, is one of the possible pathological mechanisms. Here, we show that (G4C2) n repeat RNA predominantly associates with essential paraspeckle proteins SFPQ, NONO, RBM14, FUS and hnRNPH and colocalizes with known paraspeckle-associated RNA hLinc-p21. As formation of paraspeckles in motor neurons has been associated with early phases of ALS, we investigated the extent of similarity between paraspeckles and (G4C2) n RNA foci. Overexpression of (G4C2)72 RNA results in their increased number and colocalization with SFPQ-stained nuclear bodies. These paraspeckle-like (G4C2)72 RNA foci form independently of the known paraspeckle scaffold, the long non-coding RNA NEAT1 Moreover, the knockdown of SFPQ protein in C9ORF72 expansion mutation-positive fibroblasts significantly reduces the number of (G4C2) n RNA foci. In conclusion, (G4C2) n RNA foci have characteristics of paraspeckles, which suggests that both RNA foci and paraspeckles play roles in FTD and ALS, and implies approaches for regulation of their formation.


Assuntos
Esclerose Lateral Amiotrófica/genética , Proteína C9orf72/genética , Demência Frontotemporal/genética , Neurônios Motores/fisiologia , Complexos Multiproteicos/metabolismo , Mutação/genética , RNA Nuclear/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Animais , Proteína C9orf72/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Demência Frontotemporal/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Espaço Intranuclear , Camundongos , Fator de Processamento Associado a PTB/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Nuclear/genética , Proteína FUS de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ratos
7.
Sci Rep ; 8(1): 5101, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29572515

RESUMO

The serotonin neurotransmitter system is widespread in the brain and implicated in modulation of neuronal responses to other neurotransmitters. Among 14 serotonin receptor subtypes, 5-HT2cR plays a pivotal role in controlling neuronal network excitability. Serotonergic activity conveyed through receptor 5-HT2cR is regulated post-transcriptionally via two mechanisms, alternative splicing and A-to-I RNA editing. Brain-specific small nucleolar RNA SNORD115 harbours a phylogenetically conserved 18-nucleotide antisense element with perfect complementarity to the region of 5ht2c primary transcript that undergoes post-transcriptional changes. Previous 5ht2c minigene studies have implicated SNORD115 in fine-tuning of both post-transcriptional events. We monitored post-transcriptional changes of endogenous 5ht2c transcripts during neuronal differentiation. Both SNORD115 and 5ht2c were upregulated upon neuronal commitment. We detected increased 5ht2c alternative exon Vb inclusion already at the stage of neuronal progenitors, and more extensive A-to-I editing of non-targeted sites A and B compared to adjacent adenosines at sites E, C and D throughout differentiation. As the extent of editing is known to positively correlate with exon Vb usage while it reduces receptor functionality, our data support the model where SNORD115 directly promotes alternative exon inclusion without the requirement for conversion of key adenosines to inosines, thereby favouring production of full-length receptor isoforms with higher potency.


Assuntos
Neurogênese , Neurônios/citologia , Células-Tronco Pluripotentes/citologia , RNA Mensageiro/genética , RNA Nucleolar Pequeno/genética , Receptor 5-HT2C de Serotonina/genética , Processamento Alternativo , Animais , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Neurônios/metabolismo , Células-Tronco Pluripotentes/metabolismo , Transcriptoma
8.
Glia ; 66(2): 413-427, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29119608

RESUMO

Astrocytes, the most abundant cells in the mammalian brain, perform key functions and are involved in several neurodegenerative diseases. The human immunodeficiency virus (HIV) can persist in astrocytes, contributing to the HIV burden and neurological dysfunctions in infected individuals. While a comprehensive approach to HIV cure must include the targeting of HIV-1 in astrocytes, dedicated tools for this purpose are still lacking. Here we report a novel Adeno-associated virus-based vector (AAV9P1) with a synthetic surface peptide for transduction of astrocytes. Analysis of AAV9P1 transduction efficiencies with single brain cell populations, including primary human brain cells, as well as human brain organoids demonstrated that AAV9P1 targeted terminally differentiated human astrocytes much more efficiently than neurons. We then investigated whether AAV9P1 can be used to deliver HIV-inhibitory genes to astrocytes. To this end we generated AAV9P1 vectors containing genes for HIV-1 proviral editing by CRISPR/Cas9. Latently HIV-1 infected astrocytes transduced with these vectors showed significantly diminished reactivation of proviruses, compared with untransduced cultures. Sequence analysis identified mutations/deletions in key HIV-1 transcriptional control regions. We conclude that AAV9P1 is a promising tool for gene delivery to astrocytes and may facilitate inactivation/destruction of persisting HIV-1 proviruses in astrocyte reservoirs.


Assuntos
Astrócitos/fisiologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/fisiologia , Dependovirus/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , Vetores Genéticos/administração & dosagem , HIV-1/fisiologia , Astrócitos/efeitos dos fármacos , Astrócitos/virologia , Linhagem Celular Transformada , Células Cultivadas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/efeitos dos fármacos , Prepúcio do Pênis/citologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Células HEK293 , HIV-1/efeitos dos fármacos , Humanos , Masculino
9.
Oncotarget ; 8(65): 109575-109586, 2017 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-29312630

RESUMO

Hematopoietic Stem Cells (HSCs) generate blood and immune cells through a hierarchical process of differentiation. Genes that regulate this process are of great interest for understanding normal and also malignant hematopoiesis. Surprisingly, however, very little is known about long-non-coding RNAs (lncRNA) in HSCs. Neat1 is a lncRNA that plays a major role in the formation of sub-nuclear structures called paraspeckles, and was reported to regulate proliferation and differentiation in other cells types. We detected Neat1 expression using RNA-seq data and RT-qPCR in HSCs, progenitors and effector immune cells, by specific detection of its isoforms. Neat1 is highly expressed in stem and progenitor cells, yet it shows significant reduction in granulocytes. Microscopically, Neat1 is detected as sharp nuclear foci. Paraspeckle proteins NONO and PSPC1 are detected as aggregated nuclear foci in fresh primary hematopoietic cells, and in cultured cells. Induction of differentiation in vitro was found to enhance Neat1 expression. Taken together, our data demonstrate for the first time the expression of Neat1 and paraspeckles formation in HSCs and along hematopoiesis.

10.
Artigo em Inglês | MEDLINE | ID: mdl-25984235

RESUMO

Since the discovery of induced pluripotent stem cells (iPSC) in 2006, the symptoms of many human diseases have been reversed in animal models with iPSC therapy, setting the stage for future clinical development. From the animal data it is clear that iPSC are rapidly becoming the lead cell type for cell replacement therapy and for the newly developing field of iPSC-derived body organ transplantation. The first human pathology that might be treated in the near future with iPSC is age-related macular degeneration (AMD), which has recently passed the criteria set down by regulators for phase I clinical trials with allogeneic human embryonic stem cell-derived cell transplantation in humans. Given that iPSC are currently in clinical trial in Japan (RIKEN) to treat AMD, the establishment of a set of international criteria to make clinical-grade iPSC and their differentiated progeny is the next step in order to prepare for future autologous cell therapy clinical trials. Armed with clinical-grade iPSC, we can then specifically test for their threat of cancer, for proper and efficient differentiation to the correct cell type to treat human disease and then to determine their immunogenicity. Such a rigorous approach sets a far more relevant paradigm for their intended future use than non-clinical-grade iPSC. This review focuses on the latest developments regarding the first possible use of iPSC-derived retinal pigment epithelial cells in treating human disease, covers data gathered on animal models to date and methods to make clinical-grade iPSC, suggests techniques to ensure quality control and discusses possible clinical immune responses.

11.
Science ; 348(6232): aaa2151, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25883361

RESUMO

Dermal fibroblasts represent a heterogeneous population of cells with diverse features that remain largely undefined. We reveal the presence of at least two fibroblast lineages in murine dorsal skin. Lineage tracing and transplantation assays demonstrate that a single fibroblast lineage is responsible for the bulk of connective tissue deposition during embryonic development, cutaneous wound healing, radiation fibrosis, and cancer stroma formation. Lineage-specific cell ablation leads to diminished connective tissue deposition in wounds and reduces melanoma growth. Using flow cytometry, we identify CD26/DPP4 as a surface marker that allows isolation of this lineage. Small molecule-based inhibition of CD26/DPP4 enzymatic activity during wound healing results in diminished cutaneous scarring. Identification and isolation of these lineages hold promise for translational medicine aimed at in vivo modulation of fibrogenic behavior.


Assuntos
Separação Celular/métodos , Cicatriz/patologia , Fibroblastos/fisiologia , Pele/patologia , Cicatrização , Animais , Linhagem da Célula/genética , Cicatriz/metabolismo , Modelos Animais de Doenças , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Fibroblastos/patologia , Expressão Gênica , Proteínas de Homeodomínio/genética , Camundongos , Boca/lesões , Boca/patologia , Boca/cirurgia , Pele/lesões , Pesquisa Translacional Biomédica
12.
Methods Mol Biol ; 1029: 17-31, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23756939

RESUMO

The potential to develop into any cell type makes human pluripotent stem cells (hPSCs) one of the most promising sources for regenerative treatments. Hurdles to their clinical applications include (1) formation of heterogeneously differentiated cultures, (2) the risk of teratoma formation from residual undifferentiated cells, and (3) immune rejection of engrafted cells. The recent production of human isogenic (genetically identical) induced PSCs (hiPSCs) has been proposed as a "solution" to the histocompatibility barrier. In theory, differentiated cells derived from patient-specific hiPSC lines should be histocompatible to their donor/recipient. However, propagation, maintenance, and non-physiologic differentiation of hPSCs in vitro may produce other, likely less powerful, immune responses. In light of recent progress towards the clinical application of hPSCs, this review focuses on two antigen presentation phenomena that may lead to rejection of isogenic hPSC derivates: namely, the expression of aberrant antigens as a result of long-term in vitro maintenance conditions or incomplete somatic cell reprogramming, and the unbalanced presentation of receptors and ligands involved in immune recognition due to accelerated differentiation. Finally, we discuss immunosuppressive approaches that could potentially address these immunological concerns.


Assuntos
Diferenciação Celular/imunologia , Imunidade/imunologia , Células-Tronco Pluripotentes/imunologia , Células-Tronco Pluripotentes/transplante , Apresentação de Antígeno/imunologia , Humanos , Terapia de Imunossupressão , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Células-Tronco Pluripotentes/citologia
13.
Proc Natl Acad Sci U S A ; 110(9): 3405-10, 2013 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-23391730

RESUMO

A goal of regenerative medicine is to identify cardiovascular progenitors from human ES cells (hESCs) that can functionally integrate into the human heart. Previous studies to evaluate the developmental potential of candidate hESC-derived progenitors have delivered these cells into murine and porcine cardiac tissue, with inconclusive evidence regarding the capacity of these human cells to physiologically engraft in xenotransplantation assays. Further, the potential of hESC-derived cardiovascular lineage cells to functionally couple to human myocardium remains untested and unknown. Here, we have prospectively identified a population of hESC-derived ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells that give rise to cardiomyocytes, endothelial cells, and vascular smooth muscle cells in vitro at a clonal level. We observed rare clusters of ROR2(+) cells and diffuse expression of KDR and PDGFRα in first-trimester human fetal hearts. We then developed an in vivo transplantation model by transplanting second-trimester human fetal heart tissues s.c. into the ear pinna of a SCID mouse. ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells were delivered into these functioning fetal heart tissues: in contrast to traditional murine heart models for cell transplantation, we show structural and functional integration of hESC-derived cardiovascular progenitors into human heart.


Assuntos
Células-Tronco Embrionárias/citologia , Feto/citologia , Coração/embriologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/transplante , Transplante de Células-Tronco , Animais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem da Célula , Separação Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Endotélio Vascular/citologia , Feto/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/citologia , Camundongos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Linha Primitiva/citologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Sobrevivência de Tecidos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Nat Biotechnol ; 29(9): 829-34, 2011 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-21841799

RESUMO

An important risk in the clinical application of human pluripotent stem cells (hPSCs), including human embryonic and induced pluripotent stem cells (hESCs and hiPSCs), is teratoma formation by residual undifferentiated cells. We raised a monoclonal antibody against hESCs, designated anti-stage-specific embryonic antigen (SSEA)-5, which binds a previously unidentified antigen highly and specifically expressed on hPSCs--the H type-1 glycan. Separation based on SSEA-5 expression through fluorescence-activated cell sorting (FACS) greatly reduced teratoma-formation potential of heterogeneously differentiated cultures. To ensure complete removal of teratoma-forming cells, we identified additional pluripotency surface markers (PSMs) exhibiting a large dynamic expression range during differentiation: CD9, CD30, CD50, CD90 and CD200. Immunohistochemistry studies of human fetal tissues and bioinformatics analysis of a microarray database revealed that concurrent expression of these markers is both common and specific to hPSCs. Immunodepletion with antibodies against SSEA-5 and two additional PSMs completely removed teratoma-formation potential from incompletely differentiated hESC cultures.


Assuntos
Anticorpos Monoclonais/metabolismo , Células-Tronco Pluripotentes/citologia , Polissacarídeos/metabolismo , Antígenos Embrionários Estágio-Específicos/metabolismo , Teratoma/patologia , Animais , Biomarcadores , Diferenciação Celular , Células Cultivadas , Biologia Computacional , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Análise em Microsséries , Células-Tronco Pluripotentes/química , Reação em Cadeia da Polimerase em Tempo Real , Teratoma/metabolismo
16.
Semin Immunopathol ; 33(6): 563-72, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21479877

RESUMO

The potential to develop into any tissue makes pluripotent stem cells (PSCs) one of the most promising sources for cellular therapeutics. However, numerous hurdles exist to their clinical applications, three of the most concerning include the inability to separate therapeutic population from heterogeneously differentiated cultures, the risk of teratoma formation from residual pluripotent cells, and immunologic rejection of engrafted cells. The recent development of induced PSCs has been proposed as a solution to the histocompatibility barrier. Theoretically, creation of patient-specific induced PSC lines would exhibit a complete histocompatibility antigen match. However, regardless of the PSC source, in vitro propagation and nonphysiologic differentiation may result in other, likely less powerful, mechanisms of immune rejection. In light of recent progress towards clinical application, this review focuses on two such potential immunologic mechanisms applicable to isogenic PSC derivates: namely, the immunogenicity of aberrant antigens resulting from long-term in vitro maintenance and alterations in immunologic properties due to rapid in vitro differentiation. These issues will be considered with attention to their relation to effector cells in the adult immune system. In addition, we highlight immunosuppressive approaches that could potentially address the immunogenicity of these proposed mechanisms.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia , Animais , Antígenos/imunologia , Células Cultivadas , Citotoxicidade Imunológica , Rejeição de Enxerto/imunologia , Humanos , Imunomodulação , Terapia de Imunossupressão , Células Matadoras Naturais/imunologia , Células-Tronco Pluripotentes/metabolismo , Antígenos Embrionários Estágio-Específicos/imunologia , Antígenos Embrionários Estágio-Específicos/metabolismo , Transplante de Células-Tronco , Linfócitos T/imunologia , Transplante Homólogo/imunologia
17.
Proc Natl Acad Sci U S A ; 108(8): 3282-7, 2011 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-21300885

RESUMO

The promise of pluripotent stem cells as a research and therapeutic tool is partly undermined by the technical challenges of generating and maintaining these cells in culture. Human embryonic stem cells (hESCs) are exquisitely sensitive to culture conditions, and require constant signaling by growth factors and cell-cell and cell-matrix interactions to prevent apoptosis, senescence, and differentiation. Previous work from our laboratory demonstrated that overexpression of the prosurvival gene BCL2 in mouse embryonic stem cells overrode the requirement of serum factors and feeder cells to maintain mESCs in culture. To determine whether this prosurvival gene could similarly protect hESCs, we generated hESC lines that constitutively or inducibly express BCL2. We find that BCL2 overexpression significantly decreases dissociation-induced apoptosis, resulting in enhanced colony formation from sorted single cells, and enhanced embryoid body formation. In addition, BCL2-hESCs exhibit normal growth in the absence of serum, but require basic fibroblast growth factor to remain undifferentiated. Furthermore, they maintain their pluripotency markers, form teratomas in vivo, and differentiate into all three germ layers. Our data suggest that the BCL2 signaling pathway plays an important role in inhibiting hESC apoptosis, such that its overexpression in hESCs offers both a survival benefit in conditions of stress by resisting apoptosis and obviates the requirement for serum or a feeder layer for maintenance.


Assuntos
Células-Tronco Embrionárias/citologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular/genética , Células-Tronco Embrionárias/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Genes bcl-2/fisiologia , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Soro , Estresse Fisiológico
18.
Cell Stem Cell ; 6(4): 361-368, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20362540

RESUMO

The lineage restriction of prospectively isolated hematopoietic progenitors has been traditionally assessed by bulk in vitro culture and transplantation of large number of cells in vivo. These methods, however, cannot distinguish between homogenous multipotent or heterogeneous lineage-restricted populations. Using clonal assays of 1 or 5 cells in vitro, single-cell quantitative gene expression analyses, and transplantation of mice with low numbers of cells, we show that a common myeloid progenitor (CMP) is Sca-1(lo)lin(-)c-Kit(+)CD27(+)Flk-2(-) (SL-CMP; Sca-1(lo) CMP) and a granulocyte/macrophage progenitor (GMP) is Sca-1(lo)lin(-)c-Kit(+)CD27(+)Flk-2(+)CD150(-/lo) (SL-GMP; Sca-1(lo) GMP). We found that mast cell progenitor potential is present in the SL-CMP fraction, but not in the more differentiated SL-GMP population, and is more closely related to megakaryocyte/erythrocyte specification. Our data provide criteria for the prospective isolation of SL-CMP and SL-GMP and support the conclusion that mast cells are specified during hematopoiesis earlier than and independently from granulocytes.


Assuntos
Bioensaio/métodos , Diferenciação Celular , Granulócitos/citologia , Mastócitos/citologia , Animais , Antígenos Ly/metabolismo , Ataxina-1 , Ataxinas , Contagem de Células , Diferenciação Celular/genética , Linhagem da Célula/genética , Regulação da Expressão Gênica , Células Progenitoras de Granulócitos e Macrófagos/citologia , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Granulócitos/metabolismo , Transplante de Células-Tronco Hematopoéticas , Mastócitos/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Reprodutibilidade dos Testes
19.
Magn Reson Med ; 62(4): 1047-54, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19526508

RESUMO

This study investigated the ability of MnCl(2) as a cellular MRI contrast agent to determine the in vitro viability of human embryonic stem cells (hESC) and human bone marrow stromal cells (hBMSC). Basic MRI parameters including T(1) and T(2) values of MnCl(2)-labeled hESC and hBMSC were measured and viability signal of manganese (Mn(2+))-labeled cells was validated. Furthermore, the biological activity of Ca(2+)-channels was modulated utilizing both Ca(2+)-channel agonist and antagonist to evaluate concomitant signal changes. Metabolic effects of MnCl(2)-labeling were also assessed using assays for cell viability, proliferation, and apoptosis. Finally, in vivo Mn(2+)-guided MRI of the transplanted hESC was successfully achieved and validated by bioluminescence imaging.


Assuntos
Cloretos , Células-Tronco Embrionárias/citologia , Aumento da Imagem/métodos , Imageamento por Ressonância Magnética/métodos , Compostos de Manganês , Células-Tronco Mesenquimais/citologia , Linhagem Celular , Sobrevivência Celular , Meios de Contraste , Humanos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
20.
PLoS One ; 3(10): e3474, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18941512

RESUMO

Human embryonic stem cells (hESCs) can serve as a potentially limitless source of cells that may enable regeneration of diseased tissue and organs. Here we investigate the use of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) in promoting recovery from cardiac ischemia reperfusion injury in a mouse model. Using microarrays, we have described the hESC-CM transcriptome within the spectrum of changes that occur between undifferentiated hESCs and fetal heart cells. The hESC-CMs expressed cardiomyocyte genes at levels similar to those found in 20-week fetal heart cells, making this population a good source of potential replacement cells in vivo. Echocardiographic studies showed significant improvement in heart function by 8 weeks after transplantation. Finally, we demonstrate long-term engraftment of hESC-CMs by using molecular imaging to track cellular localization, survival, and proliferation in vivo. Taken together, global gene expression profiling of hESC differentiation enables a systems-based analysis of the biological processes, networks, and genes that drive hESC fate decisions, and studies such as this will serve as the foundation for future clinical applications of stem cell therapies.


Assuntos
Células-Tronco Embrionárias/citologia , Perfilação da Expressão Gênica , Traumatismo por Reperfusão Miocárdica/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Eletrocardiografia , Células-Tronco Embrionárias/transplante , Sobrevivência de Enxerto , Humanos , Camundongos , Modelos Animais , Miócitos Cardíacos/transplante , Análise de Sequência com Séries de Oligonucleotídeos , Transcrição Gênica , Transplante Heterólogo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA