Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Z Rheumatol ; 83(5): 393-400, 2024 Jun.
Artigo em Alemão | MEDLINE | ID: mdl-38802503

RESUMO

Lysosomal storage diseases are a group of rare hereditary metabolic diseases. Due to a deficiency of lysosomal enzymes, complex substrates accumulate in the lysosomes of various organs. Depending on the affected enzyme, this results in clinically variable and chronic progressive multiorgan diseases. Diagnosis is often delayed. As clinical symptoms include the musculoskeletal system, an awareness of lysosomal storage diseases is of relevance to (pediatric) rheumatologists. This article is focused on Mucopolysaccharidosis type I­S, Mucolipidosis type III, Gaucher disease and Fabry disease. When suspecting a lysosomal storage disease, enzyme activity should be determined in dried blood spots or leukocytes. For some diseases, specific biomarkers can additionally be analyzed. Diagnosis should be confirmed by genetic testing. As causal treatment options are available for three of the presented diseases, a timely diagnosis is very important.


Assuntos
Doenças por Armazenamento dos Lisossomos , Doenças Reumáticas , Humanos , Doenças por Armazenamento dos Lisossomos/diagnóstico , Doenças por Armazenamento dos Lisossomos/genética , Doenças Reumáticas/diagnóstico , Doenças Reumáticas/genética , Doenças Reumáticas/sangue , Reumatologia , Diagnóstico Diferencial , Medicina Baseada em Evidências
3.
Front Neurol ; 13: 907317, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35734474

RESUMO

Gaucher Disease (GD) 2 is a rare inherited lysosomal disorder. Early-onset and rapid progression of neurovisceral symptoms lead to fatal outcome in early childhood. Treatment is symptomatic, a curative therapy is currently not available. This prospective study describes the clinical and biochemical outcome of a GD 2 patient treated with high dose ambroxol from the age of 4 months. Due to progressive hepatosplenomegaly additional enzyme replacement therapy was required 1 year after ambroxol monotherapy was initiated. Detailed clinical follow-up data demonstrated an age-appropriate neurocognitive and motor development but no clear benefit on peripheral organs. Glucosylsphingosine (Lyso-GL1) in cerebrospinal fluid decreased remarkably compared to pre-treatment, whereas Lyso-GL1 and chitotriosidase in blood increased. Ambroxol treatment of patient fibroblasts revealed a significant increase in ß-glucocerebrosidase activity in vitro. To our knowledge, this is the first report of a GD 2 patient with age-appropriate cognitive and motor development at 3 years of age. Combination of high dose ambroxol with ERT proved to be a successful approach to manage both visceral and neurological manifestations.

4.
Mol Genet Metab Rep ; 23: 100578, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32226768

RESUMO

BACKGROUND: Mucopolysaccharidosis type IIIA (MPS IIIA, Sanfilippo A syndrome) is a chronic progressive neurodegenerative storage disorder caused by a deficiency of lysosomal sulfamidase. The clinical hallmarks are sleep disturbances, behavioral abnormalities and loss of cognitive, speech and motor abilities. Affected children show developmental slowing from the second year of life, dementia occurs by the age of 5 years followed by death in the second decade of life. Only a few studies concerning HSCT in MPS IIIA have been published and do not document a clear benefit of treatment. METHODS: The present study summarizes the clinical outcome of a girl with MPS IIIA who received HSCT at the age of 2.5 years. Her clinical course was compared with the natural history of six untreated MPS IIIA patients carrying the same mutations (p.R74C and p. R245H) in the SGSH-gene. RESULTS: Eight years after successful HSCT, the patient showed a global developmental delay. However, cognitive abilities continued to develop, albeit very slowly. There was no sign of regression. She could talk in short sentences, had good motor abilities and performed basic daily living activities by herself. She did not present with sleeping problems, but behavioral abnormalities were profound. In contrast, the six untreated patients with identical mutations in the SGSH-gene showed the typical progressive course of disease with early and continuous loss of abilities. CONCLUSIONS: The present data suggest a beneficial effect of HSCT performed at an early stage of MPS IIIA on cognitive skills, motor function and quality of life.

5.
PLoS Genet ; 11(5): e1005239, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26000857

RESUMO

In vitro disease modeling based on induced pluripotent stem cells (iPSCs) provides a powerful system to study cellular pathophysiology, especially in combination with targeted genome editing and protocols to differentiate iPSCs into affected cell types. In this study, we established zinc-finger nuclease-mediated genome editing in primary fibroblasts and iPSCs generated from a mouse model for radiosensitive severe combined immunodeficiency (RS-SCID), a rare disorder characterized by cellular sensitivity to radiation and the absence of lymphocytes due to impaired DNA-dependent protein kinase (DNA-PK) activity. Our results demonstrate that gene editing in RS-SCID fibroblasts rescued DNA-PK dependent signaling to overcome radiosensitivity. Furthermore, in vitro T-cell differentiation from iPSCs was employed to model the stage-specific T-cell maturation block induced by the disease causing mutation. Genetic correction of the RS-SCID iPSCs restored T-lymphocyte maturation, polyclonal V(D)J recombination of the T-cell receptor followed by successful beta-selection. In conclusion, we provide proof that iPSC-based in vitro T-cell differentiation is a valuable paradigm for SCID disease modeling, which can be utilized to investigate disorders of T-cell development and to validate gene therapy strategies for T-cell deficiencies. Moreover, this study emphasizes the significance of designer nucleases as a tool for generating isogenic disease models and their future role in producing autologous, genetically corrected transplants for various clinical applications.


Assuntos
Diferenciação Celular , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Animais , Proteína Quinase Ativada por DNA/deficiência , Proteína Quinase Ativada por DNA/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/metabolismo , Genoma , Técnicas de Genotipagem , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Camundongos , Células NIH 3T3 , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Fenótipo , Proteínas Quinases/genética , Linfócitos T/metabolismo , Dedos de Zinco
6.
J Mol Med (Berl) ; 93(1): 39-49, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25401168

RESUMO

UNLABELLED: Two long and one truncated isoforms (termed LAP*, LAP, and LIP, respectively) of the transcription factor CCAAT enhancer binding protein beta (C/EBPß) are expressed from a single intronless Cebpb gene by alternative translation initiation. Isoform expression is sensitive to mammalian target of rapamycin (mTOR)-mediated activation of the translation initiation machinery and relayed through an upstream open reading frame (uORF) on the C/EBPß mRNA. The truncated C/EBPß LIP, initiated by high mTOR activity, has been implied in neoplasia, but it was never shown whether endogenous C/EBPß LIP may function as an oncogene. In this study, we examined spontaneous tumor formation in C/EBPß knockin mice that constitutively express only the C/EBPß LIP isoform from its own locus. Our data show that deregulated C/EBPß LIP predisposes to oncogenesis in many tissues. Gene expression profiling suggests that C/EBPß LIP supports a pro-tumorigenic microenvironment, resistance to apoptosis, and alteration of cytokine/chemokine expression. The results imply that enhanced translation reinitiation of C/EBPß LIP promotes tumorigenesis. Accordingly, pharmacological restriction of mTOR function might be a therapeutic option in tumorigenesis that involves enhanced expression of the truncated C/EBPß LIP isoform. KEY MESSAGE: Elevated C/EBPß LIP promotes cancer in mice. C/EBPß LIP is upregulated in B-NHL. Deregulated C/EBPß LIP alters apoptosis and cytokine/chemokine networks. Deregulated C/EBPß LIP may support a pro-tumorigenic microenvironment.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Carcinogênese/metabolismo , Neoplasias/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Células Cultivadas , Citocinas/metabolismo , Fibroblastos , Perfilação da Expressão Gênica , Camundongos Knockout , Neoplasias/genética , Neoplasias/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
7.
Springerplus ; 3: 588, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25332888

RESUMO

Feeder cells are essential for the establishment and culture of pluripotent rat embryonic stem cells (ESC) in vitro. Therefore, we tested several fibroblast and epithelial cell lines derived from the female genital tract as feeder cells to further improve ESC culture conditions. The immortalized tumor derived rat fibroblast TRF-O3 cells isolated from a Dnd1-deficient teratoma were identified as optimal feeder cells supporting stemness and proliferation of rat ESC. The TRF-O3 cells were characterized as myofibroblasts by expression of fibroblast specific genes alpha-2 type I collagen, collagen prolyl 4-hydroxylase alpha (II), vimentin, S100A4, and smooth muscle α-actin. Culture of inner cell masses (ICM) derived from WKY/Ztm rat blastocysts in 2i-LIF medium on TRF-O3 feeder cells lacking LIF, SCF and FGF2 expression resulted in pluripotent and germ-line competent rat ESC lines. Therein, genotyping confirmed up to 26% male ESC lines. On the other hand the TRF-O3 specific BMP4 expression was correlated with transcriptional activity of the mesodermal marker T-brachyury and the ectoderm specific nestin in the ESC line ES21 demonstrating mesodermal or ectodermal cell lineage differentiation processes within the ESC population. Substitution of 2i-LIF by serum-containing YPAC medium supplemented with TGF-ß and rho kinase inhibitors or by 4i medium in combination with TRF-O3 feeder cells led to enhanced differentiation of ES21 cells and freshly isolated ICMs. These results suggest that the ESC culture conditions using TRF-O3 feeder cells and 2i-LIF medium supported the establishment of male ESC lines from WKY/Ztm rats, which represent a favored, permissive genetic background for rat ESC culture.

8.
J Clin Invest ; 123(3): 1285-98, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23434590

RESUMO

Uniparental parthenotes are considered an unwanted byproduct of in vitro fertilization. In utero parthenote development is severely compromised by defective organogenesis and in particular by defective cardiogenesis. Although developmentally compromised, apparently pluripotent stem cells can be derived from parthenogenetic blastocysts. Here we hypothesized that nonembryonic parthenogenetic stem cells (PSCs) can be directed toward the cardiac lineage and applied to tissue-engineered heart repair. We first confirmed similar fundamental properties in murine PSCs and embryonic stem cells (ESCs), despite notable differences in genetic (allelic variability) and epigenetic (differential imprinting) characteristics. Haploidentity of major histocompatibility complexes (MHCs) in PSCs is particularly attractive for allogeneic cell-based therapies. Accordingly, we confirmed acceptance of PSCs in MHC-matched allotransplantation. Cardiomyocyte derivation from PSCs and ESCs was equally effective. The use of cardiomyocyte-restricted GFP enabled cell sorting and documentation of advanced structural and functional maturation in vitro and in vivo. This included seamless electrical integration of PSC-derived cardiomyocytes into recipient myocardium. Finally, we enriched cardiomyocytes to facilitate engineering of force-generating myocardium and demonstrated the utility of this technique in enhancing regional myocardial function after myocardial infarction. Collectively, our data demonstrate pluripotency, with unrestricted cardiogenicity in PSCs, and introduce this unique cell type as an attractive source for tissue-engineered heart repair.


Assuntos
Células-Tronco Embrionárias/fisiologia , Coração/fisiologia , Engenharia Tecidual , Potenciais de Ação , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Sinalização do Cálcio , Diferenciação Celular , Forma Celular , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Genótipo , Histocompatibilidade/genética , Antígenos de Histocompatibilidade Classe II/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos SCID , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Técnicas de Cultura de Órgãos , Organoides/transplante , Partenogênese , Fenótipo , Transplante de Células-Tronco , Transplante Homólogo
9.
Genes Chromosomes Cancer ; 52(4): 423-30, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23340989

RESUMO

Suv39h1 mediates heterochromatin formation in pericentric and telomeric regions by trimethylation of lysine 9 of histone 3 (H3K9me3). Yet, its role in the induction of chromosomal instability is poorly understood. We established a leukemia model by retrovirally expressing Myc in wild-type and histone methyltransferase Suv39h1-deficient hematopoietic cells and characterized the resulting leukemias for chromosomal instability. All mice that received cells overexpressing Myc developed myeloid leukemia with a median survival of 44 days posttransplantation. Myc-overexpressing wild-type leukemias demonstrated clones with numerical chromosomal aberrations (5/16). In secondary transplantations of these leukemic cells, structural changes, mostly end-to-end fusions of chromosomes, appeared (10/12). In contrast, leukemic cells overexpressing Myc with reduced or no Suv39h1 expression had a normal karyotype in primary, secondary, and tertiary transplantations (16/16). Myc-transduced Suv39h1-deficient cells showed less critically short telomeres (P < 0.05) compared with Myc-transduced wild-type bone marrow cells. Gene expression analysis showed upregulation of genes involved in the alternative lengthening of telomeres (ALT) mechanism. Thus, we hypothesize that loss of Suv39h1 implies activation of the ALT mechanism, in turn ensuring telomere length and stability. Our data show for the first time that Suv39h1 deficiency may prevent chromosomal instability by more efficient telomere stabilization in hematopoietic bone marrow cells overexpressing Myc.


Assuntos
Instabilidade Cromossômica , Leucemia Mieloide/genética , Metiltransferases/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Repressoras/genética , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Feminino , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hibridização in Situ Fluorescente , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Masculino , Metiltransferases/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Repressoras/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cariotipagem Espectral , Telômero/genética , Homeostase do Telômero/genética , Encurtamento do Telômero/genética
10.
Blood ; 121(5): 812-21, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23118218

RESUMO

UNLABELLED: Survival of chronic lymphocytic leukemia (CLL) cells depends on stimuli provided by a suitable microenvironment. The factors and mechanisms providing this growth support for CLL cells are not fully understood. We found that plasma levels of macrophage migration inhibitory factor (MIF), a proinflammatory and immunoregulatory chemokine, were elevated in CLL patients. Therefore, we characterized the functional role of MIF in a CLL mouse model. For this purpose, we crossed Eµ-TCL1 mice with MIF knockout (MIF-/-) mice. The resulting TCL1+/wtMIF/ mice showed a delayed onset of leukemia, reduced splenomegaly and hepatomegaly, and a longer survival than TCL1+/wtMIFwt/wt controls. Immunohistochemical examination of the lymphoid organs showed that the numbers of macrophages were significantly reduced in the spleen and bone marrow of TCL1+/wtMIF/ mice compared with TCL1+/wtMIFwt/wt controls. Mechanistic studies in vitro revealed that the absence of MIF rendered CLL cells more susceptible to apoptosis. Accordingly, incubation with an anti-MIF antibody reduced the survival of CLL cells on a macrophage feeder layer. In addition, the migratory activity of TCL1+/wtMIF/ macrophages was decreased compared with TCL1+/wtMIFwt/wt macrophages. Taken together, our results provide evidence that MIF supports the development of CLL by enhancing the interaction of CLL cells with macrophages. KEY POINTS: Targeted deletion of the gene for macrophage migration inhibitory factor (MIF) delays development of chronic lymphocytic leukemia and prolongs survival in mice. MIF recruits leukemia-associated macrophages to spleen or liver.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Comunicação Celular/imunologia , Oxirredutases Intramoleculares/imunologia , Leucemia Linfocítica Crônica de Células B/imunologia , Fatores Inibidores da Migração de Macrófagos/imunologia , Macrófagos/imunologia , Animais , Linfócitos T CD8-Positivos/patologia , Sobrevivência Celular , Células Alimentadoras , Humanos , Oxirredutases Intramoleculares/genética , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Fatores Inibidores da Migração de Macrófagos/genética , Macrófagos/patologia , Camundongos , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Células Tumorais Cultivadas
11.
Stem Cells Transl Med ; 1(8): 581-91, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23197864

RESUMO

Induced pluripotent stem cells (iPSCs) may become a promising source for the generation of patient-specific hematopoietic stem cells (HSCs) in vitro. A crucial prerequisite will be the availability of reliable protocols for the directed and efficient differentiation toward HSCs. So far, the most robust strategy for generating HSCs from pluripotent cells in vitro has been established in the mouse model involving ectopic expression of the human transcription factor HOXB4. However, most differentiation protocols include coculture on a xenogenic stroma cell line and the use of animal serum. Involvement of any of both would pose a major barrier to the translation of those protocols to human autologous iPSCs intended for clinical use. Therefore, we asked whether long-term repopulating HSCs can, in principle, be generated from embryonic stem cells without stroma cells or serum. Here, we showed that long-term multilineage engraftment could be accomplished in immunocompetent mice when HSCs were generated in serum-free medium without stroma cell support and when hypoxic conditions were used. Under those conditions, HOXB4(+) embryonic stem cell-derived hematopoietic stem and progenitor cells were immunophenotypically similar to definitive bone marrow resident E-SLAM(+) (CD150(+)CD48(-)CD45(+)CD201(+)) HSCs. Thus, our findings may ease the development of definitive, adult-type HSCs from pluripotent stem cells, entirely in vitro.


Assuntos
Diferenciação Celular , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Hipóxia/metabolismo , Células-Tronco Pluripotentes/citologia , Células Estromais/citologia , Animais , Medula Óssea/metabolismo , Células Cultivadas , Técnicas de Cocultura , Meios de Cultura Livres de Soro/farmacologia , Citocinas/farmacologia , Células-Tronco Embrionárias/metabolismo , Citometria de Fluxo , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos SCID , Células-Tronco Pluripotentes/metabolismo , Regeneração/fisiologia , Células Estromais/metabolismo
12.
Mol Ther ; 20(6): 1187-95, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22472950

RESUMO

Integrating vectors developed on the basis of various retroviruses have demonstrated therapeutic potential following genetic modification of long-lived hematopoietic stem and progenitor cells. Lentiviral vectors (LV) are assumed to circumvent genotoxic events previously observed with γ-retroviral vectors, due to their integration bias to transcription units in comparison to the γ-retroviral preference for promoter regions and CpG islands. However, recently several studies have revealed the potential for gene activation by LV insertions. Here, we report a murine acute B-lymphoblastic leukemia (B-ALL) triggered by insertional gene inactivation. LV integration occurred into the 8th intron of Ebf1, a major regulator of B-lymphopoiesis. Various aberrant splice variants could be detected that involved splice donor and acceptor sites of the lentiviral construct, inducing downregulation of Ebf1 full-length message. The transcriptome signature was compatible with loss of this major determinant of B-cell differentiation, with partial acquisition of myeloid markers, including Csf1r (macrophage colony-stimulating factor (M-CSF) receptor). This was accompanied by receptor phosphorylation and STAT5 activation, both most likely contributing to leukemic progression. Our results highlight the risk of intragenic vector integration to initiate leukemia by inducing haploinsufficiency of a tumor suppressor gene. We propose to address this risk in future vector design.


Assuntos
Vetores Genéticos , Haploinsuficiência , Lentivirus/genética , Leucemia/genética , Transativadores/genética , Integração Viral , Animais , Análise por Conglomerados , Regulação para Baixo/genética , Feminino , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Ordem dos Genes , Instabilidade Genômica , Células-Tronco Hematopoéticas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Insercional , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Processamento Pós-Transcricional do RNA , Fator de Transcrição STAT5/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Transdução Genética
13.
PLoS One ; 6(12): e28911, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22194948

RESUMO

Gene knockout in murine embryonic stem cells (ESCs) has been an invaluable tool to study gene function in vitro or to generate animal models with altered phenotypes. Gene targeting using standard techniques, however, is rather inefficient and typically does not exceed frequencies of 10(-6). In consequence, the usage of complex positive/negative selection strategies to isolate targeted clones has been necessary. Here, we present a rapid single-step approach to generate a gene knockout in mouse ESCs using engineered zinc-finger nucleases (ZFNs). Upon transient expression of ZFNs, the target gene is cleaved by the designer nucleases and then repaired by non-homologous end-joining, an error-prone DNA repair process that introduces insertions/deletions at the break site and therefore leads to functional null mutations. To explore and quantify the potential of ZFNs to generate a gene knockout in pluripotent stem cells, we generated a mouse ESC line containing an X-chromosomally integrated EGFP marker gene. Applying optimized conditions, the EGFP locus was disrupted in up to 8% of ESCs after transfection of the ZFN expression vectors, thus obviating the need of selection markers to identify targeted cells, which may impede or complicate downstream applications. Both activity and ZFN-associated cytotoxicity was dependent on vector dose and the architecture of the nuclease domain. Importantly, teratoma formation assays of selected ESC clones confirmed that ZFN-treated ESCs maintained pluripotency. In conclusion, the described ZFN-based approach represents a fast strategy for generating gene knockouts in ESCs in a selection-independent fashion that should be easily transferrable to other pluripotent stem cells.


Assuntos
Células-Tronco Embrionárias/metabolismo , Endonucleases/metabolismo , Técnicas de Inativação de Genes , Dedos de Zinco/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Cromossomos de Mamíferos/metabolismo , Células-Tronco Embrionárias/citologia , Humanos , Metáfase , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
14.
Chromosome Res ; 19(7): 857-68, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22009222

RESUMO

Induced pluripotent stem (iPS) cells have greatly provoked people's interest due to their enormous potential of clinical applications. Increasing care is taken with the genetic safety of iPS cells. However, up to now, the chromosomal integrity of murine iPS (miPS) cells has been largely unknown. We have observed recurrent trisomy and/or Robertsonian translocation (Rb) of chromosome 14 in six out of nine independent miPS cell lines from three laboratories by G-banding, fluorescence in situ hybridization (FISH) and spectral karyotyping (SKY) analyses, while all the miPS cell lines were derived from mouse embryonic fibroblasts (MEFs) or neural precursor cells (NPCs) with a normal karyotype. The miPS cells with trisomy and/or Rb of chromosome 14 showed growth advantage over the miPS cells with a normal karyotype. We found a significantly higher frequency of Rbs in the miPS cell lines induced with c-Myc than those without c-Myc. Our findings demonstrate that miPS cell lines have the propensity for chromosomal aberrations and there is an obvious correlation between the extent of chromosomal aberrations in miPS cells and the transcriptional factors used for their reprogramming. Therefore, our study raises awareness of the need for improvements of the induction conditions of miPS cells in order to avoid the chromosomal aberrations and ensure future safe applications.


Assuntos
Cromossomos/genética , Hibridização in Situ Fluorescente/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Cariotipagem/métodos , Translocação Genética , Trissomia , Animais , Diferenciação Celular , Linhagem Celular , Bandeamento Cromossômico , Cromossomos/química , Fibroblastos/citologia , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariótipo , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas c-myc/farmacologia
15.
PLoS Biol ; 9(7): e1001099, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21765802

RESUMO

Using the murine model of tyrosinemia type 1 (fumarylacetoacetate hydrolase [FAH] deficiency; FAH⁻/⁻ mice) as a paradigm for orphan disorders, such as hereditary metabolic liver diseases, we evaluated fibroblast-derived FAH⁻/⁻-induced pluripotent stem cells (iPS cells) as targets for gene correction in combination with the tetraploid embryo complementation method. First, after characterizing the FAH⁻/⁻ iPS cell lines, we aggregated FAH⁻/⁻-iPS cells with tetraploid embryos and obtained entirely FAH⁻/⁻-iPS cell-derived mice that were viable and exhibited the phenotype of the founding FAH⁻/⁻ mice. Then, we transduced FAH cDNA into the FAH⁻/⁻-iPS cells using a third-generation lentiviral vector to generate gene-corrected iPS cells. We could not detect any chromosomal alterations in these cells by high-resolution array CGH analysis, and after their aggregation with tetraploid embryos, we obtained fully iPS cell-derived healthy mice with an astonishing high efficiency for full-term development of up to 63.3%. The gene correction was validated functionally by the long-term survival and expansion of FAH-positive cells of these mice after withdrawal of the rescuing drug NTBC (2-(2-nitro-4-fluoromethylbenzoyl)-1,3-cyclohexanedione). Furthermore, our results demonstrate that both a liver-specific promoter (transthyretin, TTR)-driven FAH transgene and a strong viral promoter (from spleen focus-forming virus, SFFV)-driven FAH transgene rescued the FAH-deficiency phenotypes in the mice derived from the respective gene-corrected iPS cells. In conclusion, our data demonstrate that a lentiviral gene repair strategy does not abrogate the full pluripotent potential of fibroblast-derived iPS cells, and genetic manipulation of iPS cells in combination with tetraploid embryo aggregation provides a practical and rapid approach to evaluate the efficacy of gene correction of human diseases in mouse models.


Assuntos
Fibroblastos/efeitos dos fármacos , Teste de Complementação Genética/métodos , Terapia Genética/métodos , Vetores Genéticos/farmacologia , Hidrolases , Células-Tronco Pluripotentes Induzidas , Lentivirus , Tirosinemias , Animais , Sobrevivência Celular , Células Cultivadas , Cromossomos/química , Cicloexanonas/farmacologia , Modelos Animais de Doenças , Feminino , Feto , Fibroblastos/citologia , Humanos , Hidrolases/deficiência , Hidrolases/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Nitrobenzoatos/farmacologia , Gravidez , Regiões Promotoras Genéticas , Vírus Formadores de Foco no Baço/química , Vírus Formadores de Foco no Baço/genética , Tetraploidia , Tirosinemias/genética , Tirosinemias/metabolismo , Tirosinemias/patologia , Tirosinemias/terapia
16.
Exp Cell Res ; 317(13): 1885-94, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21570390

RESUMO

Pluripotent cells referred to as embryonic germ cells (EGCs) can be derived from the embryonic precursors of the mature gametes: the primordial germ cells (PGCs). A homozygous mutation (ter) of the dead-end homolog 1 gene (Dnd1) in the rat causes gonadal teratocarcinogenesis and sterility due to neoplastic transformation and loss of germ cells. We mated heterozygous ter/+ WKY-Dnd1(ter)/Ztm rats and were able to cultivate the first genital ridge-derived EGCs of the rat embryo at day 14.5 post coitum (pc). Genotyping revealed that ten EGC lines were Dnd1 deficient, while only one wild type cell line had survived in culture. This suggests that the inactivation of the putative tumor suppressor gene Dnd1 facilitates the immortalization of late EGCs in vitro. Injection of the wild type EGCs into blastocysts resulted in the first germ-line competent chimeras. These new pluripotent rat EGCs offer an innovative approach for studies on germ cell tumor development as well as a new tool for genetic manipulations in rats.


Assuntos
Células Germinativas/citologia , Proteínas de Ligação a RNA/metabolismo , Proteínas Supressoras de Tumor/deficiência , Animais , Células Cultivadas , Feminino , Genes Supressores de Tumor , Células Germinativas/metabolismo , Masculino , Proteínas de Ligação a RNA/genética , Ratos , Ratos Endogâmicos , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
17.
PLoS One ; 6(1): e14486, 2011 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-21245929

RESUMO

BACKGROUND: Successful treatment of acute radiation syndromes relies on immediate supportive care. In patients with limited hematopoietic recovery potential, hematopoietic stem cell (HSC) transplantation is the only curative treatment option. Because of time consuming donor search and uncertain outcome we propose MSC treatment as an alternative treatment for severely radiation-affected individuals. METHODS AND FINDINGS: Mouse mesenchymal stromal cells (mMSCs) were expanded from bone marrow, retrovirally labeled with eGFP (bulk cultures) and cloned. Bulk and five selected clonal mMSCs populations were characterized in vitro for their multilineage differentiation potential and phenotype showing no contamination with hematopoietic cells. Lethally irradiated recipients were i.v. transplanted with bulk or clonal mMSCs. We found a long-term survival of recipients with fast hematopoietic recovery after the transplantation of MSCs exclusively without support by HSCs. Quantitative PCR based chimerism analysis detected eGFP-positive donor cells in peripheral blood immediately after injection and in lungs within 24 hours. However, no donor cells in any investigated tissue remained long-term. Despite the rapidly disappearing donor cells, microarray and quantitative RT-PCR gene expression analysis in the bone marrow of MSC-transplanted animals displayed enhanced regenerative features characterized by (i) decreased proinflammatory, ECM formation and adhesion properties and (ii) boosted anti-inflammation, detoxification, cell cycle and anti-oxidative stress control as compared to HSC-transplanted animals. CONCLUSIONS: Our data revealed that systemically administered MSCs provoke a protective mechanism counteracting the inflammatory events and also supporting detoxification and stress management after radiation exposure. Further our results suggest that MSCs, their release of trophic factors and their HSC-niche modulating activity rescue endogenous hematopoiesis thereby serving as fast and effective first-line treatment to combat radiation-induced hematopoietic failure.


Assuntos
Síndrome Aguda da Radiação/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Lesões Experimentais por Radiação/terapia , Células Estromais/citologia , Síndrome Aguda da Radiação/metabolismo , Síndrome Aguda da Radiação/patologia , Animais , Adesão Celular , Ciclo Celular , Células Cultivadas , Hematopoese , Humanos , Inflamação , Células-Tronco Mesenquimais/citologia , Camundongos , Lesões Experimentais por Radiação/metabolismo , Lesões Experimentais por Radiação/patologia , Proteção Radiológica/métodos , Células Estromais/efeitos da radiação , Taxa de Sobrevida , Resultado do Tratamento
18.
Cell Reprogram ; 12(1): 55-65, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20132013

RESUMO

Fusion of terminally differentiated somatic cells with pluripotent embryonic stem cells has been proposed as model for reprogramming the somatic cell genome, and may contribute to our understanding of the underlying mechanisms of this epigenetic process. We established an interspecies cell fusion model using murine embryonic stem cells (ESCs) and porcine fibroblasts. These inter-species fusion experiments yielded much lower conversion efficiency rates than murine intraspecies fusion. Nevertheless, two double-resistant mouse-pig hybrid clones could be generated. Reactivation of the porcine OCT4 gene, an essential pluripotent stem cell marker, and demethylation of the porcine OCT4 promoter in hybrid clone 1, suggested successful reprogramming of porcine chromosomes. A rapid loss of porcine chromosomes was observed during the selection phase. Spectral karyotyping (SKY) analysis showed that fusion-hybrid clone 1 carried a tetraploid mouse chromosome complement with only few pig chromosomes and/or chromosomal fragments. Hybrid clone 2 had a diploid set of murine chromosomes complements and also contained an interspecies chromosome fusion product. Interspecies cell fusion results in hybrid cells that retained the complement of mouse chromosomes and preferentially lose porcine chromosomes during colony expansion. Neither species-specific chromosomal segregation nor reprogrammed diploid porcine cells were observed. These findings indicate that generation of reprogrammed pluripotent diploid cells by cell fusion may require additional supporting provisions.


Assuntos
Reprogramação Celular , Cromossomos de Mamíferos , Células Híbridas , Animais , Animais Geneticamente Modificados , Fusão Celular/métodos , Linhagem Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/fisiologia , Estudos de Viabilidade , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Células-Tronco Pluripotentes/fisiologia , Sus scrofa/genética
19.
Blood ; 113(19): 4690-701, 2009 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-19228926

RESUMO

Loss of neurofibromin or interferon consensus sequence binding protein (Icsbp) leads to a myeloproliferative disorder. Transcription of NF1 is directly controlled by ICSBP. It has been postulated that loss of NF1 expression resulting from loss of transcriptional activation by ICSBP contributes to human hematologic malignancies. To investigate the functional cooperation of these 2 proteins, we have established Icsbp-deficient mice with Nf1 haploinsufficiency. We here demonstrate that loss of Icsbp and Nf1 haploinsufficiency synergize to induce a forced myeloproliferation in Icsbp-deficient mice because of an expansion of a mature myeloid progenitor cell. Furthermore, Nf1 haploinsufficiency and loss of Icsbp contribute synergistically to progression of the myeloproliferative disorder toward transplantable leukemias. Leukemias are characterized by distinct phenotypes, which correlate with progressive genetic abnormalities. Loss of Nf1 heterozygosity is not mandatory for disease progression, but its occurrence with other genetic abnormalities indicates progressive genetic alterations in a defined subset of leukemias. These data show that loss of the 2 tumor suppressor genes Nf1 and Icsbp synergize in the induction of leukemias.


Assuntos
Regulação Leucêmica da Expressão Gênica , Fatores Reguladores de Interferon/fisiologia , Leucemia/etiologia , Transtornos Mieloproliferativos/etiologia , Neurofibromina 1/fisiologia , Animais , Células da Medula Óssea , Ensaio de Unidades Formadoras de Colônias , Sinergismo Farmacológico , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas , Leucemia/metabolismo , Leucemia/patologia , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mielopoese , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Pathol ; 217(5): 620-32, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19191266

RESUMO

Infiltrating lobular breast cancer (ILBC) is a clinically and biologically distinct tumour entity defined by a characteristic linear cord invasion pattern and inactivation of the CDH1 tumour suppressor gene encoding for E-cadherin. ILBCs also lack beta-catenin expression and show aberrant cytoplasmic localization of the E-cadherin binding protein p120-catenin. The lack of a well-characterized ILBC cell line has hampered the functional characterization of ILBC cells in vitro. We report the establishment of a permanent ILBC cell line, named IPH-926, which was derived from a patient with metastatic ILBC. The DNA fingerprint of IPH-926 verified genetic identity with the patient and had no match among the human cell line collections of several international biological resource banks. IPH-926 expressed various epithelial cell markers but lacked expression of E-cadherin due to a previously unreported, homozygous CDH1 241ins4 frameshift mutation. Detection of the same CDH1 241ins4 mutation in archival tumour tissue of the corresponding primary ILBC proved the clonal origin of IPH-926 from this particular tumour. IPH-926 also lacked beta-catenin expression and showed aberrant cytoplasmic localization of p120-catenin. Array-CGH analysis of IPH-926 revealed a profile of genomic imbalances that included many distinct alterations previously observed in primary ILBCs. Spectral karyotyping of IPH-926 showed a hyperdiploid chromosome complement and numerous clonal, structural aberrations. IPH-926 cells were anti-cancer drug-resistant, clonogenic in soft agar, and tumourigenic in SCID mice. In xenograft tumours, IPH-926 cells recapitulated the linear cord invasion pattern that defines ILBCs. In summary, IPH-926 significantly extends the biological spectrum of the established breast cancer cell lines and will facilitate functional analyses of genuine human ILBC cells in vitro and in vivo.


Assuntos
Neoplasias da Mama/genética , Caderinas/genética , Carcinoma Lobular/genética , Idoso , Desequilíbrio Alélico , Animais , Antígenos CD , Sequência de Bases , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/deficiência , Caderinas/metabolismo , Carcinoma Lobular/metabolismo , Carcinoma Lobular/patologia , Carcinoma Lobular/secundário , Linhagem Celular Tumoral , Feminino , Humanos , Cariotipagem , Camundongos , Camundongos Endogâmicos NOD , Mutação , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...