Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nat Immunol ; 18(1): 74-85, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27893700

RESUMEN

The cellular sources of interleukin 6 (IL-6) that are relevant for differentiation of the TH17 subset of helper T cells remain unclear. Here we used a novel strategy for the conditional deletion of distinct IL-6-producing cell types to show that dendritic cells (DCs) positive for the signaling regulator Sirpα were essential for the generation of pathogenic TH17 cells. Using their IL-6 receptor α-chain (IL-6Rα), Sirpα+ DCs trans-presented IL-6 to T cells during the process of cognate interaction. While ambient IL-6 was sufficient to suppress the induction of expression of the transcription factor Foxp3 in T cells, trans-presentation of IL-6 by DC-bound IL-6Rα (called 'IL-6 cluster signaling' here) was needed to prevent premature induction of interferon-γ (IFN-γ) expression in T cells and to generate pathogenic TH17 cells in vivo. Our findings should guide therapeutic approaches for the treatment of TH17-cell-mediated autoimmune diseases.


Asunto(s)
Sistema Nervioso Central/inmunología , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Subunidad alfa del Receptor de Interleucina-6/genética , Interleucina-6/metabolismo , Células Th17/inmunología , Animales , Autoinmunidad , Diferenciación Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
2.
Genes Dev ; 33(9-10): 524-535, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30862660

RESUMEN

The balance between proliferation and differentiation of muscle stem cells is tightly controlled, ensuring the maintenance of a cellular pool needed for muscle growth and repair. We demonstrate here that the transcriptional regulator Hes1 controls the balance between proliferation and differentiation of activated muscle stem cells in both developing and regenerating muscle. We observed that Hes1 is expressed in an oscillatory manner in activated stem cells where it drives the oscillatory expression of MyoD. MyoD expression oscillates in activated muscle stem cells from postnatal and adult muscle under various conditions: when the stem cells are dispersed in culture, when they remain associated with single muscle fibers, or when they reside in muscle biopsies. Unstable MyoD oscillations and long periods of sustained MyoD expression are observed in differentiating cells. Ablation of the Hes1 oscillator in stem cells interfered with stable MyoD oscillations and led to prolonged periods of sustained MyoD expression, resulting in increased differentiation propensity. This interfered with the maintenance of activated muscle stem cells, and impaired muscle growth and repair. We conclude that oscillatory MyoD expression allows the cells to remain in an undifferentiated and proliferative state and is required for amplification of the activated stem cell pool.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Proteína MioD/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factor de Transcripción HES-1/metabolismo , Animales , Células Cultivadas , Ratones , Proteína MioD/genética , Receptores Notch/metabolismo , Transducción de Señal , Factor de Transcripción HES-1/genética
4.
J Hepatol ; 74(2): 407-418, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32987028

RESUMEN

BACKGROUND & AIMS: Interleukin (IL)-6 cytokine family members contribute to inflammatory and regenerative processes. Engagement of the signaling receptor subunit gp130 is common to almost all members of the family. In the liver, all major cell types respond to IL-6-type cytokines, making it difficult to delineate cell type-specific effects. We therefore generated mouse models for liver cell type-specific analysis of IL-6 signaling. METHODS: We produced mice with a Cre-inducible expression cassette encoding a designed pre-dimerized constitutive active gp130 variant. We bred these mice to different Cre-drivers to induce transgenic gp130 signaling in distinct liver cell types: hepatic stellate cells, cholangiocytes/liver progenitor cells or hepatocytes. We phenotyped these mice using multi-omics approaches, immunophenotyping and a bacterial infection model. RESULTS: Hepatocyte-specific gp130 activation led to the upregulation of innate immune system components, including acute-phase proteins. Consequently, we observed peripheral mobilization and recruitment of myeloid cells to the liver. Hepatic myeloid cells, including liver-resident Kupffer cells were instructed to adopt a bactericidal phenotype which ultimately conferred enhanced resistance to bacterial infection in these mice. We demonstrate that persistent hepatocyte-specific gp130 activation resulted in amyloid A amyloidosis in aged mice. In contrast, we did not observe overt effects of hepatic stellate cell- or cholangiocyte/liver progenitor cell-specific transgenic gp130 signaling. CONCLUSIONS: Hepatocyte-specific gp130 activation alone is sufficient to trigger a robust innate immune response in the absence of NF-κB activation. We therefore conclude that gp130 engagement, e.g. by IL-6 trans-signaling, represents a safe-guard mechanism in innate immunity. LAY SUMMARY: Members of the interleukin-6 cytokine family signal via the receptor subunit gp130 and are involved in multiple processes in the liver. However, as several liver cell types respond to interleukin-6 family cytokines, it is difficult to delineate cell type-specific effects. Using a novel mouse model, we provide evidence that hepatocyte-specific gp130 activation is sufficient to trigger a robust systemic innate immune response.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Hepatocitos/metabolismo , Inmunidad Innata/fisiología , Interleucina-6/inmunología , Hígado , Factor de Transcripción STAT3/metabolismo , Reacción de Fase Aguda/inmunología , Animales , Hepatocitos/clasificación , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Transgénicos , Modelos Animales , Transducción de Señal/inmunología
5.
Proc Natl Acad Sci U S A ; 110(24): 9862-7, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23696660

RESUMEN

The microbiota is pivotal in the pathogenesis of inflammatory bowel disease (IBD)-associated inflammation-induced colorectal cancer (CRC), yet mechanisms for these effects remain poorly characterized. Here, we demonstrate that aberrant inflammasome-induced microbiota plays a critical role in CRC development, where mice deficient in the NOD-like receptor family pyrin domain containing 6 (NLRP6) inflammasome feature enhanced inflammation-induced CRC formation. Intriguingly, WT mice cohoused either with inflammasome-deficient mice or with mice lacking IL-18 feature exacerbated inflammation-induced CRC compared with singly housed WT mice. Enhanced tumorigenesis is dependent on microbiota-induced chemokine (C-C motif) ligand 5 (CCL5)-driven inflammation, which in turn promotes epithelial cell proliferation through local activation of the IL-6 pathway, leading to cancer formation. Altogether, our results mechanistically link the altered microbiota with the pathogenesis of inflammation-induced CRC and suggest that in some conditions, microbiota components may transfer CRC susceptibility between individuals.


Asunto(s)
Inflamasomas/inmunología , Inflamación/inmunología , Interleucina-6/inmunología , Metagenoma/inmunología , Neoplasias/inmunología , Animales , Quimiocina CCL5/deficiencia , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Colonoscopía , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Epitelio/inmunología , Epitelio/metabolismo , Epitelio/microbiología , Femenino , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Interleucina-18/deficiencia , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/genética , Neoplasias/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Transducción de Señal/inmunología
6.
Hum Mol Genet ; 22(7): 1328-47, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23263861

RESUMEN

F-actin bundling plastin 3 (PLS3) is a fully protective modifier of the neuromuscular disease spinal muscular atrophy (SMA), the most common genetic cause of infant death. The generation of a conditional PLS3-over-expressing mouse and its breeding into an SMA background allowed us to decipher the exact biological mechanism underlying PLS3-mediated SMA protection. We show that PLS3 is a key regulator that restores main processes depending on actin dynamics in SMA motor neurons (MNs). MN soma size significantly increased and a higher number of afferent proprioceptive inputs were counted in SMAPLS3 compared with SMA mice. PLS3 increased presynaptic F-actin amount, rescued synaptic vesicle and active zones content, restored the organization of readily releasable pool of vesicles and increased the quantal content of the neuromuscular junctions (NMJs). Most remarkably, PLS3 over-expression led to a stabilization of axons which, in turn, resulted in a significant delay of axon pruning, counteracting poor axonal connectivity at SMA NMJs. These findings together with the observation of increased endplate and muscle fiber size upon MN-specific PLS3 over-expression suggest that PLS3 significantly improves neuromuscular transmission. Indeed, ubiquitous over-expression moderately improved survival and motor function in SMA mice. As PLS3 seems to act independently of Smn, PLS3 might be a potential therapeutic target not only in SMA but also in other MN diseases.


Asunto(s)
Glicoproteínas de Membrana/fisiología , Proteínas de Microfilamentos/fisiología , Placa Motora/fisiopatología , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/patología , Actinas/metabolismo , Animales , Potenciales Evocados Motores , Expresión Génica , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Placa Motora/metabolismo , Placa Motora/patología , Neuronas Motoras/patología , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/fisiopatología , Fenotipo , Propiocepción , Transporte de Proteínas , Receptores Colinérgicos/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Sinapsis/metabolismo , Vesículas Sinápticas/metabolismo
7.
Nature ; 460(7259): 1159-63, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19641494

RESUMEN

Reactive oxygen species (ROS) produced by NADPH oxidase function as defence and signalling molecules related to innate immunity and various cellular responses. The activation of NADPH oxidase in response to plasma membrane receptor activation depends on the phosphorylation of cytoplasmic oxidase subunits, their translocation to membranes and the assembly of all NADPH oxidase components. Tumour necrosis factor (TNF) is a prominent stimulus of ROS production, but the molecular mechanisms by which TNF activates NADPH oxidase are poorly understood. Here we identify riboflavin kinase (RFK, formerly known as flavokinase) as a previously unrecognized TNF-receptor-1 (TNFR1)-binding protein that physically and functionally couples TNFR1 to NADPH oxidase. In mouse and human cells, RFK binds to both the TNFR1-death domain and to p22(phox), the common subunit of NADPH oxidase isoforms. RFK-mediated bridging of TNFR1 and p22(phox) is a prerequisite for TNF-induced but not for Toll-like-receptor-induced ROS production. Exogenous flavin mononucleotide or FAD was able to substitute fully for TNF stimulation of NADPH oxidase in RFK-deficient cells. RFK is rate-limiting in the synthesis of FAD, an essential prosthetic group of NADPH oxidase. The results suggest that TNF, through the activation of RFK, enhances the incorporation of FAD in NADPH oxidase enzymes, a critical step for the assembly and activation of NADPH oxidase.


Asunto(s)
NADPH Oxidasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Línea Celular , Grupo Citocromo b/metabolismo , Activación Enzimática , Fibroblastos , Mononucleótido de Flavina/metabolismo , Flavina-Adenina Dinucleótido/biosíntesis , Flavina-Adenina Dinucleótido/metabolismo , Células HeLa , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasas/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Unión Proteica , Estructura Terciaria de Proteína , Especies Reactivas de Oxígeno/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/química
8.
FASEB J ; 27(5): 1973-80, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23395911

RESUMEN

Loss of appetite is a hallmark of inflammatory diseases. The underlying mechanisms remain undefined, but it is known that myeloid differentiation primary response gene 88 (MyD88), an adaptor protein critical for Toll-like and IL-1 receptor family signaling, is involved. Here we addressed the question of determining in which cells the MyD88 signaling that results in anorexia development occurs by using chimeric mice and animals with cell-specific deletions. We found that MyD88-knockout mice, which are resistant to bacterial lipopolysaccharide (LPS)-induced anorexia, displayed anorexia when transplanted with wild-type bone marrow cells. Furthermore, mice with a targeted deletion of MyD88 in hematopoietic or myeloid cells were largely protected against LPS-induced anorexia and displayed attenuated weight loss, whereas mice with MyD88 deletion in hepatocytes or in neural cells or the cerebrovascular endothelium developed anorexia and weight loss of similar magnitude as wild-type mice. Furthermore, in a model for cancer-induced anorexia-cachexia, deletion of MyD88 in hematopoietic cells attenuated the anorexia and protected against body weight loss. These findings demonstrate that MyD88-dependent signaling within the brain is not required for eliciting inflammation-induced anorexia. Instead, we identify MyD88 signaling in hematopoietic/myeloid cells as a critical component for acute inflammatory-driven anorexia, as well as for chronic anorexia and weight loss associated with malignant disease.


Asunto(s)
Anorexia/fisiopatología , Encéfalo/citología , Caquexia/fisiopatología , Células Endoteliales/fisiología , Inflamación/fisiopatología , Células Mieloides/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Sarcoma Experimental/fisiopatología , Animales , Quimera/fisiología , Metilcolantreno , Ratones , Ratones Noqueados , Neuronas/citología , Sarcoma Experimental/inducido químicamente , Transducción de Señal/fisiología , Pérdida de Peso/fisiología
9.
J Immunol ; 189(10): 4770-6, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23066153

RESUMEN

CYLD was originally identified as a tumor suppressor gene mutated in familial cylindromatosis, an autosomal dominant predisposition to multiple benign neoplasms of the skin known as cylindromas. The CYLD protein is a deubiquitinating enzyme that acts as a negative regulator of NF-κB and JNK signaling through its interaction with NEMO and TNFR-associated factor 2. We have previously described a novel mouse strain that expresses solely and excessively a naturally occurring splice variant of CYLD (CYLD(ex7/8)). In this study, we demonstrate that CYLD plays a critical role in Treg development and function. T cells of CYLD(ex7/8) mice had a hyperactive phenotype manifested by increased production of inflammatory cytokines and constitutive activation of the NF-κB pathway. Furthermore, the amount of Foxp3(+) regulatory T cells in these mice was markedly enhanced in thymus and peripheral organs. Importantly, these regulatory T cells displayed decreased expression levels of CD25 and CTLA-4 associated with impaired suppressive capacity. Hence, our data emphasize an essential role of CYLD in maintaining T cell homeostasis as well as normal T regulatory cell function, thereby controlling abnormal T cell responses.


Asunto(s)
Cisteína Endopeptidasas/inmunología , Linfocitos T Reguladores/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Antígeno CTLA-4/metabolismo , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Enzima Desubiquitinante CYLD , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Mutantes , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
Cell Rep ; 43(4): 113960, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38507407

RESUMEN

GFRAL-expressing neurons actuate aversion and nausea, are targets for obesity treatment, and may mediate metformin effects by long-term GDF15-GFRAL agonism. Whether GFRAL+ neurons acutely regulate glucose and energy homeostasis is, however, underexplored. Here, we report that cell-specific activation of GFRAL+ neurons using a variety of techniques causes a torpor-like state, including hypothermia, the release of stress hormones, a shift from glucose to lipid oxidation, and impaired insulin sensitivity, glucose tolerance, and skeletal muscle glucose uptake but augmented glucose uptake in visceral fat. Metabolomic analysis of blood and transcriptomics of muscle and fat indicate alterations in ketogenesis, insulin signaling, adipose tissue differentiation and mitogenesis, and energy fluxes. Our findings indicate that acute GFRAL+ neuron activation induces endocrine and gluco- and thermoregulatory responses associated with nausea and torpor. While chronic activation of GFRAL signaling promotes weight loss in obesity, these results show that acute activation of GFRAL+ neurons causes hypothermia and hyperglycemia.


Asunto(s)
Glucosa , Hipotermia , Náusea , Neuronas , Letargo , Animales , Neuronas/metabolismo , Náusea/metabolismo , Hipotermia/metabolismo , Letargo/fisiología , Glucosa/metabolismo , Ratones , Masculino , Músculo Esquelético/metabolismo , Ratones Endogámicos C57BL , Insulina/metabolismo , Resistencia a la Insulina , Transducción de Señal
11.
Eur Heart J Open ; 4(4): oeae046, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39015379

RESUMEN

Aims: The cytokine interleukin-6 (IL-6) plays a central role in the inflammation cascade as well as cardiovascular disease progression. Since myeloid cells are a primary source of IL-6 formation, we aimed to generate a mouse model to study the role of myeloid cell-derived IL-6 in vascular disease. Methods and results: Interleukin-6-overexpressing (IL-6OE) mice were generated and crossed with LysM-Cre mice, to generate mice (LysM-IL-6OE mice) overexpressing the cytokine in myeloid cells. Eight- to 12-week-old LysM-IL-6OE mice spontaneously developed inflammatory colitis and significantly impaired endothelium-dependent aortic relaxation, increased aortic reactive oxygen species (ROS) formation, and vascular dysfunction in resistance vessels. The latter phenotype was associated with decreased survival. Vascular dysfunction was accompanied by a significant accumulation of neutrophils, monocytes, and macrophages in the aorta, increased myeloid cell reactivity (elevated ROS production), and vascular fibrosis associated with phenotypic changes in vascular smooth muscle cells. In addition to elevated Mcp1 and Cxcl1 mRNA levels, aortae from LysM-IL-6OE mice expressed higher levels of inducible NO synthase and endothelin-1, thus partially accounting for vascular dysfunction, whereas systemic blood pressure alterations were not observed. Bone marrow (BM) transplantation experiments revealed that vascular dysfunction and ROS formation were driven by BM cell-derived IL-6 in a dose-dependent manner. Conclusion: Mice with conditional overexpression of IL-6 in myeloid cells show systemic and vascular inflammation as well as endothelial dysfunction. A decrease in circulating IL-6 levels by replacing IL-6-producing myeloid cells in the BM improved vascular dysfunction in this model, underpinning the relevant role of IL-6 in vascular disease.

12.
Nat Aging ; 3(11): 1430-1445, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37946043

RESUMEN

Tissues within an organism and even cell types within a tissue can age with different velocities. However, it is unclear whether cells of one type experience different aging trajectories within a tissue depending on their spatial location. Here, we used spatial transcriptomics in combination with single-cell ATAC-seq and RNA-seq, lipidomics and functional assays to address how cells in the male murine liver are affected by age-related changes in the microenvironment. Integration of the datasets revealed zonation-specific and age-related changes in metabolic states, the epigenome and transcriptome. The epigenome changed in a zonation-dependent manner and functionally, periportal hepatocytes were characterized by decreased mitochondrial fitness, whereas pericentral hepatocytes accumulated large lipid droplets. Together, we provide evidence that changing microenvironments within a tissue exert strong influences on their resident cells that can shape epigenetic, metabolic and phenotypic outputs.


Asunto(s)
Epigenoma , Transcriptoma , Masculino , Ratones , Animales , Transcriptoma/genética , Epigenoma/genética , Hígado/metabolismo , Hepatocitos/metabolismo , Metaboloma
13.
Nat Commun ; 13(1): 4352, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35896539

RESUMEN

Obesity is a pre-disposing condition for chronic obstructive pulmonary disease, asthma, and pulmonary arterial hypertension. Accumulating evidence suggests that metabolic influences during development can determine chronic lung diseases (CLD). We demonstrate that maternal obesity causes early metabolic disorder in the offspring. Here, interleukin-6 induced bronchial and microvascular smooth muscle cell (SMC) hyperproliferation and increased airway and pulmonary vascular resistance. The key anti-proliferative transcription factor FoxO1 was inactivated via nuclear exclusion. These findings were confirmed using primary SMC treated with interleukin-6 and pharmacological FoxO1 inhibition as well as genetic FoxO1 ablation and constitutive activation. In vivo, we reproduced the structural and functional alterations in offspring of obese dams via the SMC-specific ablation of FoxO1. The reconstitution of FoxO1 using IL-6-deficient mice and pharmacological treatment did not protect against metabolic disorder but prevented SMC hyperproliferation. In human observational studies, childhood obesity was associated with reduced forced expiratory volume in 1 s/forced vital capacity ratio Z-score (used as proxy for lung function) and asthma. We conclude that the interleukin-6-FoxO1 pathway in SMC is a molecular mechanism by which perinatal obesity programs the bronchial and vascular structure and function, thereby driving CLD development. Thus, FoxO1 reconstitution provides a potential therapeutic option for preventing this metabolic programming of CLD.


Asunto(s)
Asma , Hipertensión Pulmonar , Obesidad Infantil , Animales , Asma/metabolismo , Niño , Femenino , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Humanos , Hipertensión Pulmonar/genética , Interleucina-6/metabolismo , Ratones , Miocitos del Músculo Liso/metabolismo , Obesidad Infantil/complicaciones , Obesidad Infantil/metabolismo , Embarazo
14.
Cell Prolif ; 53(10): e12892, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32918782

RESUMEN

OBJECTIVES: Genetic engineering of human-induced pluripotent stem cell-derived neural stem cells (hiPSC-NSC) may increase the risk of genomic aberrations. Therefore, we asked whether genetic modification of hiPSC-NSCs exacerbates chromosomal abnormalities that may occur during passaging and whether they may cause any functional perturbations in NSCs in vitro and in vivo. MATERIALS AND METHODS: The transgenic cassette was inserted into the AAVS1 locus, and the genetic integrity of zinc-finger nuclease (ZFN)-modified hiPSC-NSCs was assessed by the SNP-based karyotyping. The hiPSC-NSC proliferation was assessed in vitro by the EdU incorporation assay and in vivo by staining of brain slices with Ki-67 antibody at 2 and 8 weeks after transplantation of ZFN-NSCs with and without chromosomal aberration into the striatum of immunodeficient rats. RESULTS: During early passages, no chromosomal abnormalities were detected in unmodified or ZFN-modified hiPSC-NSCs. However, at higher passages both cell populations acquired duplication of the entire long arm of chromosome 1, dup(1)q. ZNF-NSCs carrying dup(1)q exhibited higher proliferation rate than karyotypically intact cells, which was partly mediated by increased expression of AKT3 located on Chr1q. Compared to karyotypically normal ZNF-NSCs, cells with dup(1)q also exhibited increased proliferation in vivo 2 weeks, but not 2 months, after transplantation. CONCLUSIONS: These results demonstrate that, independently of ZFN-editing, hiPSC-NSCs have a propensity for acquiring dup(1)q and this aberration results in increased proliferation which might compromise downstream hiPSC-NSC applications.


Asunto(s)
Cromosomas Humanos Par 1/genética , Edición Génica/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Duplicación de Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Cariotipo , Células-Madre Neurales/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Dedos de Zinc/genética
15.
PLoS One ; 9(9): e107363, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25226590

RESUMEN

Cell loss after transplantation is a major limitation for cell replacement approaches in regenerative medicine. To assess the survival kinetics of induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) we generated transgenic murine iPSC lines which, in addition to CM-specific expression of puromycin N-acetyl-transferase and enhanced green fluorescent protein (EGFP), also constitutively express firefly luciferase (FLuc) for bioluminescence (BL) in vivo imaging. While undifferentiated iPSC lines generated by random integration of the transgene into the genome retained stable FLuc activity over many passages, the BL signal intensity was strongly decreased in purified iPS-CM compared to undifferentiated iPSC. Targeted integration of FLuc-expression cassette into the ROSA26 genomic locus using zinc finger nuclease (ZFN) technology strongly reduced transgene silencing in iPS-CM, leading to a several-fold higher BL compared to iPS-CM expressing FLuc from random genomic loci. To investigate the survival kinetics of iPS-CM in vivo, purified CM obtained from iPSC lines expressing FLuc from a random or the ROSA26 locus were transplanted into cryoinfarcted hearts of syngeneic mice. Engraftment of viable cells was monitored by BL imaging over 4 weeks. Transplanted iPS-CM were poorly retained in the myocardium independently of the cell line used. However, up to 8% of cells survived for 28 days at the site of injection, which was confirmed by immunohistological detection of EGFP-positive iPS-CM in the host tissue. Transplantation of iPS-CM did not affect the scar formation or capillary density in the periinfarct region of host myocardium. This report is the first to determine the survival kinetics of drug-selected iPS-CM in the infarcted heart using BL imaging and demonstrates that transgene silencing in the course of iPSC differentiation can be greatly reduced by employing genome editing technology. FLuc-expressing iPS-CM generated in this study will enable further studies to reduce their loss, increase long-term survival and functional integration upon transplantation.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Mediciones Luminiscentes/métodos , Imagen Molecular , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Miocitos Cardíacos/trasplante , Animales , Diferenciación Celular , Línea Celular , Expresión Génica , Silenciador del Gen , Genes Reporteros , Sitios Genéticos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Regiones Promotoras Genéticas , ARN no Traducido/genética , Transducción Genética , Transgenes
16.
Sci Rep ; 3: 2605, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24008402

RESUMEN

We present electronic structure and transport calculations for hydrogen and lithium chains, using density functional theory and scattering theory on the Green's function level, to systematically study impurity effects on the transmission coefficient. To this end we address various impurity configurations. Tight-binding results allow us to interpret our the findings. We analyze under which circumstances impurities lead to level splitting and/or can be used to switch between metallic and insulating states. We also address the effects of strongly electronegative impurities.


Asunto(s)
Computadores Moleculares , Hidrógeno/química , Litio/química , Modelos Químicos , Modelos Moleculares , Simulación por Computador , Conductividad Eléctrica
17.
Artículo en Inglés | MEDLINE | ID: mdl-24241423

RESUMEN

Epstein-Barr virus (EBV) is a γ herpes virus endemic in humans and transforming human B lymphocytes. It causes a variety of human pathologies ranging from infectious mononucleosis upon acute infection to EBV-driven B-cell lymphomas. In humans, EBV-infected cells are under powerful immune surveillance by T and NK cells. If this immune surveillance is compromised as in immunosuppressed (AIDS- or posttransplantation) patients, the virus can spread from rare, EBV-containing cells and cause life-threatening pathologies. We have found that EBV immune surveillance and lymphomagenesis can be modeled in mice by targeted expression of key EBV proteins in the B-cell lineage. As EBV does not infect mouse B cells and mice have thus not coevolved with the virus, EBV exploits basic modes of the host immune response to optimize its coexistence with the host.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Vigilancia Inmunológica , Enfermedad Aguda , Animales , Linfocitos B/virología , Modelos Animales de Enfermedad , Humanos , Terapia de Inmunosupresión , Linfoma de Células B/inmunología , Ratones , Transducción de Señal , Linfocitos T/inmunología , Proteínas de la Matriz Viral/metabolismo
18.
Diabetes ; 57(8): 2074-82, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18443199

RESUMEN

OBJECTIVE: IGF-1 and the IGF-1 receptor (IGF-1R) have been implicated in the regulation of adipocyte differentiation and lipid accumulation in vitro. RESEARCH DESIGN AND METHODS: To investigate the role of IGF-1 receptor in vivo, we have inactivated the Igf-1r gene in adipose tissue (IGF-1R(aP2Cre) mice) using conditional gene targeting strategies. RESULTS: Conditional IGF-1R inactivation resulted in increased adipose tissue mass with a predominantly increased lipid accumulation in epigonadal fat pads. However, insulin-stimulated glucose uptake into adipocytes was unaffected by the deletion of the IGF-1R. Surprisingly, IGF-1R(aP2Cre) mice exhibited markedly increased somatic growth in the presence of elevated IGF-1 serum concentrations, and IGF-1 mRNA expression was significantly increased in liver and adipose tissue. IGF-1 stimulation of wild-type adipocytes significantly decreased IGF-1 mRNA expression, whereas the opposite effect was observed in IGF-1R-deficient adipocytes. CONCLUSIONS: IGF-1R signaling in adipocytes does not appear to be crucial for the development and differentiation of adipose tissue in vivo, but we identified a negative IGF-1R-mediated feedback mechanism of IGF-1 on its own gene expression in adipocytes, indicating an unexpected role for adipose tissue IGF-1 signaling in the regulation of IGF-1 serum concentrations in control of somatic growth.


Asunto(s)
Adipocitos/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptor IGF Tipo 1/fisiología , Adipocitos/citología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Peso Corporal/fisiología , Células Cultivadas , Ingestión de Alimentos/fisiología , Eliminación de Gen , Expresión Génica , Glucosa/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor IGF Tipo 1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA