Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 472(7342): 221-5, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21346760

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal human premature ageing disease, characterized by premature arteriosclerosis and degeneration of vascular smooth muscle cells (SMCs). HGPS is caused by a single point mutation in the lamin A (LMNA) gene, resulting in the generation of progerin, a truncated splicing mutant of lamin A. Accumulation of progerin leads to various ageing-associated nuclear defects including disorganization of nuclear lamina and loss of heterochromatin. Here we report the generation of induced pluripotent stem cells (iPSCs) from fibroblasts obtained from patients with HGPS. HGPS-iPSCs show absence of progerin, and more importantly, lack the nuclear envelope and epigenetic alterations normally associated with premature ageing. Upon differentiation of HGPS-iPSCs, progerin and its ageing-associated phenotypic consequences are restored. Specifically, directed differentiation of HGPS-iPSCs to SMCs leads to the appearance of premature senescence phenotypes associated with vascular ageing. Additionally, our studies identify DNA-dependent protein kinase catalytic subunit (DNAPKcs, also known as PRKDC) as a downstream target of progerin. The absence of nuclear DNAPK holoenzyme correlates with premature as well as physiological ageing. Because progerin also accumulates during physiological ageing, our results provide an in vitro iPSC-based model to study the pathogenesis of human premature and physiological vascular ageing.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Envejecimiento/metabolismo , Envejecimiento/patología , Envejecimiento/fisiología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/patología , Envejecimiento Prematuro/fisiopatología , Proteínas de Unión al Calcio/análisis , Diferenciación Celular , Línea Celular , Reprogramación Celular , Senescencia Celular , Proteína Quinasa Activada por ADN/metabolismo , Epigénesis Genética , Fibroblastos/patología , Holoenzimas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lamina Tipo A , Proteínas de Microfilamentos/análisis , Modelos Biológicos , Músculo Liso Vascular/patología , Membrana Nuclear/patología , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo , Progeria/genética , Progeria/patología , Progeria/fisiopatología , Precursores de Proteínas/análisis , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Especificidad por Sustrato , Calponinas
2.
Nature ; 471(7336): 63-7, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21368825

RESUMEN

Defined transcription factors can induce epigenetic reprogramming of adult mammalian cells into induced pluripotent stem cells. Although DNA factors are integrated during some reprogramming methods, it is unknown whether the genome remains unchanged at the single nucleotide level. Here we show that 22 human induced pluripotent stem (hiPS) cell lines reprogrammed using five different methods each contained an average of five protein-coding point mutations in the regions sampled (an estimated six protein-coding point mutations per exome). The majority of these mutations were non-synonymous, nonsense or splice variants, and were enriched in genes mutated or having causative effects in cancers. At least half of these reprogramming-associated mutations pre-existed in fibroblast progenitors at low frequencies, whereas the rest occurred during or after reprogramming. Thus, hiPS cells acquire genetic modifications in addition to epigenetic modifications. Extensive genetic screening should become a standard procedure to ensure hiPS cell safety before clinical use.


Asunto(s)
Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Mutagénesis/genética , Mutación Puntual/genética , Células Cultivadas , Análisis Mutacional de ADN , Epistasis Genética/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Persona de Mediana Edad , Modelos Genéticos , Sistemas de Lectura Abierta/genética
3.
Stem Cells ; 33(7): 2343-50, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25826782

RESUMEN

Danon disease is a familial cardiomyopathy associated with impaired autophagy due to mutations in the gene encoding lysosomal-associated membrane protein type 2 (LAMP-2). Emerging evidence has highlighted the importance of autophagy in regulating cardiomyocyte bioenergetics, function, and survival. However, the mechanisms responsible for cellular dysfunction and death in cardiomyocytes with impaired autophagic flux remain unclear. To investigate the molecular mechanisms responsible for Danon disease, we created induced pluripotent stem cells (iPSCs) from two patients with different LAMP-2 mutations. Danon iPSC-derived cardiomyocytes (iPSC-CMs) exhibited impaired autophagic flux and key features of heart failure such as increased cell size, increased expression of natriuretic peptides, and abnormal calcium handling compared to control iPSC-CMs. Additionally, Danon iPSC-CMs demonstrated excessive amounts of mitochondrial oxidative stress and apoptosis. Using the sulfhydryl antioxidant N-acetylcysteine to scavenge free radicals resulted in a significant reduction in apoptotic cell death in Danon iPSC-CMs. In summary, we have modeled Danon disease using human iPSC-CMs from patients with mutations in LAMP-2, allowing us to gain mechanistic insight into the pathogenesis of this disease. We demonstrate that LAMP-2 deficiency leads to an impairment in autophagic flux, which results in excessive oxidative stress, and subsequent cardiomyocyte apoptosis. Scavenging excessive free radicals with antioxidants may be beneficial for patients with Danon disease. In vivo studies will be necessary to validate this new treatment strategy.


Asunto(s)
Enfermedad por Depósito de Glucógeno de Tipo IIb/genética , Insuficiencia Cardíaca/genética , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/genética , Apoptosis , Autofagia , Enfermedad por Depósito de Glucógeno de Tipo IIb/patología , Insuficiencia Cardíaca/patología , Humanos , Células Madre Pluripotentes Inducidas
4.
Proc Natl Acad Sci U S A ; 109(40): 16196-201, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-22991473

RESUMEN

Generation of human induced pluripotent stem cells (hiPSCs) by the expression of specific transcription factors depends on successful epigenetic reprogramming to a pluripotent state. Although hiPSCs and human embryonic stem cells (hESCs) display a similar epigenome, recent reports demonstrated the persistence of specific epigenetic marks from the somatic cell type of origin and aberrant methylation patterns in hiPSCs. However, it remains unknown whether the use of different somatic cell sources, encompassing variable levels of selection pressure during reprogramming, influences the level of epigenetic aberrations in hiPSCs. In this work, we characterized the epigenomic integrity of 17 hiPSC lines derived from six different cell types with varied reprogramming efficiencies. We demonstrate that epigenetic aberrations are a general feature of the hiPSC state and are independent of the somatic cell source. Interestingly, we observe that the reprogramming efficiency of somatic cell lines inversely correlates with the amount of methylation change needed to acquire pluripotency. Additionally, we determine that both shared and line-specific epigenetic aberrations in hiPSCs can directly translate into changes in gene expression in both the pluripotent and differentiated states. Significantly, our analysis of different hiPSC lines from multiple cell types of origin allow us to identify a reprogramming-specific epigenetic signature comprised of nine aberrantly methylated genes that is able to segregate hESC and hiPSC lines regardless of the somatic cell source or differentiation state.


Asunto(s)
Reprogramación Celular/fisiología , Metilación de ADN/genética , Epigénesis Genética/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Línea Celular , Reprogramación Celular/genética , Islas de CpG/genética , Epigénesis Genética/genética , Epigenómica , Técnica del Anticuerpo Fluorescente , Biblioteca de Genes , Humanos , Análisis por Micromatrices , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN
5.
bioRxiv ; 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38645112

RESUMEN

Most GWAS loci are presumed to affect gene regulation, however, only ∼43% colocalize with expression quantitative trait loci (eQTLs). To address this colocalization gap, we identify eQTLs, chromatin accessibility QTLs (caQTLs), and histone acetylation QTLs (haQTLs) using molecular samples from three early developmental (EDev) tissues. Through colocalization, we annotate 586 GWAS loci for 17 traits by QTL complexity, QTL phenotype, and QTL temporal specificity. We show that GWAS loci are highly enriched for colocalization with complex QTL modules that affect multiple elements (genes and/or peaks). We also demonstrate that caQTLs and haQTLs capture regulatory variations not associated with eQTLs and explain ∼49% of the functionally annotated GWAS loci. Additionally, we show that EDev-unique QTLs are strongly depleted for colocalizing with GWAS loci. By conducting one of the largest multi-omic QTL studies to date, we demonstrate that many GWAS loci exhibit phenotypic complexity and therefore, are missed by traditional eQTL analyses.

6.
J Biol Chem ; 287(48): 40767-78, 2012 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-23019325

RESUMEN

BACKGROUND: Strategies on the basis of doxycycline-inducible lentiviruses in mouse cells allowed the examination of mechanisms governing somatic cell reprogramming. RESULTS: Using a doxycycline-inducible human reprogramming system, we identified unreported miRs enhancing reprogramming efficiency. CONCLUSION: We generated a drug-inducible human reprogramming reporter system as an invaluable tool for genetic or chemical screenings. SIGNIFICANCE: These cellular systems provide a tool to enable the advancement of reprogramming technologies in human cells. Reprogramming of somatic cells into induced pluripotent stem cells is achieved by the expression of defined transcription factors. In the last few years, reprogramming strategies on the basis of doxycycline-inducible lentiviruses in mouse cells became highly powerful for screening purposes when the expression of a GFP gene, driven by the reactivation of endogenous stem cell specific promoters, was used as a reprogramming reporter signal. However, similar reporter systems in human cells have not been generated. Here, we describe the derivation of drug-inducible human fibroblast-like cell lines that express different subsets of reprogramming factors containing a GFP gene under the expression of the endogenous OCT4 promoter. These cell lines can be used to screen functional substitutes for reprogramming factors or modifiers of reprogramming efficiency. As a proof of principle of this system, we performed a screening of a library of pluripotent-enriched microRNAs and identified hsa-miR-519a as a novel inducer of reprogramming efficiency.


Asunto(s)
Diferenciación Celular , Técnicas Citológicas/métodos , Doxiciclina/farmacología , Genes Reporteros/efectos de los fármacos , Células Madre/citología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Evaluación Preclínica de Medicamentos/métodos , Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Células Madre/metabolismo
7.
Nature ; 448(7152): 480-3, 2007 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-17581589

RESUMEN

After activation, CD4+ helper T (T(H)) cells differentiate into distinct effector subsets that are characterized by their unique cytokine expression and immunoregulatory function. During this differentiation, T(H)1 and T(H)2 cells produce interferon-gamma and interleukin (IL)-4, respectively, as autocrine factors necessary for selective lineage commitment. A distinct T(H) subset, termed T(HIL-17), T(H)17 or inflammatory T(H) (T(H)i), has been recently identified as a distinct T(H) lineage mediating tissue inflammation. T(H)17 differentiation is initiated by transforming growth factor-beta and IL-6 (refs 5-7) and reinforced by IL-23 (ref. 8), in which signal transduction and activators of transcription (STAT)3 and retinoic acid receptor-related orphan receptor (ROR)-gamma mediate the lineage specification. T(H)17 cells produce IL-17, IL-17F and IL-22, all of which regulate inflammatory responses by tissue cells but have no importance in T(H)17 differentiation. Here we show that IL-21 is another cytokine highly expressed by mouse T(H)17 cells. IL-21 is induced by IL-6 in activated T cells, a process that is dependent on STAT3 but not ROR-gamma. IL-21 potently induces T(H)17 differentiation and suppresses Foxp3 expression, which requires STAT3 and ROR-gamma, which is encoded by Rorc. IL-21 deficiency impairs the generation of T(H)17 cells and results in protection against experimental autoimmune encephalomyelitis. IL-21 is therefore an autocrine cytokine that is sufficient and necessary for T(H)17 differentiation, and serves as a target for treating inflammatory diseases.


Asunto(s)
Comunicación Autocrina , Interleucinas/inmunología , Interleucinas/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Diferenciación Celular , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Interleucinas/deficiencia , Activación de Linfocitos , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Receptores de Ácido Retinoico/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Factor de Transcripción STAT3/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología
8.
Curr Opin Hematol ; 19(4): 256-60, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22555392

RESUMEN

PURPOSE OF REVIEW: With the advent of reprogramming came the possibility of generating patient-specific clinical therapies. The purpose of this review is to discuss the recent key developments and remaining limitations in the stem cell and hematopoietic fields toward the goal of translating induced pluripotent stem cell (iPSC) technologies into the hematology clinic. RECENT FINDINGS: Recent progress in the hematopoietic and reprogramming fields has included identification of hematopoietic stem cells (HSCs) capable of long-term engraftment at the single-cell level, improvements in ex-vivo expansion of HSCs, transdifferentiation of somatic cells into hematopoietic progenitors, and the 'correction' of several disease-specific iPSCs using various gene-targeting strategies. SUMMARY: In light of recent advances, it is the hope that the hurdle of obtaining fully functional HSCs in a laboratory setting will be overcome through either in-vitro differentiation of pluripotent stem cells, ex-vivo expansion of HSCs obtained in vivo, or transdifferentiation from other somatic sources. Equally important will be for the reprogramming field to better understand the causes and consequences of the recently reported genetic/epigenetic variations present in iPSCs, especially within the context of gene-targeted strategies for correcting disease. The progress in the reprogramming and hematopoietic fields provides a strong foundation for future work toward the possible treatment of numerous hematological disorders using iPSC technologies.


Asunto(s)
Diferenciación Celular , Hematología/métodos , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes Inducidas/citología , Técnicas de Cultivo de Célula/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos
9.
Nanoscale ; 15(32): 13322-13334, 2023 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-37526009

RESUMEN

Here, rational engineering of doxorubicin prodrug loaded peptide-targeted liposomal nanoparticles to selectively target metastatic breast cancer cells in vivo is described. Glucose-regulated protein 78 (GRP78), a heat shock protein typically localized in the endoplasmic reticulum in healthy cells, has been identified to home to the cell surface in certain cancers, and thus has emerged as a promising therapeutic target. Recent reports indicated GRP78 to be expressed on the cell surface of an aggressive subpopulation of stem-like breast cancer cells that exhibit metastatic potential. In this study, a targeted nanoparticle formulation with a GRP78-binding peptide (Kd of 7.4 ± 1.0 µM) was optimized to selectively target this subpopulation. In vitro studies with breast cancer cell lines showed the targeted nanoparticle formulation (TNPGRP78pep) achieved enhanced cellular uptake, while maintaining selectivity over the control groups. In vivo, TNPGRP78pep loaded with doxorubicin prodrug was evaluated using a lung metastatic mouse model and demonstrated inhibition of breast cancer cell seeding to lungs down at the level of negative control groups. Combined, this study established that specific-targeting of surface GRP78 expressing a subpopulation of aggressive breast cancer cells was able to inhibit breast cancer metastasis to lungs, and underpinned the significance of GRP78 in breast cancer metastasis.


Asunto(s)
Neoplasias , Profármacos , Animales , Ratones , Chaperón BiP del Retículo Endoplásmico , Proteínas de la Membrana , Línea Celular Tumoral , Glucosa , Péptidos , Doxorrubicina/farmacología
10.
Blood ; 115(16): 3354-63, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20185584

RESUMEN

Neutrophil mobilization, the release of neutrophils from the bone marrow reserve into circulating blood, is important to increase peripheral neutrophil amounts during bacterial infections. Granulocyte colony-stimulating factor (G-CSF) and chemokines, such as macrophage-inflammatory protein-2 (MIP-2; CXCL2), can induce neutrophil mobilization, but the mechanism(s) they use remain unclear. Signal transducers and activator of transcription 3 (STAT3) is the principal intracellular signaling molecule activated upon G-CSF ligation of its receptor. Using a murine model with conditional STAT3 deletion in bone marrow, we demonstrated previously that STAT3 regulates acute G-CSF-responsive neutrophil mobilization and MIP-2-dependent neutrophil chemotaxis. In this study, we show STAT3 is also necessary for MIP-2-elicited neutrophil mobilization. STAT3 appears to function by controlling extracellular signal-regulated kinase (ERK) activation, which is important for MIP-2-mediated chemotaxis. In addition, we demonstrate that G-CSF stimulates the expression of the MIP-2 receptor via STAT3-dependent transcriptional activation of Il8rb. G-CSF treatment also induces STAT3-dependent changes in bone marrow chemokine expression levels which may further affect neutrophil retention and release. Taken together, our study demonstrates that STAT3 regulates multiple aspects of chemokine and chemokine receptor expression and function within the bone marrow, indicating a central role in the neutrophil mobilization response.


Asunto(s)
Quimiocina CXCL2/metabolismo , Quimiotaxis de Leucocito/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Neutrófilos/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/inmunología , Animales , Separación Celular , Quimiocina CXCL2/inmunología , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Expresión Génica , Regulación de la Expresión Génica/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Transgénicos , Neutrófilos/inmunología , Receptores de Interleucina-8 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/inmunología
11.
Blood ; 116(14): 2462-71, 2010 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-20581311

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) mediates "emergency" granulopoiesis during infection, a process that is mimicked by clinical G-CSF use, yet we understand little about the intracellular signaling cascades that control demand-driven neutrophil production. Using a murine model with conditional deletion of signal transducer and activator of transcription 3 (STAT3) in bone marrow, we investigated the cellular and molecular mechanisms of STAT3 function in the emergency granulopoiesis response to G-CSF administration or infection with Listeria monocytogenes, a pathogen that is restrained by G-CSF signaling in vivo. Our results show that STAT3 deficiency renders hematopoietic progenitor cells and myeloid precursors refractory to the growth-promoting functions of G-CSF or L monocytogenes infection. STAT3 is necessary for accelerating granulocyte cell-cycle progression and maturation in response to G-CSF. STAT3 directly controls G-CSF-dependent expression of CCAAT-enhancer-binding protein ß (C/EBPß), a crucial factor in the emergency granulopoiesis response. Moreover, STAT3 and C/EBPß coregulate c-Myc through interactions with the c-myc promoter that control the duration of C/EBPα occupancy during demand-driven granulopoiesis. These results place STAT3 as an essential mediator of emergency granulopoiesis by its regulation of transcription factors that direct G-CSF-responsive myeloid progenitor expansion.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/metabolismo , Granulocitos/citología , Células Madre Hematopoyéticas/citología , Leucopoyesis , Factor de Transcripción STAT3/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Ciclo Celular , Regulación de la Expresión Génica , Granulocitos/metabolismo , Granulocitos/microbiología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/microbiología , Listeria monocytogenes/patogenicidad , Ratones , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal
12.
Stem Cell Res ; 62: 102799, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35550987

RESUMEN

Kabuki syndrome (KS) is a rare genetic disorder typically characterized by facial abnormalities, developmental delay, cognitive dysfunction, and organ impairment. In this report, fibroblast cells obtained from a KS patient containing a heterozygous KMT2D c.12592 C>T mutation (p.R4198X) were reprogrammed using non-integrative Sendai virus to generate three induced pluripotent stem cell (iPSC) clones. The iPSC lines retained the KS patient mutation, and displayed normal karyotypes, pluripotency marker expression, and the ability to differentiate into the three germ layers.


Asunto(s)
Enfermedades Hematológicas , Células Madre Pluripotentes Inducidas , Enfermedades Vestibulares , Anomalías Múltiples , Cara/anomalías , Enfermedades Hematológicas/genética , Humanos , Mutación/genética , Enfermedades Vestibulares/genética
13.
Oncogene ; 40(23): 4050-4059, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33981001

RESUMEN

The heat shock protein GRP78 typically resides in the endoplasmic reticulum in normal tissues, but it has been shown to be expressed on the cell surface of several cancer cells, and some stem cells, where it can act as a signaling molecule by not-yet-fully defined mechanisms. Although cell surface GRP78 (sGRP78) has emerged as an attractive chemotherapeutic target, understanding how sGRP78 is functioning in cancer has been complicated by the fact that sGRP78 can function in a cell-context dependent manner, with a diverse array of reported binding partners, to regulate a variety of cellular responses. We had previously shown that sGRP78 was important in regulating pluripotent stem cell (PSC) functions, and hypothesized that embryonic-like mechanisms of GRP78 were critical to regulating aggressive breast cancer cell functions. Here, using proteomics we identify Dermcidin (DCD) as a novel sGRP78 binding partner common to both PSCs and breast cancer cells. We show that GRP78 and DCD cooperate to regulate stem cell and cancer cell migration that is dependent on the cell surface functions of these proteins. Finally, we identify Wnt/ß-catenin signaling, a critical pathway in stem cell and cancer cell biology, as an important downstream intermediate in regulating this migration phenotype.


Asunto(s)
Neoplasias de la Mama/metabolismo , Membrana Celular/metabolismo , Chaperón BiP del Retículo Endoplásmico/metabolismo , Péptidos/metabolismo , Vía de Señalización Wnt , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Femenino , Humanos , Células Madre/metabolismo
14.
J Immunol ; 181(12): 8391-401, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19050256

RESUMEN

Th17 and regulatory T (Treg) cells play opposite roles in autoimmune diseases. However, the mechanisms underlying their proper migration to inflammatory tissues are unclear. In this study, we report that these two T cell subsets both express CCR6. CCR6 expression in Th17 cells is regulated by TGF-beta and requires two nuclear receptors, RORalpha and RORgamma. Th17 cells also express the CCR6 ligand CCL20, which is induced synergistically by TGF-beta and IL-6, which requires STAT3, RORgamma and IL-21. Th17 cells, by producing CCL20, promote migration of Th17 and Treg cells in vitro in a CCR6-dependent manner. Lack of CCR6 in Th17 cells reduces the severity of experimental autoimmune encephalomyelitis and Th17 and Treg recruitment into inflammatory tissues. Similarly, CCR6 on Treg cells is also important for their recruitment into inflammatory tissues. Our data indicate an important role of CCR6 in Treg and Th17 cell migration.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Mediadores de Inflamación/fisiología , Interleucina-17/fisiología , Receptores CCR6/fisiología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Secuencia de Aminoácidos , Animales , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Células Cultivadas , Quimiotaxis de Leucocito/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores CCR6/biosíntesis , Receptores CCR6/deficiencia , Receptores CCR6/genética , Linfocitos T Reguladores/patología
15.
Stem Cell Res ; 49: 102096, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33370871

RESUMEN

Cancer-derived iPSCs have provided valuable insight into oncogenesis, but human cancer cells can often be difficult to reprogram, especially in cases of complex genetic abnormalities. Here we report, to our knowledge, the first successful generation of an iPSC line from a human immortalized acute myeloid leukemia (AML) cell line, the cell line HL-60. This iPSC line retains a majority of the leukemic genotype and displays defects in myeloid differentiation, thus providing a tool for modeling and studying AML.


Asunto(s)
Células Madre Pluripotentes Inducidas , Leucemia Mieloide Aguda , Diferenciación Celular , Células HL-60 , Hematopoyesis , Humanos , Leucemia Mieloide Aguda/genética
16.
Sci Rep ; 10(1): 3474, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-32103065

RESUMEN

Reliable approaches to identify stem cell mechanisms that mediate aggressive cancer could have great therapeutic value, based on the growing evidence of embryonic signatures in metastatic cancers. However, how to best identify and target stem-like mechanisms aberrantly acquired by cancer cells has been challenging. We harnessed the power of reprogramming to examine GRP78, a chaperone protein generally restricted to the endoplasmic reticulum in normal tissues, but which is expressed on the cell surface of human embryonic stem cells and many cancer types. We have discovered that (1) cell surface GRP78 (sGRP78) is expressed on iPSCs and is important in reprogramming, (2) sGRP78 promotes cellular functions in both pluripotent and breast cancer cells (3) overexpression of GRP78 in breast cancer cells leads to an induction of a CD24-/CD44+ tumor initiating cell (TIC) population (4) sGRP78+ breast cancer cells are enriched for stemness genes and appear to be a subset of TICs (5) sGRP78+ breast cancer cells show an enhanced ability to seed metastatic organ sites in vivo. These collective findings show that GRP78 has important functions in regulating both pluripotency and oncogenesis, and suggest that sGRP78 marks a stem-like population in breast cancer cells that has increased metastatic potential in vivo.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Proteínas de Choque Térmico/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transformación Celular Neoplásica , Reprogramación Celular , Chaperón BiP del Retículo Endoplásmico , Femenino , Células HEK293 , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Noqueados , Células Madre Neoplásicas/citología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Trasplante Heterólogo
17.
Stem Cell Res ; 41: 101587, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31739201

RESUMEN

Using iPSCs to study cancer has been complicated by the fact that many cancer cells are difficult to reprogram, which has been attributed to the genomic abnormalities present. Acute Myeloid Leukemia (AML) is a complex disease that presents with various types of genomic aberrations that affect prognosis. Here we reprogrammed CD34+ cells from an AML patient containing a rare der(7)t(7;13) translocation associated with poor prognosis, who had relapsed and was refractory to current treatments. The generated AML-iPSCs displayed normal karyotypes and myeloid differentiation potential. These findings have implications for modeling and treating AML disease.


Asunto(s)
Médula Ósea/patología , Diferenciación Celular , Resistencia a Antineoplásicos , Células Madre Pluripotentes Inducidas/patología , Leucemia Mieloide Aguda/patología , Células Mieloides/patología , Recurrencia Local de Neoplasia/patología , Anciano , Humanos , Cariotipo , Masculino , Células Tumorales Cultivadas
18.
Cytokine ; 42(3): 277-88, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18400509

RESUMEN

Neutrophils are phagocytes whose principal function is to maintain anti-bacterial immunity. Neutrophils ingest and kill invading bacteria, releasing cytotoxic, chemotactic and inflammatory mediators at sites of infection. This serves to control the immediate host immune response and attract other cells, such as macrophages and dendritic cells, which are important for establishing long-term adaptive immunity. Neutrophils thus contribute to both the initiation and the maintenance of inflammation at sites of infection. Aberrant neutrophil activity is deleterious; suppressed responses can cause extreme susceptibility to infection while overactivation can lead to excessive inflammation and tissue damage. This review will focus on neutrophil regulation by granulocyte colony-stimulating factor (G-CSF), the principal cytokine controlling neutrophil development and function. The review will emphasize the molecular aspects of G-CSF-driven granulopoiesis in steady state (healthy) conditions and during demand-driven or 'emergency' conditions elicited by infection or clinical administration of G-CSF. Understanding the molecular control of granulopoiesis will aid in the development of new approaches designed to treat disorders of neutrophil production and function.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/fisiología , Mielopoyesis/fisiología , Activación Neutrófila/fisiología , Animales , Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/fisiopatología , Proliferación Celular , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Interleucina-17/fisiología , Ratones , Neutropenia/etiología , Neutropenia/inmunología , Neutropenia/fisiopatología , Receptores de Factor Estimulante de Colonias de Granulocito/fisiología , Transducción de Señal
19.
Genome Biol ; 18(1): 135, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28728561

RESUMEN

Three recent studies analyzing large-scale collections of human induced pluripotent stem cell lines provide valuable insight into how genetic regulatory variation affects cellular and molecular traits.


Asunto(s)
Enfermedad , Variación Genética , Células Madre Pluripotentes Inducidas , Modelos Genéticos , Diferenciación Celular , Humanos
20.
Stem Cell Reports ; 8(4): 1101-1111, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28410643

RESUMEN

Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) offers the possibility of studying the molecular mechanisms underlying human diseases in cell types difficult to extract from living patients, such as neurons and cardiomyocytes. To date, studies have been published that use small panels of iPSC-derived cell lines to study monogenic diseases. However, to study complex diseases, where the genetic variation underlying the disorder is unknown, a sizable number of patient-specific iPSC lines and controls need to be generated. Currently the methods for deriving and characterizing iPSCs are time consuming, expensive, and, in some cases, descriptive but not quantitative. Here we set out to develop a set of simple methods that reduce cost and increase throughput in the characterization of iPSC lines. Specifically, we outline methods for high-throughput quantification of surface markers, gene expression analysis of in vitro differentiation potential, and evaluation of karyotype with markedly reduced cost.


Asunto(s)
Variación Genética , Ensayos Analíticos de Alto Rendimiento/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipificación/métodos , Miocitos Cardíacos/metabolismo , Neuronas/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Reprogramación Celular/genética , Análisis Costo-Beneficio , Genotipo , Ensayos Analíticos de Alto Rendimiento/economía , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Células Madre Pluripotentes Inducidas/citología , Cariotipificación/economía , Miocitos Cardíacos/citología , Neuronas/citología , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA