Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 24(11): 103326, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34805788

RESUMEN

Langerhans cells (LC) are skin-resident antigen-presenting cells that regulate immune responses to epithelial microorganisms. Human papillomavirus (HPV) infection can promote malignant epithelial transformation. As LCs are considered important for controlling HPV infection, we compared the transcriptome of murine LCs from skin transformed by K14E7 oncoprotein and from healthy skin. We identified transcriptome heterogeneity at the single cell level amongst LCs in normal skin, associated with ontogeny, cell cycle, and maturation. We identified a balanced co-existence of immune-stimulatory and immune-inhibitory LC cell states in normal skin that was significantly disturbed in HPV16 E7-transformed skin. Hyperplastic skin was depleted of immune-stimulatory LCs and enriched for LCs with an immune-inhibitory gene signature, and LC-keratinocyte crosstalk was dysregulated. We identified reduced expression of interleukin (IL)-34, a critical molecule for LC homeostasis. Enrichment of an immune-inhibitory LC gene signature and reduced levels of epithelial IL-34 were also found in human HPV-associated cervical epithelial cancers.

2.
Blood ; 136(8): 957-973, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32369597

RESUMEN

Modulators of epithelial-to-mesenchymal transition (EMT) have recently emerged as novel players in the field of leukemia biology. The mechanisms by which EMT modulators contribute to leukemia pathogenesis, however, remain to be elucidated. Here we show that overexpression of SNAI1, a key modulator of EMT, is a pathologically relevant event in human acute myeloid leukemia (AML) that contributes to impaired differentiation, enhanced self-renewal, and proliferation of immature myeloid cells. We demonstrate that ectopic expression of Snai1 in hematopoietic cells predisposes mice to AML development. This effect is mediated by interaction with the histone demethylase KDM1A/LSD1. Our data shed new light on the role of SNAI1 in leukemia development and identify a novel mechanism of LSD1 corruption in cancer. This is particularly pertinent given the current interest surrounding the use of LSD1 inhibitors in the treatment of multiple different malignancies, including AML.


Asunto(s)
Transformación Celular Neoplásica , Transición Epitelial-Mesenquimal/genética , Histona Demetilasas/metabolismo , Leucemia Mieloide Aguda/patología , Factores de Transcripción de la Familia Snail/fisiología , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células HEK293 , Células HL-60 , Histona Demetilasas/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Transgénicos , Unión Proteica , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo
3.
Sci Rep ; 9(1): 7851, 2019 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-31110193

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

4.
BMC Genomics ; 20(1): 417, 2019 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-31126231

RESUMEN

BACKGROUND: Mutations in the transcription factor, KLF1, are common within certain populations of the world. Heterozygous missense mutations in KLF1 mostly lead to benign phenotypes, but a heterozygous mutation in a DNA-binding residue (E325K in human) results in severe Congenital Dyserythropoietic Anemia type IV (CDA IV); i.e. an autosomal-dominant disorder characterized by neonatal hemolysis. RESULTS: To investigate the biochemical and genetic mechanism of CDA IV, we generated murine erythroid cell lines that harbor tamoxifen-inducible (ER™) versions of wild type and mutant KLF1 on a Klf1-/- genetic background. Nuclear translocation of wild type KLF1 results in terminal erythroid differentiation, whereas mutant KLF1 results in hemolysis without differentiation. The E to K variant binds poorly to the canonical 9 bp recognition motif (NGG-GYG-KGG) genome-wide but binds at high affinity to a corrupted motif (NGG-GRG-KGG). We confirmed altered DNA-binding specificity by quantitative in vitro binding assays of recombinant zinc-finger domains. Our results are consistent with previously reported structural data of KLF-DNA interactions. We employed 4sU-RNA-seq to show that a corrupted transcriptome is a direct consequence of aberrant DNA binding. CONCLUSIONS: Since all KLF/SP family proteins bind DNA in an identical fashion, these results are likely to be generally applicable to mutations in all family members. Importantly, they explain how certain mutations in the DNA-binding domain of transcription factors can generate neomorphic functions that result in autosomal dominant disease.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Mutación Puntual , Animales , Línea Celular , ADN/química , Regulación de la Expresión Génica , Ratones , Motivos de Nucleótidos , Unión Proteica , Transcripción Genética
5.
Sci Rep ; 8(1): 12457, 2018 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-30127368

RESUMEN

A direct interaction between the erythropoietin (EPOR) and the beta-common (ßc) receptors to form an Innate Repair Receptor (IRR) is controversial. On one hand, studies have shown a functional link between EPOR and ßc receptor in tissue protection while others have shown no involvement of the ßc receptor in tissue repair. To date there is no biophysical evidence to confirm a direct association of the two receptors either in vitro or in vivo. We investigated the existence of an interaction between the extracellular regions of EPOR and the ßc receptor in silico and in vitro (either in the presence or absence of EPO or EPO-derived peptide ARA290). Although a possible interaction between EPOR and ßc was suggested by our computational and genomic studies, our in vitro biophysical analysis demonstrates that the extracellular regions of the two receptors do not specifically associate. We also explored the involvement of the ßc receptor gene (Csf2rb) under anaemic stress conditions and found no requirement for the ßc receptor in mice. In light of these studies, we conclude that the extracellular regions of the EPOR and the ßc receptor do not directly interact and that the IRR is not involved in anaemic stress.

6.
Sci Rep ; 8(1): 12793, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-30143664

RESUMEN

Anemic Nan mice carry a mutation (E339D) in the second zinc finger of erythroid transcription factor KLF1. Nan-KLF1 fails to bind a subset of normal KLF1 targets and ectopically binds a large set of genes not normally engaged by KLF1, resulting in a corrupted fetal liver transcriptome. Here, we performed RNAseq using flow cytometric-sorted spleen erythroid precursors from adult Nan and WT littermates rendered anemic by phlebotomy to identify global transcriptome changes specific to the Nan Klf1 mutation as opposed to anemia generally. Mutant Nan-KLF1 leads to extensive and progressive transcriptome corruption in adult spleen erythroid precursors such that stress erythropoiesis is severely compromised. Terminal erythroid differentiation is defective in the bone marrow as well. Principle component analysis reveals two major patterns of differential gene expression predicting that defects in basic cellular processes including translation, cell cycle, and DNA repair could contribute to disordered erythropoiesis and anemia in Nan. Significant erythroid precursor stage specific changes were identified in some of these processes in Nan. Remarkably, however, despite expression changes in large numbers of associated genes, most basic cellular processes were intact in Nan indicating that developing red cells display significant physiological resiliency and establish new homeostatic set points in vivo.


Asunto(s)
Envejecimiento/patología , Anemia/genética , Anemia/patología , Diferenciación Celular/genética , Eritropoyesis/genética , Factores de Transcripción de Tipo Kruppel/genética , Mutación/genética , Transcriptoma/genética , Animales , Secuencia de Bases , Ciclo Celular/genética , Daño del ADN , Células Eritroides/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Ontología de Genes , Factores de Transcripción de Tipo Kruppel/metabolismo , Hígado/embriología , Hígado/metabolismo , Ratones , Ratones Mutantes , Mitofagia/genética , Anotación de Secuencia Molecular , Análisis de Componente Principal , Especies Reactivas de Oxígeno/metabolismo , Bazo/embriología , Bazo/metabolismo
8.
PLoS One ; 12(7): e0180922, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28732065

RESUMEN

Erythropoietin (EPO) acts through the dimeric erythropoietin receptor to stimulate proliferation, survival, differentiation and enucleation of erythroid progenitor cells. We undertook two complimentary approaches to find EPO-dependent pSTAT5 target genes in murine erythroid cells: RNA-seq of newly transcribed (4sU-labelled) RNA, and ChIP-seq for pSTAT5 30 minutes after EPO stimulation. We found 302 pSTAT5-occupied sites: ~15% of these reside in promoters while the rest reside within intronic enhancers or intergenic regions, some >100kb from the nearest TSS. The majority of pSTAT5 peaks contain a central palindromic GAS element, TTCYXRGAA. There was significant enrichment for GATA motifs and CACCC-box motifs within the neighbourhood of pSTAT5-bound peaks, and GATA1 and/or KLF1 co-occupancy at many sites. Using 4sU-RNA-seq we determined the EPO-induced transcriptome and validated differentially expressed genes using dynamic CAGE data and qRT-PCR. We identified known direct pSTAT5 target genes such as Bcl2l1, Pim1 and Cish, and many new targets likely to be involved in driving erythroid cell differentiation including those involved in mRNA splicing (Rbm25), epigenetic regulation (Suv420h2), and EpoR turnover (Clint1/EpsinR). Some of these new EpoR-JAK2-pSTAT5 target genes could be used as biomarkers for monitoring disease activity in polycythaemia vera, and for monitoring responses to JAK inhibitors.


Asunto(s)
Eritropoyesis/fisiología , Eritropoyetina/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Eritropoyesis/genética , Eritropoyetina/genética , Retroalimentación Fisiológica , Ratones , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT5/genética , Transducción de Señal , Transcriptoma
9.
Nucleic Acids Res ; 45(11): 6572-6588, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28541545

RESUMEN

Krüppel-like factors (KLFs) are a family of 17 transcription factors characterized by a conserved DNA-binding domain of three zinc fingers and a variable N-terminal domain responsible for recruiting cofactors. KLFs have diverse functions in stem cell biology, embryo patterning, and tissue homoeostasis. KLF1 and related family members function as transcriptional activators via recruitment of co-activators such as EP300, whereas KLF3 and related members act as transcriptional repressors via recruitment of C-terminal Binding Proteins. KLF1 directly activates the Klf3 gene via an erythroid-specific promoter. Herein, we show KLF1 and KLF3 bind common as well as unique sites within the erythroid cell genome by ChIP-seq. We show KLF3 can displace KLF1 from key erythroid gene promoters and enhancers in vivo. Using 4sU RNA labelling and RNA-seq, we show this competition results in reciprocal transcriptional outputs for >50 important genes. Furthermore, Klf3-/- mice displayed exaggerated recovery from anemic stress and persistent cell cycling consistent with a role for KLF3 in dampening KLF1-driven proliferation. We suggest this study provides a paradigm for how KLFs work in incoherent feed-forward loops or networks to fine-tune transcription and thereby control diverse biological processes such as cell proliferation.


Asunto(s)
Elementos de Facilitación Genéticos , Factores de Transcripción de Tipo Kruppel/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , Animales , Línea Celular , Técnicas de Cocultivo , Células Eritroides/metabolismo , Eritropoyesis , Ratones , Transcripción Genética
10.
Development ; 144(3): 430-440, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28143845

RESUMEN

Transcription factor control of cell-specific downstream targets can be significantly altered when the controlling factor is mutated. We show that the semi-dominant neonatal anemia (Nan) mutation in the EKLF/KLF1 transcription factor leads to ectopic expression of proteins that are not normally expressed in the red blood cell, leading to systemic effects that exacerbate the intrinsic anemia in the adult and alter correct development in the early embryo. Even when expressed as a heterozygote, the Nan-EKLF protein accomplishes this by direct binding and aberrant activation of genes encoding secreted factors that exert a negative effect on erythropoiesis and iron use. Our data form the basis for a novel mechanism of physiological deficiency that is relevant to human dyserythropoietic anemia and likely other disease states.


Asunto(s)
Anemia Neonatal/genética , Factores de Transcripción de Tipo Kruppel/genética , Mutación , Sustitución de Aminoácidos , Anemia Neonatal/sangre , Anemia Neonatal/embriología , Animales , Animales Recién Nacidos , Citocinas/sangre , ADN/genética , ADN/metabolismo , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Humanos , Factores de Transcripción de Tipo Kruppel/sangre , Factores de Transcripción de Tipo Kruppel/deficiencia , Ratones , Ratones Noqueados , Ratones Mutantes , Modelos Biológicos , Proteínas Musculares/sangre , Proteínas Mutantes/sangre , Proteínas Mutantes/genética
11.
Nucleic Acids Res ; 45(3): 1130-1143, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28180284

RESUMEN

The rules of engagement between zinc finger transcription factors and DNA have been partly defined by in vitro DNA-binding and structural studies, but less is known about how these rules apply in vivo. Here, we demonstrate how a missense mutation in the second zinc finger of Krüppel-like factor-1 (KLF1) leads to degenerate DNA-binding specificity in vivo, resulting in ectopic transcription and anemia in the Nan mouse model. We employed ChIP-seq and 4sU-RNA-seq to identify aberrant DNA-binding events genome wide and ectopic transcriptional consequences of this binding. We confirmed novel sequence specificity of the mutant recombinant zinc finger domain by performing biophysical measurements of in vitro DNA-binding affinity. Together, these results shed new light on the mechanisms by which missense mutations in DNA-binding domains of transcription factors can lead to autosomal dominant diseases.


Asunto(s)
ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Transcriptoma/genética , Dedos de Zinc/genética , Animales , Línea Celular , Supervivencia Celular/genética , Células Eritroides/metabolismo , Eritropoyesis/genética , Humanos , Factores de Transcripción de Tipo Kruppel/química , Ratones , Modelos Genéticos , Modelos Moleculares , Proteínas Mutantes/química , Mutación Missense , Unión Proteica
12.
Sci Rep ; 6: 26657, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27226347

RESUMEN

Thousands of sense-antisense mRNA-lncRNA gene pairs occur in the mammalian genome. While there is usually little doubt about the function of the coding transcript, the function of the lncRNA partner is mostly untested. Here we examine the function of the homeotic Evx1-Evx1as gene locus. Expression is tightly co-regulated in posterior mesoderm of mouse embryos and in embryoid bodies. Expression of both genes is enhanced by BMP4 and WNT3A, and reduced by Activin. We generated a suite of deletions in the locus by CRISPR-Cas9 editing. We show EVX1 is a critical downstream effector of BMP4 and WNT3A with respect to patterning of posterior mesoderm. The lncRNA, Evx1as arises from alternative promoters and is difficult to fully abrogate by gene editing or siRNA approaches. Nevertheless, we were able to generate a large 2.6 kb deletion encompassing the shared promoter with Evx1 and multiple additional exons of Evx1as. This led to an identical dorsal-ventral patterning defect to that generated by micro-deletion in the DNA-binding domain of EVX1. Thus, Evx1as has no function independent of EVX1, and is therefore unlikely to act in trans. We predict many antisense lncRNAs have no specific trans function, possibly only regulating the linked coding genes in cis.


Asunto(s)
Tipificación del Cuerpo/fisiología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/fisiología , Embrión de Mamíferos/embriología , Gastrulación/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/biosíntesis , ARN Largo no Codificante/biosíntesis , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Sistemas CRISPR-Cas , Edición Génica , Proteínas de Homeodominio/genética , Ratones , ARN Largo no Codificante/genética , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo
13.
Growth Factors ; 34(5-6): 210-216, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28209092

RESUMEN

Fibroblast growth factor-1 (FGF-1) promotes differentiation of human preadipocytes into mature adipocytes via modulation of a BMP and Activin Membrane-Bound Inhibitor (BAMBI)/Peroxisome proliferator-activated receptor (PPARγ)-dependent network. Here, we combined transcriptomic and functional investigations to identify novel downstream effectors aligned with complementary analyses of gene expression in human adipose tissue to explore relationships with insulin sensitivity. RNA-Seq and qRT-PCR analysis revealed significant down-regulation of carboxypeptidase A4 (CPA4) following FGF-1 treatment or induction of differentiation of human preadipocytes in a BAMBI/PPARγ-independent manner. siRNA-mediated knockdown of CPA4 resulted in enhanced differentiation of human preadipocytes. Furthermore, expression of CPA4 in subcutaneous adipose tissue correlated negatively with indices of local and systemic (liver and muscle) insulin sensitivity. These results identify CPA4 as a negative regulator of adipogenesis that is down-regulated by FGF-1 and a putative deleterious modulator of local and systemic insulin sensitivity. Further investigations are required to define the molecular mechanism(s) involved and potential therapeutic opportunities.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Carboxipeptidasas A/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Resistencia a la Insulina , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adulto , Carboxipeptidasas A/genética , Células Cultivadas , Regulación hacia Abajo , Humanos , Hígado/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Músculo Esquelético/metabolismo , PPAR gamma/metabolismo
14.
Blood ; 125(15): 2405-17, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25724378

RESUMEN

We describe a case of severe neonatal anemia with kernicterus caused by compound heterozygosity for null mutations in KLF1, each inherited from asymptomatic parents. One of the mutations is novel. This is the first described case of a KLF1-null human. The phenotype of severe nonspherocytic hemolytic anemia, jaundice, hepatosplenomegaly, and marked erythroblastosis is more severe than that present in congenital dyserythropoietic anemia type IV as a result of dominant mutations in the second zinc-finger of KLF1. There was a very high level of HbF expression into childhood (>70%), consistent with a key role for KLF1 in human hemoglobin switching. We performed RNA-seq on circulating erythroblasts and found that human KLF1 acts like mouse Klf1 to coordinate expression of many genes required to build a red cell including those encoding globins, cytoskeletal components, AHSP, heme synthesis enzymes, cell-cycle regulators, and blood group antigens. We identify novel KLF1 target genes including KIF23 and KIF11 which are required for proper cytokinesis. We also identify new roles for KLF1 in autophagy, global transcriptional control, and RNA splicing. We suggest loss of KLF1 should be considered in otherwise unexplained cases of severe neonatal NSHA or hydrops fetalis.


Asunto(s)
Anemia Neonatal/genética , Anemia Neonatal/patología , Eliminación de Gen , Hidropesía Fetal/genética , Hidropesía Fetal/patología , Factores de Transcripción de Tipo Kruppel/genética , Transcriptoma , Anemia Neonatal/sangre , Anemia Neonatal/complicaciones , Autofagia , Eritroblastos/metabolismo , Eritroblastos/patología , Eritropoyesis , Femenino , Regulación de la Expresión Génica , Humanos , Hidropesía Fetal/sangre , Recién Nacido , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino
15.
BMC Genomics ; 15: 1002, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25409780

RESUMEN

BACKGROUND: CRISPR-Cas9 is a revolutionary genome editing technique that allows for efficient and directed alterations of the eukaryotic genome. This relatively new technology has already been used in a large number of 'loss of function' experiments in cultured cells. Despite its simplicity and efficiency, screening for mutated clones remains time-consuming, laborious and/or expensive. RESULTS: Here we report a high-throughput screening strategy that allows parallel screening of up to 96 clones, using next-generation sequencing. As a proof of principle, we used CRISPR-Cas9 to disrupt the coding sequence of the homeobox gene, Evx1 in mouse embryonic stem cells. We screened 67 CRISPR-Cas9 transfected clones simultaneously by next-generation sequencing on the Ion Torrent PGM. We were able to identify both homozygous and heterozygous Evx1 mutants, as well as mixed clones, which must be identified to maintain the integrity of subsequent experiments. CONCLUSIONS: Our CRISPR-Cas9 screening strategy could be widely applied to screen for CRISPR-Cas9 mutants in a variety of contexts including the generation of mutant cell lines for in vitro research, the generation of transgenic organisms and for assessing the veracity of CRISPR-Cas9 homology directed repair. This technique is cost and time-effective, provides information on clonal heterogeneity and is adaptable for use on various sequencing platforms.


Asunto(s)
Sistemas CRISPR-Cas/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Mutación/genética , Animales , Células Clonales , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones
16.
Stem Cell Rev Rep ; 10(1): 60-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23955574

RESUMEN

We show that neural crest stem cells affect mouse hair follicle development. During embryogenesis hair follicle induction is regulated by complex reciprocal and functionally redundant signals between epidermis and dermis, which remain to be fully understood. Canonical Wnt signalling is a hallmark of neural crest cells and also a prerequisite for hair follicle induction prior to hair placode formation in the epidermis. As neural crest stem cells invade the epidermis during early embryonic development we aimed at determining whether neural crest cells affect hair follicle development. To attenuate, but not silence, canonical Wnt signalling specifically in neural crest cells, we analyzed Wnt1-cre(+/-)::Pygo2(-/-) mice in which the ß-catenin co-activator gene, Pygopus 2 (Pygo2), is deleted specifically in neural crest cells. Both, hair density and hair thickness were reduced in mutant mice. Furthermore, hair development was delayed and the relative ratio of hair types was affected. There was a decrease in zig-zag hairs and an increase in awl hairs. Mouse neural crest stem cells expressed ectodysplasin, an essential effector in the formation of zig-zag hair. Taken together, our data support the novel notion that neural crest cells are involved in the earliest stages of hair follicle development.


Asunto(s)
Eliminación de Gen , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Cresta Neural/citología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Folículo Piloso/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Cresta Neural/metabolismo
17.
Stem Cell Rev Rep ; 7(4): 799-814, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21455606

RESUMEN

Here we describe the isolation, characterisation and ex-vivo expansion of human epidermal neural crest stem cells (hEPI-NCSC) and we provide protocols for their directed differentiation into osteocytes and melanocytes. hEPI-NCSC are neural crest-derived multipotent stem cells that persist into adulthood in the bulge of hair follicles. Multipotency and self-renewal were determined by in vitro clonal analyses. hEPI-NCSC generate all major neural crest derivatives, including bone/cartilage cells, neurons, Schwann cells, myofibroblasts and melanocytes. Furthermore, hEPI-NCSC express additional neural crest stem cell markers and global stem cell genes. To variable degrees and in a donor-dependent manner, hEPI-NCSC express the six essential pluripotency genes C-MYC, KLF4, SOX2, LIN28, OCT-4/POU5F1 and NANOG. hEPI-NCSC can be expanded ex vivo into millions of stem cells that remain mulitpotent and continue to express stem cell genes. The novelty of hEPI-NCSC lies in the combination of their highly desirable traits. hEPI-NCSC are embryonic remnants in a postnatal location, the bulge of hair follicles. Therefore they are readily accessible in the hairy skin by minimal invasive procedure. hEPI-NCSC are multipotent somatic stem cells that can be isolated reproducibly and with high yield. By taking advantage of their migratory ability, hEPI-NCSC can be isolated as a highly pure population of stem cells. hEPI-NCSC can undergo robust ex vivo expansion and directed differentiation. As somatic stem cells, hEPI-NCSC are conducive to autologous transplantation, which avoids graft rejection. Together, these traits make hEPI-NCSC novel and attractive candidates for future cell-based therapies and regenerative medicine.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Epidérmicas , Melanocitos/citología , Cresta Neural/citología , Osteocitos/citología , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Movimiento Celular , Células Cultivadas , Criopreservación , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Factor 4 Similar a Kruppel , Células de Schwann/citología
18.
Dev Dyn ; 236(2): 502-11, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17106887

RESUMEN

Crim1 is a transmembrane protein, containing six vWF-C type cysteine-rich repeats, that tethers growth factors to the cell surface. A mouse line, KST264, generated in a LacZ insertion mutagenesis gene-trap screen, was examined to elucidate Crim1 function in development. We showed that Crim1(KST264/KST264) mice were not null for Crim1 due to the production of a shortened protein isoform. These mice are likely to represent an effective hypomorph or a dominant-negative for Crim1. Transgene expression recapitulated known Crim1 expression in lens, brain, and limb, but also revealed expression in the smooth muscle cells of the developing heart and renal vasculature, developing cartilage, mature ovary and detrusor of the bladder. Transgene expression was also observed in glomerular epithelial cells, podocytes, mesangial cells, and urothelium in the kidney. Crim1(KST264/KST264) mice displayed perinatal lethality, syndactyly, eye, and kidney abnormalities. The severe and complex phenotype observed in Crim1(KST264/KST264) mice highlights the importance of Crim1 in numerous aspects of organogenesis.


Asunto(s)
Anomalías Múltiples/embriología , Receptores de Proteínas Morfogenéticas Óseas/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de la Membrana/genética , Organogénesis/genética , Fenotipo , Anomalías Múltiples/genética , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Cartilla de ADN , Técnicas Histológicas , Immunoblotting , Proteínas de la Membrana/metabolismo , Ratones , Mutagénesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transgenes/genética
19.
Oncogene ; 23(17): 3067-79, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15021918

RESUMEN

The Wilms' tumour suppressor gene, WT1, encodes a zinc-finger protein that is mutated in Wilms' tumours and other malignancies. WT1 is one of the earliest genes expressed during kidney development. WT1 proteins can activate and repress putative target genes in vitro, although the in vivo relevance of such target genes often remains unverified. To better understand the role of WT1 in tumorigenesis and kidney development, we need to identify downstream target genes. In this study, we have expression profiled human embryonic kidney 293 cells stably transfected to allow inducible WT1 expression and mouse mesonephric M15 cells transfected with a WT1 antisense construct to abolish endogenous expression of all WT1 isoforms to identify WT1-responsive genes. The complementary overlap between the two cell lines revealed a pronounced repression of genes involved in cholesterol biosynthesis by WT1. This pathway is transcriptionally regulated by the sterol responsive element-binding proteins (SREBPs). Here, we provide evidence that the C-terminal end of the WT1 protein can directly interact with SREBP, suggesting that WT1 may modify the transcriptional function of SREBPs via a direct protein-protein interaction. Therefore, the tumour suppressor activities of WT1 may be achieved by repressing the mevalonate pathway, thereby controlling cellular proliferation and promoting terminal differentiation.


Asunto(s)
Perfilación de la Expresión Génica , Proteínas WT1/genética , Animales , Sitios de Unión , Northern Blotting , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular , División Celular , Línea Celular , Clonación Molecular , ADN sin Sentido/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Riñón , Ácido Mevalónico/metabolismo , Proteínas Recombinantes/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección , Proteínas WT1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...