Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Ther ; 27(5): 933-946, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-30879952

RESUMEN

Chimeric antigen receptor (CAR) engineering of T cells allows one to specifically target tumor cells via cell surface antigens. A candidate target in Ewing sarcoma is the ganglioside GD2, but heterogeneic expression limits its value. Here we report that pharmacological inhibition of Enhancer of Zeste Homolog 2 (EZH2) at doses reducing H3K27 trimethylation, but not cell viability, selectively and reversibly induces GD2 surface expression in Ewing sarcoma cells. EZH2 in Ewing sarcoma cells directly binds to the promoter regions of genes encoding for two key enzymes of GD2 biosynthesis, and EZH2 inhibition enhances expression of these genes. GD2 surface expression in Ewing sarcoma cells is not associated with distinct in vitro proliferation, colony formation, chemosensitivity, or in vivo tumorigenicity. Moreover, disruption of GD2 synthesis by gene editing does not affect its in vitro behavior. EZH2 inhibitor treatment sensitizes Ewing sarcoma cells to effective cytolysis by GD2-specific CAR gene-modified T cells. In conclusion, we report a clinically applicable pharmacological approach for enhancing efficacy of adoptively transferred GD2-redirected T cells against Ewing sarcoma, by enabling recognition of tumor cells with low or negative target expression.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , Gangliósidos/genética , Receptores Quiméricos de Antígenos/genética , Sarcoma de Ewing/tratamiento farmacológico , Antígenos de Superficie/efectos de los fármacos , Antígenos de Superficie/genética , Benzamidas/farmacología , Compuestos de Bifenilo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Gangliósidos/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoterapia/métodos , Inmunoterapia Adoptiva/métodos , Indoles/farmacología , Morfolinas , Regiones Promotoras Genéticas/genética , Piridonas/farmacología , Receptores Quiméricos de Antígenos/inmunología , Sarcoma de Ewing/genética , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
2.
Stem Cells ; 31(11): 2343-53, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23922292

RESUMEN

The conversion of the nuclear program of a somatic cell from a differentiated to an undifferentiated state can be accomplished by transplanting its nucleus to an enucleated oocyte (somatic cell nuclear transfer [SCNT]) in a process termed "reprogramming." This process achieves pluripotency and occasionally also totipotency. Exploiting the obstacle of tetraploidy to full development in mammals, we show that mouse ooplasts transplanted with two somatic nuclei simultaneously (double SCNT) support preimplantation development and derivation of novel tetraploid SCNT embryonic stem cells (tNT-ESCs). Although the double SCNT embryos do not recapitulate the expression pattern of the pluripotency-associated gene Oct4 in fertilized embryos, derivative tNT-ESCs have characteristics of genuine pluripotency: in vitro they differentiate into neurons, cardiomyocytes, and endodermal cells; in vivo, tNT-ESCs form teratomas, albeit at reduced rates compared to diploid counterparts. Global transcriptome analysis revealed only few specific alterations, for example, in the quantitative expression of gastrulation-associated genes. In conclusion, we have shown that the oocyte's reprogramming capacity is in excess of a single nucleus and that double nucleus-transplanted embryos and derivative ESCs are very similar to their diploid counterparts. These results have key implications for reprogramming studies based on pluripotency: while reprogramming in the tetraploid state was known from fusion-mediated reprogramming and from fetal and adult hepatocyte-derived induced pluripotent stem cells, we have now accomplished it with enucleated oocytes.


Asunto(s)
Reprogramación Celular/fisiología , Células Madre Embrionarias/fisiología , Oocitos/fisiología , Células Madre Pluripotentes/fisiología , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Transferencia Nuclear , Oocitos/citología , Oocitos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Tetraploidía
3.
Cancers (Basel) ; 14(19)2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36230742

RESUMEN

Ewing sarcoma (EwS) is a rare and highly malignant bone tumor occurring mainly in childhood and adolescence. Physiologically, the bone is a central hub for Ca2+ homeostasis, which is severely disturbed by osteolytic processes in EwS. Therefore, we aimed to investigate how ion transport proteins involved in Ca2+ homeostasis affect EwS pathophysiology. We characterized the expression of 22 candidate genes of Ca2+-permeable or Ca2+-regulated ion channels in three EwS cell lines and found the Ca2+-activated K+ channel KCa2.1 (KCNN1) to be exceptionally highly expressed. We revealed that KCNN1 expression is directly regulated by the disease-driving oncoprotein EWSR1-FL1. Due to its consistent overexpression in EwS, KCNN1 mRNA could be a prognostic marker in EwS. In a large cohort of EwS patients, however, KCNN1 mRNA quantity does not correlate with clinical parameters. Several functional studies including patch clamp electrophysiology revealed no evidence for KCa2.1 function in EwS cells. Thus, elevated KCNN1 expression is not translated to KCa2.1 channel activity in EwS cells. However, we found that the low K+ conductance of EwS cells renders them susceptible to hypoosmotic solutions. The absence of a relevant K+ conductance in EwS thereby provides an opportunity for hypoosmotic therapy that can be exploited during tumor surgery.

4.
J Proteome Res ; 10(5): 2140-53, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21344949

RESUMEN

The oocyte is the only cell of the body that can reprogram transplanted somatic nuclei and sets the gold standard for all reprogramming methods. Therefore, an in-depth characterization of its proteome holds promise to advance our understanding of reprogramming and germ cell biology. To date, limitations on oocyte numbers and proteomic technology have impeded this task, and the search for reprogramming factors has been conducted in embryonic stem (ES) cells instead. Here, we present the proteome of metaphase II mouse oocytes to a depth of 3699 proteins, which substantially extends the number of proteins identified until now in mouse oocytes and is comparable by size to the proteome of undifferentiated mouse ES cells. Twenty-eight oocyte proteins, also detected in ES cells, match the criteria of our multilevel approach to screen for reprogramming factors, namely nuclear localization, chromatin modification, and catalytic activity. Our oocyte proteome catalog thus advances the definition of the "reprogrammome", the set of molecules--proteins, RNAs, lipids, and small molecules--that enable reprogramming.


Asunto(s)
Reprogramación Celular/genética , Metafase/genética , Oocitos/metabolismo , Proteínas/metabolismo , Proteómica/métodos , Animales , Cromatografía Liquida , Biología Computacional , Cartilla de ADN/genética , Células Madre Embrionarias/metabolismo , Femenino , Espectrometría de Masas , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Técnicas de Transferencia Nuclear , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Mech Dev ; 125(1-2): 153-66, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18054470

RESUMEN

An important question in oocyte-mediated nuclear reprogramming is whether gene expression of the donor nucleus changes randomly or follows a pattern. Since cloned embryos are very heterogeneous and arrest frequently during preimplantation development, a random scenario is generally accepted. In the present study, we resolve the heterogeneity of cumulus cell-derived mouse clones by recognizing structured subsets, and we analyze their relationship to reprogramming of donor nuclei. We utilize live cell imaging of the Oct4 promoter-driven GFP transgene to resolve the populations of cloned and ICSI-fertilized morulae, and we sort them both into three subsets based on different GFP expression. Functionally, subsets of cloned but not ICSI morulae form blastocysts and ES cells proportional to Oct4-GFP expression. Regulatively, the subsets of cloned morulae are characterized by small differences of transcript level for the pluripotency-associated genes Oct4, Nanog and Sox2. Small differences of the level of select mRNAs across subsets suggest a uniform rather than random course of reprogramming from the morula stage on. Since these small differences correspond with substantial differences in developmental competence, we propose that developmental potential of clones relates to levels of gene expression in a different way than fertilized embryos.


Asunto(s)
Núcleo Celular , Embrión de Mamíferos/citología , Animales , Secuencia de Bases , Clonación de Organismos , Cartilla de ADN , Embrión de Mamíferos/ultraestructura , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/fisiología , Inmunohistoquímica , Hibridación Fluorescente in Situ , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Reacción en Cadena de la Polimerasa
6.
J Exp Med ; 215(12): 3094-3114, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30404791

RESUMEN

The tendency of mitochondria to undergo or resist BCL2-controlled apoptosis (so-called mitochondrial priming) is a powerful predictor of response to cytotoxic chemotherapy. Fully exploiting this finding will require unraveling the molecular genetics underlying phenotypic variability in mitochondrial priming. Here, we report that mitochondrial apoptosis resistance in T cell acute lymphoblastic leukemia (T-ALL) is mediated by inactivation of polycomb repressive complex 2 (PRC2). In T-ALL clinical specimens, loss-of-function mutations of PRC2 core components (EZH2, EED, or SUZ12) were associated with mitochondrial apoptosis resistance. In T-ALL cells, PRC2 depletion induced resistance to apoptosis induction by multiple chemotherapeutics with distinct mechanisms of action. PRC2 loss induced apoptosis resistance via transcriptional up-regulation of the LIM domain transcription factor CRIP2 and downstream up-regulation of the mitochondrial chaperone TRAP1 These findings demonstrate the importance of mitochondrial apoptotic priming as a prognostic factor in T-ALL and implicate mitochondrial chaperone function as a molecular determinant of chemotherapy response.


Asunto(s)
Apoptosis , Resistencia a Antineoplásicos , Proteínas de Neoplasias/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Femenino , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas de Neoplasias/genética , Complejo Represivo Polycomb 2/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
7.
Cancer Discov ; 6(7): 700-2, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27371576

RESUMEN

Histone H3 lysine 36 dimethylation (H3K36me2), a modification associated with transcriptional activation, is required for mixed-lineage leukemia-dependent transcription and leukemic transformation. In this issue of Cancer Discovery, Zhu and colleagues map the network of readers, writers, and erasers of H3K36me2 and uncover the ASH1L histone methyltransferase as a novel target for therapeutic intervention. Cancer Discov; 6(7); 700-2. ©2016 AACR.See related article by Zhu and colleagues, p. 770.


Asunto(s)
Histonas/metabolismo , Leucemia/genética , Leucemia/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Translocación Genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Hematopoyesis/genética , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia/tratamiento farmacológico , Lisina/metabolismo , Metilación , Factores de Transcripción/metabolismo
8.
JIMD Rep ; 26: 77-84, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26303607

RESUMEN

Phosphoglucomutase 1 deficiency has recently been reported as a novel disease that belongs to two different classes of metabolic disorders, congenital disorders of glycosylation (CDG) and glycogen storage diseases.This paper focuses on previously reported siblings with short stature, hypothyroidism, increased transaminases, and, in one of them, dilated cardiomyopathy (DCM). An intronic point mutation in the PGM1-gene (c.1145-222 G>T) leads to a complex alternative splicing pattern and to almost complete absence of PGM1 activity.Exercise-induced muscle fatigue, chest pain, and rhabdomyolysis persisted into adulthood. Fainting occurred during the first minutes of strong exercise due to glucose depletion and serum heart troponin was increased. A second wind phenomenon with an improvement in exercise capacity after some minutes of training was observed. Regular aerobic training improved fitness and helped to avoid acute damage. DCM improved during therapy.Glycosylation deficiency was most prominent in childhood. Glycosylation improved with age and further improved with oral galactose supplementation even in adulthood. Optimal improvement of glycosylation-dependent phenotypes should be achieved by early and permanent galactose treatment.However, in case of mutations in ZASP, DCM can develop as a consequence of impaired binding of PGM1 to the heart-specific isoform of ZASP, independently of overall glycosylation efficiency. Thus, even if mutations in PGM1 impair the function of the ZASP-PGM1 complex, supplementation of galactose cannot be expected to restore that function. Therefore, knowledge of PGM1 deficiency in a patient should prompt surveillance of early signs of DCM and specific treatment if necessary.

9.
Methods Mol Biol ; 1222: 149-59, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25287344

RESUMEN

Nuclear transfer (NT) into mouse oocytes yields a transcriptionally and functionally heterogeneous population of cloned embryos. Most studies of NT embryos consider only embryos at predefined key stages (e.g., morula or blastocyst), that is, after the bulk of reprogramming has taken place. These retrospective approaches are of limited use to elucidate mechanisms of reprogramming and to predict developmental success. Observing cloned embryo development using live embryo cinematography has the potential to reveal otherwise undetectable embryo features. However, light exposure necessary for live cell cinematography is highly toxic to cloned embryos. Here we describe a protocol for combined bright-field and fluorescence live-cell imaging of histone H2b-GFP expressing mouse embryos, to record cell divisions up to the blastocyst stage. This protocol, which can be adapted to observe other reporters such as Oct4-GFP or Nanog-GFP, allowed us to quantitatively analyze cleavage kinetics of cloned embryos.


Asunto(s)
Blastocisto/citología , Inestabilidad Cromosómica , Microscopía Fluorescente/métodos , Imagen Molecular/métodos , Técnicas de Transferencia Nuclear , Grabación en Video/métodos , Animales , Blastocisto/fisiología , División Celular , Clonación de Organismos/métodos , Medios de Cultivo , Técnicas de Cultivo de Embriones , Femenino , Proteínas Fluorescentes Verdes/genética , Histonas/genética , Ratones , Ratones Transgénicos , Micromanipulación , Microscopía Fluorescente/instrumentación , Recuperación del Oocito/métodos
10.
PLoS One ; 7(4): e35322, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22530006

RESUMEN

Establishment of totipotency after somatic cell nuclear transfer (NT) requires not only reprogramming of gene expression, but also conversion of the cell cycle from quiescence to the precisely timed sequence of embryonic cleavage. Inadequate adaptation of the somatic nucleus to the embryonic cell cycle regime may lay the foundation for NT embryo failure and their reported lower cell counts. We combined bright field and fluorescence imaging of histone H(2b)-GFP expressing mouse embryos, to record cell divisions up to the blastocyst stage. This allowed us to quantitatively analyze cleavage kinetics of cloned embryos and revealed an extended and inconstant duration of the second and third cell cycles compared to fertilized controls generated by intracytoplasmic sperm injection (ICSI). Compared to fertilized embryos, slow and fast cleaving NT embryos presented similar rates of errors in M phase, but were considerably less tolerant to mitotic errors and underwent cleavage arrest. Although NT embryos vary substantially in their speed of cell cycle progression, transcriptome analysis did not detect systematic differences between fast and slow NT embryos. Profiling of amino acid turnover during pre-implantation development revealed that NT embryos consume lower amounts of amino acids, in particular arginine, than fertilized embryos until morula stage. An increased arginine supplementation enhanced development to blastocyst and increased embryo cell numbers. We conclude that a cell cycle delay, which is independent of pluripotency marker reactivation, and metabolic restraints reduce cell counts of NT embryos and impede their development.


Asunto(s)
Ciclo Celular/fisiología , Reprogramación Celular/fisiología , Aminoácidos/metabolismo , Animales , Arginina/metabolismo , Blastocisto/metabolismo , Análisis por Conglomerados , Daño del ADN , Transferencia de Embrión , Embrión de Mamíferos/citología , Embrión de Mamíferos/fisiología , Desarrollo Embrionario/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Cinética , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Técnicas de Transferencia Nuclear , Imagen de Lapso de Tiempo , Transgenes
11.
PLoS One ; 7(6): e36850, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22693623

RESUMEN

While reprogramming a foreign nucleus after somatic cell nuclear transfer (SCNT), the enucleated oocyte (ooplasm) must signal that biomass and cellular requirements changed compared to the nucleus donor cell. Using cells expressing nuclear-encoded but mitochondria-targeted EGFP, a strategy was developed to directly distinguish maternal and embryonic products, testing ooplasm demands on transcriptional and post-transcriptional activity during reprogramming. Specifically, we compared transcript and protein levels for EGFP and other products in pre-implantation SCNT embryos, side-by-side to fertilized controls (embryos produced from the same oocyte pool, by intracytoplasmic injection of sperm containing the EGFP transgene). We observed that while EGFP transcript abundance is not different, protein levels are significantly lower in SCNT compared to fertilized blastocysts. This was not observed for Gapdh and Actb, whose protein reflected mRNA. This transcript-protein relationship indicates that the somatic nucleus can keep up with ooplasm transcript demands, whilst transcription and translation mismatch occurs after SCNT for certain mRNAs. We further detected metabolic disturbances after SCNT, suggesting a place among forces regulating post-transcriptional changes during reprogramming. Our observations ascribe oocyte-induced reprogramming with previously unsuspected regulatory dimensions, in that presence of functional proteins may no longer be inferred from mRNA, but rather depend on post-transcriptional regulation possibly modulated through metabolism.


Asunto(s)
Reprogramación Celular/fisiología , Mitocondrias/metabolismo , Oocitos/citología , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Reprogramación Celular/genética , Microscopía por Crioelectrón , Femenino , Peróxido de Hidrógeno/metabolismo , Potencial de la Membrana Mitocondrial/genética , Potencial de la Membrana Mitocondrial/inmunología , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura , Oocitos/ultraestructura
12.
Aging Cell ; 10(1): 80-95, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20969722

RESUMEN

The mammalian oocyte has the unique feature of supporting fertilization and normal development, while capable of reprogramming nuclei of somatic cells toward pluripotency, and occasionally even totipotency. While oocyte quality is known to decay with somatic aging, it is not a given that different biological functions decay concurrently. In this study, we tested whether oocyte's reprogramming ability decreases with aging. We show that oocytes isolated from mice aged beyond the usual reproductive age (climacteric) yield ooplasts that retain reprogramming capacity after somatic nuclear transfer (SCNT), giving rise to higher blastocysts rates compared to young donors ooplasts. Despite the differences in transcriptome between climacteric and young ooplasts, gene expression profiles of SCNT blastocysts were very similar. Importantly, embryonic stem cell lines with capacity to differentiate into tissues from all germ layers were derived from SCNT blastocysts obtained from climacteric ooplasts. Although apoptosis-related genes were down-regulated in climacteric ooplasts, and reprogramming by transcription factors (direct-induced pluripotency) benefits from the inhibition of p53-mediated apoptosis, reprogramming capacity of young ooplasts was not improved by blocking p53. However, more outgrowths were derived from SCNT blastocysts developed in the presence of a p53 inhibitor, indicating a beneficial effect on trophectoderm function. Results strongly suggest that oocyte-induced reprogramming outcome is determined by the availability and balance of intrinsic pro- and anti-reprogramming factors tightly regulated and even improved throughout aging, leading to the proposal that oocytes can still be a resource for somatic reprogramming when they cease to be considered safe for sexual reproduction.


Asunto(s)
Envejecimiento/metabolismo , Blastocisto/fisiología , Reprogramación Celular/genética , Embrión de Mamíferos/metabolismo , Oocitos/fisiología , Factores de Edad , Envejecimiento/genética , Animales , Apoptosis/genética , Blastocisto/citología , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Climaterio , Transferencia de Embrión , Embrión de Mamíferos/citología , Femenino , Fertilización/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/fisiología , Genes p53/fisiología , Aptitud Genética/fisiología , Caballos , Humanos , Masculino , Ratones , Técnicas de Transferencia Nuclear , Oocitos/citología , Embarazo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Int J Dev Biol ; 54(11-12): 1649-57, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21136379

RESUMEN

Cloned mouse embryo development to blastocyst stage correlates positively with the expression level of Oct4 (Pou5f1) at the morula stage, as reported previously by our laboratory. However, whether this correlation is based on a cause-effect relationship has remained unclear. To address this question, we artificially increased the level of Oct4 prior and subsequent to somatic cell nuclear transfer, by microinjection of Oct4 mRNA into ooplasts and by transgenic Oct4 induction at the morula stage, respectively. We observed higher developmental rates of cloned embryos to blastocyst when higher levels of Oct4 were superimposed with the initial reprogramming events; whereas increasing Oct4 at later stages of preimplantation development did not have a significant effect on developmental rates. Our results show that supplemental Oct4 facilitates oocyte-mediated reprogramming only during the first cleavages, implying that the higher Oct4 level observed in developmentally competent cloned morulae is a readout of reprogramming events that successfully took place earlier.


Asunto(s)
Reprogramación Celular/genética , Embrión de Mamíferos/metabolismo , Mórula/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Animales , Blastocisto/metabolismo , Separación Celular , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mórula/metabolismo , Técnicas de Transferencia Nuclear , Factor 3 de Transcripción de Unión a Octámeros/genética , Reacción en Cadena de la Polimerasa , ARN Mensajero
14.
Dev Biol ; 308(2): 309-21, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17610862

RESUMEN

The mechanisms that have evolved to maintain genome stability during cell cycle progression are challenged when a somatic cell nucleus is placed in a meiotic environment such as the ooplasm. Chromosomal spindle aberrations ensue in the majority of reconstructed oocytes within 2 h of transplantation, but it is not known if they recover or persist with the onset of embryonic divisions. We analyzed the chromosomal spindles and the karyotype of cumulus cell-derived mouse clones through the initial and hence most critical mitoses. Cloned embryos start out with less aneuploidy than fertilized embryos but surpass them after ES cell derivation, as measured by frequencies of chromosome trisomies and structural rearrangements. Despite the limited proportion of cloned mouse embryos that reach late gestation, a phenotypic mutation lacking a karyotypic mark was found in a newborn mouse cloned in 2002 and has been inherited since by its offspring. These data concur with a prevalent epigenetic, rather than genetic, basis for cloned embryo failure, but they also warn against the temptation to think that all conditions of clones are epigenetic and recover during gametogenesis. The cloning procedure is defenseless (no matter how technically refined) towards pre-existing or induced subchromosomal mutations that are below the experimental detection limit of the cytogenetic assay.


Asunto(s)
Inestabilidad Cromosómica , Clonación de Organismos , Células Madre Embrionarias/ultraestructura , Aneuploidia , Animales , Epigénesis Genética , Femenino , Hibridación Fluorescente in Situ , Técnicas In Vitro , Cariotipificación , Masculino , Meiosis/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mitosis/genética , Modelos Genéticos , Mutación , Técnicas de Transferencia Nuclear , Fenotipo , Embarazo , Inyecciones de Esperma Intracitoplasmáticas , Huso Acromático/ultraestructura
15.
Nat Protoc ; 1(1): 125-38, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17406224

RESUMEN

Although it has now been 10 years since the first cloned mammals were generated from somatic cells using nuclear transfer (NT), the success rate for producing live offspring by cloning remains < 5%. Nevertheless, the techniques have potential as important tools for future research in basic biology. We have been able to develop a stable NT method in the mouse, in which donor nuclei are directly injected into the oocyte using a piezo-actuated micromanipulator. Although manipulation of the piezo unit is complex, once mastered it is of great help not only in NT experiments but also in almost all other forms of micromanipulation. In addition to this technique, embryonic stem (ES) cell lines established from somatic cell nuclei by NT can be generated relatively easily from a variety of mouse genotypes and cell types. Such NT-ES cells can be used not only for experimental models of human therapeutic cloning but also as a backup of the donor cell's genome. Our most recent protocols for mouse cloning, as described here, will allow the production of cloned mice in > or = 3 months.


Asunto(s)
Clonación de Organismos/métodos , Ratones/genética , Técnicas de Transferencia Nuclear , Animales , Clonación de Organismos/instrumentación , Diseño de Equipo , Micromanipulación , Técnicas de Transferencia Nuclear/instrumentación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA