Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Am J Reprod Immunol ; 86(5): e13480, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34191381

RESUMEN

PROBLEM: Decidual macrophages (dMφ ) play an important role in the formation of maternal-fetal immune tolerance. However, factors that influence the immune status of dMφ and the related potential mechanisms have not been elucidated to date. METHOD OF STUDY: The gene transcription in dMφ , decidual stromal cells (DSCs), extravillous trophoblasts (EVTs), and peripheral monocytes (pMo) from human samples were measured using real-time polymerase chain reaction (PCR). Monocyte-DSC co-culture was established to explore whether DSCs influenced dMφ polarization via C-C motif ligand 2 (CCL2)-C-C chemokine receptor (CCR2) binding using flow cytometry. In vivo, changes in dMφ percentage and M1 and M2 marker expression after treatment with CCR2 or Janus kinase 2 (JAK2) inhibitor were detected with flow cytometry. Embryo resorption percentages in the above groups were also analyzed. RESULTS: We found that dMφ were an M1/M2 mixed status at the maternal-fetal interface during early pregnancy. CCL2 influenced the immune status of dMφ in an autocrine and paracrine manner. As a downstream regulator of CCR2 and triggers the Stat3 pathway, JAK2 was found to be essential for dMφ homeostasis in vivo. JAK2 inhibitor decreased the dMφ proportion and attenuated Ki67, CD36, CD86, CD206, TNF, and IL-10 expression in dMφ at E8.5 d. Moreover, CCR2-JAK2 pathway inhibition decreased the width of the placental labyrinth layer, further influencing the pregnancy outcome. CONCLUSION: The M1/M2 mixed immune status of dMφ was regulated by DSCs via CCR2, and the CCL2/CCR2/JAK2 pathway was essential for the immune status of dMφ and the outcome of early pregnancy.


Asunto(s)
Quimiocina CCL2/metabolismo , Decidua/enzimología , Histocompatibilidad Materno-Fetal , Tolerancia Inmunológica , Janus Quinasa 2/metabolismo , Macrófagos/enzimología , Receptores CCR2/metabolismo , Células del Estroma/enzimología , Adulto , Animales , Células Cultivadas , Técnicas de Cocultivo , Decidua/efectos de los fármacos , Decidua/inmunología , Pérdida del Embrión/enzimología , Pérdida del Embrión/inmunología , Femenino , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de las Cinasas Janus/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones Endogámicos C57BL , Fenotipo , Embarazo , Resultado del Embarazo , Receptores CCR2/antagonistas & inhibidores , Transducción de Señal , Células del Estroma/efectos de los fármacos , Células del Estroma/inmunología , Adulto Joven
2.
Dev Cell ; 56(8): 1182-1194.e6, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33773101

RESUMEN

Pyruvate dehydrogenase kinases (PDK1-4) inhibit the TCA cycle by phosphorylating pyruvate dehydrogenase complex (PDC). Here, we show that PDK family is dispensable for murine embryonic development and that BCKDK serves as a compensatory mechanism by inactivating PDC. First, we knocked out all four Pdk genes one by one. Surprisingly, Pdk total KO embryos developed and were born in expected ratios but died by postnatal day 4 because of hypoglycemia or ketoacidosis. Moreover, PDC was phosphorylated in these embryos, suggesting that another kinase compensates for PDK family. Bioinformatic analysis implicated branched-chain ketoacid dehydrogenase kinase (Bckdk), a key regulator of branched-chain amino acids (BCAAs) catabolism. Indeed, knockout of Bckdk and Pdk family led to the loss of PDC phosphorylation, an increase in PDC activity and pyruvate entry into the TCA cycle, and embryonic lethality. These findings reveal a regulatory crosstalk hardwiring BCAA and glucose catabolic pathways, which feed the TCA cycle.


Asunto(s)
Ciclo del Ácido Cítrico , Desarrollo Embrionario , Proteínas Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Animales , Animales Recién Nacidos , Pérdida del Embrión/enzimología , Pérdida del Embrión/patología , Eliminación de Gen , Hipoglucemia/complicaciones , Hipoglucemia/enzimología , Hipoglucemia/patología , Cetosis/complicaciones , Cetosis/enzimología , Cetosis/patología , Ratones Noqueados , Modelos Biológicos , Fosforilación , Ácido Pirúvico/metabolismo
3.
Mol Brain ; 12(1): 59, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221184

RESUMEN

Recessive mutations in the ubiquitously expressed POLR3A and POLR3B genes are the most common cause of POLR3-related hypomyelinating leukodystrophy (POLR3-HLD), a rare childhood-onset disorder characterized by deficient cerebral myelin formation and cerebellar atrophy. POLR3A and POLR3B encode the two catalytic subunits of RNA Polymerase III (Pol III), which synthesizes numerous small non-coding RNAs. We recently reported that mice homozygous for the Polr3a mutation c.2015G > A (p.Gly672Glu) have no neurological abnormalities and thus do not recapitulate the human POLR3-HLD phenotype. To determine if other POLR3-HLD mutations can cause a leukodystrophy phenotype in mouse, we characterized mice carrying the Polr3b mutation c.308G > A (p.Arg103His). Surprisingly, homozygosity for this mutation was embryonically lethal with only wild-type and heterozygous animals detected at embryonic day 9.5. Using proteomics in a human cell line, we found that the POLR3B R103H mutation severely impairs assembly of the Pol III complex. We next generated Polr3aG672E/G672E/Polr3b+/R103Hdouble mutant mice but observed that this additional mutation was insufficient to elicit a neurological or transcriptional phenotype. Taken together with our previous study on Polr3a G672E mice, our results indicate that missense mutations in Polr3a and Polr3b can variably impair mouse development and Pol III function. Developing a proper model of POLR3-HLD is crucial to gain insights into the pathophysiological mechanisms involved in this devastating neurodegenerative disease.


Asunto(s)
Pérdida del Embrión/enzimología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Mutación/genética , ARN Polimerasa III/genética , Animales , Secuencia de Bases , Pérdida del Embrión/genética , Regulación Enzimológica de la Expresión Génica , Técnicas de Sustitución del Gen , Células HEK293 , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Homocigoto , Humanos , Ratones Endogámicos C57BL , Ratones Mutantes , Actividad Motora , Vaina de Mielina/metabolismo , ARN Polimerasa III/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Oncogene ; 38(7): 998-1018, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30190546

RESUMEN

Cell cycle regulation, especially faithful DNA replication and mitosis, are crucial to maintain genome stability. Cyclin-dependent kinase (CDK)/cyclin complexes drive most processes in cellular proliferation. In response to DNA damage, cell cycle surveillance mechanisms enable normal cells to arrest and undergo repair processes. Perturbations in genomic stability can lead to tumor development and suggest that cell cycle regulators could be effective targets in anticancer therapy. However, many clinical trials ended in failure due to off-target effects of the inhibitors used. Here, we investigate in vivo the importance of WEE1- and MYT1-dependent inhibitory phosphorylation of mammalian CDK1. We generated Cdk1AF knockin mice, in which two inhibitory phosphorylation sites are replaced by the non-phosphorylatable amino acids T14A/Y15F. We uncovered that monoallelic expression of CDK1AF is early embryonic lethal in mice and induces S phase arrest accompanied by γH2AX and DNA damage checkpoint activation in mouse embryonic fibroblasts (MEFs). The chromosomal fragmentation in Cdk1AF MEFs does not rely on CDK2 and is partly caused by premature activation of MUS81-SLX4 structure-specific endonuclease complexes, as well as untimely onset of chromosome condensation followed by nuclear lamina disassembly. We provide evidence that tumor development in liver expressing CDK1AF is inhibited. Interestingly, the regulatory mechanisms that impede cell proliferation in CDK1AF expressing cells differ partially from the actions of the WEE1 inhibitor, MK-1775, with p53 expression determining the sensitivity of cells to the drug response. Thus, our work highlights the importance of improved therapeutic strategies for patients with various cancer types and may explain why some patients respond better to WEE1 inhibitors.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Pérdida del Embrión/enzimología , Embrión de Mamíferos/enzimología , Mitosis , Fase S , Sustitución de Aminoácidos , Animales , Proteína Quinasa CDC2/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Pérdida del Embrión/genética , Pérdida del Embrión/patología , Embrión de Mamíferos/patología , Activación Enzimática , Ratones , Ratones Transgénicos , Mutación Missense , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
J Cell Physiol ; 232(6): 1441-1447, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27731508

RESUMEN

Maternal infections with gram-negative bacteria are associated with miscarriage and are one of the most common complications during pregnancy. Previous studies from our group have shown that lipopolysaccharide (LPS)-activated infiltrating peripheral blood mononuclear cells (PBMC) into decidual tissue plays an important role in the establishment of a local inflammatory process that results in embryo cytotoxicity and early embryo resorption. Moreover, we have also shown that an increased endocannabinoid tone mediates LPS-induced deleterious effects during early pregnancy loss. Here, we sought to investigate whether the infiltrating PBMC modulates the decidual endocannabinoid tone and the molecular mechanisms involved. PBMC isolated from 7-day pregnant mice subjected to different treatments were co-cultured in a transwell system with decidual tissue from control 7-day pregnant mice. Decidual fatty acid amide hydrolase (FAAH) activity was measured by radioconvertion, total decidual protein nitration by Western blot (WB), and decidual FAAH nitration by immunoprecipitation followed by WB. We found that co-culture of PBMC obtained from LPS-treated mice increased the level of nitration of decidual FAAH, which resulted in a negative modulation of decidual FAAH activity. Interestingly, co-treatment with progesterone or aminoguanidine prevented this effect. We found that LPS-treated PBMC release high amounts of nitric oxide (NO) which causes tyrosine nitration of decidual FAAH, diminishing its enzymatic activity. Inactivation of FAAH, the main degrading enzyme of anandamide and similar endocannabinoids, could lead to an increased decidual endocannabinoid tone with embryotoxic effects. J. Cell. Physiol. 232: 1441-1447, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Amidohidrolasas/metabolismo , Decidua/enzimología , Regulación hacia Abajo , Pérdida del Embrión/inducido químicamente , Pérdida del Embrión/enzimología , Leucocitos Mononucleares/metabolismo , Animales , Decidua/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Pérdida del Embrión/patología , Femenino , Guanidinas/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Lipopolisacáridos/administración & dosificación , Ratones Endogámicos BALB C , Óxido Nítrico/metabolismo , Nitrosación , Progesterona/farmacología , Quercetina/farmacología
6.
BMB Rep ; 49(8): 443-8, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27345715

RESUMEN

The arginylation branch of the N-end rule pathway is a ubiquitin-mediated proteolytic system in which post-translational conjugation of Arg by ATE1-encoded Arg-tRNA-protein transferase to N-terminal Asp, Glu, or oxidized Cys residues generates essential degradation signals. Here, we characterized the ATE1-/- mice and identified the essential role of N-terminal arginylation in neural tube development. ATE1-null mice showed severe intracerebral hemorrhages and cystic space near the neural tubes. Expression of ATE1 was prominent in the developing brain and spinal cord, and this pattern overlapped with the migration path of neural stem cells. The ATE1-/- brain showed defective G-protein signaling. Finally, we observed reduced mitosis in ATE1-/- neuroepithelium and a significantly higher nitric oxide concentration in the ATE1-/- brain. Our results strongly suggest that the crucial role of ATE1 in neural tube development is directly related to proper turn-over of the RGS4 protein, which participate in the oxygen-sensing mechanism in the cells. [BMB Reports 2016; 49(8): 443-448].


Asunto(s)
Aminoaciltransferasas/metabolismo , Eliminación de Gen , Tubo Neural/anomalías , Tubo Neural/embriología , Alelos , Aminoaciltransferasas/deficiencia , Animales , Proliferación Celular , Sistema Nervioso Central/patología , Pérdida del Embrión/enzimología , Pérdida del Embrión/patología , Embrión de Mamíferos/patología , Femenino , Ratones Endogámicos C57BL , Tubo Neural/enzimología , Tubo Neural/patología , Células Neuroepiteliales/metabolismo , Células Neuroepiteliales/patología , Neuronas/patología , Embarazo , Proteolisis , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , beta-Galactosidasa/metabolismo
7.
J Biol Chem ; 291(6): 2647-63, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26677226

RESUMEN

To interpret epigenetic information, chromatin readers utilize various protein domains for recognition of DNA and histone modifications. Some readers possess multidomains for modification recognition and are thus multivalent. Bromodomain- and plant homeodomain-linked finger-containing protein 3 (BRPF3) is such a chromatin reader, containing two plant homeodomain-linked fingers, one bromodomain and a PWWP domain. However, its molecular and biological functions remain to be investigated. Here, we report that endogenous BRPF3 preferentially forms a tetrameric complex with HBO1 (also known as KAT7) and two other subunits but not with related acetyltransferases such as MOZ, MORF, TIP60, and MOF (also known as KAT6A, KAT6B, KAT5, and KAT8, respectively). We have also characterized a mutant mouse strain with a lacZ reporter inserted at the Brpf3 locus. Systematic analysis of ß-galactosidase activity revealed dynamic spatiotemporal expression of Brpf3 during mouse embryogenesis and high expression in the adult brain and testis. Brpf3 disruption, however, resulted in no obvious gross phenotypes. This is in stark contrast to Brpf1 and Brpf2, whose loss causes lethality at E9.5 and E15.5, respectively. In Brpf3-null mice and embryonic fibroblasts, RT-quantitative PCR uncovered no changes in levels of Brpf1 and Brpf2 transcripts, confirming no compensation from them. These results indicate that BRPF3 forms a functional tetrameric complex with HBO1 but is not required for mouse development and survival, thereby distinguishing BRPF3 from its paralogs, BRPF1 and BRPF2.


Asunto(s)
Embrión de Mamíferos/enzimología , Desarrollo Embrionario , Histona Acetiltransferasas/metabolismo , Complejos Multienzimáticos/metabolismo , Animales , Pérdida del Embrión/enzimología , Pérdida del Embrión/genética , Células HEK293 , Histona Acetiltransferasas/genética , Humanos , Ratones , Ratones Mutantes , Complejos Multienzimáticos/genética
8.
Development ; 141(3): 604-616, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24449838

RESUMEN

The histone deacetylases HDAC1 and HDAC2 are crucial regulators of chromatin structure and gene expression, thereby controlling important developmental processes. In the mouse brain, HDAC1 and HDAC2 exhibit different developmental stage- and lineage-specific expression patterns. To examine the individual contribution of these deacetylases during brain development, we deleted different combinations of Hdac1 and Hdac2 alleles in neural cells. Ablation of Hdac1 or Hdac2 by Nestin-Cre had no obvious consequences on brain development and architecture owing to compensation by the paralog. By contrast, combined deletion of Hdac1 and Hdac2 resulted in impaired chromatin structure, DNA damage, apoptosis and embryonic lethality. To dissect the individual roles of HDAC1 and HDAC2, we expressed single alleles of either Hdac1 or Hdac2 in the absence of the respective paralog in neural cells. The DNA-damage phenotype observed in double knockout brains was prevented by expression of a single allele of either Hdac1 or Hdac2. Strikingly, Hdac1(-/-)Hdac2(+/-) brains showed normal development and no obvious phenotype, whereas Hdac1(+/-)Hdac2(-/-) mice displayed impaired brain development and perinatal lethality. Hdac1(+/-)Hdac2(-/-) neural precursor cells showed reduced proliferation and premature differentiation mediated by overexpression of protein kinase C, delta, which is a direct target of HDAC2. Importantly, chemical inhibition or knockdown of protein kinase C delta was sufficient to rescue the phenotype of neural progenitor cells in vitro. Our data indicate that HDAC1 and HDAC2 have a common function in maintaining proper chromatin structures and show that HDAC2 has a unique role by controlling the fate of neural progenitors during normal brain development.


Asunto(s)
Alelos , Encéfalo/embriología , Encéfalo/enzimología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Homología de Secuencia de Aminoácido , Acetofenonas/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzopiranos/farmacología , Encéfalo/metabolismo , Encéfalo/patología , Proteínas Co-Represoras/metabolismo , Daño del ADN/genética , Pérdida del Embrión/enzimología , Pérdida del Embrión/patología , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
9.
Nature ; 504(7480): 460-4, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24291790

RESUMEN

Genomic imprinting is an allele-specific gene expression system that is important for mammalian development and function. The molecular basis of genomic imprinting is allele-specific DNA methylation. Although it is well known that the de novo DNA methyltransferases Dnmt3a and Dnmt3b are responsible for the establishment of genomic imprinting, how the methylation mark is erased during primordial germ cell (PGC) reprogramming remains unclear. Tet1 is one of the ten-eleven translocation family proteins, which have the capacity to oxidize 5-methylcytosine (5mC), specifically expressed in reprogramming PGCs. Here we report that Tet1 has a critical role in the erasure of genomic imprinting. We show that despite their identical genotype, progenies derived from mating between Tet1 knockout males and wild-Peg10 and Peg3, which exhibit aberrant hypermethylation in the paternal allele of differential methylated regions (DMRs). RNA-seq reveals extensive dysregulation of imprinted genes in the next generation due to paternal loss of Tet1 function. Genome-wide DNA methylation analysis of embryonic day 13.5 PGCs and sperm of Tet1 knockout mice revealed hypermethylation of DMRs of imprinted genes in sperm, which can be traced back to PGCs. Analysis of the DNA methylation dynamics in reprogramming PGCs indicates that Tet1 functions to wipe out remaining methylation, including imprinted genes, at the late reprogramming stage. Furthermore, we provide evidence supporting the role of Tet1 in the erasure of paternal imprints in the female germ line. Thus, our study establishes a critical function of Tet1 in the erasure of genomic imprinting.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Impresión Genómica , Células Germinativas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Alelos , Animales , Reprogramación Celular/genética , Cruzamientos Genéticos , Metilación de ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Dioxigenasas/deficiencia , Dioxigenasas/genética , Dioxigenasas/metabolismo , Pérdida del Embrión/enzimología , Pérdida del Embrión/genética , Embrión de Mamíferos/embriología , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/metabolismo , Femenino , Impresión Genómica/genética , Genotipo , Masculino , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Espermatozoides/metabolismo
10.
Dis Model Mech ; 6(5): 1205-12, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23744273

RESUMEN

Neural crest cells (NCCs) participate in the remodeling of the cardiac outflow tract and pharyngeal arch arteries during cardiovascular development. Integrin-linked kinase (ILK) is a serine/threonine kinase and a major regulator of integrin signaling. It links integrins to the actin cytoskeleton and recruits other adaptor molecules into a large complex to regulate actin dynamics and integrin function. Using the Cre-lox system, we deleted Ilk from NCCs of mice to investigate its role in NCC morphogenesis. The resulting mutants developed a severe aneurysmal arterial trunk that resulted in embryonic lethality during late gestation. Ilk mutants showed normal cardiac NCC migration but reduced differentiation into smooth muscle within the aortic arch arteries and the outflow tract. Within the conotruncal cushions, Ilk-deficient NCCs exhibited disorganization of F-actin stress fibers and a significantly rounder morphology, with shorter cellular projections. Additionally, absence of ILK resulted in reduced in vivo phosphorylation of Smad3 in NCCs, which correlated with reduced αSMA levels. Our findings resemble those seen in Pinch1 and ß1 integrin conditional mutant mice, and therefore support that, in neural crest-derived cells, ILK and Pinch1 act as cytoplasmic effectors of ß1 integrin in a pathway that protects against aneurysms. In addition, our conditional Ilk mutant mice might prove useful as a model to study aortic aneurysms caused by reduced Smad3 signaling, as occurs in the newly described aneurysms-osteoarthritis syndrome, for example.


Asunto(s)
Aneurisma de la Aorta/enzimología , Aneurisma de la Aorta/patología , Pérdida del Embrión/enzimología , Eliminación de Gen , Cresta Neural/enzimología , Cresta Neural/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Citoesqueleto de Actina/metabolismo , Animales , Aorta Torácica/anomalías , Aorta Torácica/embriología , Aorta Torácica/patología , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/patología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/patología , Pérdida del Embrión/patología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/patología , Integrasas/metabolismo , Ratones , Ratones Mutantes , Morfogénesis , Especificidad de Órganos , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo
11.
PLoS One ; 7(9): e44223, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22984479

RESUMEN

BACKGROUND: A plethora of biological metabolisms are regulated by the mechanisms of ubiquitination, wherein this process is balanced with the action of deubiquitination system. Dub-2 is an IL-2-inducible, immediate-early gene that encodes a deubiquitinating enzyme with growth regulatory activity. DUB-2 presumably removes ubiquitin from ubiquitin-conjugated target proteins regulating ubiquitin-mediated proteolysis, but its specific target proteins are unknown yet. METHODOLOGY/PRINCIPAL FINDINGS: To elucidate the functional role of Dub-2, we generated genetically modified mice by introducing neo cassette into the second exon of Dub-2 and then homologous recombination was done to completely abrogate the activity of DUB-2 proteins. We generated Dub-2+/- heterozygous mice showing a normal phenotype and are fertile, whereas new born mouse of Dub-2-/- homozygous alleles could not survive. In addition, Dub-2-/- embryo could not be seen between E6.5 and E12.5 stages. Furthermore, the number of embryos showing normal embryonic development for further stages is decreased in heterozygotes. Even embryonic stem cells from inner cell mass of Dub-2-/- embryos could not be established. CONCLUSIONS: Our study suggests that the targeted disruption of Dub-2 may cause embryonic lethality during early gestation, possibly due to the failure of cell proliferation during hatching process.


Asunto(s)
Pérdida del Embrión/enzimología , Pérdida del Embrión/patología , Endopeptidasas/genética , Eliminación de Gen , Marcación de Gen , Proteínas Inmediatas-Precoces/genética , Animales , Apoptosis , Masa Celular Interna del Blastocisto/metabolismo , Masa Celular Interna del Blastocisto/patología , Proliferación Celular , Supervivencia Celular , Desarrollo Embrionario , Endopeptidasas/deficiencia , Endopeptidasas/metabolismo , Fertilización In Vitro , Técnicas de Genotipaje , Heterocigoto , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/metabolismo , Tejido Linfoide/metabolismo , Tejido Linfoide/patología , Masculino , Ratones , Ratones Mutantes , Fenotipo , Motilidad Espermática , Espermatozoides/patología , Bazo/patología , Timo/patología
12.
Reproduction ; 144(4): 455-65, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22919046

RESUMEN

Female mice generating oocytes lacking complex N- and O-glycans (double mutants (DM)) produce only one small litter before undergoing premature ovarian failure (POF) by 3 months. Here we investigate the basis of the small litter by evaluating ovulation rate and embryo development in DM (Mgat1(F/F)C1galt1(F/F):ZP3Cre) and Control (Mgat1(F/F)C1galt1(F/F)) females. Surprisingly, DM ovulation rate was normal at 6 weeks, but declined dramatically by 9 weeks. In vitro development of zygotes to blastocysts was equivalent to Controls although all embryos from DM females lacked a normal zona pellucida (ZP) and ∼30% lacked a ZP entirely. In contrast, in vivo preimplantation development resulted in less embryos recovered from DM females compared with Controls at 3.5 days post coitum (dpc) (3.2±1.3 vs 7.0±0.6). Furthermore, only 45% of mated DM females contained embryos at 3.5 dpc. Of the preimplantation embryos collected from DM females, approximately half were morulae unlike Controls where the majority were blastocysts, indicating delayed embryo development in DM females. Post-implantation development in DM females was analysed to determine whether delayed preimplantation development affected subsequent development. In DM females at 5.5 dpc, only ∼40% of embryos found at 3.5 dpc had implanted. However, at 6.5 dpc, implantation sites in DM females corresponded to embryo numbers at 3.5 dpc indicating delayed implantation. At 9.5 dpc, the number of decidua corresponded to embryo numbers 6 days earlier indicating that all implanted embryos progress to midgestation. Therefore, a lack of complex N- and O-glycans in oocytes during development impairs early embryo development and viability in vivo leading to delayed implantation and a small litter.


Asunto(s)
Aciltransferasas/metabolismo , Implantación Tardía del Embrión , Pérdida del Embrión/metabolismo , Embrión de Mamíferos/metabolismo , Galactosiltransferasas/metabolismo , Polisacáridos/metabolismo , Aciltransferasas/genética , Animales , Blastocisto/enzimología , Blastocisto/metabolismo , Blastocisto/patología , Decidua/enzimología , Decidua/metabolismo , Ectogénesis , Proteínas del Huevo/genética , Proteínas del Huevo/metabolismo , Pérdida del Embrión/enzimología , Pérdida del Embrión/patología , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/patología , Femenino , Galactosiltransferasas/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Mutantes , Ratones Transgénicos , Mórula/enzimología , Mórula/metabolismo , Mórula/patología , N-Acetilglucosaminiltransferasas , Ovulación , Embarazo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Zona Pelúcida/enzimología , Zona Pelúcida/metabolismo , Glicoproteínas de la Zona Pelúcida , Cigoto/enzimología , Cigoto/metabolismo , Cigoto/patología
13.
Reproduction ; 144(4): 447-54, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22843771

RESUMEN

The initial inactivation of prostaglandins (PGs) is mediated by 15-hydroxyprostaglandin dehydrogenase (15-PGDH). PGs are potent mediators of several biological processes, including inflammation and reproduction. In uterus, PGs play a key role in infection-induced pregnancy loss, in which concentration of this mediator increased. This process is accompanied with the induction of nitric oxide synthase expression and a marked increase in uterine levels of nitric oxide. There is no information concerning nitric oxide contribution to potential changes in PG catabolism, but experimental evidence suggests that nitric oxide modulates PG pathways. The specific objectives of the study were to evaluate the protein expression of HPGD (15-PGDH) and to characterize the nitric oxide-dependent regulation of this enzyme in a model of lipopolysaccharide (LPS)-induced embryonic resorption. Results show that LPS decreased HPGD protein expression and augmented PGE synthase activity; therefore, PGE2 levels increased in uterus in this inflammatory condition. Just as LPS, the treatment with a nitric oxide donor diminished HPGD protein expression in uterine tissue. In contrast, the inhibition of nitric oxide synthesis both in control and in LPS-treated mice increased 15-PGDH levels. Also, we have found that this enzyme and PGE2 levels are not modulated by peroxynitrite, an oxidant agent derived from nitric oxide. This study suggests that LPS and nitric oxide promote a decrease in the ability of the uterus for PG catabolism during bacterially triggered pregnancy loss in mice.


Asunto(s)
Regulación hacia Abajo , Pérdida del Embrión/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Óxido Nítrico/metabolismo , Útero/metabolismo , Animales , Dinoprostona/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Pérdida del Embrión/enzimología , Pérdida del Embrión/inmunología , Inhibidores Enzimáticos/farmacología , Infecciones por Escherichia coli/enzimología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/metabolismo , Femenino , Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/metabolismo , Lipopolisacáridos , Ratones , Ratones Endogámicos BALB C , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Embarazo , Complicaciones Infecciosas del Embarazo/enzimología , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/metabolismo , Prostaglandina-E Sintasas , Distribución Aleatoria , Regulación hacia Arriba/efectos de los fármacos , Útero/efectos de los fármacos , Útero/inmunología
14.
PLoS One ; 7(5): e37070, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22606329

RESUMEN

Cullin-RING ligases (CRLs) complexes participate in the regulation of diverse cellular processes, including cell cycle progression, transcription, signal transduction and development. Serving as the scaffold protein, cullins are crucial for the assembly of ligase complexes, which recognize and target various substrates for proteosomal degradation. Mutations in human CUL4B, one of the eight members in cullin family, are one of the major causes of X-linked mental retardation. We here report the generation and characterization of Cul4b knockout mice, in which exons 3 to 5 were deleted. In contrast to the survival to adulthood of human hemizygous males with CUL4B null mutation, Cul4b null mouse embryos show severe developmental arrest and usually die before embryonic day 9.5 (E9.5). Accumulation of cyclin E, a CRL (CUL4B) substrate, was observed in Cul4b null embryos. Cul4b heterozygotes were recovered at a reduced ratio and exhibited a severe developmental delay. The placentas in Cul4b heterozygotes were disorganized and were impaired in vascularization, which may contribute to the developmental delay. As in human CUL4B heterozygotes, Cul4b null cells were selected against in Cul4b heterozygotes, leading to various degrees of skewed X-inactivation in different tissues. Together, our results showed that CUL4B is indispensable for embryonic development in the mouse.


Asunto(s)
Proteínas Cullin/fisiología , Animales , Apoptosis , Secuencia de Bases , Proliferación Celular , Proteínas Cullin/genética , ADN Complementario/genética , Pérdida del Embrión/enzimología , Pérdida del Embrión/genética , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Exones , Femenino , Hemicigoto , Heterocigoto , Humanos , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/embriología , Discapacidad Intelectual Ligada al Cromosoma X/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placenta/anomalías , Placenta/irrigación sanguínea , Placenta/enzimología , Embarazo , Eliminación de Secuencia , Especificidad de la Especie , Inactivación del Cromosoma X
15.
Cell Res ; 22(8): 1258-69, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22453236

RESUMEN

Mutations of the CUL4B ubiquitin ligase gene are causally linked to syndromic X-linked mental retardation (XLMR). However, the pathogenic role of CUL4B mutations in neuronal and developmental defects is not understood. We have generated mice with targeted disruption of Cul4b, and observed embryonic lethality with pronounced growth inhibition and increased apoptosis in extra-embryonic tissues. Cul4b, but not its paralog Cul4a, is expressed at high levels in extra-embryonic tissues post implantation. Silencing of CUL4B expression in an extra-embryonic cell line resulted in the robust accumulation of the CUL4 substrate p21(Cip1/WAF) and G2/M cell cycle arrest, which could be partially rescued by silencing of p21(Cip1/WAF). Epiblast-specific deletion of Cul4b prevented embryonic lethality and gave rise to viable Cul4b null mice. Therefore, while dispensable in the embryo proper, Cul4b performs an essential developmental role in the extra-embryonic tissues. Our study offers a strategy to generate viable Cul4b-deficient mice to model the potential neuronal and behavioral deficiencies of human CUL4B XLMR patients.


Asunto(s)
Proteínas Cullin/metabolismo , Embrión de Mamíferos/patología , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Alelos , Animales , Apoptosis , Cruzamientos Genéticos , Proteínas Cullin/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Pérdida del Embrión/enzimología , Pérdida del Embrión/genética , Embrión de Mamíferos/metabolismo , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular , Silenciador del Gen , Genotipo , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Patrón de Herencia , Puntos de Control de la Fase M del Ciclo Celular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Embarazo , Inactivación del Cromosoma X
16.
J Biol Chem ; 286(18): 15797-805, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21367862

RESUMEN

Iron-sulfur proteins play an essential role in a variety of biologic processes and exist in multiple cellular compartments. The biogenesis of these proteins has been the subject of extensive investigation, and particular focus has been placed on the pathways that assemble iron-sulfur clusters in the different cellular compartments. Iron-only hydrogenase-like protein 1 (IOP1; also known as nuclear prelamin A recognition factor like protein, or NARFL) is a human protein that is homologous to Nar1, a protein in Saccharomyces cerevisiae that, in turn, is an essential component of the cytosolic iron-sulfur protein assembly pathway in yeast. Previous siRNA-induced knockdown studies using mammalian cells point to a similar role for IOP1 in mammals. In the present studies, we pursued this further by knocking out Iop1 in Mus musculus. We find that Iop1 knock-out results in embryonic lethality before embryonic day 10.5. Acute, inducible global knock-out of Iop1 in adult mice results in lethality and significantly diminished activity of cytosolic aconitase, an iron-sulfur protein, in liver extracts. Inducible knock-out of Iop1 in mouse embryonic fibroblasts results in diminished activity of cytosolic but not mitochondrial aconitase and loss of cell viability. Therefore, just as with knock-out of Nar1 in yeast, we find that knock-out of Iop1/Narfl in mice results in lethality and defective cytosolic iron-sulfur cluster assembly. The findings demonstrate an essential role for IOP1 in this pathway.


Asunto(s)
Hidrogenasas/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Aconitato Hidratasa/genética , Aconitato Hidratasa/metabolismo , Animales , Línea Celular , Pérdida del Embrión/enzimología , Pérdida del Embrión/genética , Embrión de Mamíferos/enzimología , Fibroblastos/enzimología , Humanos , Hidrogenasas/genética , Hierro/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Hígado/enzimología , Ratones , Ratones Noqueados , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Azufre/metabolismo
17.
Nature ; 471(7338): 368-72, 2011 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-21368762

RESUMEN

Apoptosis and necroptosis are complementary pathways controlled by common signalling adaptors, kinases and proteases; among these, caspase-8 (Casp8) is critical for death receptor-induced apoptosis. This caspase has also been implicated in non-apoptotic pathways that regulate Fas-associated via death domain (FADD)-dependent signalling and other less defined biological processes as diverse as innate immune signalling and myeloid or lymphoid differentiation patterns. Casp8 suppresses RIP3-RIP1 (also known as RIPK3-RIPK1) kinase complex-dependent necroptosis that follows death receptor activation as well as a RIP3-dependent, RIP1-independent necrotic pathway that has emerged as a host defence mechanism against murine cytomegalovirus. Disruption of Casp8 expression leads to embryonic lethality in mice between embryonic days 10.5 and 11.5 (ref. 7). Thus, Casp8 may naturally hold alternative RIP3-dependent death pathways in check in addition to promoting apoptosis. We find that RIP3 is responsible for the mid-gestational death of Casp8-deficient embryos. Remarkably, Casp8(-/-)Rip3(-/-) double mutant mice are viable and mature into fertile adults with a full immune complement of myeloid and lymphoid cell types. These mice seem immunocompetent but develop lymphadenopathy by four months of age marked by accumulation of abnormal T cells in the periphery, a phenotype reminiscent of mice with Fas-deficiency (lpr/lpr; also known as Fas). Thus, Casp8 contributes to homeostatic control in the adult immune system; however, RIP3 and Casp8 are together completely dispensable for mammalian development.


Asunto(s)
Apoptosis , Caspasa 8/genética , Caspasa 8/metabolismo , Pérdida del Embrión/genética , Pérdida del Embrión/metabolismo , Eliminación de Gen , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Inhibidores de Caspasas , Línea Celular , Pérdida del Embrión/enzimología , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Femenino , Proteínas Activadoras de GTPasa/metabolismo , Inmunocompetencia/genética , Inmunocompetencia/inmunología , Enfermedades Linfáticas/genética , Enfermedades Linfáticas/inmunología , Enfermedades Linfáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
19.
J Biol Chem ; 286(15): 13404-13, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21349843

RESUMEN

Gene mutations in the phosphoinositide-metabolizing enzymes are linked to various human diseases. In mammals, PIKfyve synthesizes PtdIns(3,5)P(2) and PtdIns5P lipids that regulate endosomal trafficking and responses to extracellular stimuli. The consequence of pikfyve gene ablation in mammals is unknown. To clarify the importance of PIKfyve and PIKfyve lipid products, in this study, we have characterized the first mouse model with global deletion of the pikfyve gene using the Cre-loxP approach. We report that nearly all PIKfyve(KO/KO) mutant embryos died before the 32-64-cell stage. Cultured fibroblasts derived from PIKfyve(flox/flox) embryos and rendered pikfyve-null by Cre recombinase expression displayed severely reduced DNA synthesis, consistent with impaired cell division causing early embryo lethality. The heterozygous PIKfyve(WT/KO) mice were born at the expected Mendelian ratio and developed into adulthood. PIKfyve(WT/KO) mice were ostensibly normal by several other in vivo, ex vivo, and in vitro criteria despite the fact that their levels of the PIKfyve protein and in vitro enzymatic activity in cells and tissues were 50-55% lower than those of wild-type mice. Consistently, steady-state levels of the PIKfyve products PtdIns(3,5)P(2) and PtdIns5P selectively decreased, but this reduction (35-40%) was 10-15% less than that expected based on PIKfyve protein reduction. The nonlinear decrease of the PIKfyve protein versus PIKfyve lipid products, the potential mechanism(s) discussed herein, may explain how one functional allele in PIKfyve(WT/KO) mice is able to support the demands for PtdIns(3,5)P(2)/PtdIns5P synthesis during life. Our data also shed light on the known human disorder linked to PIKFYVE mutations.


Asunto(s)
Blastocisto/enzimología , ADN/biosíntesis , Heterocigoto , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/biosíntesis , Animales , Blastocisto/citología , Células Cultivadas , ADN/genética , Pérdida del Embrión/enzimología , Pérdida del Embrión/genética , Femenino , Fibroblastos/enzimología , Expresión Génica , Humanos , Integrasas , Errores Innatos del Metabolismo Lipídico/enzimología , Errores Innatos del Metabolismo Lipídico/genética , Masculino , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfatos de Fosfatidilinositol/genética
20.
J Pharmacol Exp Ther ; 334(1): 69-77, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20375200

RESUMEN

A mouse model termed Cpr-low (CL) was recently generated, in which the expression of the cytochrome P450 reductase (Cpr) gene was globally down-regulated. The decreased CPR expression was accompanied by phenotypical changes, including reduced embryonic survival, decreases in circulating cholesterol, increases in hepatic P450 expression, and female infertility (accompanied by elevated serum testosterone and progesterone levels). In the present study, a complementary mouse model [named reversible-CL (r-CL)] was generated, in which the reduced CPR expression can be reversed in an organ-specific fashion. The neo cassette, which was inserted into the last Cpr intron in r-CL mice, can be deleted by Cre recombinase, thus returning the structure of the Cpr gene (and hence CPR expression) to normal in Cre-expressing cells. All previously identified phenotypes of the CL mice were preserved in the r-CL mice. As a first application of the r-CL model, we have generated an extrahepatic-CL (xh-CL) mouse for testing of the functions of CPR-dependent enzymes in all extrahepatic tissues. The xh-CL mice, generated by mating of r-CL mice with albumin-Cre mice, had normal CPR expression in hepatocytes but down-regulated CPR expression elsewhere. They were indistinguishable from wild-type mice in body and liver weights, circulating cholesterol levels, and hepatic microsomal P450 expression and activities; however, they still showed elevated serum testosterone and progesterone levels and sterility in females. Embryonic lethality was prevented in males, but apparently not in females, indicating a critical role for fetal hepatic CPR-dependent enzymes in embryonic development, at least in males.


Asunto(s)
Hígado/enzimología , Modelos Animales , NADPH-Ferrihemoproteína Reductasa/genética , NADPH-Ferrihemoproteína Reductasa/fisiología , Animales , Peso Corporal , Colesterol/sangre , Pérdida del Embrión/enzimología , Pérdida del Embrión/genética , Femenino , Desarrollo Fetal/genética , Hepatocitos/enzimología , Infertilidad Femenina/enzimología , Infertilidad Femenina/genética , Hígado/embriología , Masculino , Ratones , Ratones Endogámicos , Microsomas Hepáticos/enzimología , NADPH-Ferrihemoproteína Reductasa/biosíntesis , Tamaño de los Órganos , Especificidad de Órganos , Progesterona/sangre , Caracteres Sexuales , Testosterona/sangre , Xenobióticos/metabolismo , Xenobióticos/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA