Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 68(1): 89-103.e7, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28943313

RESUMEN

Genomic imprinting is an allelic gene expression phenomenon primarily controlled by allele-specific DNA methylation at the imprinting control region (ICR), but the underlying mechanism remains largely unclear. N-α-acetyltransferase 10 protein (Naa10p) catalyzes N-α-acetylation of nascent proteins, and mutation of human Naa10p is linked to severe developmental delays. Here we report that Naa10-null mice display partial embryonic lethality, growth retardation, brain disorders, and maternal effect lethality, phenotypes commonly observed in defective genomic imprinting. Genome-wide analyses further revealed global DNA hypomethylation and enriched dysregulation of imprinted genes in Naa10p-knockout embryos and embryonic stem cells. Mechanistically, Naa10p facilitates binding of DNA methyltransferase 1 (Dnmt1) to DNA substrates, including the ICRs of the imprinted allele during S phase. Moreover, the lethal Ogden syndrome-associated mutation of human Naa10p disrupts its binding to the ICR of H19 and Dnmt1 recruitment. Our study thus links Naa10p mutation-associated Ogden syndrome to defective DNA methylation and genomic imprinting.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Discapacidades del Desarrollo/genética , Epigénesis Genética , Impresión Genómica , Acetiltransferasa A N-Terminal/genética , Acetiltransferasa E N-Terminal/genética , ARN Largo no Codificante/genética , Animales , ADN/genética , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Eliminación de Gen , Genes Letales , Estudio de Asociación del Genoma Completo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/patología , Acetiltransferasa A N-Terminal/deficiencia , Acetiltransferasa E N-Terminal/deficiencia , Unión Proteica , ARN Largo no Codificante/metabolismo , Fase S/genética
2.
Circulation ; 146(25): 1950-1967, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36420731

RESUMEN

BACKGROUND: Cardiac regeneration after injury is limited by the low proliferative capacity of adult mammalian cardiomyocytes (CMs). However, certain animals readily regenerate lost myocardium through a process involving dedifferentiation, which unlocks their proliferative capacities. METHODS: We bred mice with inducible, CM-specific expression of the Yamanaka factors, enabling adult CM reprogramming and dedifferentiation in vivo. RESULTS: Two days after induction, adult CMs presented a dedifferentiated phenotype and increased proliferation in vivo. Microarray analysis revealed that upregulation of ketogenesis was central to this process. Adeno-associated virus-driven HMGCS2 overexpression induced ketogenesis in adult CMs and recapitulated CM dedifferentiation and proliferation observed during partial reprogramming. This same phenomenon was found to occur after myocardial infarction, specifically in the border zone tissue, and HMGCS2 knockout mice showed impaired cardiac function and response to injury. Finally, we showed that exogenous HMGCS2 rescues cardiac function after ischemic injury. CONCLUSIONS: Our data demonstrate the importance of HMGCS2-induced ketogenesis as a means to regulate metabolic response to CM injury, thus allowing cell dedifferentiation and proliferation as a regenerative response.


Asunto(s)
Infarto del Miocardio , Miocitos Cardíacos , Ratones , Animales , Miocitos Cardíacos/metabolismo , Corazón , Miocardio/metabolismo , Ratones Noqueados , Regeneración/genética , Proliferación Celular , Mamíferos
3.
Proc Natl Acad Sci U S A ; 117(13): 7418-7429, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32170006

RESUMEN

The striatal complex of basal ganglia comprises two functionally distinct districts. The dorsal district controls motor and cognitive functions. The ventral district regulates the limbic function of motivation, reward, and emotion. The dorsoventral parcellation of the striatum also is of clinical importance as differential striatal pathophysiologies occur in Huntington's disease, Parkinson's disease, and drug addiction disorders. Despite these striking neurobiologic contrasts, it is largely unknown how the dorsal and ventral divisions of the striatum are set up. Here, we demonstrate that interactions between the two key transcription factors Nolz-1 and Dlx1/2 control the migratory paths of striatal neurons to the dorsal or ventral striatum. Moreover, these same transcription factors control the cell identity of striatal projection neurons in both the dorsal and the ventral striata including the D1-direct and D2-indirect pathways. We show that Nolz-1, through the I12b enhancer, represses Dlx1/2, allowing normal migration of striatal neurons to dorsal and ventral locations. We demonstrate that deletion, up-regulation, and down-regulation of Nolz-1 and Dlx1/2 can produce a striatal phenotype characterized by a withered dorsal striatum and an enlarged ventral striatum and that we can rescue this phenotype by manipulating the interactions between Nolz-1 and Dlx1/2 transcription factors. Our study indicates that the two-tier system of striatal complex is built by coupling of cell-type identity and migration and suggests that the fundamental basis for divisions of the striatum known to be differentially vulnerable at maturity is already encoded by the time embryonic striatal neurons begin their migrations into developing striata.


Asunto(s)
Ganglios Basales/citología , Cuerpo Estriado/citología , Estriado Ventral/citología , Animales , Ganglios Basales/metabolismo , Diferenciación Celular , Cuerpo Estriado/metabolismo , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Interneuronas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Estriado Ventral/metabolismo
4.
Int J Mol Sci ; 24(3)2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36768628

RESUMEN

Activating transcription factor 3 (ATF3) is a stress-induced transcription factor and a familiar neuronal marker for nerve injury. This factor has been shown to protect neurons from hypoxic insult in vitro by suppressing carboxyl-terminal modulator protein (CTMP) transcription, and indirectly activating the anti-apoptotic Akt/PKB cascade. Despite prior studies in vitro, whether this neuroprotective pathway also exists in the brain in vivo after ischemic insult remains to be determined. In the present study, we showed a rapid and marked induction of ATF3 mRNA throughout ischemia-reperfusion in a middle cerebral artery (MCA) occlusion model. Although the level of CTMP mRNA was quickly induced upon ischemia, its level showed only a mild increase after reperfusion. With the gain-of-function approach, both pre- and post-ischemic administration of Ad-ATF3 ameliorated brain infarct and neurological deficits. Whereas, with the loss-of-function approach, ATF3 knockout (KO) mice showed bigger infarct and worse functional outcome after ischemia. In addition, these congenital defects were rescued upon reintroducing ATF3 to the brain of KO mice. ATF3 overexpression led to a lower level of CTMP and a higher level of p-Akt(473) in the ischemic brain. On the contrary, ATF3 KO resulted in upregulation of CTMP and downregulation of p-Akt(473) instead. Furthermore, post-ischemic CTMP siRNA knockdown led to smaller infarct and better behaviors. CTMP siRNA knockdown increased the level of p-Akt(473), but did not alter the ATF3 level in the ischemic brain, upholding the ATF3→CTMP signal cascade. In summary, our proof-of-principle experiments support the existence of neuroprotective ATF3→CTMP signal cascade regulating the ischemic brain. Furthermore, these results suggest the therapeutic potential for both ATF3 overexpression and CTMP knockdown for stroke treatment.


Asunto(s)
Isquemia Encefálica , Proteínas Proto-Oncogénicas c-akt , Ratones , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Proteínas Portadoras/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Ratones Noqueados , Infarto Encefálico/genética , ARN Interferente Pequeño/genética , Infarto Cerebral , Palmitoil-CoA Hidrolasa/metabolismo
5.
Mol Med ; 28(1): 71, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35739477

RESUMEN

BACKGROUND: Complete healing of diabetic wounds continues to be a clinically unmet need. Although robust therapies such as stem cell therapy and growth factor treatment are clinically applied, these treatments are costly for most diabetic wound patients. Therefore, a cheaper alternative is needed. Cobalt protoporphyrin (CoPP) has recently been demonstrated to promote tissue regeneration. In this study, the therapeutic benefits of CoPP in diabetic wound healing were examined. METHODS: An in vitro wound healing model that mimics re-epithelialization was established to examine the effect of CoPP on the migratory capability of human keratinocytes (HaCaT) in either normal glucose (NG) or high glucose (HG) media, as well as in the presence of either H2O2 or lipopolysaccharide (LPS). At the end of the migration assays, cells were collected and subjected to Western blotting analysis and immunostaining. RESULTS: HaCaT were found to migrate significantly more slowly in the HG media compared to the NG media. CoPP treatment was found to enhance cell migration in HG media, but was found to decrease cell migration and proliferation when HaCaT were cultured in NG media. CoPP treatment induced high levels of expression of Nrf-2/HO-1 and FoxO1 in HaCaT cultured in either glucose concentration, although the FoxO1 expression was found to be significantly higher in HaCaT that underwent the migration assay in NG media compared to those in HG media. The higher level of FoxO1 expression seen in CoPP-treated HaCaT cultured in NG media resulted in upregulation of CCL20 and downregulation of TGFß1. In contrast, HaCaT migrated in HG media were found to have high levels of expression of TGFß1, and low levels of expression of CCL20. Interestingly, in the presence of H2O2, CoPP-pretreated HaCaT cultured in either NG or HG media had similar expression level of Nrf-2/HO-1 and FoxO1 to each other. Moreover, the anti-apoptotic effect of CoPP pretreatment was noticed in HaCaT cultured in either glucose concentration. Additionally, CoPP pretreatment was shown to promote tight junction formation in HaCaT suffering from LPS-induced damage. CONCLUSIONS: CoPP enhances cell migratory capacity under hyperglycemic conditions, and protects cells from oxidative and LPS-induced cellular damage in HG media containing either H2O2 or LPS.


Asunto(s)
Peróxido de Hidrógeno , Lipopolisacáridos , Movimiento Celular , Glucosa/metabolismo , Glucosa/farmacología , Humanos , Peróxido de Hidrógeno/metabolismo , Queratinocitos , Protoporfirinas
6.
PLoS Biol ; 17(10): e3000508, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31593566

RESUMEN

CDGSH iron-sulfur domain-containing protein 2 (Cisd2) is pivotal to mitochondrial integrity and intracellular Ca2+ homeostasis. In the heart of Cisd2 knockout mice, Cisd2 deficiency causes intercalated disc defects and leads to degeneration of the mitochondria and sarcomeres, thereby impairing its electromechanical functioning. Furthermore, Cisd2 deficiency disrupts Ca2+ homeostasis via dysregulation of sarco/endoplasmic reticulum Ca2+-ATPase (Serca2a) activity, resulting in an increased level of basal cytosolic Ca2+ and mitochondrial Ca2+ overload in cardiomyocytes. Most strikingly, in Cisd2 transgenic mice, a persistently high level of Cisd2 is sufficient to delay cardiac aging and attenuate age-related structural defects and functional decline. In addition, it results in a younger cardiac transcriptome pattern during old age. Our findings indicate that Cisd2 plays an essential role in cardiac aging and in the heart's electromechanical functioning. They highlight Cisd2 as a novel drug target when developing therapies to delay cardiac aging and ameliorate age-related cardiac dysfunction.


Asunto(s)
Envejecimiento Prematuro/genética , Envejecimiento/fisiología , Bloqueo Atrioventricular/genética , Proteínas Relacionadas con la Autofagia/genética , Corazón/fisiopatología , Proteínas del Tejido Nervioso/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/fisiopatología , Animales , Bloqueo Atrioventricular/diagnóstico por imagen , Bloqueo Atrioventricular/metabolismo , Bloqueo Atrioventricular/fisiopatología , Proteínas Relacionadas con la Autofagia/deficiencia , Calcio/metabolismo , Electrocardiografía , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Corazón/fisiología , Homeostasis/fisiología , Masculino , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Proteínas del Tejido Nervioso/deficiencia , Sarcómeros/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transcriptoma
7.
Eur J Haematol ; 105(3): 326-334, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32421883

RESUMEN

BACKGROUND: In multiple myeloma (MM), impact of specific chromosomal translocations involving IgH (14q21 locus, including t(4;14), t(11;14), and t(14;16)) has been explored extensively. However, over 15% MM patients harboring IgH translocation with undefined partners have long been ignored. METHODS: A prospective non-randomized cohort study with a total of 715 newly-diagnosed MM cases was conducted, 13.6% of whom were t(14;undefined) positive. The whole cohort was divided into four groups: no IgH split (47.7%); t(14;undefined) (13.6%); t(11;14) (17.6%); and t(4;14) or t(14;16) group (21.1%). RESULTS: Median OS for the four groups was 84.2, not reached (NR), 58.7, and 44.2 months, respectively, with P values for t(14;undefined) vs no IgH split, t(11;14), and t(4;14)/t(14;16) groups of 0.197, 0.022, and 0.001, respectively. In bortezomib-based group, the survival advantage gained by t(14;undefined) group was much more significant compared to t(11;14) and t(4;14)/t(14;16) groups. Importantly, t(14;undefined) turned out to be an independent predictive factor for longer OS of MM patients in multivariate analysis, especially in the context of bortezomib treatment. Similar results were also observed in the PUMCH external validation cohort. CONCLUSION: Collectively, our data confirmed and externally validated the favorable prognosis of the t(14;undefined) groups, especially in the era of novel agents.


Asunto(s)
Cadenas Pesadas de Inmunoglobulina/genética , Mieloma Múltiple/genética , Mieloma Múltiple/mortalidad , Translocación Genética , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 14 , Cromosomas Humanos Par 16 , Cromosomas Humanos Par 4 , Femenino , Frecuencia de los Genes , Humanos , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/terapia , Pronóstico , Modelos de Riesgos Proporcionales
8.
PLoS Genet ; 13(8): e1006984, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28827800

RESUMEN

HSPB7 is a member of the small heat-shock protein (HSPB) family and is expressed in the cardiomyocytes from cardiogenesis onwards. A dramatic increase in HSPB7 is detected in the heart and blood plasma immediately after myocardial infarction. Additionally, several single-nucleotide polymorphisms of HSPB7 have been identified to be associated with heart failure caused by cardiomyopathy in human patients. Although a recent study has shown that HSPB7 is required for maintaining myofiber structure in skeletal muscle, its molecular and physiological functions in the heart remain unclear. In the present study, we generated a cardiac-specific inducible HSPB7 knockout mouse and demonstrated that the loss of HSPB7 in cardiomyocytes results in rapid heart failure and sudden death. The electrocardiogram showed cardiac arrhythmia with abnormal conduction in the HSPB7 mutant mice before death. In HSPB7 CKO cardiomyocytes, no significant defect was detected in the organization of contractile proteins in sarcomeres, but a severe structural disruption was observed in the intercalated discs. The expression of connexin 43, a gap-junction protein located at the intercalated discs, was downregulated in HSPB7 knockout cardiomyocytes. Mislocalization of desmoplakin, and N-cadherin, the intercalated disc proteins, was also observed in the HSPB7 CKO hearts. Furthermore, filamin C, the interaction protein of HSPB7, was upregulated and aggregated in HSPB7 mutant cardiomyocytes. In conclusion, our findings characterize HSPB7 as an intercalated disc protein and suggest it has an essential role in maintaining intercalated disc integrity and conduction function in the adult heart.


Asunto(s)
Cardiomiopatías/genética , Proteínas de Choque Térmico HSP27/genética , Insuficiencia Cardíaca/genética , Miocitos Cardíacos/metabolismo , Animales , Síndrome de Brugada/genética , Síndrome de Brugada/patología , Cadherinas/genética , Trastorno del Sistema de Conducción Cardíaco , Cardiomiopatías/fisiopatología , Conexina 43/genética , Modelos Animales de Enfermedad , Electrocardiografía , Sistema de Conducción Cardíaco/metabolismo , Sistema de Conducción Cardíaco/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Sarcómeros/metabolismo , Sarcómeros/patología
9.
J Cell Sci ; 129(8): 1661-70, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26929074

RESUMEN

HSPB7 belongs to the small heat-shock protein (sHSP) family, and its expression is restricted to cardiac and skeletal muscles from embryonic stages to adulthood. Here, we found that skeletal-muscle-specific ablation of the HspB7 does not affect myogenesis during embryonic stages to postnatal day 1 (P1), but causes subsequent postnatal death owing to a respiration defect, with progressive myopathy phenotypes in the diaphragm. Deficiency of HSPB7 in the diaphragm muscle resulted in muscle fibrosis, sarcomere disarray and sarcolemma integrity loss. We identified dimerized filamin C (FLNC) as an interacting partner of HSPB7. Immunofluorescence studies demonstrated that the aggregation and mislocalization of FLNC occurred in the muscle of HspB7 mutant adult mice. Furthermore, the components of dystrophin glycoprotein complex, γ- and δ-sarcoglycan, but not dystrophin, were abnormally upregulated and mislocalized in HSPB7 mutant muscle. Collectively, our findings suggest that HSPB7 is essential for maintaining muscle integrity, which is achieved through its interaction with FLNC, in order to prevent the occurrence and progression of myopathy.


Asunto(s)
Diafragma/patología , Filaminas/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Músculo Esquelético/fisiología , Enfermedades Musculares/metabolismo , Animales , Células Cultivadas , Dimerización , Fibrosis , Proteínas de Choque Térmico HSP27/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos/genética , Enfermedades Musculares/genética , Unión Proteica , Transporte de Proteínas/genética , Respiración/genética , Sarcoglicanos/metabolismo
10.
PLoS Genet ; 11(3): e1005043, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25760041

RESUMEN

Production of protein containing lengthy stretches of polyglutamine encoded by multiple repeats of the trinucleotide CAG is a hallmark of Huntington's disease (HD) and of a variety of other inherited degenerative neurological and neuromuscular disorders. Earlier work has shown that interference with production of the transcription elongation protein SUPT4H results in decreased cellular capacity to transcribe mutant huntingtin gene (Htt) alleles containing long CAG expansions, but has little effect on expression of genes containing short CAG stretches. zQ175 and R6/2 are genetically engineered mouse strains whose genomes contain human HTT alleles that include greatly expanded CAG repeats and which are used as animal models for HD. Here we show that reduction of SUPT4H expression in brains of zQ175 mice by intracerebroventricular bolus injection of antisense 2'-O-methoxyethyl oligonucleotides (ASOs) directed against Supt4h, or in R6/2 mice by deletion of one copy of the Supt4h gene, results in a decrease in mRNA and protein encoded specifically by mutant Htt alleles. We further show that reduction of SUPT4H in mouse brains is associated with decreased HTT protein aggregation, and in R6/2 mice, also with prolonged lifespan and delay of the motor impairment that normally develops in these animals. Our findings support the view that targeting of SUPT4H function may be useful as a therapeutic countermeasure against HD.


Asunto(s)
Proteínas de Unión al ADN/genética , Enfermedad de Huntington/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Expansión de Repetición de Trinucleótido/genética , Alelos , Animales , Conducta Animal , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Unión al ADN/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Ratones , Mutación , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/metabolismo , Neuronas/patología , Proteínas Nucleares/biosíntesis , Agregación Patológica de Proteínas/genética , ARN Mensajero/genética
11.
J Cell Sci ; 128(22): 4196-209, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26430214

RESUMEN

Nuclear receptor interaction protein (NRIP, also known as DCAF6 and IQWD1) is a Ca(2+)-dependent calmodulin-binding protein. In this study, we newly identify NRIP as a Z-disc protein in skeletal muscle. NRIP-knockout mice were generated and found to have reduced muscle strength, susceptibility to fatigue and impaired adaptive exercise performance. The mechanisms of NRIP-regulated muscle contraction depend on NRIP being downstream of Ca(2+) signaling, where it stimulates activation of both 'calcineurin-nuclear factor of activated T-cells, cytoplasmic 1' (CaN-NFATc1; also known as NFATC1) and calmodulin-dependent protein kinase II (CaMKII) through interaction with calmodulin (CaM), resulting in the induction of mitochondrial activity and the expression of genes encoding the slow class of myosin, and in the regulation of Ca(2+) homeostasis through the internal Ca(2+) stores of the sarcoplasmic reticulum. Moreover, NRIP-knockout mice have a delayed regenerative capacity. The amount of NRIP can be enhanced after muscle injury and is responsible for muscle regeneration, which is associated with the increased expression of myogenin, desmin and embryonic myosin heavy chain during myogenesis, as well as for myotube formation. In conclusion, NRIP is a novel Z-disc protein that is important for skeletal muscle strength and regenerative capacity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Calmodulina/metabolismo , Músculo Esquelético/fisiología , Proteínas Nucleares/metabolismo , Regeneración/fisiología , Animales , Ratones , Ratones Noqueados , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Transducción de Señal
12.
Hum Mol Genet ; 23(21): 5579-96, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24895408

RESUMEN

OVCA1/DPH1 (OVCA1) encodes a component of the diphthamide biosynthesis pathway and is located on chromosome 17p13.3. Deletions in this region are associated with Miller-Dieker syndrome (MDS). Ovca1/Dph1 (Ovca1)-null mice exhibit multiple developmental defects, including cleft palate, growth restriction and perinatal lethality, suggesting a role in the craniofacial abnormalities associated with MDS. Conditional ablation of Ovca1 in neural crest cells, but not in cranial paraxial mesoderm, also results in cleft palate and shortened lower jaw phenotypes, similar to Ovca1-null embryos. Expression of transgenic myc-tagged Ovca1 in craniofacial structures can partially rescue the cleft palate and shortened mandible of Ovca1-null embryos. Interestingly, Ovca1-null mutants are resistant to conditional expression of diphtheria toxin subunit A in both neural crest cell and paraxial mesoderm derivatives. However, OVCA1-dependent diphthamide biosynthesis is essential for neural crest cell-derived craniofacial development but that is dispensable for paraxial mesodermal-derived craniofacial structures in mammals. These findings suggest that OVCA1 deficiency in the neural crest contributes to the craniofacial abnormalities in patients with MDS. Also, our findings provide new insights into the molecular and cellular mechanisms that lead to the craniofacial defects of MDS.


Asunto(s)
Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/genética , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/patología , Estudios de Asociación Genética , Fenotipo , Proteínas Supresoras de Tumor/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Mapeo Cromosómico , Fisura del Paladar/embriología , Fisura del Paladar/genética , Fisura del Paladar/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Expresión Génica , Ligamiento Genético , Genotipo , Masculino , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor , Mutación , Cresta Neural/embriología , Cresta Neural/metabolismo , Organogénesis/genética , Proteína Wnt1/genética
13.
Stem Cells ; 33(12): 3468-80, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26303070

RESUMEN

Leukemia inhibitory factor (LIF) regulates mouse embryonic stem cell (mESC) pluripotency through STAT3 activation, but the downstream signaling remains largely unelucidated. Using cDNA microarrays, we verified B cell leukemia/lymphoma 3 (Bcl3) as the most significantly downregulated factor following LIF withdrawal in mESCs. Bcl3 knockdown altered mESC morphology, reduced expression of pluripotency genes including Oct4, Sox2, and Nanog, and downregulated DNA binding of acetylated histone 3 and RNA polymerase II on the Oct4 promoter. Conversely, Bcl3 overexpression partially prevented cell differentiation and promoted Oct4 and Nanog promoter activities. Furthermore, coimmunoprecipitation and chromatin immunoprecipitation experiments demonstrated that Bcl3 regulation of mESC pluripotency may be through its association with Oct4 and ß-catenin and its promoter binding capability. These results establish that Bcl3 positively regulates pluripotency genes and thus shed light on the mechanism of Bcl3 as a downstream molecule of LIF/STAT3 signaling in pluripotency maintenance.


Asunto(s)
Factor Inhibidor de Leucemia/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Proteínas del Linfoma 3 de Células B , Regulación de la Expresión Génica , Factor Inhibidor de Leucemia/genética , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Proteínas Proto-Oncogénicas/genética , Factor de Transcripción STAT3/genética , Factores de Transcripción/genética
14.
J Cell Sci ; 125(Pt 14): 3402-11, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22467851

RESUMEN

We have previously shown that DDA3 - also known as proline/serine-rich coiled-coil protein 1 (PSRC1) - is a microtubule-associated protein that promotes cell growth by stimulating the ß-catenin pathway. Here, we report that DDA3 can bundle and stabilize microtubules in vivo and in vitro. We found that overexpression of DDA3 increased the abundance of acetylated and tyrosinated microtubules. We employed PC12 and N2a cell lines, as well as cultured hippocampal neurons, and demonstrated that overexpression of DDA3 suppressed neurite/axon outgrowth, whereas its depletion accelerated neurite/axon formation and elongation. Knockdown of DDA3 reduced ß3-tubulin levels in N2a cells, which contributed to the spontaneous neurite formation caused by DDA3 depletion. Consistent with its role in suppressing neuritogenesis, DDA3 was downregulated during induced neuronal differentiation. Moreover, expression of DDA3 was detected in the rat brain at embryonic (E) day E15 and in the cortical region at E17, the period of active neurogenesis. Levels of cortical DDA3 decreased at the beginning of E19, when active neuritogenesis is completed. Overall our results demonstrate that DDA3 is a so-far-unknown microtubule-stabilizing protein that is involved in regulating neurite formation and elongation.


Asunto(s)
Microtúbulos/metabolismo , Neuritas/metabolismo , Neuronas/metabolismo , Fosfoproteínas/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular , Ratones , Células 3T3 NIH , Neuronas/citología , Células PC12 , Ratas , Ratas Sprague-Dawley
15.
Huan Jing Ke Xue ; 45(3): 1439-1447, 2024 Mar 08.
Artículo en Zh | MEDLINE | ID: mdl-38471859

RESUMEN

The Yangtze River, the largest river in China, has not been comprehensively studied for its basin's microplastic pollution status. Therefore, a comprehensive investigation and assessment system of microplastics was developed at the river basin scale to characterize the spatial distribution and composition of microplastics in the Yangtze River Basin in order to analyze their influencing factors and assess their ecological risks. The results showed that the microplastic abundance in the study area ranged from 21 to 44 080 n·m-3, with an average abundance of 4 483 n·m-3. The spatial distribution of microplastic abundance was higher in the tributaries than in the main streams (except the Ganjiang Basin), with the Chengdu of the Minjiang Basin being the tributary area with the highest abundance of microplastics detected. The size of microplastics in the river basin was concentrated in the 0-1 mm range; the shapes were mainly fiber and fragment; and the colors were mainly colored and transparent. Further, introducing the diversity index of microplastics, it was found that both the Simpson index and the Shannon-Wiener index could quantify the diversity of microplastic characteristic composition in the river basin, but there were certain differences in the changing trends between the two. Regression analysis showed that anthropogenic activities were significantly and positively correlated with microplastic abundance (P<0.05), and among the eight anthropogenic activity factors, civilian vehicle ownership and tourism income were the most strongly correlated with microplastic abundance, indicating that transportation and tourism were the main factors influencing microplastic distribution. From the perspective of the potential ecological risk index of microplastics, microplastics in the Yangtze River Basin posed a certain ecological risk, with 68.97% of the area falling within risk zones III and IV, with the ecological risk of microplastics in Taihu Lake warranting more widespread attention.


Asunto(s)
Microplásticos , Contaminantes Químicos del Agua , Ríos , Plásticos , Contaminantes Químicos del Agua/análisis , Monitoreo del Ambiente/métodos , China , Medición de Riesgo
16.
Hum Mol Genet ; 20(3): 455-65, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21075835

RESUMEN

Glycogen storage disease type IV (GSD-IV) is an autosomal recessive disease caused by a deficiency in glycogen-branching enzyme (GBE1) activity that results in the accumulation of amylopectin-like polysaccharide, which presumably leads to osmotic swelling and cell death. This disease is extremely heterogeneous in terms of tissue involvement, age of onset and clinical manifestation. The most severe fetal form presents as hydrops fetalis; however, its pathogenetic mechanisms are largely unknown. In this study, mice carrying a stop codon mutation (E609X) in the Gbe1 gene were generated using a gene-driven mutagenesis approach. Homozygous mutants (Gbe(-/-) mice) recapitulated the clinical features of hydrops fetalis and the embryonic lethality of the severe fetal form of GSD-IV. However, contrary to conventional expectations, little amylopectin accumulation and no cell degeneration were found in Gbe(-/-) embryonic tissues. Glycogen accumulation was reduced in developing hearts of Gbe(-/-)embryos, and abnormal cardiac development, including hypertrabeculation and noncompaction of the ventricular wall, was observed. Further, Gbe1 ablation led to poor ventricular function in late gestation and ultimately caused heart failure, fetal hydrops and embryonic lethality. We also found that the cell-cycle regulators cyclin D1 and c-Myc were highly expressed in cardiomyocytes and likely contributed to cardiomyocyte proliferation and trabeculation/compaction of the ventricular wall. Our results reveal that early molecular events associated with Gbe1 deficiency contribute to abnormal cardiac development and fetal hydrops in the fetal form of GSD-IV.


Asunto(s)
Enzima Ramificadora de 1,4-alfa-Glucano/deficiencia , Enzima Ramificadora de 1,4-alfa-Glucano/genética , Enfermedad del Almacenamiento de Glucógeno Tipo IV/genética , Glucógeno/metabolismo , Cardiopatías Congénitas/genética , Corazón/embriología , Amilopectina/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular , Codón de Terminación , Ciclina D1/genética , Pérdida del Embrión , Técnica del Anticuerpo Fluorescente , Genes myc , Enfermedad del Almacenamiento de Glucógeno Tipo IV/embriología , Enfermedad del Almacenamiento de Glucógeno Tipo IV/metabolismo , Cardiopatías Congénitas/metabolismo , Insuficiencia Cardíaca , Frecuencia Cardíaca , Hidropesía Fetal , Ratones , Miocitos Cardíacos , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN , Función Ventricular
17.
Int J Ophthalmol ; 16(9): 1521-1526, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37724262

RESUMEN

AIM: To describe the clinical and radiologic features of retrolaminar migration silicone oil (SiO) and observe the dynamic position of ventricular oil accumulation in supine and prone. METHODS: For this retrospective study, 29 patients who had a history of SiO injection treatment and underwent unenhanced head computed tomography (CT) were included from January 2019 to October 2022. The patients were divided into migration-positive and negative groups. Clinical history and CT features were compared using Whitney U and Fisher's exact tests. The dynamic position of SiO was observed within the ventricular system in supine and prone. CT images were visually assessed for SiO migration along the retrolaminar involving pathways for vision (optic nerve, chiasm, and tract) and ventricular system. RESULTS: Intraocular SiO migration was found in 5 of the 29 patients (17.24%), with SiO at the optic nerve head (n=1), optic nerve (n=4), optic chiasm (n=1), optic tract (n=1), and within lateral ventricles (n=1). The time interval between SiO injection and CT examination of migration-positive cases was significantly higher than that of migration-negative patients (22.8±16.5mo vs 13.1±2.6mo, P<0.001). The hyperdense lesion located in the frontal horns of the right lateral ventricle migrated to the fourth ventricle when changing the position from supine to prone. CONCLUSION: Although SiO retrolaminar migration is unusual, the clinician and radiologist should be aware of migration routes. The supine combined with prone examination is the first-choice method to confirm the presence of SiO in the ventricular system.

18.
Cell Rep ; 42(11): 113416, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37967007

RESUMEN

Differentiated cardiomyocytes (CMs) must undergo diverse morphological and functional changes during postnatal development. However, the mechanisms underlying initiation and coordination of these changes remain unclear. Here, we delineate an integrated, time-ordered transcriptional network that begins with expression of genes for cell-cell connections and leads to a sequence of structural, cell-cycle, functional, and metabolic transitions in mouse postnatal hearts. Depletion of histone H2B ubiquitin ligase RNF20 disrupts this gene network and impairs CM polarization. Subsequently, assay for transposase-accessible chromatin using sequencing (ATAC-seq) analysis confirmed that RNF20 contributes to chromatin accessibility in this context. As such, RNF20 is likely to facilitate binding of transcription factors at the promoters of genes involved in cell-cell connections and actin organization, which are crucial for CM polarization and functional integration. These results suggest that CM polarization is one of the earliest events during postnatal heart development and provide insights into how RNF20 regulates CM polarity and the postnatal gene program.


Asunto(s)
Miocitos Cardíacos , Ubiquitina-Proteína Ligasas , Animales , Ratones , Miocitos Cardíacos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Histonas/metabolismo , Cromatina , Epigénesis Genética , Expresión Génica
19.
Circ J ; 76(9): 2226-33, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22785082

RESUMEN

BACKGROUND: Heat shock proteins (HSPs) act as chaperones and have a protective function in cardiovascular diseases. The clinical association of a novel small HSPB7 with cardiovascular disease, however, has not been reported. The aim of this study was to investigate the potential biological functions of HSPB7 and its relationship with acute coronary syndrome (ACS). METHODS AND RESULTS: A mouse myocardial infarction (MI) model and samples from clinical human subjects were used to determine plasma HSPB7 concentration after acute MI. The associations of plasma HSPB7 concentration with ACS and other risk factors of coronary artery disease were analyzed. Plasma HSPB7 concentration was found to be rapidly elevated in mice after coronary artery ligation. In addition, plasma HSPB7 concentration was significantly higher in patients with ACS than in control patients with non-cardiac chest pain (5.1 ng/ml vs. 2.9 ng/ml, P<0.001). Plasma HSPB7 was detected as early as 1-3 h after the onset of symptoms and remained detectable up to 24h. Furthermore, in patients presenting to the emergency department with acute chest pain, HSPB7 level was an independent risk factor of ACS (adjusted odds ratio, 7.44; 95% confidence interval: 1.91-28.93, P<0.01). CONCLUSIONS: HSPB7 is a potential early biomarker after MI and serves as an independent risk factor of ACS in patients with acute chest pain.


Asunto(s)
Síndrome Coronario Agudo/sangre , Proteínas de Choque Térmico HSP27/sangre , Infarto del Miocardio/sangre , Anciano , Animales , Biomarcadores/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Factores de Riesgo , Factores de Tiempo
20.
NPJ Regen Med ; 7(1): 37, 2022 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-35906243

RESUMEN

The balance among quiescence, differentiation, and self-renewal of skeletal muscle stem cells (MuSCs) is tightly regulated by their intrinsic and extrinsic properties from the niche. How the niche controls MuSC fate remains unclear. Ribonucleotide reductase M2B (Rrm2b) modulates MuSC quiescence/differentiation in muscle in response to injury. Rrm2b knockout in myofibers, but not in MuSCs, led to weakness of muscles, such as a loss of muscle mass and strength. After muscle injury, damaged myofibers were more efficiently repaired in the Rrm2b myofiber-specific knockout mice than the control mice, but these myofibers were thinner and showed weak functioning. Rrm2b-deleted myofibers released several myokines, which trigger MuSCs to differentiate but not re-enter the quiescent stage to replenish the stem cell pool. Overall, Rrm2b in the myofibers plays a critical role in modulating the MuSC fate by modifying the microenvironment, and it may lead to a possible strategy to treat muscle disorders.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA