Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Blood ; 138(21): 2117-2128, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34115847

RESUMEN

Shwachman-Diamond syndrome (SDS; OMIM #260400) is caused by variants in SBDS (Shwachman-Bodian-Diamond syndrome gene), which encodes a protein that plays an important role in ribosome assembly. Recent reports suggest that recessive variants in EFL1 are also responsible for SDS. However, the precise genetic mechanism that leads to EFL1-induced SDS remains incompletely understood. Here we present 3 unrelated Korean SDS patients who carry biallelic pathogenic variants in EFL1 with biased allele frequencies, resulting from a bone marrow-specific somatic uniparental disomy in chromosome 15. The recombination events generated cells that were homozygous for the relatively milder variant, allowing for the evasion of catastrophic physiologic consequences. However, the milder EFL1 variant was still solely able to impair 80S ribosome assembly and induce SDS features in cell line and animal models. The loss of EFL1 resulted in a pronounced inhibition of terminal oligopyrimidine element-containing ribosomal protein transcript 80S assembly. Therefore, we propose a more accurate pathogenesis mechanism of EFL1 dysfunction that eventually leads to aberrant translational control and ribosomopathy.


Asunto(s)
Factores de Elongación de Péptidos/genética , Ribonucleoproteína Nuclear Pequeña U5/genética , Síndrome de Shwachman-Diamond/genética , Disomía Uniparental/genética , Adulto , Alelos , Animales , Niño , Preescolar , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Modelos Moleculares , Mutación Puntual
2.
Biochem Biophys Res Commun ; 534: 359-366, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33256983

RESUMEN

Paired Box (Pax) gene family, a group of transcription regulators have been implicated in diverse physiological processes. However, their role during hematopoiesis which generate a plethora of blood cells remains largely unknown. Using a previously reported single cell transcriptomics data, we analyzed the expression of individual Pax family members in hematopoietic cells in zebrafish. We have identified that Pax9, which is an essential regulator for odontogenesis and palatogenesis, is selectively localized within a single cluster of the hematopoietic lineage. To further analyze the function of Pax9 in hematopoiesis, we generated two independent pax9 knock-out mutants using the CRISPR-Cas9 technique. We found that Pax9 appears to be an essential regulator for granulopoiesis but dispensable for erythropoiesis during development, as lack of pax9 selectively decreased the number of neutrophils with a concomitant decrease in the expression level of neutrophil markers. In addition, embryos, where pax9 was functionally disrupted by injecting morpholinos, failed to increase the number of neutrophils in response to pathogenic bacteria, suggesting that Pax9 is not only essential for developmental granulopoiesis but also emergency granulopoiesis. Due to the inability to initiate emergency granulopoiesis, innate immune responses were severely compromised in pax9 morpholino-mediated embryos, increasing their susceptibility and mortality. Taken together, our data indicate that Pax9 is essential for granulopoiesis and promotes innate immunity in zebrafish larvae.


Asunto(s)
Eritropoyesis/inmunología , Mielopoyesis/inmunología , Factor de Transcripción PAX9/inmunología , Proteínas de Pez Cebra/inmunología , Pez Cebra/inmunología , Animales , Animales Modificados Genéticamente , Infecciones Bacterianas/inmunología , Sistemas CRISPR-Cas , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Granulocitos/inmunología , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Mielopoyesis/genética , Factor de Transcripción PAX9/deficiencia , Factor de Transcripción PAX9/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
3.
Circulation ; 135(23): 2288-2298, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28356442

RESUMEN

BACKGROUND: Bone morphogenetic protein (BMP) signaling has multiple roles in the development and function of the blood vessels. In humans, mutations in BMP receptor type 2 (BMPR2), a key component of BMP signaling, have been identified in the majority of patients with familial pulmonary arterial hypertension (PAH). However, only a small subset of individuals with BMPR2 mutation develops PAH, suggesting that additional modifiers of BMPR2 function play an important role in the onset and progression of PAH. METHODS: We used a combination of studies in zebrafish embryos and genetically engineered mice lacking endothelial expression of Vegfr3 to determine the interaction between vascular endothelial growth factor receptor 3 (VEGFR3) and BMPR2. Additional in vitro studies were performed by using human endothelial cells, including primary lung endothelial cells from subjects with PAH. RESULTS: Attenuation of Vegfr3 in zebrafish embryos abrogated Bmp2b-induced ectopic angiogenesis. Endothelial cells with disrupted VEGFR3 expression failed to respond to exogenous BMP stimulation. Mechanistically, VEGFR3 is physically associated with BMPR2 and facilitates ligand-induced endocytosis of BMPR2 to promote phosphorylation of SMADs and transcription of ID genes. Conditional, endothelial-specific deletion of Vegfr3 in mice resulted in impaired BMP signaling responses, and significantly worsened hypoxia-induced pulmonary hypertension. Consistent with these data, we found significant decrease in VEGFR3 expression in pulmonary arterial endothelial cells from human PAH subjects, and reconstitution of VEGFR3 expression in PAH pulmonary arterial endothelial cells restored BMP signaling responses. CONCLUSIONS: Our findings identify VEGFR3 as a key regulator of endothelial BMPR2 signaling and a potential determinant of PAH penetrance in humans.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/biosíntesis , Endotelio Vascular/metabolismo , Hipertensión Pulmonar/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Células Cultivadas , Endotelio Vascular/patología , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Pez Cebra
4.
Circ Res ; 114(1): 56-66, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24122719

RESUMEN

RATIONALE: The emergence of lymphatic endothelial cells (LECs) seems to be highly regulated during development. Although several factors that promote the differentiation of LECs in embryonic development have been identified, those that negatively regulate this process are largely unknown. OBJECTIVE: Our aim was to delineate the role of bone morphogenetic protein (BMP) 2 signaling in lymphatic development. METHODS AND RESULTS: BMP2 signaling negatively regulates the formation of LECs. Developing LECs lack any detectable BMP signaling activity in both zebrafish and mouse embryos, and excess BMP2 signaling in zebrafish embryos and mouse embryonic stem cell-derived embryoid bodies substantially decrease the emergence of LECs. Mechanistically, BMP2 signaling induces expression of miR-31 and miR-181a in a SMAD-dependent mechanism, which in turn results in attenuated expression of prospero homeobox protein 1 during development. CONCLUSIONS: Our data identify BMP2 as a key negative regulator for the emergence of the lymphatic lineage during vertebrate development.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Endotelio Linfático/embriología , Endotelio Linfático/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Animales , Proteína Morfogenética Ósea 2/genética , Diferenciación Celular , Línea Celular , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Vasos Linfáticos/embriología , Vasos Linfáticos/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Smad/metabolismo , Transcripción Genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética
5.
Circ Res ; 113(1): 22-31, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23603510

RESUMEN

RATIONALE: The peptide ligand apelin and its receptor APJ constitute a signaling pathway with numerous effects on the cardiovascular system, including cardiovascular development in model organisms such as xenopus and zebrafish. OBJECTIVE: This study aimed to characterize the embryonic lethal phenotype of the Apj-/- mice and to define the involved downstream signaling targets. METHODS AND RESULTS: We report the first characterization of the embryonic lethality of the Apj-/- mice. More than half of the expected Apj-/- embryos died in utero because of cardiovascular developmental defects. Those succumbing to early embryonic death had markedly deformed vasculature of the yolk sac and the embryo, as well as poorly looped hearts with aberrantly formed right ventricles and defective atrioventricular cushion formation. Apj-/- embryos surviving to later stages demonstrated incomplete vascular maturation because of a deficiency of vascular smooth muscle cells and impaired myocardial trabeculation and ventricular wall development. The molecular mechanism implicates a novel, noncanonical signaling pathway downstream of apelin-APJ involving Gα13, which induces histone deacetylase (HDAC) 4 and HDAC5 phosphorylation and cytoplasmic translocation, resulting in activation of myocyte enhancer factor 2. Apj-/- mice have greater endocardial Hdac4 and Hdac5 nuclear localization and reduced expression of the myocyte enhancer factor 2 (MEF2) transcriptional target Krüppel-like factor 2. We identify a number of commonly shared transcriptional targets among apelin-APJ, Gα13, and MEF2 in endothelial cells, which are significantly decreased in the Apj-/- embryos and endothelial cells. CONCLUSIONS: Our results demonstrate a novel role for apelin-APJ signaling as a potent regulator of endothelial MEF2 function in the developing cardiovascular system.


Asunto(s)
Anomalías Cardiovasculares/embriología , Sistema Cardiovascular/embriología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Factores Reguladores Miogénicos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transporte Activo de Núcleo Celular , Adipoquinas , Animales , Apelina , Receptores de Apelina , Anomalías Cardiovasculares/genética , Endocardio/embriología , Endocardio/metabolismo , Endotelio Vascular/metabolismo , Femenino , Corazón Fetal/anomalías , Subunidades alfa de la Proteína de Unión al GTP G12-G13/fisiología , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Histona Desacetilasas/metabolismo , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción MEF2 , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Transcripción Genética
6.
Arterioscler Thromb Vasc Biol ; 34(9): 1838-45, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25060789

RESUMEN

Endothelial cells are a highly diverse group of cells which display distinct cellular responses to exogenous stimuli. Although the aptly named vascular endothelial growth factor-A signaling pathway is hailed as the most important signaling input for endothelial cells, additional factors also participate in regulating diverse aspects of endothelial behaviors and functions. Given this heterogeneity, these additional factors seem to play a critical role in creating a custom-tailored environment to regulate behaviors and functions of distinct subgroups of endothelial cells. For instance, molecular cues that modulate morphogenesis of arterial vascular beds can be distinct from those that govern morphogenesis of venous vascular beds. Recently, we have found that bone morphogenetic protein signaling selectively promotes angiogenesis from venous vascular beds without eliciting similar responses from arterial vascular beds in zebrafish, indicating that bone morphogenetic protein signaling functions as a context-dependent regulator during vascular morphogenesis. In this review, we will provide an overview of the molecular mechanisms that underlie proangiogenic effects of bone morphogenetic protein signaling on venous vascular beds in the context of endothelial heterogeneity and suggest a more comprehensive picture of the molecular mechanisms of vascular morphogenesis during development.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Venas/embriología , Proteínas de Pez Cebra/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas/fisiología , Mesodermo/citología , Mesodermo/fisiología , Ratones , Especificidad de Órganos , Receptores Notch/fisiología , Proteínas Smad/fisiología , Especificidad de la Especie , Factor A de Crecimiento Endotelial Vascular/fisiología , Pez Cebra/embriología
7.
Arterioscler Thromb Vasc Biol ; 34(2): 338-45, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24311379

RESUMEN

OBJECTIVE: Apelin and its cognate receptor Aplnr/Apj are essential for diverse biological processes. However, the function of Apelin signaling in lymphatic development remains to be identified, despite the preferential expression of Apelin and Aplnr within developing blood and lymphatic endothelial cells in vertebrates. In this report, we aim to delineate the functions of Apelin signaling during lymphatic development. APPROACH AND RESULTS: We investigated the functions of Apelin signaling during lymphatic development using zebrafish embryos and found that attenuation of Apelin signaling substantially decreased the formation of the parachordal vessel and the number of lymphatic endothelial cells within the developing thoracic duct, indicating an essential role of Apelin signaling during the early phase of lymphatic development. Mechanistically, we found that abrogation of Apelin signaling selectively attenuates lymphatic endothelial serine-threonine kinase Akt 1/2 phosphorylation without affecting the phosphorylation status of extracellular signal-regulated kinase 1/2. Moreover, lymphatic abnormalities caused by the reduction of Apelin signaling were significantly exacerbated by the concomitant partial inhibition of serine-threonine kinase Akt/protein kinase B signaling. Apelin and vascular endothelial growth factor-C (VEGF-C) signaling provide a nonredundant activation of serine-threonine kinase Akt/protein kinase B during lymphatic development because overexpression of VEGF-C or apelin was unable to rescue the lymphatic defects caused by the lack of Apelin or VEGF-C, respectively. CONCLUSIONS: Taken together, our data present compelling evidence suggesting that Apelin signaling regulates lymphatic development by promoting serine-threonine kinase Akt/protein kinase B activity in a VEGF-C/VEGF receptor 3-independent manner during zebrafish embryogenesis.


Asunto(s)
Quimiocinas/metabolismo , Linfangiogénesis , Transducción de Señal , Conducto Torácico/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Apelina , Receptores de Apelina , Células Cultivadas , Quimiocinas/genética , Células Endoteliales/metabolismo , Endotelio Linfático/embriología , Endotelio Linfático/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Conducto Torácico/embriología , Factores de Tiempo , Transfección , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
8.
J Neurosci ; 32(24): 8201-7, 2012 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-22699901

RESUMEN

Oligodendrocytes are myelinating glial cells in the CNS and are essential for proper neuronal function. During development, oligodendrocyte progenitor cells (OPCs) are specified from the motor neuron precursor domain of the ventral spinal cord and differentiate into myelinating oligodendrocytes after migration to the white matter of the neural tube. Cell cycle control of OPCs influences the balance between immature OPCs and myelinating oligodendrocytes, but the precise mechanism regulating the differentiation of OPCs into myelinating oligodendrocytes is unclear. To understand the mechanisms underlying oligodendrocyte differentiation, an N-ethyl-N-nitrosourea-based mutagenesis screen was performed and a zebrafish leo1 mutant, dalmuri (dal(knu6)) was identified in the current study. Leo1 is a component of the evolutionarily conserved RNA polymerase II-associated factor 1 complex (PAF1C), which is a positive regulator of transcription elongation. The dal(knu6) mutant embryos specified motor neurons and OPCs normally, and at the appropriate time, but OPCs subsequently failed to differentiate into myelinating oligodendrocytes and were eliminated by apoptosis. A loss-of-function study of cdc73, another member of PAF1C, showed the same phenotype in the CNS, indicating that PAF1C function is required for oligodendrocyte differentiation. Interestingly, inhibition of positive transcription elongation factor b (p-TEFb), rescued downregulated gene expression and impaired oligodendrocyte differentiation in the dal(knu6) mutant and Cdc73-deficient embryos. Together, these results indicate that antagonistic regulation of gene expression by PAF1C and p-TEFb plays a crucial role in oligodendrocyte development in the CNS.


Asunto(s)
Proteínas Portadoras/fisiología , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Oligodendroglía/fisiología , Células Madre/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Proteínas Portadoras/genética , Sistema Nervioso Central/citología , Sistema Nervioso Central/fisiología , Técnicas de Silenciamiento del Gen/métodos , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Oligodendroglía/citología , Factor B de Elongación Transcripcional Positiva/metabolismo , Células Madre/citología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
9.
Biochem Biophys Res Commun ; 430(4): 1212-6, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23266606

RESUMEN

Segregation of two axial vessels, the dorsal aorta and caudal vein, is one of the earliest patterning events occur during development of vasculature. Despite the importance of this process and recent advances in our understanding on vascular patterning during development, molecular mechanisms that coordinate the segregation of axial vessels remain largely elusive. In this report, we find that vascular endothelial growth factor-A (Vegf-A) signaling regulates the segregation of dorsal aorta and axial vein during development. Inhibition of Vegf-A pathway components including ligand Vegf-A and its cognate receptor Kdrl, caused failure in segregation of axial vessels in zebrafish embryos. Similarly, chemical inhibition of Mitogen-activated protein kinase kinase (Map2k1)/Extracellular-signal-regulated kinases (Erk) and phosphatidylinositol 3-kinases (PI3K), which are downstream effectors of Vegf-A signaling pathway, led to the fusion of two axial vessels. Moreover, we find that restoring Erk activity by over-expression of constitutively active MEK in embryos with a reduced level of Vegf-A signaling can rescue the defects in axial vessel segregation. Taken together, our data show that segregation of axial vessels requires the function of Vegf-A signaling, and Erk may function as the major downstream effector in this process.


Asunto(s)
Aorta/crecimiento & desarrollo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/fisiología , Venas/crecimiento & desarrollo , Animales , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Pez Cebra/crecimiento & desarrollo
10.
Nat Commun ; 14(1): 2390, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37185814

RESUMEN

A comprehensive understanding of endothelial cell lineage specification will advance cardiovascular regenerative medicine. Recent studies found that unique epigenetic signatures preferentially regulate cell identity genes. We thus systematically investigate the epigenetic landscape of endothelial cell lineage and identify MECOM to be the leading candidate as an endothelial cell lineage regulator. Single-cell RNA-Seq analysis verifies that MECOM-positive cells are exclusively enriched in the cell cluster of bona fide endothelial cells derived from induced pluripotent stem cells. Our experiments demonstrate that MECOM depletion impairs human endothelial cell differentiation, functions, and Zebrafish angiogenesis. Through integrative analysis of Hi-C, DNase-Seq, ChIP-Seq, and RNA-Seq data, we find MECOM binds enhancers that form chromatin loops to regulate endothelial cell identity genes. Further, we identify and verify the VEGF signaling pathway to be a key target of MECOM. Our work provides important insights into epigenetic regulation of cell identity and uncovered MECOM as an endothelial cell lineage regulator.


Asunto(s)
Células Endoteliales , Epigénesis Genética , Animales , Humanos , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Endoteliales/metabolismo , Proteína del Locus del Complejo MDS1 y EV11/genética , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
11.
Biochem Biophys Res Commun ; 423(1): 140-6, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22634317

RESUMEN

During vertebrate heart valve formation, Wnt/ß-catenin signaling induces BMP signals in atrioventricular canal (AVC) myocardial cells and underlying AVC endocardial cells then undergo endothelial-mesenchymal transdifferentiation (EMT) by receiving this BMP signals. Histone deacetylases (HDACs) have been implicated in numerous developmental processes by regulating gene expression. However, their specific roles in controlling heart valve development are largely unexplored. To investigate the role of HDACs in vertebrate heart valve formation, we treated zebrafish embryos with trichostatin A (TSA), an inhibitor of class I and II HDACs, from 36 to 48 h post-fertilization (hpf) during which heart looping and valve formation occur. Following TSA treatment, abnormal linear heart tube development was observed. In these embryos, expression of AVC myocardial bmp4 and AVC endocardial notch1b genes was markedly reduced with subsequent failure of EMT in the AVC endocardial cells. However, LiCl-mediated activation of Wnt/ß-catenin signaling was able to rescue defective heart tube formation, bmp4 and notch1b expression, and EMT in the AVC region. Taken together, our results demonstrated that HDAC activity plays a pivotal role in vertebrate heart tube formation by activating Wnt/ß-catenin signaling which induces bmp4 expression in AVC myocardial cells.


Asunto(s)
Válvulas Cardíacas/embriología , Histona Desacetilasas/metabolismo , Vía de Señalización Wnt , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , beta Catenina/metabolismo , Acetilación , Animales , Proteína Morfogenética Ósea 4/biosíntesis , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Cojinetes Endocárdicos/embriología , Endocardio/embriología , Endocardio/metabolismo , Expresión Génica/efectos de los fármacos , Válvulas Cardíacas/anomalías , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histonas/metabolismo , Ácidos Hidroxámicos/farmacología , Cloruro de Litio/farmacología , Miocardio/metabolismo , Organogénesis/efectos de los fármacos , Organogénesis/genética , Receptor Notch1/biosíntesis , Pez Cebra/metabolismo , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/genética
12.
Cells ; 11(22)2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36429071

RESUMEN

Accumulating evidence indicates that the APOA1 binding protein (AIBP)-a secreted protein-plays a profound role in lipid metabolism. Interestingly, AIBP also functions as an NAD(P)H-hydrate epimerase to catalyze the interconversion of NAD(P)H hydrate [NAD(P)HX] epimers and is renamed as NAXE. Thus, we call it NAXE hereafter. We investigated its role in NAD(P)H-involved metabolism in murine cardiomyocytes, focusing on the metabolism of hexose, lipids, and amino acids as well as mitochondrial redox function. Unbiased metabolite profiling of cardiac tissue shows that NAXE knockout markedly upregulates the ketone body 3-hydroxybutyric acid (3-HB) and increases or trends increasing lipid-associated metabolites cholesterol, α-linolenic acid and deoxycholic acid. Paralleling greater ketone levels, ChemRICH analysis of the NAXE-regulated metabolites shows reduced abundance of hexose despite similar glucose levels in control and NAXE-deficient blood. NAXE knockout reduces cardiac lactic acid but has no effect on the content of other NAD(P)H-regulated metabolites, including those associated with glucose metabolism, the pentose phosphate pathway, or Krebs cycle flux. Although NAXE is present in mitochondria, it has no apparent effect on mitochondrial oxidative phosphorylation. Instead, we detected more metabolites that can potentially improve cardiac function (3-HB, adenosine, and α-linolenic acid) in the Naxe-/- heart; these mice also perform better in aerobic exercise. Our data reveal a new role of NAXE in cardiac ketone and lipid metabolism.


Asunto(s)
Cetonas , NAD , Animales , Ratones , Ácido alfa-Linolénico , NAD/metabolismo , Racemasas y Epimerasas , Respiración de la Célula
13.
PLoS One ; 16(4): e0248964, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33793635

RESUMEN

Emerging studies indicate that APOA-I binding protein (AIBP) is a secreted protein and functions extracellularly to promote cellular cholesterol efflux, thereby disrupting lipid rafts on the plasma membrane. AIBP is also present in the mitochondria and acts as an epimerase, facilitating the repair of dysfunctional hydrated NAD(P)H, known as NAD(P)H(X). Importantly, AIBP deficiency contributes to lethal neurometabolic disorder, reminiscent of the Leigh syndrome in humans. Whereas cyclic NADPHX production is proposed to be the underlying cause, we hypothesize that an unbiased metabolic profiling may: 1) reveal new clues for the lethality, e.g., changes of mitochondrial metabolites., and 2) identify metabolites associated with new AIBP functions. To this end, we performed unbiased and profound metabolic studies of plasma obtained from adult AIBP knockout mice and control littermates of both genders. Our systemic metabolite profiling, encompassing 9 super pathways, identified a total of 640 compounds. Our studies demonstrate a surprising sexual dimorphism of metabolites affected by AIBP deletion, with more statistically significant changes in the AIBP knockout female vs male when compared with the corresponding controls. AIBP knockout trends to reduce cholesterol but increase the bile acid precursor 7-HOCA in female but not male. Complex lipids, phospholipids, sphingomyelin and plasmalogens were reduced, while monoacylglycerol, fatty acids and the lipid soluble vitamins E and carotene diol were elevated in AIBP knockout female but not male. NAD metabolites were not significantly different in AIBP knockout vs control mice but differed for male vs female mice. Metabolites associated with glycolysis and the Krebs cycle were unchanged by AIBP knockout. Importantly, polyamine spermidine, critical for many cellular functions including cerebral cortex synapses, was reduced in male but not female AIBP knockout. This is the first report of a systemic metabolite profile of plasma samples from AIBP knockout mice, and provides a metabolic basis for future studies of AIBP regulation of cellular metabolism and the pathophysiological presentation of AIBP deficiency in patients.


Asunto(s)
Fosfoproteínas/metabolismo , Racemasas y Epimerasas/metabolismo , Factores Sexuales , Animales , Colesterol/metabolismo , Femenino , Metabolismo de los Lípidos , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NAD/metabolismo
14.
Front Cell Dev Biol ; 8: 603306, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33330499

RESUMEN

Unpaired fins, which are the most ancient form of locomotory appendages in chordates, had emerged at least 500 million years ago. While it has been suggested that unpaired fins and paired fins share structural similarities, cellular and molecular mechanisms that regulate the outgrowth of the former have not been fully elucidated yet. Using the ventral fin fold in zebrafish as a model, here, we investigate how the outgrowth of the unpaired fin is modulated. We show that Bone Morphogenetic Protein (BMP) signaling restricts extension of the ventral fin fold along the proximodistal axis by modulating diverse aspects of cellular behaviors. We find that lack of BMP signaling, either caused by genetic or chemical manipulation, prolongs the proliferative capacity of epithelial cells and substantially increases the number of cells within the ventral fin fold. In addition, inhibition of BMP signaling attenuates the innate propensity of cell division along the anteroposterior axis and shifts the orientation of cell division toward the proximodistal axis. Moreover, abrogating BMP signaling appears to induce excessive distal migration of cells within the ventral fin fold, and therefore precipitates extension along the proximodistal axis. Taken together, our data suggest that BMP signaling restricts the outgrowth of the ventral fin fold during zebrafish development.

15.
Front Cell Dev Biol ; 8: 589717, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33330468

RESUMEN

Endothelial cells appear to emerge from diverse progenitors. However, to which extent their developmental origin contributes to define their cellular and molecular characteristics remains largely unknown. Here, we report that a subset of endothelial cells that emerge from the tailbud possess unique molecular characteristics that set them apart from stereotypical lateral plate mesoderm (LPM)-derived endothelial cells. Lineage tracing shows that these tailbud-derived endothelial cells arise at mid-somitogenesis stages, and surprisingly do not require Npas4l or Etsrp function, indicating that they have distinct spatiotemporal origins and are regulated by distinct molecular mechanisms. Microarray and single cell RNA-seq analyses reveal that somitogenesis- and neurogenesis-associated transcripts are over-represented in these tailbud-derived endothelial cells, suggesting that they possess a unique transcriptomic signature. Taken together, our results further reveal the diversity of endothelial cells with respect to their developmental origin and molecular properties, and provide compelling evidence that the molecular characteristics of endothelial cells may reflect their distinct developmental history.

16.
Biochem Biophys Res Commun ; 381(4): 717-21, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19258008

RESUMEN

During normal forebrain development in vertebrates, rostral neural tissue must be protected from Wnt signals via the actions of locally expressed Wnt antagonistic factors. In zebrafish zygotic oep (Zoep) mutants, forebrain structure is severely disrupted with reduced expression of the Wnt antagonists secreted frizzled related protein1 and dickkopf1. To analyze the temporal effects of Wnt antagonism on forebrain development, we generated transgenic zebrafish that overexpressed the dominant negative form of frizzled8a (DNfz8a) in wild-type and Zoep mutants under the control of a heat-inducible promoter. This model allowed for assessment of the dynamics of Wnt antagonistic signaling during forebrain development. Our results demonstrated that overexpression of DNfz8a in Zoep embryos between 7 and 16hpf increased putative forebrain region demarcated by anf and distal-less2 expressions. These results suggest that normal forebrain development requires continual Wnt antagonism from the early gastrula to the mid-somitogenesis stage.


Asunto(s)
Receptores Frizzled/biosíntesis , Prosencéfalo/crecimiento & desarrollo , Receptores Acoplados a Proteínas G/biosíntesis , Somitos/crecimiento & desarrollo , Proteínas Wnt/antagonistas & inhibidores , Proteínas de Pez Cebra/biosíntesis , Pez Cebra/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Proteínas de Homeodominio/metabolismo , Prosencéfalo/metabolismo , Somitos/metabolismo , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
17.
Sci Rep ; 9(1): 4152, 2019 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842432

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

18.
Science ; 363(6431): 1085-1088, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30705153

RESUMEN

Hypercholesterolemia, the driving force of atherosclerosis, accelerates the expansion and mobilization of hematopoietic stem and progenitor cells (HSPCs). The molecular determinants connecting hypercholesterolemia with hematopoiesis are unclear. Here, we report that a somite-derived prohematopoietic cue, AIBP, orchestrates HSPC emergence from the hemogenic endothelium, a type of specialized endothelium manifesting hematopoietic potential. Mechanistically, AIBP-mediated cholesterol efflux activates endothelial Srebp2, the master transcription factor for cholesterol biosynthesis, which in turn transactivates Notch and promotes HSPC emergence. Srebp2 inhibition impairs hypercholesterolemia-induced HSPC expansion. Srebp2 activation and Notch up-regulation are associated with HSPC expansion in hypercholesterolemic human subjects. Genome-wide chromatin immunoprecipitation followed by sequencing (ChIP-seq), RNA sequencing (RNA-seq), and assay for transposase-accessible chromatin using sequencing (ATAC-seq) indicate that Srebp2 transregulates Notch pathway genes required for hematopoiesis. Our studies outline an AIBP-regulated Srebp2-dependent paradigm for HSPC emergence in development and HPSC expansion in atherosclerotic cardiovascular disease.


Asunto(s)
Colesterol/biosíntesis , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Hipercolesterolemia/metabolismo , Animales , Anticolesterolemiantes/farmacología , Atorvastatina/farmacología , Secuencia de Bases , Inmunoprecipitación de Cromatina , Enfermedad de la Arteria Coronaria/metabolismo , Regulación de la Expresión Génica , Hematopoyesis/genética , Racemasas y Epimerasas/metabolismo , Receptores Notch/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
19.
Sci Rep ; 8(1): 9840, 2018 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-29959335

RESUMEN

Intussusceptive angiogenesis (IA) is a complementary method to sprouting angiogenesis (SA). The hallmark of IA is formation of trans-capillary tissue pillars, their fusion and remodeling of the vascular plexus. In this study, we investigate the formation of the zebrafish caudal vein plexus (CVP) in Tg(fli1a:eGFP) y7 and the synergistic interaction of IA and SA in crafting the archetypical angio-architecture of the CVP. Dynamic in vivo observations and quantitative analyses revealed that the primitive CVP during development was initiated through SA. Further vascular growth and remodeling occurred by IA. Intussusception contributed to the expansion of the CVP by formation of new pillars. Those pillars arose in front of the already existing ones; and in a subsequent step the serried pillars elongated and fused together. This resulted in segregation of larger vascular segments and remodelling of the disorganized vascular meshwork into hierarchical tree-like arrangement. Blood flow was the main driving force for IA, particularly shear stress geometry at the site of pillar formation and fusion. Computational simulations based on hemodynamics showed drop in shear stress levels at locations of new pillar formation, pillar elongation and fusion. Correlative 3D serial block face scanning electron microscopy confirmed the morphological substrate of the phenomena of the pillar formation observed in vivo. The data obtained demonstrates that after the sprouting phase and formation of the primitive capillary meshwork, the hemodynamic conditions enhance intussusceptive segregation of hierarchical vascular tree i.e. intussusceptive arborization resulting in complex vascular structures with specific angio-architecture.


Asunto(s)
Hemodinámica , Morfogénesis , Neovascularización Fisiológica , Venas/crecimiento & desarrollo , Pez Cebra/fisiología , Animales , Intususcepción , Venas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA