Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dev Biol ; 490: 53-65, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35853502

RESUMEN

Mammalian KMT2C, KMT2D, and HCFC1 are expressed during heart development and have been associated with congenital heart disease, but their roles in heart development remain elusive. We found that the Drosophila Lpt and trr genes encode the N-terminal and C-terminal homologs, respectively, of mammalian KMT2C or KMT2D. Lpt and trr mutant embryos showed reduced cardiac progenitor cells. Silencing of Lpt, trr, or both simultaneously in the heart led to similar abnormal cardiac morphology, tissue fibrosis, and cardiac functional defects. Like KMT2D, Lpt and trr were found to modulate histone H3K4 mono- and dimethylation, but not trimethylation. Investigation of downstream genes regulated by mouse KMT2D in the heart showed that their fly homologs are similarly regulated by Lpt or trr in the fly heart, suggesting that Lpt and trr regulate an evolutionarily conserved transcriptional network for heart development. Moreover, we showed that cardiac silencing of Hcf, the fly homolog of mammalian HCFC1, leads to heart defects similar to those observed in Lpt and trr silencing, as well as reduced H3K4 monomethylation. Our findings suggest that Lpt and trr function together to execute the conserved function of mammalian KMT2C and KMT2D in histone H3 lysine K4 mono- and dimethylation required for heart development. Possibly aided by Hcf, which we show plays a related role in H3K4 methylation during fly heart development.


Asunto(s)
Proteínas de Drosophila , N-Metiltransferasa de Histona-Lisina , Histonas , Coactivadores de Receptor Nuclear , Animales , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Metilación , Ratones , Coactivadores de Receptor Nuclear/genética , Coactivadores de Receptor Nuclear/metabolismo
2.
Int J Mol Sci ; 24(24)2023 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-38139143

RESUMEN

Highly evolutionarily conserved multiprotein complexes termed Complex of Proteins Associated with Set1 (COMPASS) are required for histone 3 lysine 4 (H3K4) methylation. Drosophila Set1, Trx, and Trr form the core subunits of these complexes. We show that flies deficient in any of these three subunits demonstrated high lethality at eclosion (emergence of adult flies from their pupal cases) and significantly shortened lifespans for the adults that did emerge. Silencing Set1, trx, or trr in the heart led to a reduction in H3K4 monomethylation (H3K4me1) and dimethylation (H3K4me2), reflecting their distinct roles in H3K4 methylation. Furthermore, we studied the gene expression patterns regulated by Set1, Trx, and Trr. Each of the COMPASS core subunits controls the methylation of different sets of genes, with many metabolic pathways active early in development and throughout, while muscle and heart differentiation processes were methylated during later stages of development. Taken together, our findings demonstrate the roles of COMPASS series complex core subunits Set1, Trx, and Trr in regulating histone methylation during heart development and, given their implication in congenital heart diseases, inform research on heart disease.


Asunto(s)
Proteínas de Drosophila , Epigénesis Genética , Animales , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Corazón/crecimiento & desarrollo
3.
Molecules ; 28(13)2023 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-37446834

RESUMEN

Euphorbia fischeriana has a long-standing history of use in traditional medicine for the treatment of tuberculosis diseases. However, the plant's therapeutic potential extends beyond this specific ailment. The present study aimed to investigate the antioxidant properties of Euphorbia fischeriana and lay the groundwork for further research on its potential therapeutic applications. Phytochemical tests were performed on the plant, and 11 types of phytochemicals were identified. Ultraviolet-visible spectrophotometry was used to evaluate the active components and antioxidant properties of eight different solvent extracts, ultimately selecting acetone extract for further research. UHPLC-ESI-Q-TOF-MS identified 43 compounds in the acetone extract, and chemical calculations were used to isolate those with high content and antioxidant activity. Three stability experiments confirmed the extract's stability, while cell viability and oral acute toxicity studies demonstrated its relatively low toxicity. In rats, the acetone extract showed significant protective effects against D-galactosamine-induced liver damage through histopathological examination and biochemical analysis. These results suggest that Euphorbia fischeriana's acetone extract has potential in treating diseases related to oxidative imbalances. Therefore, this study highlights the plant's potential therapeutic applications while providing insight into its antioxidant properties.


Asunto(s)
Antioxidantes , Euphorbia , Ratas , Animales , Antioxidantes/farmacología , Antioxidantes/química , Extractos Vegetales/química , Euphorbia/química , Acetona , Fitoquímicos/farmacología
4.
BMC Microbiol ; 21(1): 18, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33419388

RESUMEN

BACKGROUND: Continuous cropping of ginseng (Panax ginseng Meyer) cultivated in farmland for an extended period gives rise to soil-borne disease. The change in soil microbial composition is a major cause of soil-borne diseases and an obstacle to continuous cropping. The impact of cultivation modes and ages on the diversity and composition of the P. ginseng rhizosphere microbial community and technology suitable for cropping P. ginseng in farmland are still being explored. METHODS: Amplicon sequencing of bacterial 16S rRNA genes and fungal ITS regions were analyzed for microbial community composition and diversity. RESULTS: The obtained sequencing data were reasonable for estimating soil microbial diversity. We observed significant variations in richness, diversity, and relative abundances of microbial taxa between farmland, deforestation field, and different cultivation years. The bacterial communities of LCK (forest soil where P. ginseng was not grown) had a much higher richness and diversity than those in NCK (farmland soil where P. ginseng was not grown). The increase in cultivation years of P. ginseng in farmland and deforestation field significantly changed the diversity of soil microbial communities. In addition, the accumulation of P. ginseng soil-borne pathogens (Monographella cucumerina, Ilyonectria mors-panacis, I. robusta, Fusarium solani, and Nectria ramulariae) varied with the cropping age of P. ginseng. CONCLUSION: Soil microbial diversity and function were significantly poorer in farmland than in the deforestation field and were affected by P. ginseng planting years. The abundance of common soil-borne pathogens of P. ginseng increased with the cultivation age and led to an imbalance in the microbial community.


Asunto(s)
Bacterias/clasificación , Hongos/clasificación , Panax/crecimiento & desarrollo , Análisis de Secuencia de ADN/métodos , Agricultura , Bacterias/genética , Bacterias/crecimiento & desarrollo , Bacterias/aislamiento & purificación , ADN Bacteriano/genética , ADN de Hongos/genética , Hongos/crecimiento & desarrollo , Hongos/aislamiento & purificación , Panax/microbiología , Filogenia , Raíces de Plantas/crecimiento & desarrollo , Raíces de Plantas/microbiología , ARN Ribosómico 16S/genética , Rizosfera , Microbiología del Suelo
5.
J Am Soc Nephrol ; 31(5): 1024-1034, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32238475

RESUMEN

BACKGROUND: Studies have linked mutations in genes encoding the eight-protein exocyst protein complex to kidney disease, but the underlying mechanism is unclear. Because Drosophila nephrocytes share molecular and structural features with mammalian podocytes, they provide an efficient model for studying this issue. METHODS: We silenced genes encoding exocyst complex proteins specifically in Drosophila nephrocytes and studied the effects on protein reabsorption by lacuna channels and filtration by the slit diaphragm. We performed nephrocyte functional assays, carried out super-resolution confocal microscopy of slit diaphragm proteins, and used transmission electron microscopy to analyze ultrastructural changes. We also examined the colocalization of slit diaphragm proteins with exocyst protein Sec15 and with endocytosis and recycling regulators Rab5, Rab7, and Rab11. RESULTS: Silencing exocyst genes in nephrocytes led to profound changes in structure and function. Abolition of cellular accumulation of hemolymph proteins with dramatically reduced lacuna channel membrane invaginations offered a strong indication of reabsorption defects. Moreover, the slit diaphragm's highly organized surface structure-essential for filtration-was disrupted, and key proteins were mislocalized. Ultrastructural analysis revealed that exocyst gene silencing led to the striking appearance of novel electron-dense structures that we named "exocyst rods," which likely represent accumulated membrane proteins following defective exocytosis or recycling. The slit diaphragm proteins partially colocalized with Sec15, Rab5, and Rab11. CONCLUSIONS: Our findings suggest that the slit diaphragm of Drosophila nephrocytes requires balanced endocytosis and recycling to maintain its structural integrity and that impairment of the exocyst complex leads to disruption of the slit diaphragm and nephrocyte malfunction. This model may help identify therapeutic targets for treating kidney diseases featuring molecular defects in vesicle endocytosis, exocytosis, and recycling.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/citología , Endocitosis , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/fisiología , Podocitos/metabolismo , Proteínas de Transporte Vesicular/fisiología , Animales , Animales Modificados Genéticamente , Factor Natriurético Atrial/metabolismo , Forma de la Célula , Dextranos/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Silenciador del Gen , Hemolinfa/metabolismo , Ratones , Complejos Multiproteicos/genética , Podocitos/ultraestructura , Proteínas de Transporte Vesicular/genética
6.
Development ; 144(5): 866-875, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28246214

RESUMEN

miR-1 is a small noncoding RNA molecule that modulates gene expression in heart and skeletal muscle. Loss of Drosophila miR-1 produces defects in somatic muscle and embryonic heart development, which have been partly attributed to miR-1 directly targeting Delta to decrease Notch signaling. Here, we show that overexpression of miR-1 in the fly wing can paradoxically increase Notch activity independently of its effects on Delta. Analyses of potential miR-1 targets revealed that miR-1 directly regulates the 3'UTR of the E3 ubiquitin ligase Nedd4 Analysis of embryonic and adult fly heart revealed that the Nedd4 protein regulates heart development in Drosophila Larval fly hearts overexpressing miR-1 have profound defects in actin filament organization that are partially rescued by concurrent overexpression of Nedd4. These results indicate that miR-1 and Nedd4 act together in the formation and actin-dependent patterning of the fly heart. Importantly, we have found that the biochemical and genetic relationship between miR-1 and the mammalian ortholog Nedd4-like (Nedd4l) is evolutionarily conserved in the mammalian heart, potentially indicating a role for Nedd4L in mammalian postnatal maturation. Thus, miR-1-mediated regulation of Nedd4/Nedd4L expression may serve to broadly modulate the trafficking or degradation of Nedd4/Nedd4L substrates in the heart.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , MicroARNs/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Regiones no Traducidas 3' , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Tipificación del Cuerpo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Proteínas Fluorescentes Verdes/metabolismo , Corazón/fisiología , Ubiquitina-Proteína Ligasas Nedd4 , Fenotipo , Fosforilación , Transporte de Proteínas , Receptores Notch/metabolismo , Transducción de Señal , Ubiquitinación , Alas de Animales/metabolismo
7.
J Am Soc Nephrol ; 30(5): 840-853, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30910934

RESUMEN

BACKGROUND: Studies have identified mutations in >50 genes that can lead to monogenic steroid-resistant nephrotic syndrome (SRNS). The NUP160 gene, which encodes one of the protein components of the nuclear pore complex nucleoporin 160 kD (Nup160), is expressed in both human and mouse kidney cells. Knockdown of NUP160 impairs mouse podocytes in cell culture. Recently, siblings with SRNS and proteinuria in a nonconsanguineous family were found to carry compound-heterozygous mutations in NUP160. METHODS: We identified NUP160 mutations by whole-exome and Sanger sequencing of genomic DNA from a young girl with familial SRNS and FSGS who did not carry mutations in other genes known to be associated with SRNS. We performed in vivo functional validation studies on the NUP160 mutations using a Drosophila model. RESULTS: We identified two compound-heterozygous NUP160 mutations, NUP160R1173× and NUP160E803K . We showed that silencing of Drosophila NUP160 specifically in nephrocytes (fly renal cells) led to functional abnormalities, reduced cell size and nuclear volume, and disorganized nuclear membrane structure. These defects were completely rescued by expression of the wild-type human NUP160 gene in nephrocytes. By contrast, expression of the NUP160 mutant allele NUP160R1173× completely failed to rescue nephrocyte phenotypes, and mutant allele NUP160E803K rescued only nuclear pore complex and nuclear lamin localization defects. CONCLUSIONS: Mutations in NUP160 are implicated in SRNS. Our findings indicate that NUP160 should be included in the SRNS diagnostic gene panel to identify additional patients with SRNS and homozygous or compound-heterozygous NUP160 mutations and further strengthen the evidence that NUP160 mutations can cause SRNS.


Asunto(s)
Resistencia a Medicamentos , Mutación/genética , Síndrome Nefrótico/genética , Proteínas de Complejo Poro Nuclear/genética , Proteinuria/genética , Esteroides/administración & dosificación , Niño , Femenino , Predisposición Genética a la Enfermedad , Humanos , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/tratamiento farmacológico , Fenotipo , Pronóstico , Medición de Riesgo
8.
Hum Mol Genet ; 26(4): 768-780, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28164240

RESUMEN

Many genetic mutations have been identified as monogenic causes of nephrotic syndrome (NS), but important knowledge gaps exist in the roles of these genes in kidney cell biology and renal diseases. More animal models are needed to assess the functions of these genes in vivo, and to determine how they cause NS in a timely manner. Drosophila nephrocytes and human podocytes share striking similarities, but to what degree these known NS genes play conserved roles in nephrocytes remains unknown. Here we systematically studied 40 genes associated with NS, including 7 that have not previously been analysed for renal function in an animal model. We found that 85% of these genes are required for nephrocyte functions, suggesting that a majority of human genes known to be associated with NS play conserved roles in renal function from flies to humans. To investigate functional conservation in more detail, we focused on Cindr, the fly homolog of the human NS gene CD2AP. Silencing Cindr in nephrocytes led to dramatic nephrocyte functional impairment and shortened life span, as well as collapse of nephrocyte lacunar channels and effacement of nephrocyte slit diaphragms. These phenotypes could be rescued by expression of a wild-type human CD2AP gene, but not a mutant allele derived from a patient with CD2AP-associated NS. We conclude that the Drosophila nephrocyte can be used to elucidate clinically relevant molecular mechanisms underlying the pathogenesis of most monogenic forms of NS, and to efficiently generate personalized in vivo models of genetic renal diseases bearing patient-specific mutations.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas del Citoesqueleto/genética , Proteínas de Drosophila/genética , Riñón/fisiopatología , Proteínas de Microfilamentos/genética , Síndrome Nefrótico/genética , Animales , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Silenciador del Gen , Humanos , Mutación , Síndrome Nefrótico/fisiopatología , Fenotipo , Podocitos/metabolismo , Podocitos/patología
9.
J Physiol ; 596(10): 1965-1979, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29512156

RESUMEN

KEY POINTS: Sensory information processing in hippocampal circuits is critical for numerous hippocampus-dependent functions, but the underlying synaptic mechanism remains elusive. We performed whole-cell recording in vivo to examine visually evoked synaptic activity in hippocampal CA1 pyramidal cells (PCs). We first found that at resting potentials, ∼30% of CA1 PCs showed synaptic responses to a flash of visual stimulation. Interestingly, at depolarizing potentials, nearly all CA1 PCs were found to exhibit NMDA receptor-dependent responses, indicating the presence of NMDA receptor-mediated gating of CA1 responses. The NMDA receptor-gated CA1 responses may play important roles in the hippocampal function that depends on sensory information processing. ABSTRACT: Hippocampal processing of environmental information is critical for hippocampus-dependent brain functions that result from experience-induced hippocampal plasticity, such as memory acquisition and storage. Hippocampal responses to sensory stimulation have been extensively investigated, particularly with respect to spike activity. However, the synaptic mechanism for hippocampal processing of sensory stimulation has been much less understood. Here, we performed in vivo whole-cell recording on hippocampal CA1 pyramidal cells (PCs) from adult rodents to examine CA1 responses to a flash of visual stimulation. We first found in recordings obtained at resting potentials that ∼30% of CA1 PCs exhibited significant excitatory/inhibitory membrane-potential (MP) or membrane-current (MC) responses to the flash stimulus. Remarkably, in the other (∼70%) CA1 PCs, although no responses could be detected at resting potentials, clear excitatory MP or MC responses to the same flash stimulus were observed at depolarizing potentials, and these responses were further found to depend on NMDA receptors. Our findings demonstrate the presence of NMDA receptor-mediated gating of visual responses in hippocampal CA1 neurons, a synaptic mechanism for hippocampal processing of sensory information that may play important roles in hippocampus-dependent functions such as learning and memory.


Asunto(s)
Región CA1 Hipocampal/fisiología , Potenciales Postsinápticos Excitadores , Neuronas/fisiología , Células Piramidales/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Percepción Visual/fisiología , Animales , Región CA1 Hipocampal/citología , Potenciales Evocados , Femenino , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Células Piramidales/citología , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica
10.
J Am Soc Nephrol ; 28(9): 2607-2617, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28428331

RESUMEN

Clinical studies have identified patients with nephrotic syndrome caused by mutations in genes involved in the biosynthesis of coenzyme Q10 (CoQ10), a lipid component of the mitochondrial electron transport chain and an important antioxidant. However, the cellular mechanisms through which these mutations induce podocyte injury remain obscure. Here, we exploited the striking similarities between Drosophila nephrocytes and human podocytes to develop a Drosophila model of these renal diseases, and performed a systematic in vivo analysis assessing the role of CoQ10 pathway genes in renal function. Nephrocyte-specific silencing of Coq2, Coq6, and Coq8, which are genes involved in the CoQ10 pathway that have been associated with genetic nephrotic syndrome in humans, induced dramatic adverse changes in these cells. In particular, silencing of Coq2 led to an abnormal localization of slit diaphragms, collapse of lacunar channels, and more dysmorphic mitochondria. In addition, Coq2-deficient nephrocytes showed elevated levels of autophagy and mitophagy, increased levels of reactive oxygen species, and increased sensitivity to oxidative stress. Dietary supplementation with CoQ10 at least partially rescued these defects. Furthermore, expressing the wild-type human COQ2 gene specifically in nephrocytes rescued the defective protein uptake, but expressing the mutant allele derived from a patient with COQ2 nephropathy did not. We conclude that transgenic Drosophila lines carrying mutations in the CoQ10 pathway genes are clinically relevant models with which to explore the pathogenesis of podocyte injury and could serve as a new platform to test novel therapeutic approaches.


Asunto(s)
Transferasas Alquil y Aril/genética , Síndrome Nefrótico/genética , Síndrome Nefrótico/metabolismo , Ubiquinona/análogos & derivados , Vitaminas/farmacología , Transferasas Alquil y Aril/deficiencia , Alelos , Animales , Autofagia/efectos de los fármacos , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Silenciador del Gen , Humanos , Mitocondrias/ultraestructura , Mitofagia/efectos de los fármacos , Organismos Modificados Genéticamente , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética , Ubiquinona/biosíntesis , Ubiquinona/genética , Ubiquinona/farmacología , Vitaminas/biosíntesis
11.
J Am Soc Nephrol ; 28(4): 1106-1116, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27864430

RESUMEN

People of African ancestry carrying certain APOL1 mutant alleles are at elevated risk of developing renal diseases. However, the mechanisms underlying APOL1-associated renal diseases are unknown. Because the APOL1 gene is unique to humans and some primates, new animal models are needed to understand the function of APOL1 in vivo We generated transgenic Drosophila fly lines expressing the human APOL1 wild type allele (G0) or the predominant APOL1 risk allele (G1) in different tissues. Ubiquitous expression of APOL1 G0 or G1 in Drosophila induced lethal phenotypes, and G1 was more toxic than was G0. Selective expression of the APOL1 G0 or G1 transgene in nephrocytes, fly cells homologous to mammalian podocytes, induced increased endocytic activity and accumulation of hemolymph proteins, dextran particles, and silver nitrate. As transgenic flies with either allele aged, nephrocyte function declined, cell size increased, and nephrocytes died prematurely. Compared with G0-expressing cells, however, G1-expressing cells showed more dramatic phenotypes, resembling those observed in cultured mammalian podocytes overexpressing APOL1-G1. Expressing the G0 or G1 APOL1 transgene in nephrocytes also impaired the acidification of organelles. We conclude that expression of an APOL1 transgene initially enhances nephrocyte function, causing hypertrophy and subsequent cell death. This new Drosophila model uncovers a novel mechanism by which upregulated expression of APOL1-G1 could precipitate renal disease in humans. Furthermore, this model may facilitate the identification of APOL1-interacting molecules that could serve as new drug targets to treat APOL1-associated renal diseases.


Asunto(s)
Apolipoproteínas/genética , Muerte Celular/fisiología , Enfermedades Renales/genética , Riñón/patología , Lipoproteínas HDL/genética , Alelos , Animales , Animales Modificados Genéticamente , Apolipoproteína L1 , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Drosophila , Regulación de la Expresión Génica , Humanos , Hipertrofia/genética , Enfermedades Renales/patología
12.
Dev Biol ; 413(2): 188-98, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26994311

RESUMEN

The Drosophila ostia are valve-like structures in the heart with functional similarity to vertebrate cardiac valves. The Wnt/ß-catenin signaling pathway is critical for valve development in zebrafish and mouse, but the key ligand(s) for valve induction remains unclear. We observed high levels of Wnt4 gene expression in Drosophila ostia progenitor cells, immediately prior to morphological differentiation of these cells associated with ostia formation. This differentiation was blocked in Wnt4 mutants and in flies expressing canonical Wnt signaling pathway inhibitors but not inhibitors of the planar cell polarity pathway. High levels of Wnt4 dependent activation of a canonical Wnt signaling reporter was observed specifically in ostia progenitor cells. In vertebrate valve formation Wnt signaling is active in cells undergoing early endothelial-mesenchymal transition (EMT) and the Wnt9 homolog of Drosophila Wnt4 is expressed in valve progenitors. In demonstrating an essential role for Wnt4 in ostia development we have identified similarities between molecular and cellular events associated with early EMT during vertebrate valve development and the differentiation and partial delamination of ostia progenitor cells in the process of ostia formation.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/embriología , Glicoproteínas/fisiología , Proteínas Wnt/fisiología , Animales , Corazón/embriología , Morfogénesis , Transducción de Señal , Células Madre/citología
13.
Dev Biol ; 414(1): 100-7, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26994946

RESUMEN

G-protein signaling is known to be required for cell-cell contacts during the development of the Drosophila dorsal vessel. However, the identity of the G protein-coupled receptor (GPCR) that regulates this signaling pathway activity is unknown. Here we describe the identification of a novel cardiac specific GPCR, called Gia, for "GPCR in aorta". Gia is the only heart-specific GPCR identified in Drosophila to date and it is specifically expressed in cardioblasts that fuse at the dorsal midline to become the aorta. Gia is the only Drosophila gene so far identified for which expression is entirely restricted to cells of the aorta. Deletion of Gia led to a broken-hearted phenotype, characterized by pericardial cells dissociated from cardioblasts and abnormal distribution of cell junction proteins. Both phenotypes were similar to those observed in mutants of the heterotrimeric cardiac G proteins. Lack of Gia also led to defects in the alignment and fusion of cardioblasts in the aorta. Gia forms a protein complex with G-αo47A, the alpha subunit of the heterotrimeric cardiac G proteins and interacts genetically with G-αo47A during cardiac morphogenesis. Our study identified Gia as an essential aorta-specific GPCR that functions upstream of cardiac heterotrimeric G proteins and is required for morphological integrity of the aorta during heart tube formation. These studies lead to a redefinition of the bro phenotype, to encompass morphological integrity of the heart tube as well as cardioblast-pericardial cell spatial interactions.


Asunto(s)
Aorta/embriología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/embriología , Corazón/embriología , Pericardio/embriología , Receptores Acoplados a Proteínas G/fisiología , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Morfogénesis , Pericardio/citología , Fenotipo , Mapeo de Interacción de Proteínas , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusión/metabolismo
14.
Cell Tissue Res ; 368(3): 615-627, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28180992

RESUMEN

The Drosophila nephrocyte is a critical component of the fly renal system and bears structural and functional homology to podocytes and proximal tubule cells of the mammalian kidney. Investigations of nephrocyte cell biological processes are fundamental to understanding the insect renal system. Nephrocytes are highly active in endocytosis and vesicle trafficking. Rab GTPases regulate endocytosis and trafficking but specific functions of nephrocyte Rabs remain undefined. We analyzed Rab GTPase expression and function in Drosophila nephrocytes and found that 11 out of 27 Drosophila Rabs were required for normal activity. Rabs 1, 5, 7, 11 and 35 were most important. Gene silencing of the nephrocyte-specific Rab5 eliminated all intracellular vesicles and the specialized plasma membrane structures essential for nephrocyte function. Rab7 silencing dramatically increased clear vacuoles and reduced lysosomes. Rab11 silencing increased lysosomes and reduced clear vacuoles. Our results suggest that Rab5 mediates endocytosis that is essential for the maintenance of functionally critical nephrocyte plasma membrane structures and that Rabs 7 and 11 mediate alternative downstream vesicle trafficking pathways leading to protein degradation and membrane recycling, respectively. Elucidating molecular pathways underlying nephrocyte function has the potential to yield important insights into human kidney cell physiology and mechanisms of cell injury that lead to disease. The Drosophila nephrocyte is emerging as a useful in vivo model system for molecular target identification and initial testing of therapeutic approaches in humans.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/enzimología , Proteínas de Unión al GTP rab/metabolismo , Animales , Tamaño de la Célula , Vesículas Citoplasmáticas , Drosophila/citología , Drosophila/ultraestructura , Femenino , Silenciador del Gen , Riñón/citología , Riñón/enzimología , Riñón/ultraestructura , Lisosomas/enzimología , Masculino , Podocitos/enzimología , Podocitos/ultraestructura , Proteínas de Unión al GTP rab/genética
15.
J Craniofac Surg ; 28(6): 1559-1562, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28708658

RESUMEN

Due to the anatomic and functional complexity, reconstruction of the condylar process after mandibular tumor extirpation remains a surgical challenge. The aim of this study was to present the results and experience of condyle head reimplantation combined with vascularized free flap for mandibular reconstruction in our institution. In the current study, the authors evaluated the clinical features of condyle head reimplantation combined with vascularized free flap for mandibular reconstruction in 5 patients with benign mandibular intraosseous lesions from December 2013 to March 2015 in our institution. All patients showed nearly normal mouth opening, with minimal mandibular deviation and joint symptoms. The radiograph data showed that 4 patients had no obvious bone resorption of condyles while only 1 patient had prominent condyle resorption. All the condyles were cut above the condylar neck, with the biggest remaining condyle height being 2.8 cm and the smallest one being 1.1 cm. Moreover, the original vertical height of mandibular ramus was decreased by 2 to 3 mm during the surgery. In conclusion, this study revealed that combining the condyle reimplantation with vascularized free flap constitutes a reliable method in condylar reconstruction and a slight reduction of the vertical height of condyles may help to diminish unfavorable outcomes.


Asunto(s)
Colgajos Tisulares Libres/cirugía , Cóndilo Mandibular/cirugía , Reconstrucción Mandibular/métodos , Humanos , Neoplasias Mandibulares/cirugía
16.
J Cardiovasc Pharmacol ; 66(3): 261-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26348824

RESUMEN

Endothelial microparticles (EMPs) are complex vesicular structures with great significance in vascular pathophysiology. Here, we aimed to determine the impact of therapeutic drugs for infantile hemangioma, a common vascular tumor of infancy, on the biochemical features of EMPs. We exposed human umbilical vein endothelial cells to propranolol (Pro), dexamethasone (Dex), or rapamycin (Rap). Compared with controls, Pro and Rap dramatically augmented EMP release, whereas Dex significantly suppressed EMP generation. Drug-stimulated EMPs could inherit but tended to lose specific endothelial surface antigens from their parental cells. On the one hand, markedly distinct messenger RNA expression patterns were observed within and between drug-stimulated endothelial cells and derived EMPs. On the other hand, Rap-treated endothelial cells and Pro-induced EMPs displayed downregulation of multiple angiogenesis-related molecules at messenger RNA level compared with corresponding controls. Meanwhile, among tested angiogenesis-associated microRNAs, twelve microRNAs were downregulated in drug-induced EMPs, whereas only let-7b and miR-133a were markedly upregulated. Collectively, these data may indicate selective and distinctive package of biomolecules into EMPs depending on specific drugs. Our findings may provide novel insights into the underlying mechanisms of pharmacological therapy for infantile hemangioma.


Asunto(s)
Dexametasona/farmacología , Endotelio Vascular/efectos de los fármacos , Hemangioma/tratamiento farmacológico , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Propranolol/farmacología , Sirolimus/farmacología , Células Cultivadas , Endotelio Vascular/metabolismo , Endotelio Vascular/ultraestructura , Citometría de Flujo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , MicroARNs/genética , Microscopía Electrónica de Transmisión , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Factor A de Crecimiento Endotelial Vascular/genética
17.
Angew Chem Int Ed Engl ; 54(3): 1036-40, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25412570

RESUMEN

Cell-derived microparticles (MPs) have been recently recognized as critical intercellular information conveyors. However, further understanding of their biological behavior and potential application has been hampered by the limitations of current labeling techniques. Herein, a universal donor-cell-assisted membrane biotinylation strategy was proposed for labeling MPs by skillfully utilizing the natural membrane phospholipid exchange of their donor cells. This innovative strategy conveniently led to specific, efficient, reproducible, and biocompatible quantum dot (QD) labeling of MPs, thereby reliably conferring valuable traceability on MPs. By further loading with small interference RNA, QD-labeled MPs that had inherent cell-targeting and biomolecule-conveying ability were successfully employed for combined bioimaging and tumor-targeted therapy. This study provides the first reliable and biofriendly strategy for transforming biogenic MPs into functionalized nanovectors.


Asunto(s)
Antineoplásicos/química , Micropartículas Derivadas de Células/química , Puntos Cuánticos/química , ARN Interferente Pequeño/química , Animales , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis , Biotinilación , Línea Celular Tumoral , Proliferación Celular , Portadores de Fármacos/química , Fluoresceínas/química , Colorantes Fluorescentes/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Desnudos , Microscopía Fluorescente , Nanopartículas/química , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Estreptavidina/química , Succinimidas/química , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Dis Model Mech ; 17(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38602042

RESUMEN

Diabetes is a metabolic disorder characterized by high blood glucose levels and is a leading cause of kidney disease. Diabetic nephropathy has been attributed to dysfunctional mitochondria. However, many questions remain about the exact mechanism. The structure, function and molecular pathways are highly conserved between mammalian podocytes and Drosophila nephrocytes; therefore, we used flies on a high-sucrose diet to model type 2 diabetic nephropathy. The nephrocytes from flies on a high-sucrose diet showed a significant functional decline and decreased cell size, associated with a shortened lifespan. Structurally, the nephrocyte filtration structure, known as the slit diaphragm, was disorganized. At the cellular level, we found altered mitochondrial dynamics and dysfunctional mitochondria. Regulating mitochondrial dynamics by either genetic modification of the Pink1-Park (mammalian PINK1-PRKN) pathway or treatment with BGP-15, mitigated the mitochondrial defects and nephrocyte functional decline. These findings support a role for Pink1-Park-mediated mitophagy and associated control of mitochondrial dynamics in diabetic nephropathy, and demonstrate that targeting this pathway might provide therapeutic benefits for type 2 diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas , Proteínas de Drosophila , Drosophila melanogaster , Mitocondrias , Dinámicas Mitocondriales , Transducción de Señal , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/metabolismo , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Mitocondrias/metabolismo , Podocitos/patología , Podocitos/metabolismo , Mitofagia , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas/metabolismo
19.
bioRxiv ; 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38496548

RESUMEN

Background: People carrying two APOL1 risk alleles (RA) G1 or G2 are at greater risk of developing HIV-associated nephropathy (HIVAN). Studies in transgenic mice showed that the expression of HIV-1 genes in podocytes, and nef in particular, led to HIVAN. However, it remains unclear whether APOL1-RA and HIV-1 Nef interact to induce podocyte cell death. Method: We generated transgenic (Tg) flies that express APOL1-G1 (derived from a child with HIVAN) and HIV-1 nef specifically in the nephrocytes, the fly equivalent of mammalian podocytes, and assessed their individual and combined effects on the nephrocyte filtration structure and function. Results: We found that HIV-1 Nef acts in synergy with APOL1-G1 resulting in nephrocyte structural and functional defects. Specifically, HIV-1 Nef itself can induce endoplasmic reticulum (ER) stress without affecting autophagy. Furthermore, Nef exacerbates the organelle acidification defects and autophagy reduction induced by APOL1-G1. The synergy between HIV-1 Nef and APOL1-G1 is built on their joint effects on elevating ER stress, triggering nephrocyte dysfunction and ultimately cell death. Conclusions: Using a new Drosophila model of HIV-1-related kidney diseases, we identified ER stress as the converging point for the synergy between HIV-1 Nef and APOL1-G1 in inducing nephrocyte cell death. Given the high relevance between Drosophila nephrocytes and human podocytes, this finding suggests ER stress as a new therapeutic target for HIV-1 and APOL1-associated nephropathies.

20.
J Cardiovasc Dev Dis ; 10(7)2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37504561

RESUMEN

Epigenetic marks regulate the transcriptomic landscape by facilitating the structural packing and unwinding of the genome, which is tightly folded inside the nucleus. Lysine-specific histone methylation is one such mark. It plays crucial roles during development, including in cell fate decisions, in tissue patterning, and in regulating cellular metabolic processes. It has also been associated with varying human developmental disorders. Heart disease has been linked to deregulated histone lysine methylation, and lysine-specific methyltransferases (KMTs) are overrepresented, i.e., more numerous than expected by chance, among the genes with variants associated with congenital heart disease. This review outlines the available evidence to support a role for individual KMTs in heart development and/or disease, including genetic associations in patients and supporting cell culture and animal model studies. It concludes with new advances in the field and new opportunities for treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA