Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.127
Filtrar
1.
Cell Mol Life Sci ; 81(1): 213, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727814

RESUMEN

Trimeric G proteins transduce signals from a superfamily of receptors and each G protein controls a wide range of cellular and systemic functions. Their highly conserved alpha subunits fall in five classes, four of which have been well investigated (Gs, Gi, G12, Gq). In contrast, the function of the fifth class, Gv is completely unknown, despite its broad occurrence and evolutionary ancient origin (older than metazoans). Here we show a dynamic presence of Gv mRNA in several organs during early development of zebrafish, including the hatching gland, the pronephros and several cartilage anlagen, employing in situ hybridisation. Next, we generated a Gv frameshift mutation in zebrafish and observed distinct phenotypes such as reduced oviposition, premature hatching and craniofacial abnormalities in bone and cartilage of larval zebrafish. These phenotypes could suggest a disturbance in ionic homeostasis as a common denominator. Indeed, we find reduced levels of calcium, magnesium and potassium in the larvae and changes in expression levels of the sodium potassium pump atp1a1a.5 and the sodium/calcium exchanger ncx1b in larvae and in the adult kidney, a major osmoregulatory organ. Additionally, expression of sodium chloride cotransporter slc12a3 and the anion exchanger slc26a4 is altered in complementary ways in adult kidney. It appears that Gv may modulate ionic homeostasis in zebrafish during development and in adults. Our results constitute the first insight into the function of the fifth class of G alpha proteins.


Asunto(s)
Homeostasis , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/metabolismo , Homeostasis/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/genética , Larva/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , Calcio/metabolismo , Riñón/metabolismo , Magnesio/metabolismo
2.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38731894

RESUMEN

Triptolide is a natural compound in herbal remedies with anti-inflammatory and anti-proliferative properties. We studied its effects on critical signaling processes within the cell, including Notch1 and STAT3 signaling. Our research showed that triptolide reduces cancer cell proliferation by decreasing the expression of downstream targets of these signals. The levels of each signal-related protein and mRNA were analyzed using Western blot and qPCR methods. Interestingly, inhibiting one signal with a single inhibitor alone did not significantly reduce cancer cell proliferation. Instead, MTT assays showed that the simultaneous inhibition of Notch1 and STAT3 signaling reduced cell proliferation. The effect of triptolide was similar to a combination treatment with inhibitors for both signals. When we conducted a study on the impact of triptolide on zebrafish larvae, we found that it inhibited muscle development and interfered with muscle cell proliferation, as evidenced by differences in the staining of myosin heavy chain and F-actin proteins in confocal fluorescence microscopy. Additionally, we noticed that inhibiting a single type of signaling did not lead to any significant muscle defects. This implies that triptolide obstructs multiple signals simultaneously, including Notch1 and STAT3, during muscle development. Chemotherapy is commonly used to treat cancer, but it may cause muscle loss due to drug-related adverse reactions or other complex mechanisms. Our study suggests that anticancer agents like triptolide, inhibiting essential signaling pathways including Notch1 and STAT3 signaling, may cause muscle atrophy through anti-proliferative activity.


Asunto(s)
Proliferación Celular , Diterpenos , Compuestos Epoxi , Fenantrenos , Receptor Notch1 , Factor de Transcripción STAT3 , Transducción de Señal , Pez Cebra , Animales , Compuestos Epoxi/farmacología , Fenantrenos/farmacología , Diterpenos/farmacología , Factor de Transcripción STAT3/metabolismo , Proliferación Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Humanos , Receptor Notch1/metabolismo , Receptor Notch1/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Línea Celular Tumoral , Receptores Notch/metabolismo
4.
Mol Biol Rep ; 51(1): 604, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38700644

RESUMEN

BACKGROUND: The healing process after a myocardial infarction (MI) in humans involves complex events that replace damaged tissue with a fibrotic scar. The affected cardiac tissue may lose its function permanently. In contrast, zebrafish display a remarkable capacity for scar-free heart regeneration. Previous studies have revealed that syndecan-4 (SDC4) regulates inflammatory response and fibroblast activity following cardiac injury in higher vertebrates. However, whether and how Sdc4 regulates heart regeneration in highly regenerative zebrafish remains unknown. METHODS AND RESULTS: This study showed that sdc4 expression was differentially regulated during zebrafish heart regeneration by transcriptional analysis. Specifically, sdc4 expression increased rapidly and transiently in the early regeneration phase upon ventricular cryoinjury. Moreover, the knockdown of sdc4 led to a significant reduction in extracellular matrix protein deposition, immune cell accumulation, and cell proliferation at the lesion site. The expression of tgfb1a and col1a1a, as well as the protein expression of Fibronectin, were all down-regulated under sdc4 knockdown. In addition, we verified that sdc4 expression was required for cardiac repair in zebrafish via in vivo electrocardiogram analysis. Loss of sdc4 expression caused an apparent pathological Q wave and ST elevation, which are signs of human MI patients. CONCLUSIONS: Our findings support that Sdc4 is required to mediate pleiotropic repair responses in the early stage of zebrafish heart regeneration.


Asunto(s)
Corazón , Regeneración , Sindecano-4 , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/metabolismo , Sindecano-4/genética , Sindecano-4/metabolismo , Regeneración/genética , Corazón/fisiología , Corazón/fisiopatología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Proliferación Celular/genética , Miocardio/metabolismo , Miocardio/patología , Técnicas de Silenciamiento del Gen
5.
Anat Histol Embryol ; 53(3): e13044, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38695121

RESUMEN

The vitamin D receptor (VDR) signalling has been implicated in vertebrate limb or fin formation. However, the involvement of VDR signalling in the early stages of limb/fin development remains to be elucidated. In this study, the role of VDR signalling in pectoral fin development was investigated in zebrafish embryos. Knockdown of vdr induced the severe impairment of pectoral fin development. The zebrafish larvae lacking vdr exhibited reduced pectoral fins with no skeletal elements. In situ hybridization revealed depletion of vdr downregulated fibroblast growth factor 24 (fgf24), a marker of early pectoral fin bud mesenchyme, in the presumptive fin field even before fin buds were visible. Moreover, a perturbed expression pattern of bone morphogenetic protein 4 (bmp4), a marker of the pectoral fin fold, was observed in the developing fin buds of zebrafish embryos that lost the vdr function. These findings suggest that VDR signalling is crucial in the early stages of fin development, potentially influencing the process by regulating other signalling molecules such as Fgf24 and Bmp4.


Asunto(s)
Aletas de Animales , Proteína Morfogenética Ósea 4 , Factores de Crecimiento de Fibroblastos , Receptores de Calcitriol , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/embriología , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Aletas de Animales/embriología , Aletas de Animales/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/genética , Técnicas de Silenciamiento del Gen , Transducción de Señal , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ
6.
Science ; 384(6695): 573-579, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38696577

RESUMEN

Neurons on the left and right sides of the nervous system often show asymmetric properties, but how such differences arise is poorly understood. Genetic screening in zebrafish revealed that loss of function of the transmembrane protein Cachd1 resulted in right-sided habenula neurons adopting left-sided identity. Cachd1 is expressed in neuronal progenitors, functions downstream of asymmetric environmental signals, and influences timing of the normally asymmetric patterns of neurogenesis. Biochemical and structural analyses demonstrated that Cachd1 can bind simultaneously to Lrp6 and Frizzled family Wnt co-receptors. Consistent with this, lrp6 mutant zebrafish lose asymmetry in the habenulae, and epistasis experiments support a role for Cachd1 in modulating Wnt pathway activity in the brain. These studies identify Cachd1 as a conserved Wnt receptor-interacting protein that regulates lateralized neuronal identity in the zebrafish brain.


Asunto(s)
Habénula , Neurogénesis , Neuronas , Vía de Señalización Wnt , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Habénula/metabolismo , Habénula/embriología , Neuronas/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Receptores Frizzled/metabolismo , Receptores Frizzled/genética , Receptores Wnt/metabolismo , Receptores Wnt/genética , Encéfalo/metabolismo , Mutación con Pérdida de Función , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética
7.
Development ; 151(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38722096

RESUMEN

During embryonic development, lymphatic endothelial cell (LEC) precursors are distinguished from blood endothelial cells by the expression of Prospero-related homeobox 1 (Prox1), which is essential for lymphatic vasculature formation in mouse and zebrafish. Prox1 expression initiation precedes LEC sprouting and migration, serving as the marker of specified LECs. Despite its crucial role in lymphatic development, Prox1 upstream regulation in LECs remains to be uncovered. SOX18 and COUP-TFII are thought to regulate Prox1 in mice by binding its promoter region. However, the specific regulation of Prox1 expression in LECs remains to be studied in detail. Here, we used evolutionary conservation and chromatin accessibility to identify enhancers located in the proximity of zebrafish prox1a active in developing LECs. We confirmed the functional role of the identified sequences through CRISPR/Cas9 mutagenesis of a lymphatic valve enhancer. The deletion of this region results in impaired valve morphology and function. Overall, our results reveal an intricate control of prox1a expression through a collection of enhancers. Ray-finned fish-specific distal enhancers drive pan-lymphatic expression, whereas vertebrate-conserved proximal enhancers refine expression in functionally distinct subsets of lymphatic endothelium.


Asunto(s)
Células Endoteliales , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Vasos Linfáticos , Proteínas Supresoras de Tumor , Proteínas de Pez Cebra , Pez Cebra , Animales , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Pez Cebra/genética , Pez Cebra/embriología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Elementos de Facilitación Genéticos/genética , Vasos Linfáticos/metabolismo , Vasos Linfáticos/embriología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Células Endoteliales/metabolismo , Linfangiogénesis/genética , Sistemas CRISPR-Cas/genética , Regiones Promotoras Genéticas/genética , Ratones
8.
Mol Vis ; 30: 123-136, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38601019

RESUMEN

Purpose: Danio rerio zebrafish constitute a popular model for studying lens development and congenital cataracts. However, the specific deletion of a gene with a Cre/LoxP system in the zebrafish lens is unavailable because of the lack of a lens-Cre-transgenic zebrafish. This study aimed to generate a transgenic zebrafish line in which Cre recombinase was specifically expressed in the lens. Methods: The pTol2 cryaa:Cre-polyA-cryaa:EGFP (enhanced green fluorescent protein) plasmid was constructed and co-injected with Tol2-transposase into one-to-two-cell-stage wild-type (WT) zebrafish embryos. Whole-mount in situ hybridization (ISH), tissue section, hematoxylin and eosin staining, a Western blot, a split-lamp observation, and a grid transmission assay were used to analyze the Cre expression, lens structure, and lens transparency of the transgenic zebrafish. Results: In this study, we generated a transgenic zebrafish line, zTg(cryaa:Cre-cryaa:EGFP), in which Cre recombinase and EGFP were driven by the lens-specific cryaa promoter. zTg(cryaa:Cre-cryaa:EGFP) began to express Cre and EGFP specifically in the lens at the 22 hpf stage, and this ectopic Cre could efficiently and specifically delete the red fluorescent protein (RFP) signal from the lens when zTg(cryaa:Cre-cryaa:EGFP) embryos were injected with the loxP-flanked RFP plasmid. The overexpression of Cre and EGFP did not impair zebrafish development or lens transparency. Accordingly, this zTg(cryaa:Cre-cryaa:EGFP) zebrafish line is a useful tool for gene editing, specifically with zebrafish lenses. Conclusions: We established a zTg(cryaa:Cre-cryaa:EGFP) zebrafish line that can specifically express an active Cre recombinase in lens tissues. This transgenic zebrafish line can be used as a tool to specifically manipulate a gene in zebrafish lenses.


Asunto(s)
Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/metabolismo , Animales Modificados Genéticamente/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/genética , Plásmidos , Regiones Promotoras Genéticas
9.
Zebrafish ; 21(2): 149-154, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38621206

RESUMEN

Rising in popularity as a model organism in the classroom, zebrafish have numerous characteristics that make them ideal for teaching. In this study, we describe an experiment that helps students better understand the concept of tissue regeneration and the genes that control it. This experiment utilizes a dominant negative transgene for fgfr1 and allows students to observe the consequences of its activation. The first part of the laboratory is hands-on, and includes details of the amputation of caudal fins, heat shocking, general fish care, and visual observations. Over the course of a week, students observed the differences between the activated and unactivated transgene in the zebrafish. The second part was literature based, in which students tried to determine which gene is responsible for inhibiting regeneration. This encouraged students to sharpen their skills of deductive reasoning and critical thinking as they conduct research based on the information they receive about dominant negative receptors and transgenes. Having both a hands-on and critical thinking component in the laboratory helped synthesize the learning goals and allowed students to actively participate.


Asunto(s)
Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética , Cola (estructura animal)/fisiología , Aletas de Animales/fisiología
10.
Zebrafish ; 21(2): 128-136, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38621212

RESUMEN

Coordinated signaling pathway activity directs early patterning to set up the vertebrate body plan. Perturbations in the timing or location of signal molecule expression impacts embryo morphology and organ formation. In this study, we present a laboratory course to use zebrafish for studying the role of Wnt signaling in specifying the early embryonic axes. Students are exposed to basic techniques in molecular and developmental biology, including embryo manipulation, fluorescence microscopy, image processing, and data analysis. Furthermore, this course incorporates student-designed experiments to stimulate independent inquiry and improve scientific learning, providing an experience resembling graduate-level laboratory research. Students appreciated following vertebrate development in real-time, and principles of embryogenesis were reinforced by observing the morphological changes that arise due to signaling alterations. Scientific and research skills were enhanced through practice in experimental design, interpretation, and presentation.


Asunto(s)
Vía de Señalización Wnt , Pez Cebra , Humanos , Animales , Pez Cebra/genética , Tipificación del Cuerpo , Desarrollo Embrionario , Proteínas de Pez Cebra/metabolismo , Regulación del Desarrollo de la Expresión Génica , Embrión no Mamífero/metabolismo
11.
Cells ; 13(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38667299

RESUMEN

It has been known for a long time that epileptic seizures provoke brain neuroinflammation involving the activation of microglial cells. However, the role of these cells in this disease context and the consequences of their inflammatory activation on subsequent neuron network activity remain poorly understood so far. To fill this gap of knowledge and gain a better understanding of the role of microglia in the pathophysiology of epilepsy, we used an established zebrafish Dravet syndrome epilepsy model based on Scn1Lab sodium channel loss-of-function, combined with live microglia and neuronal Ca2+ imaging, local field potential (LFP) recording, and genetic microglia ablation. Data showed that microglial cells in scn1Lab-deficient larvae experiencing epileptiform seizures displayed morphological and biochemical changes characteristic of M1-like pro-inflammatory activation; i.e., reduced branching, amoeboid-like morphology, and marked increase in the number of microglia expressing pro-inflammatory cytokine Il1ß. More importantly, LFP recording, Ca2+ imaging, and swimming behavior analysis showed that microglia-depleted scn1Lab-KD larvae displayed an increase in epileptiform seizure-like neuron activation when compared to that seen in scn1Lab-KD individuals with microglia. These findings strongly suggest that despite microglia activation and the synthesis of pro-inflammatory cytokines, these cells provide neuroprotective activities to epileptic neuronal networks, making these cells a promising therapeutic target in epilepsy.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsias Mioclónicas , Microglía , Neuronas , Pez Cebra , Animales , Microglía/metabolismo , Microglía/patología , Epilepsias Mioclónicas/patología , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/metabolismo , Epilepsias Mioclónicas/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Interleucina-1beta/metabolismo , Larva , Calcio/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
12.
Cell Rep ; 43(4): 114092, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38607913

RESUMEN

Macrophages conduct critical roles in heart repair, but the niche required to nurture and anchor them is poorly studied. Here, we investigated the macrophage niche in the regenerating heart. We analyzed cell-cell interactions through published single-cell RNA sequencing datasets and identified a strong interaction between fibroblast/epicardial (Fb/Epi) cells and macrophages. We further visualized the association of macrophages with Fb/Epi cells and the blockage of macrophage response without Fb/Epi cells in the regenerating zebrafish heart. Moreover, we found that ptx3a+ epicardial cells associate with reparative macrophages, and their depletion resulted in fewer reparative macrophages. Further, we identified csf1a expression in ptx3a+ cells and determined that pharmacological inhibition of the csf1a pathway or csf1a knockout blocked the reparative macrophage response. Moreover, we found that genetic overexpression of csf1a enhanced the reparative macrophage response with or without heart injury. Altogether, our studies illuminate a cardiac Fb/Epi niche, which mediates a beneficial macrophage response after heart injury.


Asunto(s)
Fibroblastos , Corazón , Macrófagos , Regeneración , Pez Cebra , Animales , Proteína C-Reactiva/metabolismo , Proteína C-Reactiva/genética , Fibroblastos/metabolismo , Corazón/fisiología , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/patología , Macrófagos/metabolismo , Pericardio/metabolismo , Pericardio/citología , Regeneración/fisiología , Componente Amiloide P Sérico/metabolismo , Componente Amiloide P Sérico/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
13.
Life Sci Space Res (Amst) ; 41: 127-135, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38670639

RESUMEN

Understanding how skeletal tissues respond to microgravity is ever more important with the increased interest in human space travel. Here, we exposed larval Danio rerio at 3.5 dpf to simulated microgravity (SMG) using a 3D mode of rotation in a ground-based experiment and then studied different cellular, molecular, and morphological bone responses both immediately after exposure and one week later. Our results indicate an overall decrease in ossification in several developing skeletal elements immediately after SMG exposure with the exception of the otoliths, however ossification returns to normal levels seven days after exposure. Coincident with the reduction in overall ossification tnfsf11 (RANKL) expression is highly elevated after 24 h of SMG exposure and also returns to normal levels seven days after exposure. We also show that genes associated with osteoblasts are unaffected immediately after SMG exposure. Thus, the observed reduction in ossification is primarily the result of a high level of bone resorption. This study sheds insight into the nuances of how osteoblasts and osteoclasts in the skeleton of a vertebrate organism respond to an external environmental disturbance, in this case simulated microgravity.


Asunto(s)
Larva , Osteogénesis , Simulación de Ingravidez , Pez Cebra , Animales , Larva/crecimiento & desarrollo , Larva/fisiología , Osteoblastos/fisiología , Osteoclastos/fisiología , Ligando RANK/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Ingravidez/efectos adversos
14.
Proc Natl Acad Sci U S A ; 121(17): e2314201121, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38635631

RESUMEN

To effectively protect the host from viral infection while avoiding excessive immunopathology, the innate immune response must be tightly controlled. However, the precise regulation of antiviral innate immunity and the underlying mechanisms remain unclear. Here, we find that sirtuin3 (SIRT3) interacts with mitochondrial antiviral signaling protein (MAVS) to catalyze MAVS deacetylation at lysine residue 7 (K7), which promotes MAVS aggregation, as well as TANK-binding kinase I and IRF3 phosphorylation, resulting in increased MAVS activation and enhanced type I interferon signaling. Consistent with these findings, loss of Sirt3 in mice and zebrafish renders them more susceptible to viral infection compared to their wild-type (WT) siblings. However, Sirt3 and Sirt5 double-deficient mice exhibit the same viral susceptibility as their WT littermates, suggesting that loss of Sirt5 in Sirt3-deficient mice may counteract the increased viral susceptibility displayed in Sirt3-deficient mice. Thus, we not only demonstrate that SIRT3 positively regulates antiviral immunity in vitro and in vivo, likely via MAVS, but also uncover a previously unrecognized mechanism by which SIRT3 acts as an accelerator and SIRT5 as a brake to orchestrate antiviral innate immunity.


Asunto(s)
Sirtuina 3 , Sirtuinas , Virosis , Animales , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Inmunidad Innata , Lisina , Sirtuina 3/genética , Sirtuinas/genética , Pez Cebra , Proteínas de Pez Cebra
15.
PeerJ ; 12: e17234, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38666079

RESUMEN

Background: Post-translational modification by Small Ubiquitin-like MOdifier (SUMO) is an important mechanism to regulate protein activity, protein stability, and localization of substrates. Zbtb21 is a zinc finger and BTB (Broad-complex, Tram-track and Bric à brac) domain-containing transcription factor. Bioinformatic prediction suggests several putative SUMOylated sites in Zbtb21 protein. Methods: Two evolutionarily conserved lysine residues in Zbtb21 protein were mutated alone or in combination to disrupt the binding with SUMO molecules. Western blot and co-immunoprecipitation analyses were performed to detect the SUMOylation state of wild type and mutant Zbtb21 proteins, respectively. Luciferase reporter assays were conducted to evaluate their transcription activities. Meanwhile, immunofluorescence staining was carried out to show their sub-nuclear localizations. Finally, co-immunoprecipitation was performed to detect the interaction between Zbtb21 and its partners. Results: Phylogenetically conserved lysines 419 and 845 of zebrafish Zbtb21 protein can be conjugated with SUMO molecules. SUMOylation does not affect the subcellular localization and protein stability of Zbtb21, as well as the interaction with Zbtb14 or Zbtb21. Nevertheless, luciferase reporter assays revealed that Zbtb21 is a dual-function transcription factor which exerts activation or repression effect on different promoters, and SUMOylation can modulate the transcriptional activity of Zbtb21 in regulating downstream target genes. Hence, Zbtb21 is identified as a novel substrate of SUMOylation, which would be important for its function. Conclusions: Zebrafish Zbtb21 protein can be SUMOylated on lysines 419 and 845, which is evolutionary conserved. SUMOylation affects the dual role of Zbtb21 on transcription.


Asunto(s)
Sumoilación , Proteínas de Pez Cebra , Pez Cebra , Sumoilación/genética , Animales , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Transcripción Genética/genética , Humanos
16.
PLoS Genet ; 20(4): e1011139, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38669217

RESUMEN

As essential components of gene expression networks, transcription factors regulate neural circuit assembly. The homeobox transcription factor encoding gene, gs homeobox 1 (gsx1), is expressed in the developing visual system; however, no studies have examined its role in visual system formation. In zebrafish, retinal ganglion cell (RGC) axons that transmit visual information to the brain terminate in ten arborization fields (AFs) in the optic tectum (TeO), pretectum (Pr), and thalamus. Pretectal AFs (AF1-AF9) mediate distinct visual behaviors, yet we understand less about their development compared to AF10 in the TeO. Using gsx1 zebrafish mutants, immunohistochemistry, and transgenic lines, we observed that gsx1 is required for vesicular glutamate transporter, Tg(slc17a6b:DsRed), expression in the Pr, but not overall neuron number. gsx1 mutants have normal eye morphology, yet they exhibit impaired visual ability during prey capture. RGC axon volume in the gsx1 mutant Pr and TeO is reduced, and AF7 that is active during feeding is missing which is consistent with reduced hunting performance. Timed laser ablation of Tg(slc17a6b:DsRed)-positive cells reveals that they are necessary for AF7 formation. This work is the first to implicate gsx1 in establishing cell identity and functional neural circuits in the visual system.


Asunto(s)
Animales Modificados Genéticamente , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Células Ganglionares de la Retina , Proteínas de Pez Cebra , Pez Cebra , Animales , Axones/metabolismo , Axones/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Mutación , Células Ganglionares de la Retina/metabolismo , Colículos Superiores/metabolismo , Colículos Superiores/crecimiento & desarrollo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vías Visuales/crecimiento & desarrollo , Vías Visuales/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
17.
Genes (Basel) ; 15(4)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38674426

RESUMEN

Haploinsufficiency of the PRR12 gene is implicated in a human neuro-ocular syndrome. Although identified as a nuclear protein highly expressed in the embryonic mouse brain, PRR12 molecular function remains elusive. This study explores the spatio-temporal expression of zebrafish PRR12 co-orthologs, prr12a and prr12b, as a first step to elucidate their function. In silico analysis reveals high evolutionary conservation in the DNA-interacting domains for both orthologs, with significant syntenic conservation observed for the prr12b locus. In situ hybridization and RT-qPCR analyses on zebrafish embryos and larvae reveal distinct expression patterns: prr12a is expressed early in zygotic development, mainly in the central nervous system, while prr12b expression initiates during gastrulation, localizing later to dopaminergic telencephalic and diencephalic cell clusters. Both transcripts are enriched in the ganglion cell and inner neural layers of the 72 hpf retina, with prr12b widely distributed in the ciliary marginal zone. In the adult brain, prr12a and prr12b are found in the cerebellum, amygdala and ventral telencephalon, which represent the main areas affected in autistic patients. Overall, this study suggests PRR12's potential involvement in eye and brain development, laying the groundwork for further investigations into PRR12-related neurobehavioral disorders.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Retina/metabolismo , Retina/crecimiento & desarrollo , Humanos
18.
Yi Chuan ; 46(4): 319-332, 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38632094

RESUMEN

Granulopoiesis is a highly ordered and precisely regulated process in which hematopoietic-related transcription factors play crucial roles. These transcription factors form complex regulatory networks through interactions with their co-factors or with each other, and anomalies in these networks can lead to the onset of leukemia. While the structures and functions of dozens of transcription factors involved in this process have been extensively studied, research on the regulatory relationships between these factors remains relatively limited. PU.1 and cMYB participate in multiple stages of neutrophil development, and their abnormalities are often associated with hematologic disorders. However, the regulatory relationship between these factors in vivo and their mode of interaction remain unclear. In this study, zebrafish models with cMyb overexpression (cmybhyper) and Pu.1 deficiency (pu.1G242D/G242D) were utilized to systematically investigate the interaction between Pu.1 and cMyb during granulopoiesis through whole-mount in situ hybridization, qRT-PCR, fluorescence reporting systems, and rescue experiments. The results showed a significant increase in cmyb expression in neutrophils of the pu.1G242D/G242D mutant, while there was no apparent change in pu.1 expression in cmybhyper. Further experiments involving injection of morpholino (MO) to decrease cmyb expression in pu.1G242D/G242D mutants, followed by SB and BrdU staining to assess neutrophil quantity and proliferation, revealed that reducing cmyb expression could rescue the abnormal proliferation phenotype of neutrophils in the pu.1G242D/G242D mutant. These findings suggest that Pu.1 negatively regulates the expression of cMyb during neutrophil development. Finally, through the construction of multi-site mutation plasmids and a fluorescent reporter system, confirmed that Pu.1 directly binds to the +72 bp site in the cmyb promoter, exerting negative regulation on its expression. In conclusion, this study delineates that Pu.1 participates in neutrophil development by regulating cmyb expression. This provides new insights into the regulatory relationship between these two factors and their roles in diseases.


Asunto(s)
Neutrófilos , Proteínas Proto-Oncogénicas c-myb , Transactivadores , Pez Cebra , Animales , Hematopoyesis , Neutrófilos/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Pez Cebra/genética , Proteínas Proto-Oncogénicas c-myb/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
19.
Sci Adv ; 10(14): eadj9637, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38578990

RESUMEN

Animals evolve diverse pigment patterns to adapt to the natural environment. Countershading, characterized by a dark-colored dorsum and a light-colored ventrum, is one of the most prevalent pigment patterns observed in vertebrates. In this study, we reveal a mechanism regulating xanthophore countershading in zebrafish embryos. We found that csf1a and csf1b mutants altered xanthophore countershading differently: csf1a mutants lack ventral xanthophores, while csf1b mutants have reduced dorsal xanthophores. Further study revealed that csf1a is expressed throughout the trunk, whereas csf1b is expressed dorsally. Ectopic expression of csf1a or csf1b in neurons attracted xanthophores into the spinal cord. Blocking csf1 signaling by csf1ra mutants disrupts spinal cord distribution and normal xanthophores countershading. Single-cell RNA sequencing identified two col1a2+ populations: csf1ahighcsf1bhigh muscle progenitors and csf1ahighcsf1blow fibroblast progenitors. Ablation of col1a2+ fibroblast and muscle progenitors abolished xanthophore patterns. Our study suggests that fibroblast and muscle progenitors differentially express csf1a and csf1b to modulate xanthophore patterning, providing insights into the mechanism of countershading.


Asunto(s)
Pigmentación , Pez Cebra , Animales , Pez Cebra/metabolismo , Pigmentación/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Músculos
20.
Sci Signal ; 17(832): eadf4299, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38626007

RESUMEN

Cell-to-cell communication through secreted Wnt ligands that bind to members of the Frizzled (Fzd) family of transmembrane receptors is critical for development and homeostasis. Wnt9a signals through Fzd9b, the co-receptor LRP5 or LRP6 (LRP5/6), and the epidermal growth factor receptor (EGFR) to promote early proliferation of zebrafish and human hematopoietic stem cells during development. Here, we developed fluorescently labeled, biologically active Wnt9a and Fzd9b fusion proteins to demonstrate that EGFR-dependent endocytosis of the ligand-receptor complex was required for signaling. In human cells, the Wnt9a-Fzd9b complex was rapidly endocytosed and trafficked through early and late endosomes, lysosomes, and the endoplasmic reticulum. Using small-molecule inhibitors and genetic and knockdown approaches, we found that Wnt9a-Fzd9b endocytosis required EGFR-mediated phosphorylation of the Fzd9b tail, caveolin, and the scaffolding protein EGFR protein substrate 15 (EPS15). LRP5/6 and the downstream signaling component AXIN were required for Wnt9a-Fzd9b signaling but not for endocytosis. Knockdown or loss of EPS15 impaired hematopoietic stem cell development in zebrafish. Other Wnt ligands do not require endocytosis for signaling activity, implying that specific modes of endocytosis and trafficking may represent a method by which Wnt-Fzd specificity is established.


Asunto(s)
Pez Cebra , beta Catenina , Animales , Humanos , beta Catenina/metabolismo , Endocitosis , Receptores ErbB/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA