Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Dis Model Mech ; 17(5)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38736327

RESUMO

Heterotopic ossification is the inappropriate formation of bone in soft tissues of the body. It can manifest spontaneously in rare genetic conditions or as a response to injury, known as acquired heterotopic ossification. There are several experimental models for studying acquired heterotopic ossification from different sources of damage. However, their tenuous mechanistic relevance to the human condition, invasive and laborious nature and/or lack of amenability to chemical and genetic screens, limit their utility. To address these limitations, we developed a simple zebrafish injury model that manifests heterotopic ossification with high penetrance in response to clinically emulating injuries, as observed in human myositis ossificans traumatica. Using this model, we defined the transcriptional response to trauma, identifying differentially regulated genes. Mutant analyses revealed that an increase in the activity of the potassium channel Kcnk5b potentiates injury response, whereas loss of function of the interleukin 11 receptor paralogue (Il11ra) resulted in a drastically reduced ossification response. Based on these findings, we postulate that enhanced ionic signalling, specifically through Kcnk5b, regulates the intensity of the skeletogenic injury response, which, in part, requires immune response regulated by Il11ra.


Assuntos
Ossificação Heterotópica , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Peixe-Zebra/genética , Ossificação Heterotópica/genética , Ossificação Heterotópica/patologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Regulação da Expressão Gênica , Envelhecimento/genética , Envelhecimento/patologia , Ferimentos e Lesões/complicações , Ferimentos e Lesões/genética , Ferimentos e Lesões/patologia , Modelos Animais de Doenças , Mutação/genética
2.
Nature ; 618(7965): 543-549, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37225983

RESUMO

The development of paired appendages was a key innovation during evolution and facilitated the aquatic to terrestrial transition of vertebrates. Largely derived from the lateral plate mesoderm (LPM), one hypothesis for the evolution of paired fins invokes derivation from unpaired median fins via a pair of lateral fin folds located between pectoral and pelvic fin territories1. Whilst unpaired and paired fins exhibit similar structural and molecular characteristics, no definitive evidence exists for paired lateral fin folds in larvae or adults of any extant or extinct species. As unpaired fin core components are regarded as exclusively derived from paraxial mesoderm, any transition presumes both co-option of a fin developmental programme to the LPM and bilateral duplication2. Here, we identify that the larval zebrafish unpaired pre-anal fin fold (PAFF) is derived from the LPM and thus may represent a developmental intermediate between median and paired fins. We trace the contribution of LPM to the PAFF in both cyclostomes and gnathostomes, supporting the notion that this is an ancient trait of vertebrates. Finally, we observe that the PAFF can be bifurcated by increasing bone morphogenetic protein signalling, generating LPM-derived paired fin folds. Our work provides evidence that lateral fin folds may have existed as embryonic anlage for elaboration to paired fins.


Assuntos
Nadadeiras de Animais , Evolução Biológica , Mesoderma , Peixe-Zebra , Animais , Nadadeiras de Animais/anatomia & histologia , Nadadeiras de Animais/embriologia , Nadadeiras de Animais/crescimento & desenvolvimento , Larva/anatomia & histologia , Larva/crescimento & desenvolvimento , Mesoderma/anatomia & histologia , Mesoderma/embriologia , Mesoderma/crescimento & desenvolvimento , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/embriologia , Peixe-Zebra/crescimento & desenvolvimento , Proteínas Morfogenéticas Ósseas/metabolismo
3.
Wound Repair Regen ; 30(6): 665-680, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36148505

RESUMO

The underlying mechanisms of appendage regeneration remain largely unknown and uncovering these mechanisms in capable organisms has far-reaching implications for potential treatments in humans. Recent studies implicate a requirement for metabolic reprogramming reminiscent of the Warburg effect during successful appendage and organ regeneration. As changes are thus predicted to be highly dynamic, methods permitting direct, real-time visualisation of metabolites at the tissue and organismal level would offer a significant advance in defining the influence of metabolism on regeneration and healing. We sought to examine whether glycolytic activity was altered during larval fin regeneration, utilising the genetically encoded biosensor, Laconic, enabling the spatiotemporal assessment of lactate levels in living zebrafish. We present evidence for a rapid increase in lactate levels within min following injury, with a role of aerobic glycolysis in actomyosin contraction and wound closure. We also find a second wave of lactate production, associated with overall larval tail regeneration. Chemical inhibition of glycolysis attenuates both the contraction of the wound and regrowth of tissue following tail amputation, suggesting aerobic glycolysis is necessary at two distinct stages of regeneration.


Assuntos
Cicatrização , Peixe-Zebra , Animais , Humanos , Peixe-Zebra/fisiologia , Larva , Cicatrização/fisiologia , Regeneração/fisiologia , Glicólise , Lactatos/farmacologia
4.
EMBO Rep ; 23(8): e54464, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35679135

RESUMO

Immigration of mesenchymal cells into the growing fin and limb buds drives distal outgrowth, with subsequent tensile forces between these cells essential for fin and limb morphogenesis. Morphogens derived from the apical domain of the fin, orientate limb mesenchyme cell polarity, migration, division and adhesion. The zebrafish mutant stomp displays defects in fin morphogenesis including blister formation and associated loss of orientation and adhesion of immigrating fin mesenchyme cells. Positional cloning of stomp identifies a mutation in the gene encoding the axon guidance ligand, Slit3. We provide evidence that Slit ligands derived from immigrating mesenchyme act via Robo receptors at the apical ectodermal ridge (AER) to promote release of sphingosine-1-phosphate (S1P). S1P subsequently diffuses back to the mesenchyme to promote their polarisation, orientation, positioning and adhesion to the interstitial matrix of the fin fold. We thus demonstrate the coordination of the Slit-Robo and S1P signalling pathways in fin fold morphogenesis. Our work introduces a mechanism regulating the orientation, positioning and adhesion of its constituent cells.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Peixe-Zebra , Animais , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lisofosfolipídeos , Mesoderma/metabolismo , Esfingosina/análogos & derivados , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
5.
J Biomed Opt ; 26(11)2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34773396

RESUMO

SIGNIFICANCE: Real-time monitoring of the heart rate and blood flow is crucial for studying cardiovascular dysfunction, which leads to cardiovascular diseases. AIM: This study aims at in-depth understanding of high-speed cardiovascular dynamics in a zebrafish embryo model for various biomedical applications via frequency-comb-referenced quantitative phase imaging (FCR-QPI). APPROACH: Quantitative phase imaging (QPI) has emerged as a powerful technique in the field of biomedicine but has not been actively applied to the monitoring of circulatory/cardiovascular parameters, due to dynamic speckles and low frame rates. We demonstrate FCR-QPI to measure heart rate and blood flow in a zebrafish embryo. FCR-QPI utilizes a high-speed photodetector instead of a conventional camera, so it enables real-time monitoring of individual red blood cell (RBC) flow. RESULTS: The average velocity of zebrafish's RBCs was measured from 192.5 to 608.8 µm / s at 24 to 28 hour-post-fertilization (hpf). In addition, the number of RBCs in a pulsatile blood flow was revealed to 16 cells/pulse at 48 hpf. The heart rates corresponded to 94 and 142 beats-per-minute at 24 and 48 hpf. CONCLUSIONS: This approach will newly enable in-depth understanding of the cardiovascular dynamics in the zebrafish model and possible usage for drug discovery applications in biomedicine.


Assuntos
Hemodinâmica , Peixe-Zebra , Animais , Diagnóstico por Imagem , Embrião de Mamíferos , Embrião não Mamífero , Frequência Cardíaca
6.
PLoS One ; 16(11): e0260372, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34847198

RESUMO

Collective cell migration is essential for embryonic development and homeostatic processes. During zebrafish development, the posterior lateral line primordium (pLLP) navigates along the embryo flank by collective cell migration. The chemokine receptors, Cxcr4b and Cxcr7b, as well as their cognate ligand, Cxcl12a, are essential for this process. We corroborate that knockdown of the zebrafish cd9 tetraspanin orthologue, cd9b, results in mild pLL abnormalities. Through generation of CRISPR and TALEN mutants, we show that cd9a and cd9b function partially redundantly in pLLP migration, which is delayed in the cd9b single and cd9a; cd9b double mutants. This delay led to a transient reduction in neuromast numbers. Loss of both Cd9a and Cd9b sensitized embryos to reduced Cxcr4b and Cxcl12a levels. Together these results provide evidence that Cd9 modulates collective cell migration of the pLLP during zebrafish development. One interpretation of these observations is that Cd9 contributes to more effective chemokine signalling.


Assuntos
Movimento Celular , Quimiocina CXCL12/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais , Tetraspanina 29/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/genética , Quimiocina CXCL12/genética , Técnicas de Silenciamento de Genes , Receptores CXCR4/genética , Tetraspanina 29/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
7.
Elife ; 102021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34165081

RESUMO

Epithelial tissues are primed to respond to insults by activating epithelial cell motility and rapid inflammation. Such responses are also elicited upon overexpression of the membrane-bound protease, Matriptase, or mutation of its inhibitor, Hai1. Unrestricted Matriptase activity also predisposes to carcinoma. How Matriptase leads to these cellular outcomes is unknown. We demonstrate that zebrafish hai1a mutants show increased H2O2, NfκB signalling, and IP3R -mediated calcium flashes, and that these promote inflammation, but do not generate epithelial cell motility. In contrast, inhibition of the Gq subunit in hai1a mutants rescues both the inflammation and epithelial phenotypes, with the latter recapitulated by the DAG analogue, PMA. We demonstrate that hai1a has elevated MAPK pathway activity, inhibition of which rescues the epidermal defects. Finally, we identify RSK kinases as MAPK targets disrupting adherens junctions in hai1a mutants. Our work maps novel signalling cascades mediating the potent effects of Matriptase on epithelia, with implications for tissue damage response and carcinoma progression.


Cancer occurs when normal processes in the cell become corrupted or unregulated. Many proteins can contribute, including one enzyme called Matriptase that cuts other proteins at specific sites. Matriptase activity is tightly controlled by a protein called Hai1. In mice and zebrafish, when Hai1 cannot adequately control Matriptase activity, invasive cancers with severe inflammation develop. However, it is unclear how unregulated Matriptase leads to both inflammation and cancer invasion. One outcome of Matriptase activity is removal of proteins called Cadherins from the cell surface. These proteins have a role in cell adhesion: they act like glue to stick cells together. Without them, cells can dissociate from a tissue and move away, a critical step in cancer cells invading other organs. However, it is unknown exactly how Matriptase triggers the removal of Cadherins from the cell surface to promote invasion. Previous work has shown that Matriptase switches on a receptor called Proteinase-activated receptor 2, or Par2 for short, which is known to activate many enzymes, including one called phospholipase C. When activated, this enzyme releases two signals into the cell: a sugar called inositol triphosphate, IP3; and a lipid or fat called diacylglycerol, DAG. It is possible that these two signals have a role to play in how Matriptase removes Cadherins from the cell surface. To find out, Ma et al. mapped the effects of Matriptase in zebrafish lacking the Hai1 protein. This revealed that Matriptase increases IP3 and DAG levels, which initiate both inflammation and invasion. IP3 promotes inflammation by switching on pro-inflammatory signals inside the cell such as the chemical hydrogen peroxide. At the same time, DAG promotes cell invasion by activating a well-known cancer signalling pathway called MAPK. This pathway activates a protein called RSK. Ma et al. show that this protein is required to remove Cadherins from the surface of cells, thus connecting Matriptase's activation of phospholipase C with its role in disrupting cell adhesion. An increase in the ratio of Matriptase to HAI-1 (the human equivalent of Hai1) is present in many cancers. For this reason, the signal cascades described by Ma et al. may be of interest in developing treatments for these cancers. Understanding how these signals work together could lead to more direct targeted anti-cancer approaches in the future.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Serina Endopeptidases/metabolismo , Animais , Animais Geneticamente Modificados , Cálcio/metabolismo , Sinalização do Cálcio , DNA/genética , Embrião não Mamífero , Ativação Enzimática , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio , Inflamação/metabolismo , Mutação , Neutrófilos/fisiologia , Peptídeos Cíclicos , Reação em Cadeia da Polimerase , RNA/genética , Serina Endopeptidases/genética , Peixe-Zebra
8.
Dis Model Mech ; 13(9)2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32988985

RESUMO

Heterotopic ossification (HO) is a disorder characterised by the formation of ectopic bone in soft tissue. Acquired HO typically occurs in response to trauma and is relatively common, yet its aetiology remains poorly understood. Genetic forms, by contrast, are very rare, but provide insights into the mechanisms of HO pathobiology. Fibrodysplasia ossificans progressiva (FOP) is the most debilitating form of HO. All patients reported to date carry heterozygous gain-of-function mutations in the gene encoding activin A receptor type I (ACVR1). These mutations cause dysregulated bone morphogenetic protein (BMP) signalling, leading to HO at extraskeletal sites including, but not limited to, muscles, ligaments, tendons and fascia. Ever since the identification of the causative gene, developing a cure for FOP has been a focus of investigation, and studies have decoded the pathophysiology at the molecular and cellular levels, and explored novel management strategies. Based on the established role of BMP signalling throughout HO in FOP, therapeutic modalities that target multiple levels of the signalling cascade have been designed, and some drugs have entered clinical trials, holding out hope of a cure. A potential role of other signalling pathways that could influence the dysregulated BMP signalling and present alternative therapeutic targets remains a matter of debate. Here, we review the recent FOP literature, including pathophysiology, clinical aspects, animal models and current management strategies. We also consider how this research can inform our understanding of other types of HO and highlight some of the remaining knowledge gaps.


Assuntos
Miosite Ossificante/patologia , Pesquisa Translacional Biomédica , Receptores de Ativinas Tipo I/química , Receptores de Ativinas Tipo I/genética , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Modelos Animais de Doenças , Humanos , Mutação/genética , Miosite Ossificante/diagnóstico , Miosite Ossificante/genética , Miosite Ossificante/fisiopatologia
9.
Sci Rep ; 9(1): 15240, 2019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31645615

RESUMO

Advances in gene editing now allow reverse genetics to be applied to a broad range of biological systems. Ultimately, any modification to coding sequences requires confirmation at the protein level, although immunoblotting is often hampered by antibody quality or availability especially in non-model species. Sequential Window Acquisition of All Theoretical Spectra (SWATH), a mass spectrometry (MS) technology with exceptional quantitative reproducibility and accuracy, offers an ideal alternative for protein-based confirmation. Here, using genome edits in mouse, zebrafish and Bicyclus anynana butterflies produced using either homologous recombination or targeted nucleases, we demonstrate absence of the targeted proteins using SWATH, thus confirming successful editing. We show that SWATH is a robust antibody-independent alternative for monitoring gene editing at the protein level and broadly applicable across diverse organisms and targeted genome manipulation techniques. Moreover, SWATH concomitantly defines the global proteome response in the edited organism, which may provide pertinent biological insights.


Assuntos
Edição de Genes , Espectrometria de Massas/métodos , Proteínas/genética , Sequência de Aminoácidos , Animais , Borboletas , Edição de Genes/métodos , Recombinação Homóloga , Camundongos , Proteínas/análise , Proteoma/análise , Proteoma/genética , Proteômica/métodos , Peixe-Zebra
10.
Dis Model Mech ; 12(9)2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31383797

RESUMO

Reduced bone quality or mineral density predict susceptibility to fracture and also attenuate subsequent repair. Bone regrowth is also compromised by bacterial infection, which exacerbates fracture site inflammation. Because of the cellular complexity of fracture repair, as well as genetic and environmental influences, there is a need for models that permit visualisation of the fracture repair process under clinically relevant conditions. To characterise the process of fracture repair in zebrafish, we employed a crush fracture of fin rays, coupled with histological and transgenic labelling of cellular responses; the results demonstrate a strong similarity to the phased response in humans. We applied our analysis to a zebrafish model of osteogenesis imperfecta (OI), which shows reduced bone quality, spontaneous fractures and propensity for non-unions. We found deficiencies in the formation of a bone callus during fracture repair in our OI model and showed that clinically employed antiresorptive bisphosphonates can reduce spontaneous fractures in OI fish and also measurably reduce fracture callus remodelling in wild-type fish. The csf1ra mutant, which has reduced osteoclast numbers, also showed reduced callus remodelling. Exposure to excessive bisphosphonate, however, disrupted callus repair. Intriguingly, neutrophils initially colonised the fracture site, but were later completely excluded. However, when fractures were infected with Staphylococcus aureus, neutrophils were retained and compromised repair. This work elevates the zebrafish bone fracture model and indicates its utility in assessing conditions of relevance to an orthopaedic setting with medium throughput.This article has an associated First Person interview with the first author of the paper.


Assuntos
Fraturas Ósseas/patologia , Peixe-Zebra/fisiologia , Alendronato/farmacologia , Alendronato/uso terapêutico , Nadadeiras de Animais/patologia , Animais , Calo Ósseo/efeitos dos fármacos , Calo Ósseo/patologia , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Modelos Animais de Doenças , Consolidação da Fratura/efeitos dos fármacos , Fraturas Ósseas/tratamento farmacológico , Fraturas Ósseas/microbiologia , Fraturas não Consolidadas/patologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteogênese Imperfeita/patologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia
11.
Trends Genet ; 34(5): 362-378, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29429760

RESUMO

Transgenic approaches are instrumental for labeling and manipulating cells and cellular machineries in vivo. Transgenes have traditionally been static entities that remained unaltered following genome integration, limiting their versatility. The development of DNA recombinase-based methods to modify, excise, or rearrange transgene cassettes has introduced versatile control of transgene activity and function. In particular, recombinase-controlled transgenes enable regulation of exogenous gene expression, conditional mutagenesis, and genetic lineage tracing. In zebrafish, transgenesis-based recombinase genetics using Cre/lox, Flp/FRT, and ΦC31 are increasingly applied to study development and homeostasis, and to generate disease models. Intersected with the versatile imaging capacity of the zebrafish model and recent breakthroughs in genome editing, we review and discuss past, current, and potential future approaches and resources for recombinase-based techniques in zebrafish.


Assuntos
Técnicas de Transferência de Genes , Recombinases/genética , Recombinação Genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Genoma/genética , Mutagênese Insercional/genética , Transgenes/genética , Peixe-Zebra/crescimento & desenvolvimento
12.
J Biol Chem ; 292(43): 17760-17776, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-28912276

RESUMO

Angiogenesis is a highly regulated process for formation of new blood vessels from pre-existing ones. Angiogenesis is dysregulated in various pathologies, including age-related macular degeneration, arthritis, and cancer. Inhibiting pathological angiogenesis therefore represents a promising therapeutic strategy for treating these disorders, highlighting the need to study angiogenesis in more detail. To this end, identifying the genes essential for blood vessel formation and elucidating their function are crucial for a complete understanding of angiogenesis. Here, focusing on potential candidate genes for angiogenesis, we performed a morpholino-based genetic screen in zebrafish and identified Cavin-2, a membrane-bound phosphatidylserine-binding protein and critical organizer of caveolae (small microdomains in the plasma membrane), as a regulator of angiogenesis. Using endothelial cells, we show that Cavin-2 is required for in vitro angiogenesis and also for endothelial cell proliferation, migration, and invasion. We noted a high level of Cavin-2 expression in the neovascular tufts in the mouse model of oxygen-induced retinopathy, suggesting a role for Cavin-2 in pathogenic angiogenesis. Interestingly, we also found that Cavin-2 regulates the production of nitric oxide (NO) in endothelial cells by controlling the stability and activity of the endothelial nitric-oxide synthase (eNOS) and that Cavin-2 knockdown cells produce much less NO than WT cells. Also, mass spectrometry, flow cytometry, and electron microscopy analyses indicated that Cavin-2 is secreted in endothelial microparticles (EMPs) and is required for EMP biogenesis. Taken together, our results indicate that in addition to its function in caveolae biogenesis, Cavin-2 plays a critical role in endothelial cell maintenance and function by regulating eNOS activity.


Assuntos
Proteínas de Membrana/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Neovascularização Retiniana/metabolismo , Retinopatia da Prematuridade/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Modelos Animais de Doenças , Estabilidade Enzimática , Proteínas de Membrana/genética , Camundongos , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Neovascularização Retiniana/genética , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
13.
Toxicol Sci ; 156(1): 133-148, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28069987

RESUMO

Organ toxicity, particularly liver toxicity, remains one of the major reasons for the termination of drug candidates in the development pipeline as well as withdrawal or restrictions of marketed drugs. A screening-amenable alternative in vivo model such as zebrafish would, therefore, find immediate application in the early prediction of unacceptable organ toxicity. To identify highly upregulated genes as biomarkers of toxic responses in the zebrafish model, a set of well-characterized reference drugs that cause drug-induced liver injury (DILI) in the clinic were applied to zebrafish larvae and adults. Transcriptome microarray analysis was performed on whole larvae or dissected adult livers. Integration of data sets from different drug treatments at different stages identified common upregulated detoxification pathways. Within these were candidate biomarkers which recurred in multiple treatments. We prioritized 4 highly upregulated genes encoding enzymes acting in distinct phases of the drug metabolism pathway. Through promoter isolation and fosmid recombineering, eGFP reporter transgenic zebrafish lines were generated and evaluated for their response to DILI drugs. Three of the 4 generated reporter lines showed a dose and time-dependent induction in endodermal organs to reference drugs and an expanded drug set. In conclusion, through integrated transcriptomics and transgenic approaches, we have developed parallel independent zebrafish in vivo screening platforms able to predict organ toxicities of preclinical drugs.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Drogas em Investigação/efeitos adversos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Fígado/efeitos dos fármacos , Testes de Toxicidade/métodos , Xenobióticos/toxicidade , Animais , Animais Geneticamente Modificados , Biomarcadores/metabolismo , Relação Dose-Resposta a Droga , Drogas em Investigação/administração & dosagem , Endoderma/efeitos dos fármacos , Endoderma/crescimento & desenvolvimento , Endoderma/metabolismo , Feminino , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Larva/efeitos dos fármacos , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Masculino , Organogênese/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Teratogênicos/toxicidade , Xenobióticos/administração & dosagem , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
14.
Arch Toxicol ; 91(3): 1187-1197, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27485346

RESUMO

Understanding and predicting whether new drug candidates will be safe in the clinic is a critical hurdle in pharmaceutical development, that relies in part on absorption, distribution, metabolism, excretion and toxicology studies in vivo. Zebrafish is a relatively new model system for drug metabolism and toxicity studies, offering whole organism screening coupled with small size and potential for high-throughput screening. Through toxicity and absorption analyses of a number of drugs, we find that zebrafish is generally predictive of drug toxicity, although assay outcomes are influenced by drug lipophilicity which alters drug uptake. In addition, liver microsome assays reveal specific differences in metabolism of compounds between human and zebrafish livers, likely resulting from the divergence of the cytochrome P450 superfamily between species. To reflect human metabolism more accurately, we generated a transgenic "humanized" zebrafish line that expresses the major human phase I detoxifying enzyme, CYP3A4, in the liver. Here, we show that this humanized line shows an elevated metabolism of CYP3A4-specific substrates compared to wild-type zebrafish. The generation of this first described humanized zebrafish liver suggests such approaches can enhance the accuracy of the zebrafish model for toxicity prediction.


Assuntos
Citocromo P-450 CYP3A/genética , Fígado/efeitos dos fármacos , Farmacocinética , Testes de Toxicidade/métodos , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP3A/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Inativação Metabólica , Fígado/metabolismo , Espectrometria de Massas , Preparações Farmacêuticas/química , Solubilidade
15.
PLoS One ; 11(4): e0152989, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27077909

RESUMO

Mutant Estrogen Receptor (ERT2) ligand-binding domain fusions with Cre recombinase are a key tool for spatio-temporally controlled genetic recombination with the Cre/lox system. CreERT2 is efficiently activated in a concentration-dependent manner by the Tamoxifen metabolite trans-4-OH-Tamoxifen (trans-4-OHT). Reproducible and efficient Cre/lox experimentation is hindered by the gradual loss of CreERT2 induction potency upon prolonged storage of dissolved trans-4-OHT, which potentially results from gradual trans-to-cis isomerization or degradation. Here, we combined zebrafish CreERT2 recombination experiments and cell culture assays to document the gradual activity loss of trans-4-OHT and describe the alternative Tamoxifen metabolite Endoxifen as more stable alternative compound. Endoxifen retains potent activation upon prolonged storage (3 months), yet consistently induces half the ERT2 domain fusion activity compared to fresh trans-4-OHT. Using 1H-NMR analysis, we reveal that trans-4-OHT isomerization is undetectable upon prolonged storage in either DMSO or Ethanol, ruling out isomer transformation as cause for the gradual loss of trans-4-OHT activity. We further establish that both trans-4-OHT and Endoxifen are insensitive to light exposure under regular laboratory handling conditions. We attribute the gradual loss of trans-4-OHT potency to precipitation over time, and show that heating of aged trans-4-OHT aliquots reinstates their CreERT2 induction potential. Our data establish Endoxifen as potent and reproducible complementary compound to 4-OHT to control ERT2 domain fusion proteins in vivo, and provide a framework for efficient chemically controlled recombination experiments.


Assuntos
Integrases/genética , Receptores de Estrogênio/genética , Tamoxifeno/metabolismo , Tamoxifeno/farmacologia , Animais , Linhagem Celular , Estabilidade de Medicamentos , Temperatura Alta , Humanos , Estrutura Terciária de Proteína , Receptores de Estrogênio/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Recombinação Genética/efeitos dos fármacos , Estereoisomerismo , Tamoxifeno/análogos & derivados , Tamoxifeno/química , Fatores de Tempo , Peixe-Zebra
16.
J Proteome Res ; 13(12): 5536-50, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25230361

RESUMO

Zebrafish is a popular system for studying vertebrate development and disease that shows high genetic conservation with humans. Molecular level studies at different stages of development are essential for understanding the processes deployed during ontogeny. Here, we performed comparative analysis of the whole proteome and transcriptome of the early stage (24 h post-fertilization) zebrafish embryo. We identified 8363 proteins with their approximate cellular abundances (the largest number of zebrafish embryo proteins quantified thus far), through a combination of thorough deyolking and extensive fractionation procedures, before resolving the peptides by mass spectrometry. We performed deep sequencing of the transcripts and found that the expressed proteome and transcriptome displayed a moderate correlation for the majority of cellular processes. Integrative functional mapping of the quantified genes demonstrated that embryonic developmental systems differentially exploit transcriptional and post-transcriptional regulatory mechanisms to modulate protein abundance. Using network mapping of the low-abundance proteins, we identified various signal transduction pathways important in embryonic development and also revealed genes that may be regulated at the post-transcriptional level. Our data set represents a deep coverage of the functional proteome and transcriptome of the developing zebrafish, and our findings unveil molecular regulatory mechanisms that underlie embryonic development.


Assuntos
Embrião não Mamífero/metabolismo , Proteoma/metabolismo , Transcriptoma , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas , Proteoma/genética , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
17.
BMC Biol ; 11: 37, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23587183

RESUMO

First discovered in Drosophila, the Hedgehog signaling pathway controls a wide range of developmental processes and is implicated in a variety of cancers. The success of a screen for chemical modulators of this pathway, published in 2002, opened a new chapter in the quest to translate the results of basic developmental biology research into therapeutic applications. Small molecule pathway agonists are now used to program stem cells, whilst antagonists are proving effective as anti-cancer therapies.


Assuntos
Antineoplásicos/farmacologia , Proteínas Hedgehog/metabolismo , Transdução de Sinais/efeitos dos fármacos , Pesquisa Translacional Biomédica , Animais , Proteínas Hedgehog/antagonistas & inibidores , Humanos , Bibliotecas de Moléculas Pequenas/farmacologia
18.
Development ; 130(12): 2809-18, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12736222

RESUMO

The transcription factor Sox10 is required for the specification, migration and survival of all nonectomesenchymal neural crest derivatives including melanophores. sox10(-/-) zebrafish lack expression of the transcription factor mitfa, which itself is required for melanophore development. We demonstrate that the zebrafish mitfa promoter has sox10 binding sites necessary for activity in vitro, consistent with studies using mammalian cell cultures that have shown that Sox10 directly regulates Mitf expression. In addition, we demonstrate that these sites are necessary for promoter activity in vivo. We show that reintroduction of mitfa expression in neural crest cells can rescue melanophore development in sox10(-/-) embryos. This rescue of melanophores in sox10(-/-) embryos is quantitatively indistinguishable from rescue in mitfa(-/-) embryos. These findings show that the essential function of sox10 in melanophore development is limited to transcriptional regulation of mitfa. We propose that the dominant melanophore phenotype in Waardenburg syndrome IV individuals with SOX10 mutations is likely to result from failure to activate MITF in the normal number of melanoblasts.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Melanóforos/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra/embriologia , Animais , Cruzamentos Genéticos , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator de Transcrição Associado à Microftalmia , Mutação , Regiões Promotoras Genéticas , Fatores de Transcrição SOXE , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA