Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Org Biomol Chem ; 22(26): 5296-5300, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38896038

RESUMO

The synthesis of medicinally relevant N-aryl-substituted 2-aminobenzothiazoles often uses 2-aminothiophenol derivatives, which are not commercially abundant, as starting materials. Herein, we report a method for the annulation of C3-substituted nitroarenes and aryl isothiocyanates towards the synthesis of 2-aminobenzothiazoles. Reactions proceeded in the presence of cobalt ferrite nanoparticles as a catalyst, DABCO as a base, and DMF as a promoter. The cobalt ferrite nanoparticles could be recovered after each run and reused up to 3 times while the product yield was not diminished. Our method appears to be the first example of the direct use of substituted nitroarenes for yielding 2-aminobenzothiazoles.

2.
Science ; 380(6646): 758-764, 2023 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-37200435

RESUMO

Zebrafish hearts can regenerate by replacing damaged tissue with new cardiomyocytes. Although the steps leading up to the proliferation of surviving cardiomyocytes have been extensively studied, little is known about the mechanisms that control proliferation and redifferentiation to a mature state. We found that the cardiac dyad, a structure that regulates calcium handling and excitation-contraction coupling, played a key role in the redifferentiation process. A component of the cardiac dyad called leucine-rich repeat-containing 10 (Lrrc10) acted as a negative regulator of proliferation, prevented cardiomegaly, and induced redifferentiation. We found that its function was conserved in mammalian cardiomyocytes. This study highlights the importance of the underlying mechanisms required for heart regeneration and their application to the generation of fully functional cardiomyocytes.


Assuntos
Cálcio , Coração , Miócitos Cardíacos , Regeneração , Sarcômeros , Peixe-Zebra , Animais , Cálcio/fisiologia , Proliferação de Células , Coração/fisiologia , Miócitos Cardíacos/fisiologia , Sarcômeros/fisiologia , Peixe-Zebra/fisiologia
3.
Mol Cell ; 82(10): 1956-1970.e14, 2022 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-35366395

RESUMO

Recent advances in single-cell sequencing technologies have enabled simultaneous measurement of multiple cellular modalities, but the combined detection of histone post-translational modifications and transcription at single-cell resolution has remained limited. Here, we introduce EpiDamID, an experimental approach to target a diverse set of chromatin types by leveraging the binding specificities of single-chain variable fragment antibodies, engineered chromatin reader domains, and endogenous chromatin-binding proteins. Using these, we render the DamID technology compatible with the genome-wide identification of histone post-translational modifications. Importantly, this includes the possibility to jointly measure chromatin marks and transcription at the single-cell level. We use EpiDamID to profile single-cell Polycomb occupancy in mouse embryoid bodies and provide evidence for hierarchical gene regulatory networks. In addition, we map H3K9me3 in early zebrafish embryogenesis, and detect striking heterochromatic regions specific to notochord. Overall, EpiDamID is a new addition to a vast toolbox to study chromatin states during dynamic cellular processes.


Assuntos
Código das Histonas , Histonas , Animais , Cromatina/genética , Histonas/genética , Histonas/metabolismo , Camundongos , Processamento de Proteína Pós-Traducional , Transcriptoma , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
4.
Development ; 148(18)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34397091

RESUMO

Zebrafish are excellent at regenerating their heart by reinitiating proliferation in pre-existing cardiomyocytes. Studying how zebrafish achieve this holds great potential in developing new strategies to boost mammalian heart regeneration. Nevertheless, the lack of appropriate live-imaging tools for the adult zebrafish heart has limited detailed studies into the dynamics underlying cardiomyocyte proliferation. Here, we address this by developing a system in which cardiac slices of the injured zebrafish heart are cultured ex vivo for several days while retaining key regenerative characteristics, including cardiomyocyte proliferation. In addition, we show that the cardiac slice culture system is compatible with live timelapse imaging and allows manipulation of regenerating cardiomyocytes with drugs that normally would have toxic effects that prevent their use. Finally, we use the cardiac slices to demonstrate that adult cardiomyocytes with fully assembled sarcomeres can partially disassemble their sarcomeres in a calpain- and proteasome-dependent manner to progress through nuclear division and cytokinesis. In conclusion, we have developed a cardiac slice culture system, which allows imaging of native cardiomyocyte dynamics in real time to discover cellular mechanisms during heart regeneration.


Assuntos
Proliferação de Células/fisiologia , Miócitos Cardíacos/fisiologia , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados/metabolismo , Animais Geneticamente Modificados/fisiologia , Calpaína/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/fisiologia , Células Cultivadas , Citocinese/fisiologia , Feminino , Coração/fisiologia , Masculino , Mamíferos/metabolismo , Mamíferos/fisiologia , Miócitos Cardíacos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Regeneração/fisiologia , Sarcômeros/metabolismo , Sarcômeros/fisiologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
5.
Cell Mol Life Sci ; 78(12): 5107-5122, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33950316

RESUMO

Cardiac regeneration is the outcome of the highly regulated interplay of multiple processes, including the inflammatory response, cardiomyocyte dedifferentiation and proliferation, neovascularization and extracellular matrix turnover. Species-specific traits affect these injury-induced processes, resulting in a wide variety of cardiac regenerative potential between species. Indeed, while mammals are generally considered poor regenerators, certain amphibian and fish species like the zebrafish display robust regenerative capacity post heart injury. The species-specific traits underlying these differential injury responses are poorly understood. In this review, we will compare the injury induced processes of the mammalian and zebrafish heart, describing where these processes overlap and diverge. Additionally, by examining multiple species across the animal kingdom, we will highlight particular traits that either positively or negatively affect heart regeneration. Last, we will discuss the possibility of overcoming regeneration-limiting traits to induce heart regeneration in mammals.


Assuntos
Evolução Biológica , Cardiopatias/terapia , Coração/embriologia , Miócitos Cardíacos/citologia , Regeneração , Animais , Humanos , Miócitos Cardíacos/fisiologia
6.
Nature ; 591(7849): 281-287, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33568815

RESUMO

Skeletal muscle regenerates through the activation of resident stem cells. Termed satellite cells, these normally quiescent cells are induced to proliferate by wound-derived signals1. Identifying the source and nature of these cues has been hampered by an inability to visualize the complex cell interactions that occur within the wound. Here we use muscle injury models in zebrafish to systematically capture the interactions between satellite cells and the innate immune system after injury, in real time, throughout the repair process. This analysis revealed that a specific subset of macrophages 'dwell' within the injury, establishing a transient but obligate niche for stem cell proliferation. Single-cell profiling identified proliferative signals that are secreted by dwelling macrophages, which include the cytokine nicotinamide phosphoribosyltransferase (Nampt, which is also known as visfatin or PBEF in humans). Nampt secretion from the macrophage niche is required for muscle regeneration, acting through the C-C motif chemokine receptor type 5 (Ccr5), which is expressed on muscle stem cells. This analysis shows that in addition to their ability to modulate the immune response, specific macrophage populations also provide a transient stem-cell-activating niche, directly supplying proliferation-inducing cues that govern the repair process that is mediated by muscle stem cells. This study demonstrates that macrophage-derived niche signals for muscle stem cells, such as NAMPT, can be applied as new therapeutic modalities for skeletal muscle injury and disease.


Assuntos
Macrófagos/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/lesões , Mioblastos/citologia , Nicotinamida Fosforribosiltransferase/metabolismo , Nicho de Células-Tronco , Peixe-Zebra/metabolismo , Animais , Proliferação de Células , Modelos Animais de Doenças , Humanos , Macrófagos/citologia , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mioblastos/metabolismo , Nicotinamida Fosforribosiltransferase/genética , Fator de Transcrição PAX7/metabolismo , RNA-Seq , Receptores CCR5/genética , Receptores CCR5/metabolismo , Regeneração/fisiologia , Análise de Célula Única , Peixe-Zebra/imunologia
7.
Dev Biol ; 471: 106-118, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33309949

RESUMO

Adult zebrafish are frequently described to be able to "completely" regenerate the heart. Yet, the extent to which cardiomyocytes lost to injury are replaced is unknown, since existing evidence for cardiomyocyte proliferation is indirect or non-quantitative. We established stereological methods to quantify the number of cardiomyocytes at several time-points post cryoinjury. Intriguingly, after cryoinjuries that killed about 1/3 of the ventricular cardiomyocytes, pre-injury cardiomyocyte numbers were restored already within 30 days. Yet, many hearts retained small residual scars, and a subset of cardiomyocytes bordering these fibrotic areas remained smaller, lacked differentiated sarcomeric structures, and displayed defective calcium signaling. Thus, a subset of regenerated cardiomyocytes failed to fully mature. While lineage-tracing experiments have shown that regenerating cardiomyocytes are derived from differentiated cardiomyocytes, technical limitations have previously made it impossible to test whether cardiomyocyte trans-differentiation contributes to regeneration of non-myocyte cell lineages. Using Cre responder lines that are expressed in all major cell types of the heart, we found no evidence for cardiomyocyte transdifferentiation into endothelial, epicardial, fibroblast or immune cell lineages. Overall, our results imply a refined answer to the question whether zebrafish can completely regenerate the heart: in response to cryoinjury, preinjury cardiomyocyte numbers are indeed completely regenerated by proliferation of lineage-restricted cardiomyocytes, while restoration of cardiomyocyte differentiation and function, as well as resorption of scar tissue, is less robustly achieved.


Assuntos
Coração/fisiologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Regeneração , Peixe-Zebra/metabolismo , Animais , Fibrose , Miocárdio/patologia , Miócitos Cardíacos/patologia
8.
Elife ; 82019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31868166

RESUMO

While the heart regenerates poorly in mammals, efficient heart regeneration occurs in zebrafish. Studies in zebrafish have resulted in a model in which preexisting cardiomyocytes dedifferentiate and reinitiate proliferation to replace the lost myocardium. To identify which processes occur in proliferating cardiomyocytes we have used a single-cell RNA-sequencing approach. We uncovered that proliferating border zone cardiomyocytes have very distinct transcriptomes compared to the nonproliferating remote cardiomyocytes and that they resemble embryonic cardiomyocytes. Moreover, these cells have reduced expression of mitochondrial genes and reduced mitochondrial activity, while glycolysis gene expression and glucose uptake are increased, indicative for metabolic reprogramming. Furthermore, we find that the metabolic reprogramming of border zone cardiomyocytes is induced by Nrg1/ErbB2 signaling and is important for their proliferation. This mechanism is conserved in murine hearts in which cardiomyocyte proliferation is induced by activating ErbB2 signaling. Together these results demonstrate that glycolysis regulates cardiomyocyte proliferation during heart regeneration.


Assuntos
Proliferação de Células , Reprogramação Celular/fisiologia , Coração/fisiologia , Miócitos Cardíacos/metabolismo , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Análise de Célula Única/métodos , Peixe-Zebra/crescimento & desenvolvimento , Animais , Animais Geneticamente Modificados , Reprogramação Celular/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes erbB-2/genética , Genes erbB-2/fisiologia , Glicólise , Coração/embriologia , Hexoquinase/genética , Hexoquinase/metabolismo , Masculino , Camundongos , Modelos Animais , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Neuregulina-1/genética , Regeneração/genética , Transdução de Sinais/genética , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
9.
Methods ; 150: 63-67, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30392565

RESUMO

Labelling cells and following their progeny, also known as lineage tracing, has provided important insights into the cellular origins of tissues. Traditional lineage tracing experiments have been limited to following single or small groups of cells with classic techniques such as dye injections and Cre/LoxP labelling of cells of interest. Brainbow is a fluorescent dependent, lineage tracing technique that allows a broader visualization and analysis of multiple cells within a tissue, initially deployed to examine lineages within neural tissues. This technique has now been adapted to zebrafish (Zebrabow) and takes advantages of the imaging capabilities that this system provides over other animal models. In this paper we shall describe how Zebrabow is performed as well as some guides on some of the common pitfalls encountered when using this labelling strategy.


Assuntos
Rastreamento de Células/normas , Coloração e Rotulagem/normas , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Linhagem da Célula/genética , Rastreamento de Células/métodos , Embrião não Mamífero , Genes Reporter/genética , Guias como Assunto , Integrases/genética , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Modelos Animais , Coloração e Rotulagem/métodos
10.
Neural Dev ; 13(1): 24, 2018 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-30447699

RESUMO

BACKGROUND: Despite conserved developmental processes and organization of the vertebrate central nervous system, only some vertebrates including zebrafish can efficiently regenerate neural damage including after spinal cord injury. The mammalian spinal cord shows very limited regeneration and neurogenesis, resulting in permanent life-long functional impairment. Therefore, there is an urgent need to identify the cellular and molecular mechanisms that can drive efficient vertebrate neurogenesis following injury. A key pathway implicated in zebrafish neurogenesis is fibroblast growth factor signaling. METHODS: In the present study we investigated the roles of distinct fibroblast growth factor members and their receptors in facilitating different aspects of neural development and regeneration at different timepoints following spinal cord injury. After spinal cord injury in adults and during larval development, loss and/or gain of Fgf signaling was combined with immunohistochemistry, in situ hybridization and transgenes marking motor neuron populations in in vivo zebrafish and in vitro mammalian PC12 cell culture models. RESULTS: Fgf3 drives neurogenesis of Islet1 expressing motor neuron subtypes and mediate axonogenesis in cMet expressing motor neuron subtypes. We also demonstrate that the role of Fgf members are not necessarily simple recapitulating development. During development Fgf2, Fgf3 and Fgf8 mediate neurogenesis of Islet1 expressing neurons and neuronal sprouting of both, Islet1 and cMet expressing motor neurons. Strikingly in mammalian PC12 cells, all three Fgfs increased cell proliferation, however, only Fgf2 and to some extent Fgf8, but not Fgf3 facilitated neurite outgrowth. CONCLUSIONS: This study demonstrates differential Fgf member roles during neural development and adult regeneration, including in driving neural proliferation and neurite outgrowth of distinct spinal cord neuron populations, suggesting that factors including Fgf type, age of the organism, timing of expression, requirements for different neuronal populations could be tailored to best drive all of the required regenerative processes.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Regeneração Nervosa/fisiologia , Neurogênese/fisiologia , Traumatismos da Medula Espinal/metabolismo , Animais , Animais Geneticamente Modificados , Proliferação de Células , Neurônios Motores/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Transdução de Sinais/fisiologia , Medula Espinal/citologia , Medula Espinal/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
11.
Methods Mol Biol ; 1556: 117-125, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28247347

RESUMO

Understanding muscle stem cell behaviors can potentially provide insights into how these cells act and respond during normal growth and diseased contexts. The zebrafish is an ideal model organism to examine these behaviors in vivo where it would normally be technically challenging in other mammalian models. This chapter will describe the procedures required to successfully conduct live imaging of zebrafish transgenics that has specifically been adapted for skeletal muscle.


Assuntos
Músculos/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Biomarcadores , Microscopia Confocal , Imagem Molecular/métodos , Músculo Esquelético/citologia , Fenótipo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo
12.
Nature ; 535(7613): 542-6, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27437584

RESUMO

The transition from fins to limbs was an important terrestrial adaptation, but how this crucial evolutionary shift arose developmentally is unknown. Current models focus on the distinct roles of the apical ectodermal ridge (AER) and the signaling molecules that it secretes during limb and fin outgrowth. In contrast to the limb AER, the AER of the fin rapidly transitions into the apical fold and in the process shuts off AER-derived signals that stimulate proliferation of the precursors of the appendicular skeleton. The differing fates of the AER during fish and tetrapod development have led to the speculation that fin-fold formation was one of the evolutionary hurdles to the AER-dependent expansion of the fin mesenchyme required to generate the increased appendicular structure evident within limbs. Consequently, a heterochronic shift in the AER-to-apical-fold transition has been postulated to be crucial for limb evolution. The ability to test this model has been hampered by a lack of understanding of the mechanisms controlling apical fold induction. Here we show that invasion by cells of a newly identified somite-derived lineage into the AER in zebrafish regulates apical fold induction. Ablation of these cells inhibits apical fold formation, prolongs AER activity and increases the amount of fin bud mesenchyme, suggesting that these cells could provide the timing mechanism proposed in Thorogood's clock model of the fin-to-limb transition. We further demonstrate that apical-fold inducing cells are progressively lost during gnathostome evolution;the absence of such cells within the tetrapod limb suggests that their loss may have been a necessary prelude to the attainment of limb-like structures in Devonian sarcopterygian fish.


Assuntos
Nadadeiras de Animais/embriologia , Nadadeiras de Animais/metabolismo , Ectoderma/embriologia , Ectoderma/metabolismo , Somitos/embriologia , Somitos/metabolismo , Peixe-Zebra/embriologia , Animais , Evolução Biológica , Linhagem da Célula , Ectoderma/citologia , Feminino , Botões de Extremidades/citologia , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/metabolismo , Somitos/citologia
13.
Am J Physiol Cell Physiol ; 303(12): C1220-8, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22932682

RESUMO

Long-term culture of primary neonatal rat cardiomyocytes is limited by the loss of spontaneous contractile phenotype within weeks in culture. This may be due to loss of contractile cardiomyocytes from the culture or overgrowth of the non-cardiomyocyte population. Using the mitochondria specific fluorescent dye, tetramethylrhodamine methyl ester perchlorate (TMRM), we showed that neonatal rat cardiomyocytes enriched by fluorescence-activated cell sorting can be maintained as contractile cultures for long periods (24-wk culture vs. 2 wk for unsorted cardiomyocytes). Long-term culture of this purified cardiomyocyte (TMRM high) population retained the expression of cardiomyocyte markers, continued calcium cycling, and displayed cyclic electrical activity that could be regulated pharmacologically. These findings suggest that non-cardiomyocyte populations can negatively influence contractility of cardiomyocytes in culture and that by purifying cardiomyocytes, the cultures retain potential as an experimental model for longitudinal studies of cardiomyocyte biology in vitro.


Assuntos
Contração Miocárdica , Miócitos Cardíacos/fisiologia , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Citometria de Fluxo , Corantes Fluorescentes , Ratos , Ratos Sprague-Dawley , Rodaminas/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA