Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
EMBO Rep ; 23(8): e55563, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35836403

RESUMO

Development of vertebrate limbs and fins requires that tissue growth is directed outwards, away from the body. How such directed growth is achieved is a fascinating biological problem. For limb/fin formation and outgrowth, signaling between mesenchymal cells and the overlying epithelium is essential. In particular, the epithelium at the distal margin of the growing limb/fin bud, termed the apical ectodermal ridge (AER), promotes directed outgrowth of the underlying mesenchyme, e.g., by providing polarization cues for mesenchymal cell migration. Several classical signaling pathways, such as fibroblast growth factor (Fgf), hedgehog, and Wnt signaling, are involved in the regulation of the cellular events that shape the limb/fin bud (Iovine, 2007). In this issue of EMBO Reports, Carney and colleagues surprisingly find that the Slit-Robo pathway, which is best known for its function in axon guidance, regulates the polarity of developing zebrafish fins (Mahabaleshwar et al, 2007). Intriguingly, they identify an intricate back and forth of signals between the mesenchyme and the AER. Slit ligands derived from mesenchyme act on Robo receptors in the AER to stimulate the production of sphingosine-1-phosphate, which then acts back on the mesenchyme to regulate cell polarity and orientation.


Assuntos
Botões de Extremidades , Peixe-Zebra , Animais , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Botões de Extremidades/metabolismo , Mesoderma/metabolismo , Morfogênese , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
2.
Cell ; 136(6): 1136-47, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19303855

RESUMO

Interactions between developmental signaling pathways govern the formation and function of stem cells. Prostaglandin (PG) E2 regulates vertebrate hematopoietic stem cells (HSC). Similarly, the Wnt signaling pathway controls HSC self-renewal and bone marrow repopulation. Here, we show that wnt reporter activity in zebrafish HSCs is responsive to PGE2 modulation, demonstrating a direct interaction in vivo. Inhibition of PGE2 synthesis blocked wnt-induced alterations in HSC formation. PGE2 modified the wnt signaling cascade at the level of beta-catenin degradation through cAMP/PKA-mediated stabilizing phosphorylation events. The PGE2/Wnt interaction regulated murine stem and progenitor populations in vitro in hematopoietic ES cell assays and in vivo following transplantation. The relationship between PGE2 and Wnt was also conserved during regeneration of other organ systems. Our work provides in vivo evidence that Wnt activation in stem cells requires PGE2, and suggests the PGE2/Wnt interaction is a master regulator of vertebrate regeneration and recovery.


Assuntos
Dinoprostona/metabolismo , Desenvolvimento Embrionário , Células-Tronco Hematopoéticas/metabolismo , Proteínas Wnt/metabolismo , Peixe-Zebra/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Células-Tronco Embrionárias/metabolismo , Fígado/fisiologia , Camundongos , Regeneração , Transdução de Sinais , Peixe-Zebra/embriologia , beta Catenina/metabolismo
3.
PLoS Genet ; 17(11): e1009890, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34723970

RESUMO

In contrast to mammals, the zebrafish maintains its cardiomyocyte proliferation capacity throughout adulthood. However, neither the molecular mechanisms that orchestrate the proliferation of cardiomyocytes during developmental heart growth nor in the context of regeneration in the adult are sufficiently defined yet. We identified in a forward genetic N-ethyl-N-nitrosourea (ENU) mutagenesis screen the recessive, embryonic-lethal zebrafish mutant baldrian (bal), which shows severely impaired developmental heart growth due to diminished cardiomyocyte proliferation. By positional cloning, we identified a missense mutation in the zebrafish histone deacetylase 1 (hdac1) gene leading to severe protein instability and the loss of Hdac1 function in vivo. Hdac1 inhibition significantly reduces cardiomyocyte proliferation, indicating a role of Hdac1 during developmental heart growth in zebrafish. To evaluate whether developmental and regenerative Hdac1-associated mechanisms of cardiomyocyte proliferation are conserved, we analyzed regenerative cardiomyocyte proliferation after Hdac1 inhibition at the wound border zone in cryoinjured adult zebrafish hearts and we found that Hdac1 is also essential to orchestrate regenerative cardiomyocyte proliferation in the adult vertebrate heart. In summary, our findings suggest an important and conserved role of Histone deacetylase 1 (Hdac1) in developmental and adult regenerative cardiomyocyte proliferation in the vertebrate heart.


Assuntos
Coração/fisiologia , Histona Desacetilase 1/metabolismo , Miócitos Cardíacos/citologia , Regeneração/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Proliferação de Células
4.
Dev Biol ; 481: 226-237, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34748730

RESUMO

Zebrafish can achieve scar-free healing of heart injuries, and robustly replace all cardiomyocytes lost to injury via dedifferentiation and proliferation of mature cardiomyocytes. Previous studies suggested that Wnt/ß-catenin signaling is active in the injured zebrafish heart, where it induces fibrosis and prevents cardiomyocyte cell cycling. Here, via targeting the destruction complex of the Wnt/ß-catenin pathway with pharmacological and genetic tools, we demonstrate that Wnt/ß-catenin activity is required for cardiomyocyte proliferation and dedifferentiation, as well as for maturation of the scar during regeneration. Using cardiomyocyte-specific conditional inhibition of the pathway, we show that Wnt/ß-catenin signaling acts cell-autonomously to promote cardiomyocyte proliferation. Our results stand in contrast to previous reports and rather support a model in which Wnt/ß-catenin signaling plays a positive role during heart regeneration in zebrafish.


Assuntos
Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Regeneração , Via de Sinalização Wnt , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , beta Catenina/genética
5.
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
6.
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
7.
Int J Mol Sci ; 23(9)2022 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-35562899

RESUMO

Rising incidences and mortalities have drawn attention to Clostridioides difficile infections (CDIs) in recent years. The main virulence factors of this bacterium are the exotoxins TcdA and TcdB, which glucosylate Rho-GTPases and thereby inhibit Rho/actin-mediated processes in cells. This results in cell rounding, gut barrier disruption and characteristic clinical symptoms. So far, treatment of CDIs is limited and mainly restricted to some antibiotics, often leading to a vicious circle of antibiotic-induced disease recurrence. Here, we demonstrate the protective effect of the human antimicrobial peptide α-defensin-6 against TcdA, TcdB and the combination of both toxins in vitro and in vivo and unravel the underlying molecular mechanism. The defensin prevented toxin-mediated glucosylation of Rho-GTPases in cells and protected human cells, model epithelial barriers as well as zebrafish embryos from toxic effects. In vitro analyses revealed direct binding to TcdB in an SPR approach and the rapid formation of TcdB/α-defensin-6 complexes, as analyzed with fluorescent TcdB by time-lapse microscopy. In conclusion, the results imply that α-defensin-6 rapidly sequesters the toxin into complexes, which prevents its cytotoxic activity. These findings extend the understanding of how human peptides neutralize bacterial protein toxins and might be a starting point for the development of novel therapeutic options against CDIs.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Infecções por Clostridium , alfa-Defensinas , Animais , Antibacterianos/farmacologia , Anticorpos Antibacterianos , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Infecções por Clostridium/microbiologia , Enterotoxinas/química , Humanos , Peixe-Zebra/metabolismo , alfa-Defensinas/farmacologia , Proteínas rho de Ligação ao GTP/metabolismo
8.
Int J Mol Sci ; 22(16)2021 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-34445098

RESUMO

Granulysin is an antimicrobial peptide (AMP) expressed by human T-lymphocytes and natural killer cells. Despite a remarkably broad antimicrobial spectrum, its implementation into clinical practice has been hampered by its large size and off-target effects. To circumvent these limitations, we synthesized a 29 amino acid fragment within the putative cytolytic site of Granulysin (termed "Gran1"). We evaluated the antimicrobial activity of Gran1 against the major human pathogen Mycobacterium tuberculosis (Mtb) and a panel of clinically relevant non-tuberculous mycobacteria which are notoriously difficult to treat. Gran1 efficiently inhibited the mycobacterial proliferation in the low micro molar range. Super-resolution fluorescence microscopy and scanning electron microscopy indicated that Gran1 interacts with the surface of Mtb, causing lethal distortions of the cell wall. Importantly, Gran1 showed no off-target effects (cytokine release, chemotaxis, cell death) in primary human cells or zebrafish embryos (cytotoxicity, developmental toxicity, neurotoxicity, cardiotoxicity). Gran1 was selectively internalized by macrophages, the major host cell of Mtb, and restricted the proliferation of the pathogen. Our results demonstrate that the hypothesis-driven design of AMPs is a powerful approach for the identification of small bioactive compounds with specific antimicrobial activity. Gran1 is a promising component for the design of AMP-containing nanoparticles with selective activity and favorable pharmacokinetics to be pushed forward into experimental in vivo models of infectious diseases, most notably tuberculosis.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T/química , Células Cultivadas , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/microbiologia , Mycobacterium tuberculosis/fisiologia , Peptídeos/química , Peptídeos/imunologia , Tuberculose/microbiologia , Peixe-Zebra
9.
Trends Genet ; 31(6): 336-43, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25929514

RESUMO

In contrast to mammals, adult salamanders and fish can completely regenerate their appendages after amputation. The cellular and molecular mechanisms underlying this fascinating phenomenon are beginning to emerge, including substantial progress in the identification of signals that control regenerative growth of the zebrafish caudal fin. Despite the fairly simple architecture of the fin, the regulation of its regeneration is complex. Many signals, including fibroblast growth factor (FGF), Wnt, Hedgehog (Hh), retinoic acid (RA), Notch, bone morphogenic protein (BMP), activin, and insulin-like growth factor (IGF), are required for regeneration. Much work needs to be done to dissect tissue-specific functions of these pathways and how they interact, but Wnt/ß-catenin signaling is already emerging as a central player. Surprisingly, Wnt/ß-catenin signaling appears to largely indirectly control epidermal patterning, progenitor cell proliferation, and osteoblast maturation via regulation of a multitude of secondary signals.


Assuntos
Nadadeiras de Animais/metabolismo , Redes Reguladoras de Genes , Regeneração/genética , Transdução de Sinais/genética , Peixe-Zebra/genética , Amputação Cirúrgica , Nadadeiras de Animais/fisiologia , Nadadeiras de Animais/cirurgia , Animais , Expressão Gênica , Modelos Genéticos , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética
10.
Development ; 141(11): 2225-34, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24821985

RESUMO

Zebrafish have an unlimited capacity to regenerate bone after fin amputation. In this process, mature osteoblasts dedifferentiate to osteogenic precursor cells and thus represent an important source of newly forming bone. By contrast, differentiated osteoblasts do not appear to contribute to repair of bone injuries in mammals; rather, osteoblasts form anew from mesenchymal stem cells. This raises the question whether osteoblast dedifferentiation is specific to appendage regeneration, a special feature of the lepidotrichia bone of the fish fin, or a process found more generally in fish bone. Here, we show that dedifferentiation of mature osteoblasts is not restricted to fin regeneration after amputation, but also occurs during repair of zebrafish fin fractures and skull injuries. In both models, mature osteoblasts surrounding the injury downregulate the expression of differentiation markers, upregulate markers of the pre-osteoblast state and become proliferative. Making use of photoconvertible Kaede protein as well as Cre-driven genetic fate mapping, we show that osteoblasts migrate to the site of injury to replace damaged tissue. Our findings suggest a fundamental role for osteoblast dedifferentiation in reparative bone formation in fish and indicate that adult fish osteoblasts display elevated cellular plasticity compared with mammalian bone-forming cells.


Assuntos
Nadadeiras de Animais/patologia , Osso e Ossos/lesões , Osso e Ossos/patologia , Diferenciação Celular , Osteoblastos/citologia , Crânio/patologia , Animais , Animais Geneticamente Modificados , Regeneração Óssea , Proliferação de Células , Corantes Fluorescentes , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/química , Proteínas Luminescentes/metabolismo , Necrose , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Cicatrização , Peixe-Zebra
11.
Development ; 141(18): 3529-39, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25183871

RESUMO

Canonical ß-catenin-dependent Wnt signal transduction is important for several biological phenomena, such as cell fate determination, cell proliferation, stem cell maintenance and anterior-posterior axis formation. The hallmark of canonical Wnt signaling is the translocation of ß-catenin into the nucleus where it activates gene transcription. However, the mechanisms regulating ß-catenin nuclear localization are poorly understood. We show that Simplet/Fam53B (Smp) is required for Wnt signaling by positively regulating ß-catenin nuclear localization. In the zebrafish embryo, the loss of smp blocks the activity of two ß-catenin-dependent reporters and the expression of Wnt target genes, and prevents nuclear accumulation of ß-catenin. Conversely, overexpression of smp increases ß-catenin nuclear localization and transcriptional activity in vitro and in vivo. Expression of mutant Smp proteins lacking either the nuclear localization signal or the ß-catenin interaction domain reveal that the translocation of Smp into the nucleus is essential for ß-catenin nuclear localization and Wnt signaling in vivo. We also provide evidence that mammalian Smp is involved in regulating ß-catenin nuclear localization: the protein colocalizes with ß-catenin-dependent gene expression in mouse intestinal crypts; siRNA knockdown of Smp reduces ß-catenin nuclear localization and transcriptional activity; human SMP mediates ß-catenin transcriptional activity in a dose-dependent manner; and the human SMP protein interacts with human ß-catenin primarily in the nucleus. Thus, our findings identify the evolutionary conserved SMP protein as a regulator of ß-catenin-dependent Wnt signal transduction.


Assuntos
Núcleo Celular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Western Blotting , Humanos , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ , Luciferases , Camundongos , Camundongos Transgênicos , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Wnt/genética
12.
Development ; 140(7): 1412-23, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23462472

RESUMO

Zebrafish can completely regenerate amputated fins via formation of a blastema, a proliferative mass of undifferentiated precursor cells. During regenerative growth, blastema proliferation must be tightly coordinated with cellular differentiation, but little is known about how this is achieved. Here, we show that Notch signaling is essential for maintenance of blastema cells in a proliferative undifferentiated state. We found that the Notch pathway is activated in response to fin amputation in the highly proliferative region of the blastema. Chemical interference with Notch signaling resulted in a complete block of regeneration. Notch signaling was not required for the earliest known cellular processes during blastema formation, i.e. dedifferentiation and migration of osteoblasts, but specifically interfered with proliferation of blastema cells. Interestingly, overactivation of the pathway via misexpression of the intracellular domain of the Notch receptor (NICD) likewise inhibited regenerative outgrowth. In NICD-overexpressing fins, overall blastemal cell proliferation was not enhanced, but expanded into proximal regions where cellular differentiation normally occurs. Similarly, blastemal and epidermal gene expression territories invaded proximal regions upon sustained Notch activation. Concomitantly, NICD overexpression suppressed differentiation of osteoblasts and caused an expansion of the undifferentiated blastema. Together, these data suggest that Notch signaling activity maintains blastemal cells in a proliferative state and thus coordinates proliferation with differentiation during regenerative growth.


Assuntos
Nadadeiras de Animais/fisiologia , Diferenciação Celular/genética , Proliferação de Células , Receptores Notch/fisiologia , Regeneração/genética , Peixe-Zebra , Alanina/análogos & derivados , Alanina/farmacologia , Nadadeiras de Animais/efeitos dos fármacos , Nadadeiras de Animais/metabolismo , Animais , Animais Geneticamente Modificados , Azepinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Modelos Biológicos , Receptores Notch/genética , Receptores Notch/metabolismo , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Cicatrização/efeitos dos fármacos , Cicatrização/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia
13.
Blood ; 121(13): 2462-73, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23365460

RESUMO

Regulated migration of hematopoietic stem cells is fundamental for hematopoiesis. The molecular mechanisms underlying stem cell trafficking are poorly defined. Based on a short hairpin RNA library and stromal cell-derived factor-1 (SDF-1) migration screening assay, we identified the histone 3 lysine 27 demethylase UTX (Kdm6a) as a novel regulator for hematopoietic cell migration. Using hematopoietic stem and progenitor cells from our conditional UTX knockout (KO) mice, we were able to confirm the regulatory function of UTX on cell migration. Moreover, adult female conditional UTX KO mice displayed myelodysplasia and splenic erythropoiesis, whereas UTX KO males showed no phenotype. During development, all UTX KO female and a portion of UTX KO male embryos developed a cardiac defect, cranioschisis, and died in utero. Therefore, UTY, the male homolog of UTX, can compensate for UTX in adults and partially during development. Additionally, we found that UTX knockdown in zebrafish significantly impairs SDF-1/CXCR4-dependent migration of primordial germ cells. Our data suggest that UTX is a critical regulator for stem cell migration and hematopoiesis.


Assuntos
Movimento Celular/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/fisiologia , Histona Desmetilases/fisiologia , Animais , Células Cultivadas , Embrião não Mamífero , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Células HEK293 , Células-Tronco Hematopoéticas/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peixe-Zebra/embriologia , Peixe-Zebra/genética
14.
Cell Rep ; 43(5): 114162, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38678558

RESUMO

Zebrafish have a lifelong cardiac regenerative ability after damage, whereas mammals lose this capacity during early postnatal development. This study investigated whether the declining expression of growth factors during postnatal mammalian development contributes to the decrease of cardiomyocyte regenerative potential. Besides confirming the proliferative ability of neuregulin 1 (NRG1), interleukin (IL)1b, receptor activator of nuclear factor kappa-Β ligand (RANKL), insulin growth factor (IGF)2, and IL6, we identified other potential pro-regenerative factors, with BMP7 exhibiting the most pronounced efficacy. Bmp7 knockdown in neonatal mouse cardiomyocytes and loss-of-function in adult zebrafish during cardiac regeneration reduced cardiomyocyte proliferation, indicating that Bmp7 is crucial in the regenerative stages of mouse and zebrafish hearts. Conversely, bmp7 overexpression in regenerating zebrafish or administration at post-mitotic juvenile and adult mouse stages, in vitro and in vivo following myocardial infarction, enhanced cardiomyocyte cycling. Mechanistically, BMP7 stimulated proliferation through BMPR1A/ACVR1 and ACVR2A/BMPR2 receptors and downstream SMAD5, ERK, and AKT signaling. Overall, BMP7 administration is a promising strategy for heart regeneration.


Assuntos
Proteína Morfogenética Óssea 7 , Proliferação de Células , Miócitos Cardíacos , Regeneração , Peixe-Zebra , Animais , Feminino , Masculino , Camundongos , Proteína Morfogenética Óssea 7/metabolismo , Proteína Morfogenética Óssea 7/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Neuregulina-1/metabolismo , Neuregulina-1/genética , Transdução de Sinais , Proteína Smad5/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
15.
Dev Biol ; 366(2): 327-40, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22546689

RESUMO

The creation of molecular tools able to unravel in vivo spatiotemporal activation of specific cell signaling events during cell migration, differentiation and morphogenesis is of great relevance to developmental cell biology. Here, we describe the generation, validation and applications of two transgenic reporter lines for Wnt/ß-catenin signaling, named TCFsiam, and show that they are reliable and sensitive Wnt biosensors for in vivo studies. We demonstrate that these lines sensitively detect Wnt/ß-catenin pathway activity in several cellular contexts, from sensory organs to cardiac valve patterning. We provide evidence that Wnt/ß-catenin activity is involved in the formation and maintenance of the zebrafish CNS blood vessel network, on which sox10 neural crest-derived cells migrate and proliferate. We finally show that these transgenic lines allow for screening of Wnt signaling modifying compounds, tissue regeneration assessment as well as evaluation of potential Wnt/ß-catenin genetic modulators.


Assuntos
Via de Sinalização Wnt , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Técnicas Biossensoriais , Diferenciação Celular , Movimento Celular , Neurônios/citologia , Neurônios/fisiologia , Peixe-Zebra/embriologia
16.
Proc Natl Acad Sci U S A ; 107(46): 19933-8, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21041642

RESUMO

Systems for spatial and temporal control of gene expression are essential for developmental studies and are of particular importance for research in adult model organisms. We present two modified dually inducible TetON systems for tissue-specific conditional control of gene expression in zebrafish based on (i) a tetracycline inducible transcriptional activator (TetActivator) fused to the ligand binding domain of a mutated glucocorticoid receptor (TetA-GBD) and (ii) a TetActivator fused with a domain of the Ecdysone receptor (TetA-EcR). Both systems showed strong induction of tetracycline-responsive promoters upon administration of the appropriate ligands (doxycycline and dexamethasone for TetA-GBD, and doxycycline and tebufenozide for TetA-EcR), and undetectable leakiness when compared with classical TetActivators. Combinations of transgenic lines expressing TetA-GBD specifically in the heart or the CNS with different Tet-responsive transgenic lines allows conditional and tissue-specific control of gene expression in embryos and adults. Importantly, induction is fully reversible and tunable by the doses of drugs used. The TetA-EcR system avoids the possible side effects of dexamethasone and displays improved sensitivity both in zebrafish and in mammalian cells. These results show that dually inducible TetON systems are convenient tools for reversible and very tightly controlled conditional gene expression in zebrafish.


Assuntos
Expressão Gênica/efeitos dos fármacos , Técnicas Genéticas , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Tetraciclina/farmacologia , Peixe-Zebra/genética , Envelhecimento/efeitos dos fármacos , Animais , Embrião não Mamífero , Receptores de Glucocorticoides/metabolismo , Receptores de Esteroides/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transgenes/genética , Peixe-Zebra/embriologia
17.
J Med Chem ; 66(22): 15189-15204, 2023 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-37940118

RESUMO

EPI-X4, a natural peptide CXCR4 antagonist, shows potential for treating inflammation and cancer, but its short plasma stability limits its clinical application. We aimed to improve the plasma stability of EPI-X4 analogues without compromising CXCR4 antagonism. Our findings revealed that only the peptide N-terminus is prone to degradation. Consequently, incorporating d-amino acids or acetyl groups in this region enhanced peptide stability in plasma. Notably, EPI-X4 leads 5, 27, and 28 not only retained their CXCR4 binding and antagonism but also remained stable in plasma for over 8 h. Molecular dynamic simulations showed that these modified analogues bind similarly to CXCR4 as the original peptide. To further increase their systemic half-lives, we conjugated these stabilized analogues with large polymers and albumin binders. These advances highlight the potential of the optimized EPI-X4 analogues as promising CXCR4-targeted therapeutics and set the stage for more detailed preclinical assessments.


Assuntos
Infecções por HIV , HIV-1 , Humanos , HIV-1/metabolismo , Peptídeos/química , Receptores CXCR4/metabolismo , Albuminas/metabolismo , Transdução de Sinais , Aminas/metabolismo
18.
Artigo em Inglês | MEDLINE | ID: mdl-34649924

RESUMO

The zebrafish caudal fin has become a popular model to study cellular and molecular mechanisms of regeneration due to its high regenerative capacity, accessibility for experimental manipulations, and relatively simple anatomy. The formation of a regenerative epidermis and blastema are crucial initial events and tightly regulated. Both the regenerative epidermis and the blastema are highly organized structures containing distinct domains, and several signaling pathways regulate the formation and interaction of these domains. Bone is the major tissue regenerated from the progenitor cells of the blastema. Several cellular mechanisms can provide source cells for blastemal (pre-)osteoblasts, including dedifferentiation of differentiated osteoblasts and de novo formation from other cell types, providing intriguing examples of cellular plasticity. In recent years, omics analyses and single-cell approaches have elucidated genetic and epigenetic regulation, increasing our knowledge of the surprisingly complex coordination of various mechanisms to achieve successful restoration of a seemingly simple structure.


Assuntos
Epigênese Genética , Peixe-Zebra , Animais , Diferenciação Celular/genética , Transdução de Sinais/fisiologia , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra
19.
Elife ; 112022 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-35748539

RESUMO

Successful regeneration requires the coordinated execution of multiple cellular responses to injury. In amputated zebrafish fins, mature osteoblasts dedifferentiate, migrate towards the injury, and form proliferative osteogenic blastema cells. We show that osteoblast migration is preceded by cell elongation and alignment along the proximodistal axis, which require actomyosin, but not microtubule (MT) turnover. Surprisingly, osteoblast dedifferentiation and migration can be uncoupled. Using pharmacological and genetic interventions, we found that NF-ĸB and retinoic acid signalling regulate dedifferentiation without affecting migration, while the complement system and actomyosin dynamics affect migration but not dedifferentiation. Furthermore, by removing bone at two locations within a fin ray, we established an injury model containing two injury sites. We found that osteoblasts dedifferentiate at and migrate towards both sites, while accumulation of osteogenic progenitor cells and regenerative bone formation only occur at the distal-facing injury. Together, these data indicate that osteoblast dedifferentiation and migration represent generic injury responses that are differentially regulated and can occur independently of each other and of regenerative growth. We conclude that successful fin bone regeneration appears to involve the coordinated execution of generic and regeneration-specific responses of osteoblasts to injury.


Assuntos
Actomiosina , Peixe-Zebra , Nadadeiras de Animais/fisiologia , Animais , Osteoblastos , Osteogênese , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
20.
Birth Defects Res A Clin Mol Teratol ; 91(6): 435-40, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21567896

RESUMO

In the developing heart, the atrioventricular canal (AVC) is essential for separation and alignment of the cardiac chambers, for valve formation, and serves to delay the electrical impulse from the atria to the ventricles. Defects in various aspects of its formation are the most common form of congenital heart defects. Using mutant and transgenic approaches in zebrafish, this study demonstrates that Wnt/ß-catenin signaling is both sufficient and required for the induction of BMP4 and Tbx2b expression in the AVC and consequently the proper patterning of the myocardium. Furthermore, genetic analysis shows that Wnt/ß-catenin signaling is upstream and in a linear pathway with BMP and Tbx2 during AVC specification.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Proteínas com Domínio T/genética , Via de Sinalização Wnt , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Receptores de Ativinas Tipo I , Animais , Proteína Morfogenética Óssea 4/genética , Receptores de Proteínas Morfogenéticas Ósseas/genética , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Átrios do Coração/embriologia , Valvas Cardíacas/embriologia , Mutação , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , beta Catenina/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA