Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Am Chem Soc ; 145(41): 22659-22670, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37812759

RESUMEN

Lipid nanoparticles (LNPs) are becoming widely adopted as vectors for the delivery of therapeutic payloads but generally lack intrinsic tissue-homing properties. These extracellular vesicle (EV) mimetics can be targeted toward the liver, lung, or spleen via charge modification of their lipid headgroups. Homing to other tissues has only been achieved via covalent surface modification strategies using small-molecule ligands, peptides, or monoclonal antibodies─methods that are challenging to couple with large-scale manufacturing. Herein, we design a novel modular artificial membrane-binding protein (AMBP) platform for the modification of LNPs postformation. The system is composed of two protein modules that can be readily coupled using bioorthogonal chemistry to yield the AMBP. The first is a membrane anchor module comprising a supercharged green fluorescent protein (scGFP) electrostatically conjugated to a dynamic polymer surfactant corona. The second is a functional module containing a cardiac tissue fibronectin homing sequence from the bacterial adhesin CshA. We demonstrate that LNPs modified using the AMBP exhibit a 20-fold increase in uptake by fibronectin-rich C2C12 cells under static conditions and a 10-fold increase under physiologically relevant shear stresses, with no loss of cell viability. Moreover, we show targeted localization of the AMBP-modified LNPs in zebrafish hearts, highlighting their therapeutic potential as a vector for the treatment of cardiac disease and, more generally, as a smart vector.


Asunto(s)
Fibronectinas , Nanopartículas , Animales , Pez Cebra , Liposomas , Nanopartículas/química , ARN Interferente Pequeño/química
2.
BMC Biol ; 20(1): 21, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35057801

RESUMEN

BACKGROUND: Scales are mineralised exoskeletal structures that are part of the dermal skeleton. Scales have been mostly lost during evolution of terrestrial vertebrates whilst bony fish have retained a mineralised dermal skeleton in the form of fin rays and scales. Each scale is a mineralised collagen plate that is decorated with both matrix-building and resorbing cells. When removed, an ontogenetic scale is quickly replaced following differentiation of the scale pocket-lining cells that regenerate a scale. Processes promoting de novo matrix formation and mineralisation initiated during scale regeneration are poorly understood. Therefore, we performed transcriptomic analysis to determine gene networks and their pathways involved in dermal scale regeneration. RESULTS: We defined the transcriptomic profiles of ontogenetic and regenerating scales of zebrafish and identified 604 differentially expressed genes (DEGs). These were enriched for extracellular matrix, ossification, and cell adhesion pathways, but not in enamel or dentin formation processes indicating that scales are reminiscent to bone. Hypergeometric tests involving monogenetic skeletal disorders showed that DEGs were strongly enriched for human orthologues that are mutated in low bone mass and abnormal bone mineralisation diseases (P< 2× 10-3). The DEGs were also enriched for human orthologues associated with polygenetic skeletal traits, including height (P< 6× 10-4), and estimated bone mineral density (eBMD, P< 2× 10-5). Zebrafish mutants of two human orthologues that were robustly associated with height (COL11A2, P=6× 10-24) or eBMD (SPP1, P=6× 10-20) showed both exo- and endo- skeletal abnormalities as predicted by our genetic association analyses; col11a2Y228X/Y228X mutants showed exoskeletal and endoskeletal features consistent with abnormal growth, whereas spp1P160X/P160X mutants predominantly showed mineralisation defects. CONCLUSION: We show that scales have a strong osteogenic expression profile comparable to other elements of the dermal skeleton, enriched in genes that favour collagen matrix growth. Despite the many differences between scale and endoskeletal developmental processes, we also show that zebrafish scales express an evolutionarily conserved sub-population of genes that are relevant to human skeletal disease.


Asunto(s)
Proteínas de Pez Cebra , Pez Cebra , Animales , Perfilación de la Expresión Génica , Humanos , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 41(9): 2454-2468, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34261327

RESUMEN

Objective: Extracellular vesicles (EVs) facilitate molecular transport across extracellular space, allowing local and systemic signaling during homeostasis and in disease. Extensive studies have described functional roles for EV populations, including during cardiovascular disease, but the in vivo characterization of endogenously produced EVs is still in its infancy. Because of their genetic tractability and live imaging amenability, zebrafish represent an ideal but under-used model to investigate endogenous EVs. We aimed to establish a transgenic zebrafish model to allow the in vivo identification, tracking, and extraction of endogenous EVs produced by different cell types. Approach and Results: Using a membrane-tethered fluorophore reporter system, we show that EVs can be fluorescently labeled in larval and adult zebrafish and demonstrate that multiple cell types including endothelial cells and cardiomyocytes actively produce EVs in vivo. Cell-type specific EVs can be tracked by high spatiotemporal resolution light-sheet live imaging and modified flow cytometry methods allow these EVs to be further evaluated. Additionally, cryo electron microscopy reveals the full morphological diversity of larval and adult EVs. Importantly, we demonstrate the utility of this model by showing that different cell types exchange EVs in the adult heart and that ischemic injury models dynamically alter EV production. Conclusions: We describe a powerful in vivo zebrafish model for the investigation of endogenous EVs in all aspects of cardiovascular biology and pathology. A cell membrane fluorophore labeling approach allows cell-type specific tracing of EV origin without bias toward the expression of individual protein markers and will allow detailed future examination of their function.


Asunto(s)
Sistema Cardiovascular/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Sistema Cardiovascular/embriología , Separación Celular , Microscopía por Crioelectrón , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Vesículas Extracelulares/genética , Vesículas Extracelulares/ultraestructura , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Larva/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
4.
Histochem Cell Biol ; 154(5): 533-548, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32926230

RESUMEN

The study of heart repair post-myocardial infarction has historically focused on the importance of cardiomyocyte proliferation as the major factor limiting adult mammalian heart regeneration. However, there is mounting evidence that a narrow focus on this one cell type discounts the importance of a complex cascade of cell-cell communication involving a whole host of different cell types. A major difficulty in the study of heart regeneration is the rarity of this process in adult animals, meaning a mammalian template for how this can be achieved is lacking. Here, we review the adult zebrafish as an ideal and unique model in which to study the underlying mechanisms and cell types required to attain complete heart regeneration following cardiac injury. We provide an introduction to the role of the cardiac microenvironment in the complex regenerative process and discuss some of the key advances using this in vivo vertebrate model that have recently increased our understanding of the vital roles of multiple different cell types. Due to the sheer number of exciting studies describing new and unexpected roles for inflammatory cell populations in cardiac regeneration, this review will pay particular attention to these important microenvironment participants.


Asunto(s)
Corazón/crecimiento & desarrollo , Miocitos Cardíacos/metabolismo , Regeneración , Pez Cebra , Animales , Microambiente Celular , Modelos Animales de Enfermedad , Miocitos Cardíacos/patología
5.
Nat Genet ; 38(11): 1329-34, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17041603

RESUMEN

The epidermis is a highly organized structure, the integrity of which is central to the protection of an organism. Development and subsequent maintenance of this tissue depends critically on the intricate balance between proliferation and differentiation of a resident stem cell population; however, the signals controlling the proliferation-differentiation switch in vivo remain elusive. Here, we show that mice carrying a homozygous missense mutation in interferon regulatory factor 6 (Irf6), the homolog of the gene mutated in the human congenital disorders Van der Woude syndrome and popliteal pterygium syndrome, have a hyperproliferative epidermis that fails to undergo terminal differentiation, resulting in soft tissue fusions. We further demonstrate that mice that are compound heterozygotes for mutations in Irf6 and the gene encoding the cell cycle regulator protein stratifin (Sfn; also known as 14-3-3sigma) show similar defects of keratinizing epithelia. Our results indicate that Irf6 is a key determinant of the keratinocyte proliferation-differentiation switch and that Irf6 and Sfn interact genetically in this process.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular , Factores Reguladores del Interferón/fisiología , Queratinocitos/citología , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Proteínas 14-3-3/fisiología , Animales , Células Cultivadas , Fisura del Paladar/genética , Epidermis/embriología , Femenino , Genes de Cambio/fisiología , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/fisiología , Factores Reguladores del Interferón/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Anomalías de la Boca/genética , Mutación Missense , Embarazo
6.
Nat Genet ; 32(2): 285-9, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12219090

RESUMEN

Interferon regulatory factor 6 (IRF6) belongs to a family of nine transcription factors that share a highly conserved helix-turn-helix DNA-binding domain and a less conserved protein-binding domain. Most IRFs regulate the expression of interferon-alpha and -beta after viral infection, but the function of IRF6 is unknown. The gene encoding IRF6 is located in the critical region for the Van der Woude syndrome (VWS; OMIM 119300) locus at chromosome 1q32-q41 (refs 2,3). The disorder is an autosomal dominant form of cleft lip and palate with lip pits, and is the most common syndromic form of cleft lip or palate. Popliteal pterygium syndrome (PPS; OMIM 119500) is a disorder with a similar orofacial phenotype that also includes skin and genital anomalies. Phenotypic overlap and linkage data suggest that these two disorders are allelic. We found a nonsense mutation in IRF6 in the affected twin of a pair of monozygotic twins who were discordant for VWS. Subsequently, we identified mutations in IRF6 in 45 additional unrelated families affected with VWS and distinct mutations in 13 families affected with PPS. Expression analyses showed high levels of Irf6 mRNA along the medial edge of the fusing palate, tooth buds, hair follicles, genitalia and skin. Our observations demonstrate that haploinsufficiency of IRF6 disrupts orofacial development and are consistent with dominant-negative mutations disturbing development of the skin and genitalia.


Asunto(s)
Labio Leporino/genética , Fisura del Paladar/genética , Proteínas de Unión al ADN/genética , Genitales/anomalías , Anomalías Cutáneas/genética , Factores de Transcripción/genética , Animales , Sitios de Unión/genética , Sitios de Unión/fisiología , Northern Blotting , ADN/metabolismo , Enfermedades en Gemelos/genética , Femenino , Humanos , Hibridación in Situ , Factores Reguladores del Interferón , Masculino , Ratones , Mutación Missense , Linaje , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Relación Estructura-Actividad , Síndrome , Gemelos Monocigóticos/genética
7.
Hum Mol Genet ; 18(14): 2632-42, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19439425

RESUMEN

In mammals, adhesion and fusion of the palatal shelves are essential mechanisms during the development of the secondary palate; failure of these processes leads to the congenital anomaly, cleft palate. The mechanisms that prevent pathological adhesion between the oral and palatal epithelia while permitting adhesion and subsequent fusion of the palatal shelves via their medial edge epithelia remain obscure. In humans, mutations in the transcription factor interferon regulatory factor 6 (IRF6) underlie Van der Woude syndrome and popliteal pterygium syndrome. Recently, we have demonstrated that mice homozygous for a mutation in Irf6 exhibit abnormalities of epithelial differentiation that results in cleft palate as a consequence of adhesion between the palatal shelves and the tongue. In the current paper, we demonstrate that Irf6 is essential for oral epithelial differentiation and that IRF6 and the Notch ligand Jagged2 function in convergent molecular pathways during this process. We further demonstrate that IRF6 plays a key role in the formation and maintenance of the oral periderm, spatio-temporal regulation of which is essential for ensuring appropriate palatal adhesion.


Asunto(s)
Fisura del Paladar/metabolismo , Factores Reguladores del Interferón/metabolismo , Proteínas de la Membrana/metabolismo , Hueso Paladar/crecimiento & desarrollo , Transducción de Señal , Animales , Adhesión Celular , Fisura del Paladar/embriología , Fisura del Paladar/genética , Femenino , Humanos , Factores Reguladores del Interferón/genética , Proteína Jagged-2 , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Hueso Paladar/embriología , Hueso Paladar/metabolismo
8.
Immunohorizons ; 4(8): 464-474, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32769178

RESUMEN

Transgenic zebrafish that express fluorophores under the control of mpeg1.1 (mpeg1) and csf1ra (c-fms) promoters have been widely used to study the dynamics and functions of mononuclear phagocytes (MNPs) in larval zebrafish, unveiling crucial roles for these innate immune cells in many processes, including tissue repair. Adult zebrafish are also being increasingly used as a model organism for such studies because of their regenerative capacity and presence of innate and adaptive immune cells. For example, recent investigations highlight roles of MNPs in the regulation of diverse cellular processes during heart regeneration, including scarring, cardiomyocyte proliferation, and neovascularization. However, transgenic lines that stratify MNP subpopulations (monocytes, macrophages, and dendritic cells) are not yet available, preventing functional analysis of these populations. In an attempt to better segregate cardiac MNPs, we assessed the coexpression of mpeg1.1 and csf1ra reporter transgenes in adult zebrafish hearts. Unexpectedly, this also identified a discrete population of mpeg1.1 + csf1ra - lymphoid-like cells, which respond to cardiac cryoinjury in a different temporal pattern to mpeg1.1 + MNPs. mpeg1.1 + lymphoid cells were also abundant in the skin, spleen, and blood, and their frequency was unaffected in the hearts of csf1raj4e1/j4e1 mutant zebrafish, which display deficiencies in MNP populations. Flow cytometry, imaging, and cytological and gene expression analyses collectively indicate that these cells comprise a mixed population of B cells and NK-like cells. Our study therefore highlights the need to identify novel MNP lineage markers but also suggests undetermined roles of B cells and NK-like cells in cardiac homeostasis and repair in adult zebrafish.


Asunto(s)
Linfocitos/metabolismo , Proteínas de la Membrana/metabolismo , Miocardio/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Células Dendríticas/metabolismo , Regulación de la Expresión Génica , Macrófagos/metabolismo , Proteínas de la Membrana/genética , Monocitos/metabolismo , Mutación , Miocardio/citología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas de Pez Cebra/genética
9.
Cardiovasc Res ; 116(7): 1357-1371, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31566660

RESUMEN

AIMS: A robust inflammatory response to tissue injury is a necessary part of the repair process but the deposition of scar tissue is a direct downstream consequence of this response in many tissues including the heart. Adult zebrafish not only possess the capacity to regenerate lost cardiomyocytes but also to remodel and resolve an extracellular scar within tissues such as the heart, but this scar resolution process remains poorly understood. This study aims to characterize the scarring and inflammatory responses to cardiac damage in adult zebrafish in full and investigate the role of different inflammatory subsets specifically in scarring and scar removal. METHODS AND RESULTS: Using stable transgenic lines, whole organ imaging and genetic and pharmacological interventions, we demonstrate that multiple inflammatory cell lineages respond to cardiac injury in adult zebrafish. In particular, macrophage subsets (tnfα+ and tnfα-) play prominent roles with manipulation of different phenotypes suggesting that pro-inflammatory (tnfα+) macrophages promote scar deposition following cardiac injury whereas tnfα- macrophages facilitate scar removal during regeneration. Detailed analysis of these specific macrophage subsets reveals crucial roles for Csf1ra in promoting pro-inflammatory macrophage-mediated scar deposition. Additionally, the multifunctional cytokine Osteopontin (Opn) (spp1) is important for initial scar deposition but also for resolution of the inflammatory response and in late-stage ventricular collagen remodelling. CONCLUSIONS: This study demonstrates the importance of a correctly balanced inflammatory response to facilitate scar deposition during repair but also to allow subsequent scar resolution, and full cardiac regeneration, to occur. We have identified Opn as having both pro-fibrotic but also potentially pro-regenerative roles in the adult zebrafish heart, driving Collagen deposition but also controlling inflammatory cell resolution.


Asunto(s)
Linaje de la Célula , Cicatriz/patología , Lesiones Cardíacas/patología , Macrófagos/patología , Miocardio/patología , Remodelación Ventricular , Animales , Animales Modificados Genéticamente , Cicatriz/metabolismo , Cicatriz/fisiopatología , Colágeno/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/fisiopatología , Macrófagos/metabolismo , Miocardio/metabolismo , Osteopontina/genética , Osteopontina/metabolismo , Fenotipo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
10.
Front Cell Dev Biol ; 7: 12, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30805338

RESUMEN

The zebrafish has emerged as an exciting vertebrate model to study different aspects of immune system development, particularly due to its transparent embryonic development, the availability of multiple fluorescent reporter lines, efficient genetic tools and live imaging capabilities. However, the study of immunity in zebrafish has largely been limited to early larval stages due to an incomplete knowledge of the full repertoire of immune cells and their specific markers, in particular, a lack of cell surface antibodies to detect and isolate such cells in living tissues. Here we focus on tissue resident or associated immunity beyond development, in the adult zebrafish. It is our view that, with our increasing knowledge and the development of improved tools and protocols, the adult zebrafish will be increasingly appreciated for offering valuable insights into the role of immunity in tissue repair and maintenance, in both health and disease throughout the lifecourse.

11.
NPJ Regen Med ; 3: 21, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30416753

RESUMEN

The cellular events that contribute to tissue healing of non-sterile wounds to the skin and ischaemic injury to internal organs such as the heart share remarkable similarities despite the differences between these injury types and organs. In adult vertebrates, both injuries are characterised by a complex series of overlapping events involving multiple different cell types and cellular interactions. In adult mammals both tissue-healing processes ultimately lead to the permanent formation of a fibrotic, collagenous scar, which can have varying effects on tissue function depending on the site and magnitude of damage. Extensive scarring in the heart as a result of a severe myocardial infarction contributes to ventricular dysfunction and the progression of heart failure. Some vertebrates such as adult zebrafish, however, retain a more embryonic capacity for scar-free tissue regeneration in many tissues including the skin and heart. In this review, the similarities and differences between these different types of wound healing are discussed, with special attention on recent advances in regenerative, non-scarring vertebrate models such as the zebrafish.

12.
J Clin Invest ; 124(9): 3891-900, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25133425

RESUMEN

Appropriate development of stratified, squamous, keratinizing epithelia, such as the epidermis and oral epithelia, generates an outer protective permeability barrier that prevents water loss, entry of toxins, and microbial invasion. During embryogenesis, the immature ectoderm initially consists of a single layer of undifferentiated, cuboidal epithelial cells that stratifies to produce an outer layer of flattened periderm cells of unknown function. Here, we determined that periderm cells form in a distinct pattern early in embryogenesis, exhibit highly polarized expression of adhesion complexes, and are shed from the outer surface of the embryo late in development. Mice carrying loss-of-function mutations in the genes encoding IFN regulatory factor 6 (IRF6), IκB kinase-α (IKKα), and stratifin (SFN) exhibit abnormal epidermal development, and we determined that mutant animals exhibit dysfunctional periderm formation, resulting in abnormal intracellular adhesions. Furthermore, tissue from a fetus with cocoon syndrome, a lethal disorder that results from a nonsense mutation in IKKA, revealed an absence of periderm. Together, these data indicate that periderm plays a transient but fundamental role during embryogenesis by acting as a protective barrier that prevents pathological adhesion between immature, adhesion-competent epithelia. Furthermore, this study suggests that failure of periderm formation underlies a series of devastating birth defects, including popliteal pterygium syndrome, cocoon syndrome, and Bartsocas-Papas syndrome.


Asunto(s)
Desarrollo Embrionario , Epidermis/embriología , Proteínas 14-3-3/fisiología , Animales , Adhesión Celular , Polaridad Celular , Ectodermo/embriología , Células Epidérmicas , Epitelio/embriología , Epitelio/fisiología , Humanos , Quinasa I-kappa B/fisiología , Factores Reguladores del Interferón/fisiología , Ratones , Mutación
13.
J Invest Dermatol ; 134(5): 1313-1322, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24232570

RESUMEN

Fraser syndrome (FS) is a phenotypically variable, autosomal recessive disorder characterized by cryptophthalmus, cutaneous syndactyly, and other malformations resulting from mutations in FRAS1, FREM2, and GRIP1. Transient embryonic epidermal blistering causes the characteristic defects of the disorder. Fras1, Frem1, and Frem2 form the extracellular Fraser complex, which is believed to stabilize the basement membrane. However, several cases of FS could not be attributed to mutations in FRAS1, FREM2, or GRIP1, and FS displays high clinical variability, suggesting that there is an additional genetic, possibly modifying contribution to this disorder. An extracellular matrix protein containing VWA-like domains related to those in matrilins and collagens (AMACO), encoded by the VWA2 gene, has a very similar tissue distribution to the Fraser complex proteins in both mouse and zebrafish. Here, we show that AMACO deposition is lost in Fras1-deficient zebrafish and mice and that Fras1 and AMACO interact directly via their chondroitin sulfate proteoglycan (CSPG) and P2 domains. Knockdown of vwa2, which alone causes no phenotype, enhances the phenotype of hypomorphic Fras1 mutant zebrafish. Together, our data suggest that AMACO represents a member of the Fraser complex.


Asunto(s)
Membrana Basal/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Síndrome de Fraser/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biomarcadores de Tumor , Proteínas de Unión al Calcio , Matriz Extracelular/metabolismo , Femenino , Síndrome de Fraser/genética , Técnicas de Silenciamiento del Gen , Genes Recesivos , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fenotipo , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA