Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell Mol Life Sci ; 80(3): 79, 2023 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-36867267

RESUMEN

Pulmonary neuroendocrine (NE) cells represent a small population in the airway epithelium, but despite this, hyperplasia of NE cells is associated with several lung diseases, such as congenital diaphragmatic hernia and bronchopulmonary dysplasia. The molecular mechanisms causing the development of NE cell hyperplasia remains poorly understood. Previously, we showed that the SOX21 modulates the SOX2-initiated differentiation of epithelial cells in the airways. Here, we show that precursor NE cells start to develop in the SOX2 + SOX21 + airway region and that SOX21 suppresses the differentiation of airway progenitors to precursor NE cells. During development, clusters of NE cells start to form and NE cells mature by expressing neuropeptide proteins, such as CGRP. Deficiency in SOX2 resulted in decreased clustering, while deficiency in SOX21 increased both the numbers of NE ASCL1 + precursor cells early in development, and the number of mature cell clusters at E18.5. In addition, at the end of gestation (E18.5), a number of NE cells in Sox2 heterozygous mice, did not yet express CGRP suggesting a delay in maturation. In conclusion, SOX2 and SOX21 function in the initiation, migration and maturation of NE cells.


Asunto(s)
Células Neuroendocrinas , Factores de Transcripción SOXB1 , Factores de Transcripción SOXB2 , Animales , Ratones , Péptido Relacionado con Gen de Calcitonina , Diferenciación Celular/genética , Epitelio , Hiperplasia , Células Neuroendocrinas/citología , Células Neuroendocrinas/metabolismo
2.
Int J Mol Sci ; 23(21)2022 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-36361852

RESUMEN

The lung originates from the ventral foregut and develops into an intricate branched structure of airways, alveoli, vessels and support tissue. As the lung develops, cells become specified and differentiate into the various cell lineages. This process is controlled by specific transcription factors, such as the SRY-related HMG-box genes SOX2 and SOX21, that are activated or repressed through intrinsic and extrinsic signals. Disturbances in any of these processes during the development of the lung may lead to various pediatric lung disorders, such as Congenital Diaphragmatic Hernia (CDH), Congenital Pulmonary Airway Malformation (CPAM) and Broncho-Pulmonary Dysplasia (BPD). Changes in the composition of the airways and the alveoli may result in reduced respiratory function and eventually lead to chronic lung disorders. In this concise review, we describe different intrinsic and extrinsic cellular processes required for proper differentiation of the epithelium during development and regeneration, and the influence of the microenvironment on this process with special focus on SOX2 and SOX21.


Asunto(s)
Hernias Diafragmáticas Congénitas , Enfermedades Pulmonares , Humanos , Niño , Diferenciación Celular/genética , Factores de Transcripción , Alveolos Pulmonares , Pulmón , Enfermedades Pulmonares/genética , Factores de Transcripción SOXB1/genética
3.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L775-L786, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34378410

RESUMEN

Air-liquid interface (ALI) cultures are frequently used in lung research but require substantial cell numbers that cannot readily be obtained from patients. We explored whether organoid expansion [three-dimensional (3D)] can be used to establish ALI cultures from clinical samples with low epithelial cell numbers. Airway epithelial cells were obtained from tracheal aspirates (TA) from preterm newborns and from bronchoalveolar lavage (BAL) or bronchial tissue (BT) from adults. TA and BAL cells were 3D-expanded, whereas cells from BT were expanded in 3D and 2D. Following expansion, cells were cultured at ALI to induce differentiation. The impact of cell origin and 2D or 3D expansion was assessed with respect to 1) cellular composition, 2) response to cigarette smoke exposure, and 3) effect of Notch inhibition or IL-13 stimulation on cellular differentiation. We established well-differentiated ALI cultures from all samples. Cellular compositions (basal, ciliated, and goblet cells) were comparable. All 3D-expanded cultures showed a similar stress response following cigarette smoke exposure but differed from the 2D-expanded cultures. Higher peak levels of antioxidant genes HMOX1 and NQO1 and a more rapid return to baseline, and a lower unfolded protein response was observed after cigarette smoke exposure in 3D-derived cultures compared to 2D-derived cultures. In addition, TA- and BAL-derived cultures were less sensitive to modulation by DAPT or IL-13 than BT-derived cultures. Organoid-based expansion of clinical samples with low cell numbers, such as TA from preterm newborns is a valid method and tool to establish ALI cultures.


Asunto(s)
Bronquios/citología , Células Epiteliales/citología , Organoides/citología , Mucosa Respiratoria/citología , Humo/efectos adversos , Adulto , Líquido del Lavado Bronquioalveolar/citología , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Células Cultivadas , Hemo-Oxigenasa 1/metabolismo , Humanos , Recién Nacido , Interleucina-13/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Receptores Notch/antagonistas & inhibidores , Productos de Tabaco/efectos adversos , Respuesta de Proteína Desplegada/efectos de los fármacos
4.
J Cell Sci ; 129(6): 1260-70, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26818841

RESUMEN

The accumulation of ubiquitylated proteinaceous inclusions represents a complex process, reflecting the disequilibrium between aggregate formation and aggregate clearance. Although decreasing aggregate formation or augmenting aggregate clearance will ultimately lead to a diminished aggregate burden, in terms of disease pathogenesis, the different approaches can have distinct outcomes. Using a novel cell-based assay that can distinguish newly formed versus preformed inclusions, we demonstrate that two proteins previously implicated in the autophagic clearance of expanded polyglutamine inclusions, HspB7 and Alfy (also known as WDFY3), actually affect very distinct cellular processes to affect aggregate burden. Using this cell-based assay, we also establish that constitutive expression of the aggregation-prone protein can measurably slow the elimination of protein aggregates, given that not all aggregates appear to be available for degradation. This new assay can therefore not only determine at what step a modifier might influence aggregate burden, but also can be used to provide new insights into how protein aggregates are targeted for degradation.


Asunto(s)
Bioquímica/métodos , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de la Membrana/metabolismo , Agregado de Proteínas , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Autofagia , Proteínas Relacionadas con la Autofagia , Proteínas de Choque Térmico HSP27/genética , Células HeLa , Humanos , Cuerpos de Inclusión , Proteínas de la Membrana/genética , Ratones , Proteolisis , Factores de Transcripción/genética
5.
Transgenic Res ; 27(1): 75-85, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29383478

RESUMEN

Sox2 is a Sry-box containing family member of related transcription factors sharing homology in their DNA binding domain. Sox2 is important during different stages of development, and previously we showed that Sox2 plays an important role in branching morphogenesis and epithelial cell differentiation in lung development. The transcriptional activity of Sox2 depends on its interaction with other proteins, leading to 'complex-specific' DNA binding and transcriptional regulation. In this study, we generated a mouse model containing a biotinylatable-tag targeted at the translational start site of the endogenous Sox2 gene (bioSox2). This tag was biotinylated by the bacterial birA protein and the resulting bioSox2 protein was used to identify associating partners of Sox2 at different phases of lung development in vivo (the Sox2 interactome). Homozygous bioSox2 mice are viable and fertile irrespective of the biotinylation of the bio tag, indicating that the bioSox2 gene is normally expressed and the protein is functional in all tissues. This suggests that partners of Sox2 are most likely able to associate with the bioSox2 protein. BioSox2 complexes were isolated with high affinity using streptavidin beads and analysed by MALDI-ToF mass spectrometry analysis. Several of the identified binding partners are already shown to have a respiratory phenotype. Two of these partners, Wdr5 and Tcf3, were validated to confirm their association in Sox2 complexes. This bioSox2 mouse model will be a valuable tool for isolating in vivo Sox2 complexes from different tissues.


Asunto(s)
Pulmón/embriología , Ratones Transgénicos , Factores de Transcripción SOXB1/metabolismo , Proteínas de Anclaje a la Quinasa A/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biotinilación , Encéfalo/embriología , Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Pulmón/metabolismo , Ratones Endogámicos C57BL , Complejos Multiproteicos , Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción SOXB1/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
6.
Respir Res ; 17: 44, 2016 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-27107715

RESUMEN

Inspired by the increasing burden of lung associated diseases in society and an growing demand to accommodate patients, great efforts by the scientific community produce an increasing stream of data that are focused on delineating the basic principles of lung development and growth, as well as understanding the biomechanical properties to build artificial lung devices. In addition, the continuing efforts to better define the disease origin, progression and pathology by basic scientists and clinicians contributes to insights in the basic principles of lung biology. However, the use of different model systems, experimental approaches and readout systems may generate somewhat conflicting or contradictory results. In an effort to summarize the latest developments in the lung epithelial stem cell biology, we provide an overview of the current status of the field. We first describe the different stem cells, or progenitor cells, residing in the homeostatic lung. Next, we focus on the plasticity of the different cell types upon several injury-induced activation or repair models, and highlight the regenerative capacity of lung cells. Lastly, we summarize the generation of lung mimics, such as air-liquid interface cultures, organoids and lung on a chip, that are required to test emerging hypotheses. Moreover, the increasing collaboration between distinct specializations will contribute to the eventual development of an artificial lung device capable of assisting reduced lung function and capacity in human patients.


Asunto(s)
Órganos Bioartificiales , Trasplante de Pulmón/instrumentación , Pulmón/citología , Pulmón/crecimiento & desarrollo , Regeneración/fisiología , Células Madre/citología , Animales , Biomimética/instrumentación , Humanos , Respiración Artificial/instrumentación , Trasplante de Células Madre/métodos , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos
7.
Dev Cell ; 59(10): 1269-1283.e6, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38565145

RESUMEN

Progenitor cells adapt their behavior in response to tissue demands. However, the molecular mechanisms controlling esophageal progenitor decisions remain largely unknown. Here, we demonstrate the presence of a Troy (Tnfrsf19)-expressing progenitor subpopulation localized to defined regions along the mouse esophageal axis. Lineage tracing and mathematical modeling demonstrate that Troy-positive progenitor cells are prone to undergoing symmetrical fate choices and contribute to esophageal tissue homeostasis long term. Functionally, TROY inhibits progenitor proliferation and enables commitment to differentiation without affecting fate symmetry. Whereas Troy expression is stable during esophageal homeostasis, progenitor cells downregulate Troy in response to tissue stress, enabling proliferative expansion of basal cells refractory to differentiation and reestablishment of tissue homeostasis. Our results demonstrate functional, spatially restricted progenitor heterogeneity in the esophageal epithelium and identify how dynamic regulation of Troy coordinates tissue generation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Esófago , Receptores del Factor de Necrosis Tumoral , Células Madre , Animales , Ratones , Linaje de la Célula , Epitelio/metabolismo , Mucosa Esofágica/metabolismo , Mucosa Esofágica/citología , Esófago/citología , Esófago/metabolismo , Proteínas de Homeodominio , Homeostasis , Células Madre/metabolismo , Células Madre/citología , Receptores del Factor de Necrosis Tumoral/análisis , Receptores del Factor de Necrosis Tumoral/metabolismo
8.
J Vis Exp ; (191)2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36688545

RESUMEN

Epithelial stem and progenitor cells contribute to the formation and maintenance of the epithelial barrier throughout life. Most stem and progenitor cell populations are tucked away in anatomically distinct locations, enabling exclusive interactions with niche signals that maintain stemness. While the development of epithelial organoid cultures provides a powerful tool for understanding the role of stem and progenitor cells in homeostasis and disease, the interaction within the niche environment is largely absent, thereby hindering the identification of factors influencing stem cell behavior. Fibroblasts play a key role in directing epithelial stem and progenitor fate. Here, a comprehensive organoid-fibroblast co-culture protocol enabling the delineation of fibroblast subpopulations in esophageal progenitor cell renewal and differentiation is presented. In this protocol, a method to isolate both epithelial cells and fibroblasts in parallel from the esophagus is described. Distinct fluorescence-activated cell sorting strategies to isolate both the esophageal progenitor cells as well as the fibroblast subpopulations from either transgenic reporter or wild-type mice are outlined. This protocol provides a versatile approach that can be adapted to accommodate the isolation of specific fibroblast subpopulations. Establishing and passaging esophageal epithelial organoid mono-cultures is included in this protocol, enabling a direct comparison with the co-culture system. In addition, a 3D clearing approach allowing for detailed image analysis of epithelial-fibroblast interactions is described. Collectively, this protocol describes a comparative and relatively high-throughput method for identifying and understanding esophageal stem cell niche components in vitro.


Asunto(s)
Organoides , Células Madre , Ratones , Animales , Técnicas de Cocultivo , Diferenciación Celular , Esófago , Fibroblastos
9.
Front Cell Dev Biol ; 10: 1022457, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36299482

RESUMEN

The lung is composed of a highly branched airway structure, which humidifies and warms the inhaled air before entering the alveolar compartment. In the alveoli, a thin layer of epithelium is in close proximity with the capillary endothelium, allowing for an efficient exchange of oxygen and carbon dioxide. During development proliferation and differentiation of progenitor cells generates the lung architecture, and in the adult lung a proper function of progenitor cells is needed to regenerate after injury. Malfunctioning of progenitors during development results in various congenital lung disorders, such as Congenital Diaphragmatic Hernia (CDH) and Congenital Pulmonary Adenomatoid Malformation (CPAM). In addition, many premature neonates experience continuous insults on the lung caused by artificial ventilation and supplemental oxygen, which requires a highly controlled mechanism of airway repair. Malfunctioning of airway progenitors during regeneration can result in reduction of respiratory function or (chronic) airway diseases. Pathways that are active during development are frequently re-activated upon damage. Understanding the basic mechanisms of lung development and the behavior of progenitor cell in the ontogeny and regeneration of the lung may help to better understand the underlying cause of lung diseases, especially those occurring in prenatal development or in the immediate postnatal period of life. This review provides an overview of lung development and the cell types involved in repair of lung damage with a focus on the airway.

10.
Front Pediatr ; 10: 881287, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35615634

RESUMEN

Congenital diaphragmatic hernia is a structural birth defect of the diaphragm, with lung hypoplasia and persistent pulmonary hypertension. Aside from vascular defects, the lungs show a disturbed balance of differentiated airway epithelial cells. The Sry related HMG box protein SOX2 is an important transcription factor for proper differentiation of the lung epithelium. The transcriptional activity of SOX2 depends on interaction with other proteins and the identification of SOX2-associating factors may reveal important complexes involved in the disturbed differentiation in CDH. To identify SOX2-associating proteins, we purified SOX2 complexes from embryonic mouse lungs at 18.5 days of gestation. Mass spectrometry analysis of SOX2-associated proteins identified several potential candidates, among which were the Chromodomain Helicase DNA binding protein 4 (CHD4), Cut-Like Homeobox1 (CUX1), and the Forkhead box proteins FOXP2 and FOXP4. We analyzed the expression patterns of FOXP2, FOXP4, CHD4, and CUX1 in lung during development and showed co-localization with SOX2. Co-immunoprecipitations validated the interactions of these four transcription factors with SOX2, and large-scale chromatin immunoprecipitation (ChIP) data indicated that SOX2 and CHD4 bound to unique sites in the genome, but also co-occupied identical regions, suggesting that these complexes could be involved in co-regulation of genes involved in the respiratory system.

11.
Elife ; 102021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34286693

RESUMEN

SOX2 expression levels are crucial for the balance between maintenance and differentiation of airway progenitor cells during development and regeneration. Here, we describe patterning of the mouse proximal airway epithelium by SOX21, which coincides with high levels of SOX2 during development. Airway progenitor cells in this SOX2+/SOX21+ zone show differentiation to basal cells, specifying cells for the extrapulmonary airways. Loss of SOX21 showed an increased differentiation of SOX2+ progenitor cells to basal and ciliated cells during mouse lung development. We propose a mechanism where SOX21 inhibits differentiation of airway progenitors by antagonizing SOX2-induced expression of specific genes involved in airway differentiation. Additionally, in the adult tracheal epithelium, SOX21 inhibits basal to ciliated cell differentiation. This suppressing function of SOX21 on differentiation contrasts SOX2, which mainly drives differentiation of epithelial cells during development and regeneration after injury. Furthermore, using human fetal lung organoids and adult bronchial epithelial cells, we show that SOX2+/SOX21+ regionalization is conserved. Lastly, we show that the interplay between SOX2 and SOX21 is context and concentration dependent leading to regulation of differentiation of the airway epithelium.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Epiteliales/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXB2/genética , Factores de Transcripción SOXB2/metabolismo , Animales , Humanos , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Ratones , Células Madre/metabolismo , Tráquea/metabolismo , Transcriptoma
12.
Neuron ; 105(5): 813-821.e6, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31899071

RESUMEN

Despite being an autosomal dominant disorder caused by a known coding mutation in the gene HTT, Huntington's disease (HD) patients with similar trinucleotide repeat mutations can have an age of onset that varies by decades. One likely contributing factor is the genetic heterogeneity of patients that might modify their vulnerability to disease. We report that although the heterozygous depletion of the autophagy adaptor protein Alfy/Wdfy3 has no consequence in control mice, it significantly accelerates age of onset and progression of HD pathogenesis. Alfy is required in the adult brain for the autophagy-dependent clearance of proteinaceous deposits, and its depletion in mice and neurons derived from patient fibroblasts accelerates the aberrant accumulation of this pathological hallmark shared across adult-onset neurodegenerative diseases. These findings indicate that selectively compromising the ability to eliminate aggregated proteins is a pathogenic driver, and the selective elimination of aggregates may confer disease resistance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Relacionadas con la Autofagia/genética , Enfermedad de Huntington/genética , Macroautofagia/genética , Neuronas/metabolismo , Agregación Patológica de Proteínas/genética , Edad de Inicio , Animales , Muerte Celular/genética , Modelos Animales de Enfermedad , Femenino , Fibroblastos , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Masculino , Ratones , Ratones Noqueados , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/fisiopatología
13.
Methods Mol Biol ; 1873: 157-169, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30341608

RESUMEN

Understanding the fundamental mechanism underlying the accumulation and clearance of misfolded proteins can lead to insights into the synthetic and degradative pathways that maintain the homeostasis of proteins in all cells. Given the interconnection between protein homeostasis and cell health, as well as the complexity of aggregate formation and the degradation pathways with which it is intertwined, the design of the tools that are used to examine protein aggregation and accumulation can have a profound impact on the interpretation of results. We rely on two previously published stable cell lines that use conditional expression and the ligand-receptor tag known as HaloTag, to temporally distinguish distinct pools of aggregates, and use a combination of biochemical- and imaging-based methods to measure aggregation of a canonical aggregation-prone protein. We measure aggregate load biochemically using Filter Trap Analysis, which combines a filter trap retardation assay and immunoblotting to measure detergent soluble and insoluble protein levels, and visually, using confocal microscopy to monitor simultaneously aggregate formation and growth events in the background of aggregate clearance. As a secondary screen to more simplistic screen based approaches, this method permits further insight into how aggregate load is affected.


Asunto(s)
Bioensayo , Agregado de Proteínas , Proteínas/química , Amiloide/química , Amiloide/metabolismo , Proteínas Amiloidogénicas/química , Proteínas Amiloidogénicas/metabolismo , Bioensayo/métodos , Línea Celular , Humanos , Proteínas/metabolismo , Solubilidad
14.
Sci Rep ; 8(1): 7349, 2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29743551

RESUMEN

Air-liquid interface (ALI) cultures of mouse tracheal epithelial cells (MTEC) are a well-established model to study airway epithelial cells, but current methods require large numbers of animals which is unwanted in view of the 3R principle and introduces variation. Moreover, stringent breeding schemes are frequently needed to generate sufficient numbers of genetically modified animals. Current protocols do not incorporate expansion of MTEC, and therefore we developed a protocol to expand MTEC while maintaining their differentiation capacity. MTEC were isolated and expanded using the ROCK inhibitor Y-27632 in presence or absence of the γ-secretase inhibitor DAPT, a Notch pathway inhibitor. Whereas MTEC proliferated without DAPT, growth rate and cell morphology improved in presence of DAPT. ALI-induced differentiation of expanded MTEC resulted in an altered capacity of basal cells to differentiate into ciliated cells, whereas IL-13-induced goblet cell differentiation remained unaffected. Ciliated cell differentiation improved by prolonging the ALI differentiation or by adding DAPT, suggesting that basal cells retain their ability to differentiate. This technique using expansion of MTEC and subsequent ALI differentiation drastically reduces animal numbers and costs for in vitro experiments, and will reduce biological variation. Additionally, we provide novel insights in the dynamics of basal cell populations in vitro.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Tráquea/citología , Animales , Diferenciación Celular/fisiología , Células Cultivadas/metabolismo , Cilios/metabolismo , Células Epiteliales/metabolismo , Células Caliciformes/metabolismo , Interleucina-13/metabolismo , Ratones , Ratones Endogámicos C57BL , Tráquea/metabolismo , Tráquea/fisiología
15.
Elife ; 52016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27648578

RESUMEN

The regulation of protein degradation is essential for maintaining the appropriate environment to coordinate complex cell signaling events and to promote cellular remodeling. The Autophagy linked FYVE protein (Alfy), previously identified as a molecular scaffold between the ubiquitinated cargo and the autophagic machinery, is highly expressed in the developing central nervous system, indicating that this pathway may have yet unexplored roles in neurodevelopment. To examine this possibility, we used mouse genetics to eliminate Alfy expression. We report that this evolutionarily conserved protein is required for the formation of axonal tracts throughout the brain and spinal cord, including the formation of the major forebrain commissures. Consistent with a phenotype reflecting a failure in axon guidance, the loss of Alfy in mice disrupts localization of glial guidepost cells, and attenuates axon outgrowth in response to Netrin-1. These findings further support the growing indication that macroautophagy plays a key role in the developing CNS.


Asunto(s)
Encéfalo/embriología , Vías Nerviosas/embriología , Neuronas/fisiología , Proteínas de Transporte Vesicular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Relacionadas con la Autofagia , Técnicas de Inactivación de Genes , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA