Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Development ; 140(21): 4386-97, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24067355

RESUMEN

External ear abnormalities are frequent in newborns ranging from microtia to partial auricle duplication. Little is known about the molecular mechanisms orchestrating external ear morphogenesis. In humans, HOXA2 partial loss of function induces a bilateral microtia associated with an abnormal shape of the auricle. In mice, Hoxa2 inactivation at early gestational stages results in external auditory canal (EAC) duplication and absence of the auricle, whereas its late inactivation results in a hypomorphic auricle, mimicking the human HOXA2 mutant condition. By genetic fate mapping we found that the mouse auricle (or pinna) derives from the Hoxa2-expressing neural crest-derived mesenchyme of the second pharyngeal arch, and not from a composite of first and second arch mesenchyme as previously proposed based on morphological observation of human embryos. Moreover, the mouse EAC is entirely lined by Hoxa2-negative first arch mesenchyme and does not develop at the first pharyngeal cleft, as previously assumed. Conditional ectopic Hoxa2 expression in first arch neural crest is sufficient to induce a complete duplication of the pinna and a loss of the EAC, suggesting transformation of the first arch neural crest-derived mesenchyme lining the EAC into an ectopic pinna. Hoxa2 partly controls the morphogenesis of the pinna through the BMP signalling pathway and expression of Eya1, which in humans is involved in branchio-oto-renal syndrome. Thus, Hoxa2 loss- and gain-of-function approaches in mice provide a suitable model to investigate the molecular aetiology of microtia and auricle duplication.


Asunto(s)
Anomalías Congénitas/genética , Pabellón Auricular/anomalías , Conducto Auditivo Externo/anomalías , Oído/anomalías , Proteínas de Homeodominio/genética , Morfogénesis/fisiología , Transducción de Señal/fisiología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Inmunoprecipitación de Cromatina , Microtia Congénita , Pabellón Auricular/embriología , Conducto Auditivo Externo/embriología , Inmunohistoquímica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mesodermo/citología , Ratones , Morfogénesis/genética , Mutación/genética , Cresta Neural/citología , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Tamoxifeno/administración & dosificación
2.
J Neurosci ; 32(48): 17172-85, 2012 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23197710

RESUMEN

Oligodendrocytes are the myelin-forming cells of the vertebrate CNS. Little is known about the molecular control of region-specific oligodendrocyte development. Here, we show that oligodendrogenesis in the mouse rostral hindbrain, which is organized in a metameric series of rhombomere-derived (rd) territories, follows a rhombomere-specific pattern, with extensive production of oligodendrocytes in the pontine territory (r4d) and delayed and reduced oligodendrocyte production in the prepontine region (r2d, r3d). We demonstrate that segmental organization of oligodendrocytes is controlled by Hox genes, namely Hoxa2 and Hoxb2. Specifically, Hoxa2 loss of function induced a dorsoventral enlargement of the Olig2/Nkx2.2-expressing oligodendrocyte progenitor domain, whereas conditional Hoxa2 overexpression in the Olig2(+) domain inhibited oligodendrogenesis throughout the brain. In contrast, Hoxb2 deletion resulted in a reduction of the pontine oligodendrogenic domain. Compound Hoxa2(-/-)/Hoxb2(-/-) mutant mice displayed the phenotype of Hoxb2(-/-) mutants in territories coexpressing Hoxa2 and Hoxb2 (rd3, rd4), indicating that Hoxb2 antagonizes Hoxa2 during rostral hindbrain oligodendrogenesis. This study provides the first in vivo evidence that Hox genes determine oligodendrocyte regional identity in the mammalian brain.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Homeodominio/genética , Oligodendroglía/metabolismo , Rombencéfalo/metabolismo , Factores de Transcripción/genética , Animales , Tipificación del Cuerpo/genética , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Noqueados , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Rombencéfalo/embriología , Factores de Transcripción/metabolismo
3.
Nat Commun ; 14(1): 3242, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37277355

RESUMEN

Enhancer-promoter interactions preferentially occur within boundary-insulated topologically associating domains (TADs), limiting inter-TAD interactions. Enhancer clusters in linear proximity, termed super-enhancers (SEs), ensure high target gene expression levels. Little is known about SE topological regulatory impact during craniofacial development. Here, we identify 2232 genome-wide putative SEs in mouse cranial neural crest cells (CNCCs), 147 of which target genes establishing CNCC positional identity during face formation. In second pharyngeal arch (PA2) CNCCs, a multiple SE-containing region, partitioned into Hoxa Inter-TAD Regulatory Element 1 and 2 (HIRE1 and HIRE2), establishes long-range inter-TAD interactions selectively with Hoxa2, that is required for external and middle ear structures. HIRE2 deletion in a Hoxa2 haploinsufficient background results in microtia. HIRE1 deletion phenocopies the full homeotic Hoxa2 knockout phenotype and induces PA3 and PA4 CNCC abnormalities correlating with Hoxa2 and Hoxa3 transcriptional downregulation. Thus, SEs can overcome TAD insulation and regulate anterior Hoxa gene collinear expression in a CNCC subpopulation-specific manner during craniofacial development.


Asunto(s)
Cresta Neural , Secuencias Reguladoras de Ácidos Nucleicos , Ratones , Animales , Cresta Neural/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cráneo/metabolismo , Cromatina/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo
4.
J Dev Biol ; 10(1)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35225962

RESUMEN

Providing appropriate positional identity and patterning information to distinct rostrocaudal subpopulations of cranial neural crest cells (CNCCs) is central to vertebrate craniofacial morphogenesis. Hox genes are not expressed in frontonasal and first pharyngeal arch (PA1) CNCCs, whereas a single Hox gene, Hoxa2, is necessary to provide patterning information to second pharyngeal arch (PA2) CNCCs. In frog, chick and mouse embryos, ectopic expression of Hoxa2 in Hox-negative CNCCs induced hypoplastic phenotypes of CNCC derivatives of variable severity, associated or not with homeotic transformation of a subset of PA1 structures into a PA2-like identity. Whether these different morphological outcomes are directly related to distinct Hoxa2 overexpression levels is unknown. To address this issue, we selectively induced Hoxa2 overexpression in mouse CNCCs, using a panel of mouse lines expressing different Hoxa2 ectopic expression levels, including a newly generated Hoxa2 knocked-in mouse line. While ectopic Hoxa2 expression at only 60% of its physiological levels was sufficient for pinna duplication, ectopic Hoxa2 expression at 100% of its normal level was required for complete homeotic repatterning of a subset of PA1 skeletal elements into a duplicated set of PA2-like elements. On the other hand, ectopic Hoxa2 overexpression at non-physiological levels (200% of normal levels) led to an almost complete loss of craniofacial skeletal structures. Moreover, ectopic Hoxa5 overexpression in CNCCs, while also resulting in severe craniofacial defects, did not induce homeotic changes of PA1-derived CNCCs, indicating Hoxa2 specificity in repatterning a subset of Hox-negative CNCCs. These results reconcile some discrepancies in previously published experiments and indicate that distinct subpopulations of CNCCs are differentially sensitive to ectopic levels of Hox expression.

5.
PLoS Biol ; 6(6): e142, 2008 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-18547144

RESUMEN

The pontine neurons (PN) represent a major source of mossy fiber projections to the cerebellum. During mouse hindbrain development, PN migrate tangentially and sequentially along both the anteroposterior (AP) and dorsoventral (DV) axes. Unlike DV migration, which is controlled by the Netrin-1/Dcc attractive pathway, little is known about the molecular mechanisms guiding PN migration along the AP axis. Here, we show that Hoxa2 and Hoxb2 are required both intrinsically and extrinsically to maintain normal AP migration of subsets of PN, by preventing their premature ventral attraction towards the midline. Moreover, the migration defects observed in Hoxa2 and Hoxb2 mutant mice were phenocopied in compound Robo1;Robo2, Slit1;Slit2, and Robo2;Slit2 knockout animals, indicating that these guidance molecules act downstream of Hox genes to control PN migration. Indeed, using chromatin immunoprecipitation assays, we further demonstrated that Robo2 is a direct target of Hoxa2 in vivo and that maintenance of high Robo and Slit expression levels was impaired in Hoxa2 mutant mice. Lastly, the analysis of Phox2b-deficient mice indicated that the facial motor nucleus is a major Slit signaling source required to prevent premature ventral migration of PN. These findings provide novel insights into the molecular control of neuronal migration from transcription factor to regulation of guidance receptor and ligand expression. Specifically, they address the question of how exposure to multiple guidance cues along the AP and DV axes is regulated at the transcriptional level and in turn translated into stereotyped migratory responses during tangential migration of neurons in the developing mammalian brain.


Asunto(s)
Movimiento Celular/genética , Genes Homeobox , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Receptores Inmunológicos/metabolismo , Factores de Transcripción/genética , Animales , Secuencia de Bases , Inmunohistoquímica , Ratones , Ratones Noqueados , Modelos Biológicos , Datos de Secuencia Molecular , Transducción de Señal , Proteínas Roundabout
6.
Nat Genet ; 53(3): 379-391, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33603234

RESUMEN

Rapid cellular responses to environmental stimuli are fundamental for development and maturation. Immediate early genes can be transcriptionally induced within minutes in response to a variety of signals. How their induction levels are regulated and their untimely activation by spurious signals prevented during development is poorly understood. We found that in developing sensory neurons, before perinatal sensory-activity-dependent induction, immediate early genes are embedded into a unique bipartite Polycomb chromatin signature, carrying active H3K27ac on promoters but repressive Ezh2-dependent H3K27me3 on gene bodies. This bipartite signature is widely present in developing cell types, including embryonic stem cells. Polycomb marking of gene bodies inhibits mRNA elongation, dampening productive transcription, while still allowing for fast stimulus-dependent mark removal and bipartite gene induction. We reveal a developmental epigenetic mechanism regulating the rapidity and amplitude of the transcriptional response to relevant stimuli, while preventing inappropriate activation of stimulus-response genes.


Asunto(s)
Cromatina/genética , Regulación del Desarrollo de la Expresión Génica , Genes Inmediatos-Precoces , Proteínas del Grupo Polycomb/genética , Animales , Cromatina/metabolismo , Células Madre Embrionarias/fisiología , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Histonas/metabolismo , Ratones Transgénicos , Mutación , Proteínas del Grupo Polycomb/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Rombencéfalo/efectos de los fármacos , Rombencéfalo/embriología , Células Receptoras Sensoriales/fisiología
7.
Cell Rep ; 31(11): 107767, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32553152

RESUMEN

The mammalian precerebellar pontine nucleus (PN) has a main role in relaying cortical information to the cerebellum. The molecular determinants establishing ordered connectivity patterns between cortical afferents and precerebellar neurons are largely unknown. We show that expression of Hox5 transcription factors is induced in specific subsets of postmitotic PN neurons at migration onset. Hox5 induction is achieved by response to retinoic acid signaling, resulting in Jmjd3-dependent derepression of Polycomb chromatin and 3D conformational changes. Hoxa5 drives neurons to settle posteriorly in the PN, where they are monosynaptically targeted by cortical neuron subsets mainly carrying limb somatosensation. Furthermore, Hoxa5 postmigratory ectopic expression in PN neurons is sufficient to attract cortical somatosensory inputs regardless of position and avoid visual afferents. Transcriptome analysis further suggests that Hoxa5 is involved in circuit formation. Thus, Hoxa5 coordinates postmitotic specification, migration, settling position, and sub-circuit assembly of PN neuron subsets in the cortico-cerebellar pathway.


Asunto(s)
Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Movimiento Celular/fisiología , Corteza Cerebral/metabolismo
8.
Cell Rep ; 18(1): 185-197, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28052248

RESUMEN

Tonotopy is a hallmark of auditory pathways and provides the basis for sound discrimination. Little is known about the involvement of transcription factors in brainstem cochlear neurons orchestrating the tonotopic precision of pre-synaptic input. We found that in the absence of Hoxa2 and Hoxb2 function in Atoh1-derived glutamatergic bushy cells of the anterior ventral cochlear nucleus, broad input topography and sound transmission were largely preserved. However, fine-scale synaptic refinement and sharpening of isofrequency bands of cochlear neuron activation upon pure tone stimulation were impaired in Hox2 mutants, resulting in defective sound-frequency discrimination in behavioral tests. These results establish a role for Hox factors in tonotopic refinement of connectivity and in ensuring the precision of sound transmission in the mammalian auditory circuit.


Asunto(s)
Vías Auditivas/fisiología , Percepción Auditiva/fisiología , Tronco Encefálico/fisiología , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Animales , Animales Recién Nacidos , Audiometría de Tonos Puros , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Adhesión Celular , Núcleo Coclear/fisiología , Condicionamiento Psicológico , Miedo , Perfilación de la Expresión Génica , Glutamatos/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Mutantes , Mutación/genética , Neuronas/metabolismo , Organogénesis/genética , Sinapsis/metabolismo , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Factores de Transcripción/metabolismo
9.
Nat Commun ; 8: 14172, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28155854

RESUMEN

The cerebral cortex is organized into specialized sensory areas, whose initial territory is determined by intracortical molecular determinants. Yet, sensory cortical area size appears to be fine tuned during development to respond to functional adaptations. Here we demonstrate the existence of a prenatal sub-cortical mechanism that regulates the cortical areas size in mice. This mechanism is mediated by spontaneous thalamic calcium waves that propagate among sensory-modality thalamic nuclei up to the cortex and that provide a means of communication among sensory systems. Wave pattern alterations in one nucleus lead to changes in the pattern of the remaining ones, triggering changes in thalamic gene expression and cortical area size. Thus, silencing calcium waves in the auditory thalamus induces Rorß upregulation in a neighbouring somatosensory nucleus preluding the enlargement of the barrel-field. These findings reveal that embryonic thalamic calcium waves coordinate cortical sensory area patterning and plasticity prior to sensory information processing.


Asunto(s)
Núcleos Talámicos Ventrales/anatomía & histología , Núcleos Talámicos Ventrales/embriología , Animales , Calcio/metabolismo , Femenino , Uniones Comunicantes/metabolismo , Expresión Génica , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal , Receptores Nucleares Huérfanos/genética , Embarazo , Corteza Somatosensorial/fisiología , Núcleos Talámicos Ventrales/metabolismo , Núcleos Talámicos Ventrales/fisiología , Visión Ocular
10.
Cell Rep ; 13(4): 783-797, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26489473

RESUMEN

Mouse whiskers are somatotopically mapped in brainstem trigeminal nuclei as neuronal modules known as barrelettes. Whisker-related afferents form barrelettes in ventral principal sensory (vPrV) nucleus, whereas mandibular input targets dorsal PrV (dPrV). How barrelette neuron identity and circuitry is established is poorly understood. We found that ectopic Hoxa2 expression in dPrV neurons is sufficient to attract whisker-related afferents, induce asymmetrical dendrite arbors, and allow ectopic barrelette map formation. Moreover, the thalamic area forming whisker-related barreloids is prenatally targeted by both vPrV and dPrV axons followed by perinatal large-scale pruning of dPrV axons and refinement of vPrV barrelette input. Ectopic Hoxa2 expression allows topographically directed targeting and refinement of dPrV axons with vPrV axons into a single whisker-related barreloid map. Thus, a single HOX transcription factor is sufficient to switch dPrV into a vPrV barrelette neuron program and coordinate input-output topographic connectivity of a dermatome-specific circuit module.


Asunto(s)
Axones/fisiología , Tronco Encefálico/fisiología , Proteínas de Homeodominio/metabolismo , Neuronas/fisiología , Vibrisas/fisiología , Animales , Tronco Encefálico/citología , Ratones , Neuronas/citología , Vibrisas/citología
11.
Science ; 339(6116): 204-7, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23307742

RESUMEN

We investigated the role of histone methyltransferase Ezh2 in tangential migration of mouse precerebellar pontine nuclei, the main relay between neocortex and cerebellum. By counteracting the sonic hedgehog pathway, Ezh2 represses Netrin1 in dorsal hindbrain, which allows normal pontine neuron migration. In Ezh2 mutants, ectopic Netrin1 derepression results in abnormal migration and supernumerary nuclei integrating in brain circuitry. Moreover, intrinsic topographic organization of pontine nuclei according to rostrocaudal progenitor origin is maintained throughout migration and correlates with patterned cortical input. Ezh2 maintains spatially restricted Hox expression, which, in turn, regulates differential expression of the repulsive receptor Unc5b in migrating neurons; together, they generate subsets with distinct responsiveness to environmental Netrin1. Thus, Ezh2-dependent epigenetic regulation of intrinsic and extrinsic transcriptional programs controls topographic neuronal guidance and connectivity in the cortico-ponto-cerebellar pathway.


Asunto(s)
Cerebelo/embriología , Vías Nerviosas/embriología , Neuronas/fisiología , Complejo Represivo Polycomb 2/metabolismo , Puente/embriología , Animales , Movimiento Celular , Cerebelo/citología , Cerebelo/metabolismo , Corteza Cerebral/embriología , Corteza Cerebral/fisiología , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/metabolismo , Metencéfalo/embriología , Ratones , Ratones Transgénicos , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Receptores de Netrina , Netrina-1 , Vías Nerviosas/fisiología , Complejo Represivo Polycomb 2/genética , Puente/citología , Puente/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transcripción Genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
Dev Cell ; 20(4): 469-82, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21497760

RESUMEN

In vertebrate embryos, retinoic acid (RA) synthesized in the mesoderm by Raldh2 emanates to the hindbrain neuroepithelium, where it induces anteroposterior (AP)-restricted Hox expression patterns and rhombomere segmentation. However, how appropriate spatiotemporal RA activity is generated in the hindbrain is poorly understood. By analyzing Pbx1/Pbx2 and Hoxa1/Pbx1 null mice, we found that Raldh2 is itself under the transcriptional control of these factors and that the resulting RA-deficient phenotypes can be partially rescued by exogenous RA. Hoxa1-Pbx1/2-Meis2 directly binds a specific regulatory element that is required to maintain normal Raldh2 expression levels in vivo. Mesoderm-specific Xhoxa1 and Xpbx1b knockdowns in Xenopus embryos also result in Xraldh2 downregulation and hindbrain defects similar to mouse mutants, demonstrating conservation of this Hox-Pbx-dependent regulatory pathway. These findings reveal a feed-forward mechanism linking Hox-Pbx-dependent RA synthesis during early axial patterning with the establishment of spatially restricted Hox-Pbx activity in the developing hindbrain.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Rombencéfalo/embriología , Rombencéfalo/metabolismo , Tretinoina/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Aldehído Oxidasa/genética , Aldehído Oxidasa/metabolismo , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Femenino , Proteínas de Homeodominio/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Embarazo , Retinal-Deshidrogenasa , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo
13.
Neural Dev ; 2: 19, 2007 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-17897445

RESUMEN

BACKGROUND: Little is known about the involvement of molecular determinants of segmental patterning of rhombomeres (r) in the development of rhythmic neural networks in the mouse hindbrain. Here, we compare the phenotypes of mice carrying targeted inactivations of Hoxa2, the only Hox gene expressed up to r2, and of Krox20, expressed in r3 and r5. We investigated the impact of such mutations on the neural circuits controlling jaw opening and breathing in newborn mice, compatible with Hoxa2-dependent trigeminal defects and direct regulation of Hoxa2 by Krox20 in r3. RESULTS: We found that Hoxa2 mutants displayed an impaired oro-buccal reflex, similarly to Krox20 mutants. In contrast, while Krox20 is required for the development of the rhythm-promoting parafacial respiratory group (pFRG) modulating respiratory frequency, Hoxa2 inactivation did not affect neonatal breathing frequency. Instead, we found that Hoxa2-/- but not Krox20-/- mutation leads to the elimination of a transient control of the inspiratory amplitude normally occurring during the first hours following birth. Tracing of r2-specific progenies of Hoxa2 expressing cells indicated that the control of inspiratory activity resides in rostral pontine areas and required an intact r2-derived territory. CONCLUSION: Thus, inspiratory shaping and respiratory frequency are under the control of distinct Hox-dependent segmental cues in the mammalian brain. Moreover, these data point to the importance of rhombomere-specific genetic control in the development of modular neural networks in the mammalian hindbrain.


Asunto(s)
Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Proteínas de Homeodominio/genética , Maxilares/fisiología , Red Nerviosa/crecimiento & desarrollo , Centro Respiratorio/crecimiento & desarrollo , Rombencéfalo/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Tipificación del Cuerpo/genética , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Maxilares/inervación , Músculos Masticadores/crecimiento & desarrollo , Músculos Masticadores/inervación , Ratones , Ratones Noqueados , Red Nerviosa/metabolismo , Periodicidad , Centro Respiratorio/metabolismo , Fenómenos Fisiológicos Respiratorios , Rombencéfalo/metabolismo , Nervio Trigémino/crecimiento & desarrollo , Nervio Trigémino/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA