Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
PLoS Genet ; 19(8): e1010925, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37639482

RESUMEN

The mammalian cochlea is composed of sensory hair cells as well as multiple different types of non-sensory supporting cells. Pillar cells are one type of supporting cell that form the tunnel of Corti and include two morphologically and functionally distinct subtypes: inner pillar cells (IPCs) and outer pillar cells (OPCs). The processes of specification and differentiation of inner versus outer pillar cells are still unclear. Here, we show that ß-Catenin is required for establishing IPC identity in the mammalian cochlea. To differentiate the transcriptional and adhesion roles of ß-Catenin in establishing IPC identity, we examined two different models of ß-Catenin deletion; one that deletes both transcriptional and structural functions and one which retains cell adhesion function but lacks transcriptional function. Here, we show that cochleae lacking ß-Catenin transcriptional function lost IPCs and displayed extranumerary OPCs, indicating its requirement for establishing IPC identity. Overexpression of ß-Catenin induced proliferation within IPCs but not ectopic IPCs. Single-cell transcriptomes of supporting cells lacking ß-Catenin transcriptional function show a loss of the IPC and gain of OPC signatures. Finally, targeted deletion of ß-Catenin in IPCs also led to the loss of IPC identity, indicating a cell autonomous role of ß-Catenin in establishing IPC identity. As IPCs have the capacity to regenerate sensory hair cells in the postnatal cochlea, our results will aid in future IPC-based hair cell regeneration strategies.


Asunto(s)
Cóclea , beta Catenina , Animales , beta Catenina/genética , Células Ciliadas Auditivas , Adhesión Celular/genética , Diferenciación Celular/genética , Mamíferos
2.
Development ; 148(10)2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-34032268

RESUMEN

Nephron endowment, defined during the fetal period, dictates renal and related cardiovascular health throughout life. We show here that, despite its negative effects on kidney growth, genetic increase of GDNF prolongs the nephrogenic program beyond its normal cessation. Multi-stage mechanistic analysis revealed that excess GDNF maintains nephron progenitors and nephrogenesis through increased expression of its secreted targets and augmented WNT signaling, leading to a two-part effect on nephron progenitor maintenance. Abnormally high GDNF in embryonic kidneys upregulates its known targets but also Wnt9b and Axin2, with concomitant deceleration of nephron progenitor proliferation. Decline of GDNF levels in postnatal kidneys normalizes the ureteric bud and creates a permissive environment for continuation of the nephrogenic program, as demonstrated by morphologically and molecularly normal postnatal nephron progenitor self-renewal and differentiation. These results establish that excess GDNF has a bi-phasic effect on nephron progenitors in mice, which can faithfully respond to GDNF dosage manipulation during the fetal and postnatal period. Our results suggest that sensing the signaling activity level is an important mechanism through which GDNF and other molecules contribute to nephron progenitor lifespan specification.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Nefronas/embriología , Nefronas/crecimiento & desarrollo , Organogénesis/genética , Vía de Señalización Wnt/genética , Animales , Proteína Axina/metabolismo , Diferenciación Celular/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre/citología , Proteínas Wnt/metabolismo
3.
Small ; : e2400638, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38804126

RESUMEN

The lithium deposited via the complex electrochemical heterogeneous lithium deposition reaction (LDR) process on a lithium foil-based anode (LFA) forms a high-aspect-ratio shape whenever the reaction kinetics reach its limit, threatening battery safety. Thereby, a research strategy that boosts the LDR kinetics is needed to construct a high-power and safe lithium metal anode. In this study, the kinetic limitations of the LDR process on LFA are elucidated through operando and ex situ observations using in-depth electrochemical analyses. In addition, ultra-thin (≈0.5 µm) and high modulus (≥19 GPa) double-walled carbon nanotube (DWNT) membranes with different surface properties are designed to catalyze high-safety LDRs. The oxygen-functionalized DWNT membranes introduced on the LFA top surface simultaneously induce multitudinous lithium nuclei, leading to film-like lithium deposition even at a high current density of 20 mA cm-2. More importantly, the layer-by-layer assembly of the oxygen-functionalized and pristine DWNT membranes results in different surface energies between the top and bottom surfaces, enabling selective surface LDRs underneath the high-modulus bilayer membranes. The protective LDR on the bilayer-covered LFA guarantees an invulnerable cycling process in large-area pouch cells at high current densities for more than 1000 cycles, demonstrating the practicability of LFA in a conventional liquid electrolyte system.

4.
PLoS Genet ; 15(7): e1008254, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31276493

RESUMEN

The mouse organ of Corti, housed inside the cochlea, contains hair cells and supporting cells that transduce sound into electrical signals. These cells develop in two main steps: progenitor specification followed by differentiation. Fibroblast Growth Factor (FGF) signaling is important in this developmental pathway, as deletion of FGF receptor 1 (Fgfr1) or its ligand, Fgf20, leads to the loss of hair cells and supporting cells from the organ of Corti. However, whether FGF20-FGFR1 signaling is required during specification or differentiation, and how it interacts with the transcription factor Sox2, also important for hair cell and supporting cell development, has been a topic of debate. Here, we show that while FGF20-FGFR1 signaling functions during progenitor differentiation, FGFR1 has an FGF20-independent, Sox2-dependent role in specification. We also show that a combination of reduction in Sox2 expression and Fgf20 deletion recapitulates the Fgfr1-deletion phenotype. Furthermore, we uncovered a strong genetic interaction between Sox2 and Fgf20, especially in regulating the development of hair cells and supporting cells towards the basal end and the outer compartment of the cochlea. To explain this genetic interaction and its effects on the basal end of the cochlea, we provide evidence that decreased Sox2 expression delays specification, which begins at the apex of the cochlea and progresses towards the base, while Fgf20-deletion results in premature onset of differentiation, which begins near the base of the cochlea and progresses towards the apex. Thereby, Sox2 and Fgf20 interact to ensure that specification occurs before differentiation towards the cochlear base. These findings reveal an intricate developmental program regulating organ of Corti development along the basal-apical axis of the cochlea.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Órgano Espiral/citología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Factores de Transcripción SOXB1/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Técnicas de Inactivación de Genes , Masculino , Ratones , Órgano Espiral/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
5.
Proc Natl Acad Sci U S A ; 116(42): 21054-21060, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31570588

RESUMEN

Development of multicellular organs requires the coordination of cell differentiation and patterning. Critical for sound detection, the mammalian organ of Corti contains functional units arranged tonotopically along the cochlear turns. Each unit consists of sensory hair cells intercalated by nonsensory supporting cells, both specified and radially patterned with exquisite precision during embryonic development. However, how cell identity and radial patterning are jointly controlled is poorly understood. Here we show that ß-catenin is required for specification of hair cell and supporting cell subtypes and radial patterning of the cochlea in vivo. In 2 mouse models of conditional ß-catenin deletion, early specification of Myosin7-expressing hair cells and Prox1-positive supporting cells was preserved. While ß-catenin-deficient cochleae expressed FGF8 and FGFR3, both of which are essential for pillar cell specification, the radial patterning of organ of Corti was disrupted, revealed by aberrant expression of cadherins and the pillar cell markers P75 and Lgr6. Moreover, ß-catenin ablation caused duplication of FGF8-positive inner hair cells and reduction of outer hair cells without affecting the overall hair cell density. In contrast, in another transgenic model with suppressed transcriptional activity of ß-catenin but preserved cell adhesion function, both specification and radial patterning of the organ of Corti were intact. Our study reveals specific functions of ß-catenin in governing cell identity and patterning mediated through cell adhesion in the developing cochlea.


Asunto(s)
Cóclea/metabolismo , Cóclea/fisiología , beta Catenina/metabolismo , Animales , Biomarcadores/metabolismo , Adhesión Celular/fisiología , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/fisiología , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/fisiología , Ratones , Órgano Espiral/metabolismo , Organogénesis/fisiología
6.
J Am Soc Nephrol ; 31(11): 2559-2572, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32753399

RESUMEN

BACKGROUND: Nephron progenitor cells (NPCs) give rise to all segments of functional nephrons and are of great interest due to their potential as a source for novel treatment strategies for kidney disease. Fibroblast growth factor (FGF) signaling plays pivotal roles in generating and maintaining NPCs during kidney development, but little is known about the molecule(s) regulating FGF signaling during nephron development. Sprouty 1 (SPRY1) is an antagonist of receptor tyrosine kinases. Although SPRY1 antagonizes Ret-GDNF signaling, which modulates renal branching, its role in NPCs is not known. METHODS: Spry1, Fgf9, and Fgf20 compound mutant animals were used to evaluate kidney phenotypes in mice to understand whether SPRY1 modulates FGF signaling in NPCs and whether FGF8 functions with FGF9 and FGF20 in maintaining NPCs. RESULTS: Loss of one copy of Spry1 counters effects of the loss of Fgf9 and Fgf20, rescuing bilateral renal agenesis premature NPC differentiation, NPC proliferation, and cell death defects. In the absence of SPRY1, FGF9, and FGF20, another FGF ligand, FGF8, promotes nephrogenesis. Deleting both Fgf8 and Fgf20 results in kidney agenesis, defects in NPC proliferation, and cell death. Deleting one copy of Fgf8 reversed the effect of deleting one copy of Spry1, which rescued the renal agenesis due to loss of Fgf9 and Fgf20. CONCLUSIONS: SPRY1 expressed in NPCs modulates the activity of FGF signaling and regulates NPC stemness. These findings indicate the importance of the balance between positive and negative signals during NPC maintenance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 9 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/genética , Proteínas de la Membrana/genética , Nefronas/fisiología , Células Madre/fisiología , Animales , Muerte Celular/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Anomalías Congénitas/genética , Femenino , Riñón/anomalías , Enfermedades Renales/congénito , Enfermedades Renales/genética , Ratones , Nefronas/metabolismo , Nefronas/patología , Fenotipo , Transducción de Señal/genética , Células Madre/metabolismo
7.
Genes Dev ; 27(4): 450-8, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23431057

RESUMEN

In hair follicle development, a placode-derived signal is believed to induce formation of the dermal condensation, an essential component of ectodermal organs. However, the identity of this signal is unknown. Furthermore, although induction and patterning of hair follicles are intimately linked, it is not known whether the mesenchymal condensation is necessary for inducing the initial epithelial pattern. Here, we show that fibroblast growth factor 20 (Fgf20) is expressed in hair placodes and is induced by and functions downstream from epithelial ectodysplasin (Eda)/Edar and Wnt/ß-Catenin signaling to initiate formation of the underlying dermal condensation. Fgf20 governs formation of primary and secondary dermal condensations in developing hair follicles and subsequent formation of guard, awl, and auchene hairs. Although primary dermal condensations are absent in Fgf20 mutant mice, a regular array of hair placodes is formed, demonstrating that the epithelial patterning process is independent of known histological and molecular markers of underlying mesenchymal patterns during the initial stages of hair follicle development.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Folículo Piloso/embriología , Animales , Ectodisplasinas/metabolismo , Receptor Edar/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Ratones , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
8.
Development ; 141(13): 2691-701, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24924191

RESUMEN

Murine lacrimal, harderian and meibomian glands develop from the prospective conjunctival and eyelid epithelia and produce secretions that lubricate and protect the ocular surface. Sox9 expression localizes to the presumptive conjunctival epithelium as early as E11.5 and is detected in the lacrimal and harderian glands as they form. Conditional deletion showed that Sox9 is required for the development of the lacrimal and harderian glands and contributes to the formation of the meibomian glands. Sox9 regulates the expression of Sox10 to promote the formation of secretory acinar lobes in the lacrimal gland. Sox9 and FGF signaling were required for the expression of cartilage-associated extracellular matrix components during early stage lacrimal gland development. Fgfr2 deletion in the ocular surface epithelium reduced Sox9 and eliminated Sox10 expression. Sox9 deletion from the ectoderm did not affect Fgf10 expression in the adjacent mesenchyme or Fgfr2 expression in the epithelium, but appeared to reduce FGF signaling. Sox9 heterozygotes showed a haploinsufficient phenotype, in which the exorbital branch of the lacrimal gland was absent in most cases. However, enhancement of epithelial FGF signaling by expression of a constitutively active FGF receptor only partially rescued the lacrimal gland defects in Sox9 heterozygotes, suggesting a crucial role of Sox9, downstream of FGF signaling, in regulating lacrimal gland branching and differentiation.


Asunto(s)
Glándula de Harder/embriología , Aparato Lagrimal/embriología , Glándulas Tarsales/embriología , Morfogénesis/fisiología , Factor de Transcripción SOX9/metabolismo , Transducción de Señal/fisiología , Animales , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Técnicas Histológicas , Inmunohistoquímica , Hibridación Fluorescente in Situ , Captura por Microdisección con Láser , Ratones , Análisis por Micromatrices , Morfogénesis/genética , Factores de Transcripción SOXE/metabolismo
9.
Blood ; 124(19): 2948-52, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25267199

RESUMEN

In this study, we report that OVOL2, a C2H2 zinc finger protein, is a novel binding protein of ER71, which is a critical transcription factor for blood and vessel development. OVOL2 directly interacted with ER71, but not with ETS1 or ETS2, in the nucleus. ER71-mediated activation of the Flk1 promoter was further enhanced by OVOL2, although OVOL2 alone failed to activate it. Consistently, coexpression of ER71 and OVOL2 in differentiating embryonic stem cells led to a significant augmentation of FLK1(+), endothelial, and hematopoietic cells. Such cooperative effects were impaired by the short hairpin RNA-mediated inhibition of Ovol2. Collectively, we show that ER71 directly interacts with OVOL2 and that such interaction is critical for FLK1(+) cell generation and their differentiation into downstream cell lineages.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Factores de Transcripción/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Linaje de la Célula/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Células Endoteliales/citología , Técnicas de Silenciamiento del Gen , Células Madre Hematopoyéticas/citología , Ratones , Proteómica , ARN Interferente Pequeño/genética , Transducción de Señal/fisiología , Factores de Transcripción/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
10.
PLoS Genet ; 9(1): e1003231, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23358455

RESUMEN

The acquisition of the external genitalia allowed mammals to cope with terrestrial-specific reproductive needs for internal fertilization, and thus it represents one of the most fundamental steps in evolution towards a life on land. How genitalia evolved remains obscure, and the key to understanding this process may lie in the developmental genetics that underpins the early establishment of the genital primordium, the genital tubercle (GT). Development of the GT is similar to that of the limb, which requires precise regulation from a distal signaling epithelium. However, whether outgrowth of the GT and limbs is mediated by common instructive signals remains unknown. In this study, we used comprehensive genetic approaches to interrogate the signaling cascade involved in GT formation in comparison with limb formation. We demonstrate that the FGF ligand responsible for GT development is FGF8 expressed in the cloacal endoderm. We further showed that forced Fgf8 expression can rescue limb and GT reduction in embryos deficient in WNT signaling activity. Our studies show that the regulation of Fgf8 by the canonical WNT signaling pathway is mediated in part by the transcription factor SP8. Sp8 mutants elicit appendage defects mirroring WNT and FGF mutants, and abolishing Sp8 attenuates ectopic appendage development caused by a gain-of-function ß-catenin mutation. These observations indicate that a conserved WNT-SP8-FGF8 genetic cassette is employed by both appendages for promoting outgrowth, and suggest a deep homology shared by the limb and external genitalia.


Asunto(s)
Proteínas de Unión al ADN , Factor 8 de Crecimiento de Fibroblastos , Genitales , Factores de Transcripción , Vía de Señalización Wnt/genética , Animales , Evolución Biológica , Comunicación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Extremidades/crecimiento & desarrollo , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genitales/crecimiento & desarrollo , Genitales/metabolismo , Ligandos , Ratones , Mutación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Development ; 139(17): 3189-99, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22833125

RESUMEN

Uncovering the origin and nature of phenotypic variation within species is the first step in understanding variation between species. Mouse models with altered activities of crucial signal pathways have highlighted many important genes and signal networks regulating the morphogenesis of complex structures, such as teeth. The detailed analyses of these models have indicated that the balanced actions of a few pathways regulating cell behavior modulate the shape and number of teeth. Currently, however, most mouse models studied have had gross alteration of morphology, whereas analyses of more subtle modification of morphology are required to link developmental studies to evolutionary change. Here, we have analyzed a signaling network involving ectodysplasin (Eda) and fibroblast growth factor 20 (Fgf20) that subtly affects tooth morphogenesis. We found that Fgf20 is a major downstream effector of Eda and affects Eda-regulated characteristics of tooth morphogenesis, including the number, size and shape of teeth. Fgf20 function is compensated for by other Fgfs, in particular Fgf9 and Fgf4, and is part of an Fgf signaling loop between epithelium and mesenchyme. We showed that removal of Fgf20 in an Eda gain-of-function mouse model results in an Eda loss-of-function phenotype in terms of reduced tooth complexity and third molar appearance. However, the extra anterior molar, a structure lost during rodent evolution 50 million years ago, was stabilized in these mice.


Asunto(s)
Ectodisplasinas/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Morfogénesis/fisiología , Transducción de Señal/fisiología , Diente/embriología , Animales , Evolución Biológica , Galactósidos , Regulación del Desarrollo de la Expresión Génica/genética , Hibridación in Situ , Indoles , Luciferasas , Ratones , Microscopía Confocal , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética
12.
PLoS Biol ; 10(1): e1001231, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22235191

RESUMEN

A large proportion of age-related hearing loss is caused by loss or damage to outer hair cells in the organ of Corti. The organ of Corti is the mechanosensory transducing apparatus in the inner ear and is composed of inner hair cells, outer hair cells, and highly specialized supporting cells. The mechanisms that regulate differentiation of inner and outer hair cells are not known. Here we report that fibroblast growth factor 20 (FGF20) is required for differentiation of cells in the lateral cochlear compartment (outer hair and supporting cells) within the organ of Corti during a specific developmental time. In the absence of FGF20, mice are deaf and lateral compartment cells remain undifferentiated, postmitotic, and unresponsive to Notch-dependent lateral inhibition. These studies identify developmentally distinct medial (inner hair and supporting cells) and lateral compartments in the developing organ of Corti. The viability and hearing loss in Fgf20 knockout mice suggest that FGF20 may also be a deafness-associated gene in humans.


Asunto(s)
Diferenciación Celular/genética , Cóclea/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Transducción de Señal/genética , Animales , Cóclea/citología , Cóclea/embriología , Oído Interno/citología , Oído Interno/embriología , Oído Interno/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Órgano Espiral/citología , Órgano Espiral/embriología , Órgano Espiral/metabolismo , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Development ; 137(7): 1137-47, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20215350

RESUMEN

DiGeorge syndrome (DGS) is a common genetic disease characterized by pharyngeal apparatus malformations and defects in cardiovascular, craniofacial and glandular development. TBX1 is the most likely candidate disease-causing gene and is located within a 22q11.2 chromosomal deletion that is associated with most cases of DGS. Here, we show that canonical Wnt-beta-catenin signaling negatively regulates Tbx1 expression and that mesenchymal inactivation of beta-catenin (Ctnnb1) in mice caused abnormalities within the DGS phenotypic spectrum, including great vessel malformations, hypoplastic pulmonary and aortic arch arteries, cardiac malformations, micrognathia, thymus hypoplasia and mislocalization of the parathyroid gland. In a heterozygous Fgf8 or Tbx1 genetic background, ectopic activation of Wnt-beta-catenin signaling caused an increased incidence and severity of DGS-like phenotypes. Additionally, reducing the gene dosage of Fgf8 rescued pharyngeal arch artery defects caused by loss of Ctnnb1. These findings identify Wnt-beta-catenin signaling as a crucial upstream regulator of a Tbx1-Fgf8 signaling pathway and suggest that factors that affect Wnt-beta-catenin signaling could modify the incidence and severity of DGS.


Asunto(s)
Anomalías Múltiples , Síndrome de DiGeorge , Fenotipo , Proteínas de Dominio T Box/metabolismo , beta Catenina/genética , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Anomalías Múltiples/fisiopatología , Animales , Región Branquial/anomalías , Región Branquial/anatomía & histología , Región Branquial/embriología , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/patología , Síndrome de DiGeorge/fisiopatología , Femenino , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Embarazo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Proteínas de Dominio T Box/genética , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
14.
Cell Mol Gastroenterol Hepatol ; 16(3): 325-339, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37270061

RESUMEN

BACKGROUND & AIMS: Acute and chronic gastric injury induces alterations in differentiation within the corpus of the stomach called pyloric metaplasia. Pyloric metaplasia is characterized by the death of parietal cells and reprogramming of mitotically quiescent zymogenic chief cells into proliferative, mucin-rich spasmolytic polypeptide-expressing metaplasia (SPEM) cells. Overall, pyloric metaplastic units show increased proliferation and specific expansion of mucous lineages, both by proliferation of normal mucous neck cells and recruitment of SPEM cells. Here, we identify Sox9 as a potential gene of interest in the regulation of mucous neck and SPEM cell identity in the stomach. METHODS: We used immunostaining and electron microscopy to characterize the expression pattern of SRY-box transcription factor 9 (SOX9) during murine gastric development, homeostasis, and injury in homeostasis, after genetic deletion of Sox9 and after targeted genetic misexpression of Sox9 in the gastric epithelium and chief cells. RESULTS: SOX9 is expressed in all early gastric progenitors and strongly expressed in mature mucous neck cells with minor expression in the other principal gastric lineages during adult homeostasis. After injury, strong SOX9 expression was induced in the neck and base of corpus units in SPEM cells. Adult corpus units derived from Sox9-deficient gastric progenitors lacked normal mucous neck cells. Misexpression of Sox9 during postnatal development and adult homeostasis expanded mucous gene expression throughout corpus units including within the chief cell zone in the base. Sox9 deletion specifically in chief cells blunts their reprogramming into SPEM. CONCLUSIONS: Sox9 is a master regulator of mucous neck cell differentiation during gastric development. Sox9 also is required for chief cells to fully reprogram into SPEM after injury.


Asunto(s)
Células Principales Gástricas , Animales , Ratones , Células Principales Gástricas/metabolismo , Mucosa Gástrica/metabolismo , Metaplasia/metabolismo , Células Parietales Gástricas/metabolismo , Estómago
15.
ACS Appl Mater Interfaces ; 14(7): 8693-8704, 2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35148064

RESUMEN

Decellularized nerve hydrogels (dNHs) containing bioactive molecules are promising biomaterials for peripheral nerve injury (PNI) treatment and have been extensively applied in clinical and preclinical practice. However, most previous research projects studied their influences on nerve-related cellular behaviors in two dimensions (2D) without taking hydrogel biomechanics into consideration. The molecular mechanisms underlying the beneficial microenvironment provided by dNHs also remain unclear. In this study, dNHs from rat sciatic nerves were prepared, and their effects on Schwann cell (SC) and dorsal root ganglion (DRG) neurite behaviors were evaluated and compared to commercial rat tail type I collagen (Col) hydrogels in three-dimensional (3D) environments. We found that dNHs could promote SC proliferation and neurite outgrowth, and both the hydrogel mechanics and components contributed to the dNH functionalization. Through proteomics analysis, we found that laminin (LAM) and type V collagen (COLV) exclusively and abundantly existed in dNHs. By adding exogenous LAM and COLV into Col hydrogels, we demonstrated that they regulated SC gene expression and that LAM could promote SC spreading and neurite outgrowth, while COLV improved SC proliferation. Lastly, dNHs were fabricated into paper-like, aligned nerve scaffolds through unidirectional freezing to expand the dNH applications in PNI treatment.


Asunto(s)
Ganglios Espinales , Neuritas , Animales , Ganglios Espinales/fisiología , Regeneración Nerviosa/fisiología , Ratas , Células de Schwann/fisiología , Nervio Ciático
16.
Hear Res ; 396: 108039, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32866767

RESUMEN

Mammalian cochlear development encompasses a series of morphological and molecular events that results in the formation of a highly intricate structure responsible for hearing. One remarkable event occurs during development is the cochlear lengthening that starts with cochlear outgrowth around E11 and continues throughout development. Different mechanisms contribute to this process including cochlear progenitor proliferation and convergent extension. We previously identified that FGF9 and FGF20 promote cochlear lengthening by regulating auditory sensory epithelial proliferation through FGFR1 and FGFR2 in the periotic mesenchyme. Here, we provide evidence that ETS-domain transcription factors ETV4 and ETV5 are downstream of mesenchymal FGF signaling to control cochlear lengthening. Next generation RNA sequencing identified that Etv1, Etv4 and Etv5 mRNAs are decreased in the Fgf9 and Fgf20 double mutant periotic mesenchyme. Deleting both Etv4 and Etv5 in periotic mesenchyme resulted in shortening of cochlear length but maintaining normal patterning of organ of Corti and density of hair cells and supporting cells. This recapitulates phenotype of mesenchymal-specific Fgfr1 and Fgfr2 deleted inner ear. Furthermore, analysis of Etv1/4/5 triple conditional mutants revealed that ETV1 does not contribute in this process. Our study reveals that ETV4 and ETV5 function downstream of mesenchymal FGF signaling to promote cochlear lengthening.


Asunto(s)
Cóclea , Animales , Células Ciliadas Auditivas , ARN Mensajero , Factores de Transcripción/genética
17.
Dev Biol ; 319(2): 426-36, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18533146

RESUMEN

Lung mesenchyme is a critical determinant of the shape and size of the lung, the extent and patterning of epithelial branching, and the formation of the pulmonary vasculature and interstitial mesenchymal components of the adult lung. Fibroblast growth factor 9 (FGF9) is a critical regulator of lung mesenchymal growth; however, upstream mechanisms that modulate the FGF mesenchymal signal and the downstream targets of mesenchymal FGF signaling are poorly understood. Here we have identified a robust regulatory network in which mesenchymal FGF signaling regulates beta-Catenin mediated WNT signaling in lung mesenchyme. By conditionally inactivating beta-Catenin in lung mesenchyme, we show that mesenchymal WNT-beta-Catenin signaling is essential for lung development and acts to regulate the cell cycle G1 to S transition and the FGF responsiveness of mesenchyme. Together, both FGF and WNT signaling pathways function to sustain mesenchymal growth and coordinate epithelial morphogenesis during the pseudoglandular stage of lung development.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos/genética , Pulmón/embriología , Mesodermo/fisiología , Proteínas Wnt/genética , Animales , Femenino , Factor 9 de Crecimiento de Fibroblastos/deficiencia , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , ARN Largo no Codificante , ARN no Traducido/genética , Proteínas Wnt/deficiencia
18.
Dev Cell ; 46(5): 564-580.e5, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30100263

RESUMEN

The olfactory epithelium (OE) is a neurosensory organ required for the sense of smell. Turbinates, bony projections from the nasal cavity wall, increase the surface area within the nasal cavity lined by the OE. Here, we use engineered fibroblast growth factor 20 (Fgf20) knockin alleles to identify a population of OE progenitor cells that expand horizontally during development to populate all lineages of the mature OE. We show that these Fgf20-positive epithelium-spanning progenitor (FEP) cells are responsive to Wnt/ß-Catenin signaling. Wnt signaling suppresses FEP cell differentiation into OE basal progenitors and their progeny and positively regulates Fgf20 expression. We further show that FGF20 signals to the underlying mesenchyme to regulate the growth of turbinates. These studies thus identify a population of OE progenitor cells that function to scale OE surface area with the underlying turbinates.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Mesodermo/citología , Mucosa Olfatoria/fisiología , Células Madre/fisiología , Cornetes Nasales/crecimiento & desarrollo , Vía de Señalización Wnt , Animales , Diferenciación Celular , Células Cultivadas , Femenino , Masculino , Mesodermo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucosa Olfatoria/citología , Mucosa Olfatoria/embriología , Células Madre/citología
19.
Mol Cell Biol ; 22(22): 7721-30, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12391142

RESUMEN

Heat shock protein 72 (Hsp72) is thought to protect cells against cellular stress. The protective role of Hsp72 was investigated by determining the effect of this protein on the stress-activated protein kinase signaling pathways. Prior exposure of NIH 3T3 cells to mild heat shock (43 degrees C for 20 min) resulted in inhibition of H(2)O(2)-induced activation of apoptosis signal-regulating kinase 1 (ASK1). Overexpression of Hsp72 also inhibited H(2)O(2)-induced activation of ASK1 as well as that of downstream kinases in the p38 mitogen-activated protein kinase (MAPK) signaling cascade. Recombinant Hsp72 bound directly to ASK1 and inhibited ASK1 activity in vitro. Furthermore, coimmunoprecipitation analysis revealed a physical interaction between endogenous Hsp72 and ASK1 in NIH 3T3 cells exposed to mild heat shock. Hsp72 blocked both the homo-oligomerization of ASK1 and ASK1-dependent apoptosis. Hsp72 antisense oligonucleotides prevented the inhibitory effects of mild heat shock on H(2)O(2)-induced ASK1 activation and apoptosis. These observations suggest that Hsp72 functions as an endogenous inhibitor of ASK1.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Células 3T3 , Animales , Apoptosis/fisiología , Activación Enzimática , Genes Reporteros , Proteínas del Choque Térmico HSP72 , Proteínas de Choque Térmico/genética , Calor , Humanos , Peróxido de Hidrógeno/farmacología , MAP Quinasa Quinasa Quinasa 5 , Quinasas Quinasa Quinasa PAM/genética , Ratones , Oligonucleótidos Antisentido/metabolismo , Oxidantes/farmacología , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo
20.
BMB Rep ; 50(10): 487-495, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28855028

RESUMEN

Mammalian inner ear comprises of six sensory organs; cochlea, utricle, saccule, and three semicircular canals. The cochlea contains sensory epithelium known as the organ of Corti which senses sound through mechanosensory hair cells. Mammalian inner ear undergoes series of morphogenesis during development beginning thickening of ectoderm nearby hindbrain. These events require tight regulation of multiple signaling cascades including FGF, Wnt, Notch and Bmp signaling. In this review, we will discuss the role of newly emerging signaling, FGF signaling, for its roles required for cochlear development. [BMB Reports 2017; 50(10): 487-495].


Asunto(s)
Cóclea/crecimiento & desarrollo , Cóclea/metabolismo , Factores de Crecimiento de Fibroblastos/fisiología , Animales , Diferenciación Celular/fisiología , Humanos , Morfogénesis , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA