Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
PLoS Genet ; 17(12): e1009982, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34928956

RESUMEN

Sonic Hedgehog/GLI3 signaling is critical in regulating digit number, such that Gli3-deficiency results in polydactyly and Shh-deficiency leads to digit number reductions. SHH/GLI3 signaling regulates cell cycle factors controlling mesenchymal cell proliferation, while simultaneously regulating Grem1 to coordinate BMP-induced chondrogenesis. SHH/GLI3 signaling also coordinates the expression of additional genes, however their importance in digit formation remain unknown. Utilizing genetic and molecular approaches, we identified HES1 as a downstream modifier of the SHH/GLI signaling axis capable of inducing preaxial polydactyly (PPD), required for Gli3-deficient PPD, and capable of overcoming digit number constraints of Shh-deficiency. Our data indicate that HES1, a direct SHH/GLI signaling target, induces mesenchymal cell proliferation via suppression of Cdkn1b, while inhibiting chondrogenic genes and the anterior autopod boundary regulator, Pax9. These findings establish HES1 as a critical downstream effector of SHH/GLI3 signaling in the development of PPD.


Asunto(s)
Proteínas Hedgehog/genética , Proteínas del Tejido Nervioso/genética , Factor de Transcripción PAX9/genética , Polidactilia/genética , Pulgar/anomalías , Factor de Transcripción HES-1/genética , Proteína Gli3 con Dedos de Zinc/genética , Animales , División Celular/genética , Proliferación Celular/genética , Condrogénesis/genética , Cromatina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales de Enfermedad , Humanos , Esbozos de los Miembros/crecimiento & desarrollo , Esbozos de los Miembros/metabolismo , Mesodermo/crecimiento & desarrollo , Ratones , Polidactilia/patología , Pulgar/patología
2.
J Cell Sci ; 134(1)2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33262314

RESUMEN

Osteoblasts are the principal bone-forming cells. As such, osteoblasts have enhanced demand for amino acids to sustain high rates of matrix synthesis associated with bone formation. The precise systems utilized by osteoblasts to meet these synthetic demands are not well understood. WNT signaling is known to rapidly stimulate glutamine uptake during osteoblast differentiation. Using a cell biology approach, we identified two amino acid transporters, γ(+)-LAT1 and ASCT2 (encoded by Slc7a7 and Slc1a5, respectively), as the primary transporters of glutamine in response to WNT. ASCT2 mediates the majority of glutamine uptake, whereas γ(+)-LAT1 mediates the rapid increase in glutamine uptake in response to WNT. Mechanistically, WNT signals through the canonical ß-catenin (CTNNB1)-dependent pathway to rapidly induce Slc7a7 expression. Conversely, Slc1a5 expression is regulated by the transcription factor ATF4 downstream of the mTORC1 pathway. Targeting either Slc1a5 or Slc7a7 using shRNA reduced WNT-induced glutamine uptake and prevented osteoblast differentiation. Collectively, these data highlight the critical nature of glutamine transport for WNT-induced osteoblast differentiation.This article has an associated First Person interview with the joint first authors of the paper.


Asunto(s)
Glutamina , Osteogénesis , Diferenciación Celular , Osteoblastos , Vía de Señalización Wnt , beta Catenina
3.
Curr Osteoporos Rep ; 20(1): 53-64, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35112289

RESUMEN

PURPOSE OF REVIEW: Osteoblasts are responsible for bone matrix production during bone development and homeostasis. Much is known about the transcriptional regulation and signaling pathways governing osteoblast differentiation. However, less is known about how osteoblasts obtain or utilize nutrients to fulfill the energetic demands associated with osteoblast differentiation and bone matrix synthesis. The goal of this review is to highlight and discuss what is known about the role and regulation of bioenergetic metabolism in osteoblasts with a focus on more recent studies. RECENT FINDINGS: Bioenergetic metabolism has emerged as an important regulatory node in osteoblasts. Recent studies have begun to identify the major nutrients and bioenergetic pathways favored by osteoblasts as well as their regulation during differentiation. Here, we highlight how osteoblasts obtain and metabolize glucose, amino acids, and fatty acids to provide energy and other metabolic intermediates. In addition, we highlight the signals that regulate nutrient uptake and metabolism and focus on how energetic metabolism promotes osteoblast differentiation. Bioenergetic metabolism provides energy and other metabolites that are critical for osteoblast differentiation and activity. This knowledge contributes to a more comprehensive understanding of osteoblast biology and may inform novel strategies to modulate osteoblast differentiation and bone anabolism in patients with bone disorders.


Asunto(s)
Osteoblastos , Osteogénesis , Desarrollo Óseo , Diferenciación Celular , Metabolismo Energético/fisiología , Humanos , Osteoblastos/metabolismo
4.
Development ; 144(22): 4173-4182, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28993399

RESUMEN

For organs to achieve their proper size, the processes of stem cell renewal and differentiation must be tightly regulated. We previously showed that in the developing kidney, Wnt9b regulates distinct ß-catenin-dependent transcriptional programs in the renewing and differentiating populations of the nephron progenitor cells. How ß-catenin stimulated these two distinct programs was unclear. Here, we show that ß-catenin cooperates with the transcription factor Myc to activate the progenitor renewal program. Although in multiple contexts Myc is a target of ß-catenin, our characterization of a cell type-specific enhancer for the Wnt9b/ß-catenin target gene Fam19a5 shows that Myc and ß-catenin cooperate to activate gene expression controlled by this element. This appears to be a more general phenomenon as we find that Myc is required for the expression of every Wnt9b/ß-catenin progenitor renewal target assessed as well as for proper nephron endowment in vivo This study suggests that, within the developing kidney, tissue-specific ß-catenin activity is regulated by cooperation with cell type-specific transcription factors. This finding not only provides insight into the regulation of ß-catenin target genes in the developing kidney, but will also advance our understanding of progenitor cell renewal in other cell types/organ systems in which Myc and ß-catenin are co-expressed.


Asunto(s)
Regulación de la Expresión Génica , Proteína Proto-Oncogénica N-Myc/metabolismo , Nefronas/citología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Células Madre/metabolismo , beta Catenina/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Autorrenovación de las Células/genética , Secuencia Conservada , Elementos de Facilitación Genéticos/genética , Genes Reporteros , Células HEK293 , Humanos , Ratones , Nefronas/metabolismo , Unión Proteica/genética , Células Madre/citología , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas Wnt/metabolismo
5.
Development ; 143(2): 339-47, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26657771

RESUMEN

Exogenous bone morphogenetic proteins (Bmp) are well known to induce ectopic bone formation, but the physiological effect of Bmp signaling on normal bone is not completely understood. By deleting the receptor Bmpr1a in osteoblast lineage cells with Dmp1-Cre, we observed a dramatic increase in trabecular bone mass in postnatal mice, which was due to a marked increase in osteoblast number that was likely to be driven by hyperproliferation of Sp7(+) preosteoblasts. Similarly, inducible deletion of Bmpr1a in Sp7(+) cells specifically in postnatal mice increased trabecular bone mass. However, deletion of Smad4 by the same approaches had only a minor effect, indicating that Bmpr1a signaling suppresses trabecular bone formation through effectors beyond Smad4. Besides increasing osteoblast number in the trabecular bone, deletion of Bmpr1a by Dmp1-Cre also notably reduced osteoblast activity, resulting in attenuation of periosteal bone growth. The impairment in osteoblast activity correlated with reduced mTORC1 signaling in vivo, whereas inhibition of mTORC1 activity abolished the induction of protein anabolism genes by BMP2 treatment in vitro. Thus, physiological Bmpr1a signaling in bone exerts a dual function in both restricting preosteoblast proliferation and promoting osteoblast activity.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Células Cultivadas , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Ratones , Transducción de Señal/genética , Transducción de Señal/fisiología
6.
Cell Mol Life Sci ; 74(9): 1649-1657, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27888287

RESUMEN

The adult human skeleton is a multifunctional organ undergoing continuous remodeling. Homeostasis of bone mass in a healthy adult requires an exquisite balance between bone resorption by osteoclasts and bone formation by osteoblasts; disturbance of such balance is the root cause for various bone disorders including osteoporosis. To develop effective and safe therapeutics to modulate bone formation, it is essential to elucidate the molecular mechanisms governing osteoblast differentiation and activity. Due to their specialized function in collagen synthesis and secretion, osteoblasts are expected to consume large amounts of nutrients. However, studies of bioenergetics and building blocks in osteoblasts have been lagging behind those of growth factors and transcription factors. Genetic studies in both humans and mice over the past 15 years have established Wnt signaling as a critical mechanism for stimulating osteoblast differentiation and activity. Importantly, recent studies have uncovered that Wnt signaling directly reprograms cellular metabolism by stimulating aerobic glycolysis, glutamine catabolism as well as fatty acid oxidation in osteoblast-lineage cells. Such findings therefore reveal an important regulatory axis between bone anabolic signals and cellular bioenergetics. A comprehensive understanding of osteoblast metabolism and its regulation is likely to reveal molecular targets for novel bone therapies.


Asunto(s)
Osteoblastos/citología , Osteoblastos/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Animales , Huesos/metabolismo , Humanos , Modelos Biológicos , Vía de Señalización Wnt
7.
Proc Natl Acad Sci U S A ; 112(15): 4678-83, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25825734

RESUMEN

Hedgehog (Hh) signaling is essential for osteoblast differentiation in the endochondral skeleton during embryogenesis. However, the molecular mechanism underlying the osteoblastogenic role of Hh is not completely understood. Here, we report that Hh markedly induces the expression of insulin-like growth factor 2 (Igf2) that activates the mTORC2-Akt signaling cascade during osteoblast differentiation. Igf2-Akt signaling, in turn, stabilizes full-length Gli2 through Serine 230, thus enhancing the output of transcriptional activation by Hh. Importantly, genetic deletion of the Igf signaling receptor Igf1r specifically in Hh-responding cells diminishes bone formation in the mouse embryo. Thus, Hh engages Igf signaling in a positive feedback mechanism to activate the osteogenic program.


Asunto(s)
Diferenciación Celular , Proteínas Hedgehog/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Osteoblastos/metabolismo , Animales , Western Blotting , Línea Celular , Retroalimentación Fisiológica/efectos de los fármacos , Femenino , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/genética , Hibridación in Situ , Factor II del Crecimiento Similar a la Insulina/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Noqueados , Ratones Transgénicos , Morfolinas/farmacología , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Osteoblastos/citología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/farmacología , Interferencia de ARN , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína con Dedos de Zinc GLI1
8.
J Biol Chem ; 291(25): 13028-39, 2016 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-27129247

RESUMEN

Developmental signals in metazoans play critical roles in inducing cell differentiation from multipotent progenitors. The existing paradigm posits that the signals operate directly through their downstream transcription factors to activate expression of cell type-specific genes, which are the hallmark of cell identity. We have investigated the mechanism through which Wnt signaling induces osteoblast differentiation in an osteoblast-adipocyte bipotent progenitor cell line. Unexpectedly, Wnt3a acutely suppresses the expression of a large number of genes while inducing osteoblast differentiation. The suppressed genes include Pparg and Cebpa, which encode adipocyte-specifying transcription factors and suppression of which is sufficient to induce osteoblast differentiation. The large scale gene suppression induced by Wnt3a corresponds to a global decrease in histone acetylation, an epigenetic modification that is associated with gene activation. Mechanistically, Wnt3a does not alter histone acetyltransferase or deacetylase activities but, rather, decreases the level of acetyl-CoA in the nucleus. The Wnt-induced decrease in histone acetylation is independent of ß-catenin signaling but, rather, correlates with suppression of glucose metabolism in the tricarboxylic acid cycle. Functionally, preventing histone deacetylation by increasing nucleocytoplasmic acetyl-CoA levels impairs Wnt3a-induced osteoblast differentiation. Thus, Wnt signaling induces osteoblast differentiation in part through histone deacetylation and epigenetic suppression of an alternative cell fate.


Asunto(s)
Acetilcoenzima A/metabolismo , Diferenciación Celular , Núcleo Celular/metabolismo , Osteoblastos/fisiología , Vía de Señalización Wnt , Proteína Wnt3A/fisiología , Acetilación , Animales , Línea Celular , Ácido Cítrico/metabolismo , Ciclo del Ácido Cítrico , Expresión Génica , Silenciador del Gen , Glucosa/metabolismo , Histonas/metabolismo , Ratones , Procesamiento Proteico-Postraduccional
9.
Hum Mol Genet ; 24(15): 4365-73, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25954032

RESUMEN

Adolescent idiopathic scoliosis (AIS) and pectus excavatum (PE) are common pediatric musculoskeletal disorders. Little is known about the tissue of origin for either condition, or about their genetic bases. Common variants near GPR126/ADGRG6 (encoding the adhesion G protein-coupled receptor 126/adhesion G protein-coupled receptor G6, hereafter referred to as GPR126) were recently shown to be associated with AIS in humans. Here, we provide genetic evidence that loss of Gpr126 in osteochondroprogenitor cells alters cartilage biology and spinal column development. Microtomographic and x-ray studies revealed several hallmarks of AIS, including postnatal onset of scoliosis without malformations of vertebral units. The mutants also displayed a dorsal-ward deflection of the sternum akin to human PE. At the cellular level, these defects were accompanied by failure of midline fusion within the developing annulus fibrosis of the intervertebral discs and increased apoptosis of chondrocytes in the ribs and vertebrae. Molecularly, we found that loss of Gpr126 upregulated the expression of Gal3st4, a gene implicated in human PE, encoding Galactose-3-O-sulfotransferase 4. Together, these data uncover Gpr126 as a genetic cause for the pathogenesis of AIS and PE in a mouse model.


Asunto(s)
Tórax en Embudo/genética , Receptores Acoplados a Proteínas G/genética , Escoliosis/genética , Sulfotransferasas/genética , Animales , Cartílago , Condrocitos/patología , Modelos Animales de Enfermedad , Tórax en Embudo/patología , Predisposición Genética a la Enfermedad , Humanos , Ratones , Receptores Acoplados a Proteínas G/biosíntesis , Escoliosis/patología , Esternón/patología , Sulfotransferasas/biosíntesis
10.
Dev Biol ; 406(2): 222-34, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26321050

RESUMEN

Wnt signaling is essential to many events during organogenesis, including the development of the mammalian lung. The Wnt family member Wnt4 has been shown to be required for the development of kidney, gonads, thymus, mammary and pituitary glands. Here, we show that Wnt4 is critical for proper morphogenesis and growth of the respiratory system. Using in situ hybridization in mouse embryos, we identify a previously uncharacterized site of Wnt4 expression in the anterior trunk mesoderm. This expression domain initiates as early as E8.25 in the mesoderm abutting the tracheoesophageal endoderm, between the fusing dorsal aortae and the heart. Analysis of Wnt4(-/-) embryos reveals severe lung hypoplasia and tracheal abnormalities; however, aortic fusion and esophageal development are unaffected. We find decreased cell proliferation in Wnt4(-/-) lung buds, particularly in tip domains. In addition, we observe reduction of the important lung growth factors Fgf9, Fgf10, Sox9 and Wnt2 in the lung bud during early stages of organogenesis, as well as decreased tracheal expression of the progenitor factor Sox9. Together, these data reveal a previously unknown role for the secreted protein Wnt4 in respiratory system development.


Asunto(s)
Proliferación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Pulmón/embriología , Vía de Señalización Wnt/fisiología , Proteína Wnt4/metabolismo , Animales , Cartilla de ADN/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción SOX9/metabolismo , Proteína wnt2/metabolismo , Proteína Wnt4/genética
11.
Genesis ; 53(3-4): 285-93, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25809849

RESUMEN

The ability to image living tissues with fluorescent proteins has revolutionized the fields of cell and developmental biology. Fusions between fluorescent proteins and various polypeptides are allowing scientists to image tissues with sub-cellular resolution. Here, we describe the generation and activity of a genetically engineered mouse line expressing a fusion between the green fluorescent protein (GFP) and the apically localized protein Crumbs3 (Crb3). This reporter drives Cre-inducible expression of Crb3-GFP under control of the EF1a regulatory domains. The fusion protein is broadly expressed in embryonic and adult tissues and shows apical restriction in the majority of epithelial cell types. It displays a variably penetrant gain of function activity in the neural tube. However, in several cell types, over-expression of Crb3 does not appear to have any effect on normal development or maintenance. Detailed analysis of kidneys expressing this reporter indicates normal morphology and function highlighting the utility for live imaging. Thus, the EF1a(Crb3-GFP) mouse line will be of broad use for studying membrane and/or tissue dynamics in living tissues.


Asunto(s)
Membrana Celular/metabolismo , Embrión de Mamíferos/metabolismo , Células Epiteliales/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/metabolismo , Proteínas de la Membrana/metabolismo , Factor 1 de Elongación Peptídica/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Células Epiteliales/citología , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Procesamiento de Imagen Asistido por Computador , Riñón/citología , Riñón/metabolismo , Glicoproteínas de Membrana , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Microscopía Confocal , Técnicas de Cultivo de Órganos , Factor 1 de Elongación Peptídica/genética , Plásmidos/genética
12.
PLoS Genet ; 8(3): e1002577, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22457635

RESUMEN

Notch signaling between neighboring cells controls many cell fate decisions in metazoans both during embryogenesis and in postnatal life. Previously, we uncovered a critical role for physiological Notch signaling in suppressing osteoblast differentiation in vivo. However, the contribution of individual Notch receptors and the downstream signaling mechanism have not been elucidated. Here we report that removal of Notch2, but not Notch1, from the embryonic limb mesenchyme markedly increased trabecular bone mass in adolescent mice. Deletion of the transcription factor RBPjk, a mediator of all canonical Notch signaling, in the mesenchymal progenitors but not the more mature osteoblast-lineage cells, caused a dramatic high-bone-mass phenotype characterized by increased osteoblast numbers, diminished bone marrow mesenchymal progenitor pool, and rapid age-dependent bone loss. Moreover, mice deficient in Hey1 and HeyL, two target genes of Notch-RBPjk signaling, exhibited high bone mass. Interestingly, Hey1 bound to and suppressed the NFATc1 promoter, and RBPjk deletion increased NFATc1 expression in bone. Finally, pharmacological inhibition of NFAT alleviated the high-bone-mass phenotype caused by RBPjk deletion. Thus, Notch-RBPjk signaling functions in part through Hey1-mediated inhibition of NFATc1 to suppress osteoblastogenesis, contributing to bone homeostasis in vivo.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Huesos/metabolismo , Diferenciación Celular , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas , Factores de Transcripción NFATC , Osteoblastos , Receptor Notch2 , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Huesos/embriología , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Mesodermo/embriología , Ratones , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoblastos/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptor Notch2/genética , Receptor Notch2/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Células Madre/metabolismo
13.
Development ; 138(7): 1247-57, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21350016

RESUMEN

The mammalian kidney is composed of thousands of individual epithelial tubules known as nephrons. Deficits in nephron number are associated with myriad diseases ranging from complete organ failure to congenital hypertension. A balance between differentiation and maintenance of a mesenchymal progenitor cell population determines the final number of nephrons. How this balance is struck is poorly understood. Previous studies have suggested that Wnt9b/ß-catenin signaling induced differentiation (mesenchymal-to-epithelial transition) in a subset of the progenitors but needed to be repressed in the remaining progenitors to keep them in the undifferentiated state. Here, we report that Wnt9b/ß-catenin signaling is active in the progenitors and is required for their renewal/proliferation. Using a combination of approaches, we have revealed a mechanism through which cells receiving the same Wnt9b/ß-catenin signal can respond in distinct ways (proliferate versus differentiate) depending on the cellular environment in which the signal is received. Interpretation of the signal is dependent, at least in part, on the activity of the transcription factor Six2. Six2-positive cells that receive the Wnt9b signal are maintained as progenitors whereas cells with reduced levels of Six2 are induced to differentiate by Wnt9b. Using this simple mechanism, the kidney is able to balance progenitor cell expansion and differentiation insuring proper nephron endowment. These findings provide novel insights into the molecular mechanisms that regulate progenitor cell differentiation during normal and pathological conditions.


Asunto(s)
Riñón/embriología , Nefronas/embriología , Transducción de Señal/fisiología , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Riñón/citología , Riñón/metabolismo , Ratones , Nefronas/citología , Nefronas/metabolismo , Organogénesis/fisiología , Células Madre/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
14.
Adv Nanobiomed Res ; 4(3)2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38665311

RESUMEN

Natural biomaterials are commonly used as tissue engineering scaffolds due to their biocompatibility and biodegradability. Plant-derived materials have also gained significant interest due to their abundance and as a sustainable resource. This study evaluates the corn-derived protein zein as a plant-derived substitute for animal-derived gelatin, which is widely used for its favorable cell adhesion properties. Limited studies exist evaluating pure zein for tissue engineering. Herein, fibrous zein scaffolds are evaluated in vitro for cell adhesion, growth, and infiltration into the scaffold in comparison to gelatin scaffolds and are further studied in a subcutaneous model in vivo. Human mesenchymal stem cells (MSCs) on zein scaffolds express focal adhesion kinase and integrins such as αvß3, α4, and ß1 similar to gelatin scaffolds. MSCs also infiltrate zein scaffolds with a greater penetration depth than cells on gelatin scaffolds. Cells loaded onto zein scaffolds in vivo show higher cell proliferation and CD31 expression, as an indicator of blood vessel formation. Findings also demonstrate the capability of zein scaffolds to maintain the multipotent capability of MSCs. Overall, findings demonstrate plant-derived zein may be a suitable alternative to the animalderived gelatin and demonstrates zein's potential as a scaffold for tissue engineering.

15.
Nat Metab ; 6(1): 113-126, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38167727

RESUMEN

Chronic stress and inflammation are both outcomes and major drivers of many human diseases. Sustained responsiveness despite mitigation suggests a failure to sense resolution of the stressor. Here we show that a proteolytic cleavage event of fatty acid synthase (FASN) activates a global cue for stress resolution in Caenorhabditis elegans. FASN is well established for biosynthesis of the fatty acid palmitate. Our results demonstrate FASN promoting an anti-inflammatory profile apart from palmitate synthesis. Redox-dependent proteolysis of limited amounts of FASN by caspase activates a C-terminal fragment sufficient to downregulate multiple aspects of stress responsiveness, including gene expression, metabolic programs and lipid droplets. The FASN C-terminal fragment signals stress resolution in a cell non-autonomous manner. Consistent with these findings, FASN processing is also seen in well-fed but not fasted male mouse liver. As downregulation of stress responses is critical to health, our findings provide a potential pathway to control diverse aspects of stress responses.


Asunto(s)
Ácido Graso Sintasas , Ácidos Grasos , Animales , Masculino , Ratones , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Palmitatos , Proteolisis , Caenorhabditis elegans , Acido Graso Sintasa Tipo I
16.
J Biol Chem ; 287(27): 23246-54, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22593577

RESUMEN

Secreted Wnt proteins constitute one of the largest families of intercellular signaling molecules in vertebrates with essential roles in embryonic development and adult tissue homeostasis. The functional redundancy of Wnt genes and the many forms of cellular responses they elicit, including some utilizing the transcriptional co-activator ß-catenin, has limited the ability of classical genetic strategies to uncover their roles in vivo. We had previously identified a chemical compound class termed Inhibitor of Wnt Production (or IWP) that targets Porcupine (Porcn), an acyltransferase catalyzing the addition of fatty acid adducts onto Wnt proteins. Here we demonstrate that diverse chemical structures are able to inhibit Porcn by targeting its putative active site. When deployed in concert with small molecules that modulate the activity of Tankyrase enzymes and glycogen synthase kinase 3 ß (GSK3ß), additional transducers of Wnt/ß-catenin signaling, the IWP compounds reveal an essential role for Wnt protein fatty acylation in eliciting ß-catenin-dependent and -independent forms of Wnt signaling during zebrafish development. This collection of small molecules facilitates rapid dissection of Wnt gene function in vivo by limiting the influence of redundant Wnt gene functions on phenotypic outcomes and enables temporal manipulation of Wnt-mediated signaling in vertebrates.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Regeneración Tisular Dirigida/métodos , Proteínas de la Membrana/antagonistas & inhibidores , Andamios del Tejido , Vía de Señalización Wnt/fisiología , Aciltransferasas , Animales , Animales Modificados Genéticamente , Antineoplásicos/farmacología , Células COS , Membrana Celular/enzimología , Chlorocebus aethiops , Diseño de Fármacos , Células HEK293 , Células HeLa , Humanos , Riñón/citología , Riñón/embriología , Riñón/enzimología , Proteínas de la Membrana/metabolismo , Técnicas de Cultivo de Órganos , Vía de Señalización Wnt/efectos de los fármacos , Pez Cebra , beta Catenina/metabolismo
17.
Development ; 137(19): 3233-43, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20823064

RESUMEN

The integrin-linked kinase (ILK), pinch and parvin ternary complex connects the cytoplasmic tails of beta1 integrins to the actin cytoskeleton. We recently showed that constitutive expression of ILK and alpha parvin in both the ureteric bud and the metanephric mesenchyme of the kidney is required for kidney development. In this study, we define the selective role of ILK in the ureteric bud of the mouse kidney in renal development by deleting it in the ureteric cell lineage before the onset of branching morphogenesis (E10.5). Although deleting ILK resulted in only a moderate decrease in branching, the mice died at 8 weeks of age from obstruction due to the unprecedented finding of intraluminal collecting duct cellular proliferation. ILK deletion in the ureteric bud resulted in the inability of collecting duct cells to undergo contact inhibition and to activate p38 mitogen-activated protein kinase (MAPK) in vivo and in vitro. p38 MAPK activation was not dependent on the kinase activity of ILK. Thus, we conclude that ILK plays a crucial role in activating p38 MAPK, which regulates cell cycle arrest of epithelial cells in renal tubulogenesis.


Asunto(s)
Ciclo Celular , Proteínas Serina-Treonina Quinasas/metabolismo , Uréter/embriología , Uréter/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Uréter/citología
18.
JCI Insight ; 8(16)2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37432749

RESUMEN

Reactive oxygen species (ROS) are natural products of mitochondrial oxidative metabolism and oxidative protein folding. ROS levels must be well controlled, since elevated ROS has been shown to have deleterious effects on osteoblasts. Moreover, excessive ROS is thought to underlie many of the skeletal phenotypes associated with aging and sex steroid deficiency in mice and humans. The mechanisms by which osteoblasts regulate ROS and how ROS inhibits osteoblasts are not well understood. Here, we demonstrate that de novo glutathione (GSH) biosynthesis is essential in neutralizing ROS and establish a proosteogenic reduction and oxidation reaction (REDOX) environment. Using a multifaceted approach, we demonstrate that reducing GSH biosynthesis led to acute degradation of RUNX2, impaired osteoblast differentiation, and reduced bone formation. Conversely, reducing ROS using catalase enhanced RUNX2 stability and promoted osteoblast differentiation and bone formation when GSH biosynthesis was limited. Highlighting the therapeutic implications of these findings, in utero antioxidant therapy stabilized RUNX2 and improved bone development in the Runx2+/- haplo-insufficient mouse model of human cleidocranial dysplasia. Thus, our data establish RUNX2 as a molecular sensor of the osteoblast REDOX environment and mechanistically clarify how ROS negatively impacts osteoblast differentiation and bone formation.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal , Osteogénesis , Ratones , Humanos , Animales , Osteogénesis/genética , Especies Reactivas de Oxígeno , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Oxidación-Reducción , Glutatión/metabolismo
19.
J Vis Exp ; (182)2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35499335

RESUMEN

Bone development and homeostasis is dependent upon the differentiation and activity of bone forming osteoblasts. Osteoblast differentiation is sequentially characterized by proliferation followed by protein synthesis and ultimately bone matrix secretion. Proliferation and protein synthesis require a constant supply of amino acids. Despite this, very little is known about amino acid consumption in osteoblasts. Here we describe a very sensitive protocol that is designed to measure amino acid consumption using radiolabeled amino acids. This method is optimized to quantify changes in amino acid uptake that are associated with osteoblast proliferation or differentiation, drug or growth factor treatments, or various genetic manipulations. Importantly, this method can be used interchangeably to quantify amino acid consumption in cultured cell lines or primary cells in vitro or in isolated bone shafts ex vivo. Finally, our method can be easily adapted to measure the transport of any of the amino acids as well as glucose and other radiolabeled nutrients.


Asunto(s)
Aminoácidos , Desarrollo Óseo , Línea Celular , Osteoblastos , Biosíntesis de Proteínas
20.
Front Physiol ; 13: 992679, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36213239

RESUMEN

Amino acids have recently emerged as important regulators of osteoblast differentiation and bone formation. Osteoblasts require a continuous supply of amino acids to sustain biomass production to fuel cell proliferation, osteoblast differentiation and bone matrix production. We recently identified proline as an essential amino acid for bone development by fulfilling unique synthetic demands that are associated with osteoblast differentiation. Osteoblasts rely on the amino acid transporter SLC38A2 to provide proline to fuel endochondral ossification. Despite this, very little is known about the function or substrates of SLC38A2 during bone homeostasis. Here we demonstrate that the neutral amino acid transporter SLC38A2 is expressed in osteoblast lineage cells and provides proline and alanine to osteoblast lineage cells. Genetic ablation of SLC38A2 using Prrx1Cre results in decreased bone mass in both male and female mice due to a reduction in osteoblast numbers and bone forming activity. Decreased osteoblast numbers are attributed to impaired proliferation and osteogenic differentiation of skeletal stem and progenitor cells. Collectively, these data highlight the necessity of SLC38A2-mediated proline and alanine uptake during postnatal bone formation and bone homeostasis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA