Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.210
Filtrar
1.
Mol Biol Rep ; 51(1): 632, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724827

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) play critical roles in the osteogenic differentiation of human bone mesenchymal stem cells (hBMSCs), but the mechanism by which miRNAs indirectly modulate osteogenesis remains unclear. Here, we explored the mechanism by which miRNAs indirectly modulate gene expression through histone demethylases to promote bone regeneration. METHODS AND RESULTS: Bioinformatics analysis was performed on hBMSCs after 7 days of osteogenic induction. The differentially expressed miRNAs were screened, and potential target mRNAs were identified. To determine the bioactivity and stemness of hBMSCs and their potential for bone repair, we performed wound healing, Cell Counting Kit-8 (CCK-8), real-time reverse transcription quantitative polymerase chain reaction (RT‒qPCR), alkaline phosphatase activity, alizarin red S (ARS) staining and radiological and histological analyses on SD rats with calvarial bone defects. Additionally, a dual-luciferase reporter assay was utilized to investigate the interaction between miR-26b-5p and ten-eleven translocation 3 (TET3) in human embryonic kidney 293T cells. The in vitro and in vivo results suggested that miR-26b-5p effectively promoted the migration, proliferation and osteogenic differentiation of hBMSCs, as well as the bone reconstruction of calvarial defects in SD rats. Mechanistically, miR-26b-5p bound to the 3' untranslated region of TET3 mRNA to mediate gene silencing. CONCLUSIONS: MiR-26b-5p downregulated the expression of TET3 to increase the osteogenic differentiation of hBMSCs and bone repair in rat calvarial defects. MiR-26b-5p/TET3 crosstalk might be useful in large-scale critical bone defects.


Asunto(s)
Regeneración Ósea , Diferenciación Celular , Dioxigenasas , Células Madre Mesenquimatosas , MicroARNs , Osteogénesis , Ratas Sprague-Dawley , Cráneo , MicroARNs/genética , MicroARNs/metabolismo , Animales , Células Madre Mesenquimatosas/metabolismo , Humanos , Osteogénesis/genética , Diferenciación Celular/genética , Ratas , Cráneo/patología , Cráneo/metabolismo , Femenino , Regeneración Ósea/genética , Dioxigenasas/genética , Dioxigenasas/metabolismo , Proliferación Celular/genética , Células HEK293
2.
Development ; 151(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38602508

RESUMEN

The skull roof, or calvaria, is comprised of interlocking plates of bones that encase the brain. Separating these bones are fibrous sutures that permit growth. Currently, we do not understand the instructions for directional growth of the calvaria, a process which is error-prone and can lead to skeletal deficiencies or premature suture fusion (craniosynostosis, CS). Here, we identify graded expression of fibronectin (FN1) in the mouse embryonic cranial mesenchyme (CM) that precedes the apical expansion of calvaria. Conditional deletion of Fn1 or Wasl leads to diminished frontal bone expansion by altering cell shape and focal actin enrichment, respectively, suggesting defective migration of calvarial progenitors. Interestingly, Fn1 mutants have premature fusion of coronal sutures. Consistently, syndromic forms of CS in humans exhibit dysregulated FN1 expression, and we also find FN1 expression altered in a mouse CS model of Apert syndrome. These data support a model of FN1 as a directional substrate for calvarial osteoblast migration that may be a common mechanism underlying many cranial disorders of disparate genetic etiologies.


Asunto(s)
Fibronectinas , Nacimiento Prematuro , Cráneo , Animales , Femenino , Humanos , Ratones , Señales (Psicología) , Modelos Animales de Enfermedad , Fibronectinas/metabolismo , Osteoblastos , Cráneo/citología , Cráneo/crecimiento & desarrollo , Cráneo/metabolismo , Suturas
3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 469-474, 2024 Mar 20.
Artículo en Chino | MEDLINE | ID: mdl-38645865

RESUMEN

Craniomaxillofacial development involves a series of highly ordered temporal-spatial cellular differentiation processes in which a variety of cell signaling factors, such as fibroblast growth factors, play important regulatory roles. As a classic fibroblast growth factor, fibroblast growth factor 7 (FGF7) serves a wide range of regulatory functions. Previous studies have demonstrated that FGF7 regulates the proliferation and migration of epithelial cells, protects them, and promotes their repair. Furthermore, recent findings indicate that epithelial cells are not the only ones subjected to the broad and powerful regulatory capacity of FGF7. It has potential effects on skeletal system development as well. In addition, FGF7 plays an important role in the development of craniomaxillofacial organs, such as the palate, the eyes, and the teeth. Nonetheless, the role of FGF7 in oral craniomaxillofacial development needs to be further elucidated. In this paper, we summarized the published research on the role of FGF7 in oral craniomaxillofacial development to demonstrate the overall understanding of FGF7 and its potential functions in oral craniomaxillofacial development.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos , Humanos , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Factor 7 de Crecimiento de Fibroblastos/genética , Animales , Cráneo/crecimiento & desarrollo , Cráneo/metabolismo , Desarrollo Maxilofacial/fisiología , Diente/metabolismo , Diente/crecimiento & desarrollo
4.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(5): 426-434, 2024 May 09.
Artículo en Chino | MEDLINE | ID: mdl-38636996

RESUMEN

Craniofacial hard tissue mainly includes craniofacial bone and tooth, which is one of the important parts of the mouth-jaw system. Congenital aplasia, tumors and trauma can cause large craniofacial hard tissue defects, which are detrimental to the facial appearance and function of patients, and affect the physical and mental health of patients. Histone acetylation modification is the earliest and most widely studied histone modification, which is an epigenetic modification mechanism jointly regulated by histone acetyltransferase and histone deacetylase. In this paper, we will review the research progress of histone acetylation mediated by histone acetyltransferase and histone deacetylase in the development and regeneration of craniofacial hard tissue.


Asunto(s)
Histona Acetiltransferasas , Histonas , Regeneración , Acetilación , Humanos , Histonas/metabolismo , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Epigénesis Genética , Diente/metabolismo , Huesos Faciales , Cráneo/metabolismo
5.
Carbohydr Polym ; 336: 122115, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38670750

RESUMEN

To alleviate skull defects and enhance the biological activity of taxifolin, this study utilized the thin-film dispersion method to prepare paclitaxel liposomes (TL). Thiolated chitosan (CSSH)-modified TL (CTL) was synthesized through charge interactions. Injectable hydrogels (BLG) were then prepared as hydrogel scaffolds loaded with TAX (TG), TL (TLG), and CTL (CTLG) using a Schiff base reaction involving oxidized dextran and carboxymethyl chitosan. The study investigated the bone reparative properties of CTLG through molecular docking, western blot techniques, and transcriptome analysis. The particle sizes of CTL were measured at 248.90 ± 14.03 nm, respectively, with zeta potentials of +36.68 ± 5.43 mV, respectively. CTLG showed excellent antioxidant capacity in vitro. It also has a good inhibitory effect on Escherichia coli and Staphylococcus aureus, with inhibition rates of 93.88 ± 1.59 % and 88.56 ± 2.83 % respectively. The results of 5-ethynyl-2 '-deoxyuridine staining, alkaline phosphatase staining and alizarin red staining showed that CTLG also had the potential to promote the proliferation and differentiation of mouse embryonic osteoblasts (MC3T3-E1). The study revealed that CTLG enhances the expression of osteogenic proteins by regulating the Wnt signaling pathway, shedding light on the potential application of TAX and bone regeneration mechanisms.


Asunto(s)
Proliferación Celular , Quitosano , Hidrogeles , Liposomas , Osteoblastos , Quercetina , Quercetina/análogos & derivados , Cráneo , Vía de Señalización Wnt , Animales , Quitosano/análogos & derivados , Quitosano/química , Quitosano/farmacología , Quercetina/farmacología , Quercetina/química , Liposomas/química , Vía de Señalización Wnt/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Proliferación Celular/efectos de los fármacos , Ratones , Cráneo/efectos de los fármacos , Cráneo/patología , Cráneo/metabolismo , Ratas , Regeneración Ósea/efectos de los fármacos , Ratas Sprague-Dawley , Osteogénesis/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Diferenciación Celular/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Masculino , Simulación del Acoplamiento Molecular
6.
J Bone Miner Res ; 39(4): 498-512, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38477756

RESUMEN

Mutations in the Chromodomain helicase DNA-binding protein 7 - coding gene (CHD7) cause CHARGE syndrome (CS). Although craniofacial and skeletal abnormalities are major features of CS patients, the role of CHD7 in bone and cartilage development remain largely unexplored. Here, using a zebrafish (Danio rerio) CS model, we show that chd7-/- larvae display abnormal craniofacial cartilage development and spinal deformities. The craniofacial and spine defects are accompanied by a marked reduction of bone mineralization. At the molecular level, we show that these phenotypes are associated with significant reduction in the expression levels of osteoblast differentiation markers. Additionally, we detected a marked depletion of collagen 2α1 in the cartilage of craniofacial regions and vertebrae, along with significantly reduced number of chondrocytes. Chondrogenesis defects are at least in part due to downregulation of htr2b, which we found to be also dysregulated in human cells derived from an individual with CHD7 mutation-positive CS. Overall, this study thus unveils an essential role for CHD7 in cartilage and bone development, with potential clinical relevance for the craniofacial defects associated with CS.


Patients with CHARGE syndrome exhibit skeletal defects. CHARGE syndrome is primarily caused by mutations in the chromatin remodeler-coding gene CHD7. To investigate the poorly characterized role of CHD7 in cartilage and bone development, here, we examine the craniofacial and bone anomalies in a zebrafish chd7-/- mutant model. We find that zebrafish mutant larvae exhibit striking dysmorphism of craniofacial structures and spinal deformities. Notably, we find a significant reduction in osteoblast, chondrocyte, and collagen matrix markers. This work provides important insights to improve our understanding of the role of chd7 in skeletal development.


Asunto(s)
Cartílago , ADN Helicasas , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/metabolismo , Pez Cebra/genética , Pez Cebra/embriología , Cartílago/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Humanos , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Síndrome CHARGE/genética , Síndrome CHARGE/metabolismo , Síndrome CHARGE/patología , Cráneo/metabolismo , Condrocitos/metabolismo , Condrogénesis/genética , Colágeno Tipo II/metabolismo , Colágeno Tipo II/genética
7.
J Mol Med (Berl) ; 102(5): 693-707, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38492027

RESUMEN

Physical therapy is extensively employed in clinical settings. Nevertheless, the absence of suitable animal models has resulted in an incomplete understanding of the in vivo mechanisms and cellular distribution that respond to physical stimuli. The objective of this research was to create a mouse model capable of indicating the cells affected by physical stimuli. In this study, we successfully established a mouse line based on the heat shock protein 70 (Hsp70) promoter, wherein the expression of CreERT2 can be induced by physical stimuli. Following stimulation of the mouse tail, ear, or cultured calvarias with heat shock (generated by heating, ultrasound, or laser), a distinct Cre-mediated excision was observed in cells stimulated by these physical factors with minimal occurrence of leaky reporter expression. The application of heat shock to Hsp70-CreERT2; FGFR2-P253R double transgenic mice or Hsp70-CreERT2 mice infected with AAV-BMP4 at calvarias induced the activation of Cre-dependent mutant FGFR2-P253R or BMP4 respectively, thereby facilitating the premature closure of cranial sutures or the repair of calvarial defects. This novel mouse line holds significant potential for investigating the underlying mechanisms of physical therapy, tissue repair and regeneration, lineage tracing, and targeted modulation of gene expression of cells in local tissue stimulated by physical factor at the interested time points. KEY MESSAGES: In the study, an Hsp70-CreERT2 transgenic mouse was generated for heat shock-induced gene modulation. Heat shock, ultrasound, and laser stimulation effectively activated Cre expression in Hsp70-CreERT2; reporter mice, which leads to deletion of floxed DNA sequence in the tail, ear, and cultured calvaria tissues of mice. Local laser stimuli on cultured calvarias effectively induce Fgfr2-P253R expression in Hsp70-mTmG-Fgfr2-P253R mice and result in accelerated premature closure of cranial suture. Heat shock activated AAV9-FLEX-BMP4 expression and subsequently promoted the repair of calvarial defect of Hsp70-CreERT2; Rosa26-mTmG mice.


Asunto(s)
Proteína Morfogenética Ósea 4 , Proteínas HSP70 de Choque Térmico , Ratones Transgénicos , Regiones Promotoras Genéticas , Animales , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Ratones , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/genética , Respuesta al Choque Térmico/genética , Cráneo/metabolismo , Regulación de la Expresión Génica , Integrasas/metabolismo , Integrasas/genética
8.
ACS Appl Mater Interfaces ; 16(5): 5486-5503, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38284176

RESUMEN

Cranial bone defects remain a major clinical challenge, increasing patients' life burdens. Tricarboxylic acid (TCA) cycle metabolites play crucial roles in facilitating bone tissue regeneration. However, the development of TCA cycle metabolite-modified biomimetic grafts for skull bone regeneration still needs to be improved. The mechanism underlying the release of TCA cycle metabolites from biomaterials in regulating immune responses and mesenchymal stem cell (MSC) fate (migration and differentiation) remains unknown. Herein, this work constructs biomimetic hydrogels composed of gelatin and chitosan networks covalently cross-linked by genipin (CGG hydrogels). A series of TCA cycle metabolite-coordinated CGG hydrogels with strong mechanical and antiswelling performances are subsequently developed. Remarkably, the citrate (Na3Cit, Cit)-coordinated CGG hydrogels (CGG-Cit hydrogels) with the highest mechanical modulus and strength significantly promote skull bone regeneration in rat and murine cranial defects. Mechanistically, using a transgenic mouse model, bulk RNA sequencing, and single-cell RNA sequencing, this work demonstrates that CGG-Cit hydrogels promote Gli1+ MSC migration via neutrophil-secreted oncostatin M. Results also indicate that citrate improves osteogenesis via enhanced histone H3K9 acetylation on osteogenic master genes. Taken together, the immune microenvironment- and MSC fate-regulated CGG-Cit hydrogels represent a highly efficient and facile approach toward skull bone tissue regeneration with great potential for bench-to-bedside translation.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Humanos , Ratas , Ratones , Animales , Histonas , Ciclo del Ácido Cítrico , Acetilación , Neutrófilos/metabolismo , Regeneración Ósea , Cráneo/metabolismo , Diferenciación Celular , Hidrogeles/farmacología , Hidrogeles/metabolismo , Citratos
9.
Genesis ; 62(1): e23580, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37974491

RESUMEN

Bop1 can promote cell proliferation and is a component of the Pes1-Bop1-WDR12 (PeBoW) complex that regulates ribosomal RNA processing and biogenesis. In embryos, however, bop1 mRNA is highly enriched in the neural plate, cranial neural crest and placodes, and potentially may interact with Six1, which also is expressed in these tissues. Recent work demonstrated that during development, Bop1 is required for establishing the size of the tadpole brain, retina and cranial cartilages, as well as controlling neural tissue gene expression levels. Herein, we extend this work by assessing the effects of Bop1 knockdown at neural plate and larval stages. Loss of Bop1 expanded neural plate gene expression domains (sox2, sox11, irx1) and reduced neural crest (foxd3, sox9), placode (six1, sox11, irx1, sox9) and epidermal (dlx5) expression domains. At larval stages, Bop1 knockdown reduced the expression of several otic vesicle genes (six1, pax2, irx1, sox9, dlx5, otx2, tbx1) and branchial arch genes that are required for chondrogenesis (sox9, tbx1, dlx5). The latter was not the result of impaired neural crest migration. Together these observations indicate that Bop1 is a multifunctional protein that in addition to its well-known role in ribosomal biogenesis functions during early development to establish the craniofacial precursor domains.


Asunto(s)
Cresta Neural , Factores de Transcripción , Cresta Neural/metabolismo , Factores de Transcripción/metabolismo , Cabeza , Cráneo/metabolismo , Ribosomas/metabolismo , Regulación del Desarrollo de la Expresión Génica
10.
Clin Nucl Med ; 49(1): e31-e32, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38015722

RESUMEN

ABSTRACT: An osteoid osteoma (OO) is a benign bone neoplasm, characterized by significant nocturnal pain that usually responds to nonsteroidal anti-inflammatory drugs. It occurs most commonly in the lower extremities and vertebrae. Here, we present a case of carcinoma prostate, who was referred to our department for 68 Ga-PSMA PET/CT scan, and we incidentally found out PSMA-avid OO involving frontal bone of skull, which is a rare finding. To the best of our knowledge, this is the second case in which high PSMA uptake is found in the OO, suggesting a possible PSMA expression related to osteoblastic activity.


Asunto(s)
Neoplasias Óseas , Osteoma Osteoide , Neoplasias de la Próstata , Masculino , Humanos , Tomografía Computarizada por Tomografía de Emisión de Positrones , Osteoma Osteoide/diagnóstico por imagen , Radioisótopos de Galio , Neoplasias de la Próstata/patología , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/metabolismo , Cráneo/metabolismo , Ácido Edético/metabolismo
11.
Int J Mol Sci ; 24(23)2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-38068970

RESUMEN

The long pentraxin 3 (PTX3) is a soluble glycoprotein made by immune and nonimmune cells endowed with pleiotropic functions in innate immunity, inflammation, and tissue remodeling. PTX3 has recently emerged as a mediator of bone turnover in both physiological and pathological conditions, with direct and indirect effects on osteoblasts and osteoclasts. This notwithstanding, its role in bone biology, with major regard to the osteogenic potential of osteoblasts and their interplay with osteoclasts, is at present unclear. Here, we investigated the contribution of this pentraxin to bone deposition in the osteogenic lineage by assessing collagen production, mineralization capacity, osteoblast maturation, extracellular matrix gene expression, and inflammatory mediators' production in primary osteoblasts from the calvaria of wild-type (WT) and Ptx3-deficient (Ptx3-/-) mice. Also, we evaluated the effect of PTX3 on osteoclastogenesis in cocultures of primary osteoblasts and bone marrow-derived osteoclasts. Our investigations were carried out both in physiological and inflammatory conditions to recapitulate in vitro aspects of inflammatory diseases of the bone. We found that primary osteoblasts from WT animals constitutively expressed low levels of the protein in osteogenic noninflammatory conditions, and genetic ablation of PTX3 in these cells had no major impact on collagen and hydroxyapatite deposition. However, Ptx3-/- osteoblasts had an increased RANKL/OPG ratio and CD44 expression, which resulted in in enhanced osteoclastogenesis when cocultured with bone marrow monocytes. Inflammation (modelled through administration of tumor necrosis factor-α, TNF-α) boosted the expression and accumulation of PTX3 and inflammatory mediators in WT osteoblasts. In these conditions, Ptx3 genetic depletion was associated with reduced collagen deposition and immune modulators' production. Our study shed light on the role of PTX3 in osteoblast and osteoclast biology and identified a major effect of inflammation on the bone-related properties of this pentraxin, which might be relevant for therapeutic and/or diagnostic purposes in musculoskeletal pathology.


Asunto(s)
Osteoclastos , Osteogénesis , Ratones , Animales , Osteogénesis/genética , Osteoclastos/metabolismo , Osteoblastos/metabolismo , Inflamación/metabolismo , Diferenciación Celular , Cráneo/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Colágeno/metabolismo , Mediadores de Inflamación/metabolismo , Ligando RANK/metabolismo
12.
Sci Rep ; 13(1): 22028, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-38086873

RESUMEN

Osteocytes form a cellular network by gap junctions between their cell processes. This network is important since intercellular communication via the network is essential for bone metabolism. However, the factors that influence the formation of this osteocyte network remain unknown. As the early stage of osteocyte network formation occurs on the bone surface, we observed a newly formed trabecular bone surface by orthogonal focused ion beam-scanning electron microscopy. The embedding late osteoblast processes tended to avoid bundled collagen fibrils and elongate into sparse collagen fibrils. Then, we examined whether the inhibition of bundling of collagen fibrils using a potent lysyl oxidase inhibitor, ß-aminopropionitrile (BAPN) changed the cellular network of the chick calvaria. The osteocyte shape of the control group was spindle-shape, while that of the BAPN group was sphere-shaped. In addition, the osteocyte processes of the control group were elongated vertically to the long axis of the cell body, whereas the osteocyte processes of the BAPN group were elongated radially. Therefore, it was suggested that the bundling of collagen fibrils influences normal osteocyte network formation during bone modeling.


Asunto(s)
Aminopropionitrilo , Osteocitos , Osteocitos/metabolismo , Matriz Extracelular/metabolismo , Cráneo/metabolismo , Colágeno/metabolismo
13.
J Neuroinflammation ; 20(1): 294, 2023 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-38071333

RESUMEN

Skull bone marrow is thought to be an immune tissue closely associated with the central nervous system (CNS). Recent studies have focused on the role of skull bone marrow in central nervous system disorders. In this study, we performed single-cell RNA sequencing on ipsilateral and contralateral skull bone marrow cells after experimental stroke and then performed flow cytometry and analysis of cytokine expression. Skull marrow showed lateralization in response to stroke. Lateralization is demonstrated primarily by the proliferation and differentiation of myeloid and lymphoid lineage cells in the skull bone marrow adjacent to the ischemic region, with an increased proportion of neutrophils compared to monocytes. Analysis of chemokines in the skull revealed marked differences in chemotactic signals between the ipsilateral and contralateral skull, whereas sympathetic signals innervating the skull did not affect cranial bone marrow lateralization. Osteopontin (OPN) is involved in region-specific activation of the skull marrow that promotes inflammation in the meninges, and inhibition of OPN expression improves neurological function.


Asunto(s)
Médula Ósea , Osteopontina , Accidente Cerebrovascular , Animales , Ratones , Isquemia , Osteopontina/metabolismo , Cráneo/metabolismo
14.
Front Endocrinol (Lausanne) ; 14: 1224318, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37886644

RESUMEN

Pathogenic heterozygous variants in SGMS2 cause a rare monogenic form of osteoporosis known as calvarial doughnut lesions with bone fragility (CDL). The clinical presentations of SGMS2-related bone pathology range from childhood-onset osteoporosis with low bone mineral density and sclerotic doughnut-shaped lesions in the skull to a severe spondylometaphyseal dysplasia with neonatal fractures, long-bone deformities, and short stature. In addition, neurological manifestations occur in some patients. SGMS2 encodes sphingomyelin synthase 2 (SMS2), an enzyme involved in the production of sphingomyelin (SM). This review describes the biochemical structure of SM, SM metabolism, and their molecular actions in skeletal and neural tissue. We postulate how disrupted SM gradient can influence bone formation and how animal models may facilitate a better understanding of SGMS2-related osteoporosis.


Asunto(s)
Nervio Facial , Osteoporosis , Transferasas (Grupos de Otros Fosfatos Sustitutos) , Animales , Niño , Humanos , Recién Nacido , Nervio Facial/metabolismo , Nervio Facial/patología , Osteoporosis/complicaciones , Osteoporosis/patología , Parálisis , Cráneo/metabolismo , Esfingomielinas/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo
15.
Dev Dyn ; 252(12): 1407-1427, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37597164

RESUMEN

BACKGROUND: Members of the sulfotransferase superfamily (SULT) influence the activity of a wide range of hormones, neurotransmitters, metabolites and xenobiotics. However, their roles in developmental processes are not well characterized even though they are expressed during embryogenesis. We previously found in a microarray screen that Six1 up-regulates LOC100037047, which encodes XB5850668.L, an uncharacterized sulfotransferase. RESULTS: Since Six1 is required for patterning the embryonic ectoderm into its neural plate, neural crest, preplacodal and epidermal domains, we used loss- and gain-of function assays to characterize the role of XB5850668.L during this process. Knockdown of endogenous XB5850668.L resulted in the reduction of epidermal, neural crest, cranial placode and otic vesicle gene expression domains, concomitant with neural plate expansion. Increased levels had minimal effects, but infrequently expanded neural plate and neural crest gene domains, and infrequently reduced cranial placode and otic vesicle gene domains. Mutation of two key amino acids in the sulfotransferase catalytic domain required for PAPS binding and enzymatic activity tended to reduce the effects of overexpressing the wild-type protein. CONCLUSIONS: Our analyses indicates that XB5850668.L is a member of the SULT2 family that plays important roles in patterning the embryonic ectoderm. Some aspects of its influence likely depend on sulfotransferase activity.


Asunto(s)
Ectodermo , Cresta Neural , Cresta Neural/metabolismo , Cráneo/metabolismo , Desarrollo Embrionario/genética , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Regulación del Desarrollo de la Expresión Génica
16.
Dev Biol ; 501: 39-59, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37301464

RESUMEN

The vertebrate head mesoderm provides the heart, the great vessels, some smooth and most head skeletal muscle, in addition to parts of the skull. It has been speculated that the ability to generate cardiac and smooth muscle is the evolutionary ground-state of the tissue. However, whether indeed the entire head mesoderm has generic cardiac competence, how long this may last, and what happens as cardiac competence fades, is not clear. Bone morphogenetic proteins (Bmps) are known to promote cardiogenesis. Using 41 different marker genes in the chicken embryo, we show that the paraxial head mesoderm that normally does not engage in cardiogenesis has the ability to respond to Bmp for a long time. However, Bmp signals are interpreted differently at different time points. Up to early head fold stages, the paraxial head mesoderm is able to read Bmps as signal to engage in the cardiac programme; the ability to upregulate smooth muscle markers is retained slightly longer. Notably, as cardiac competence fades, Bmp promotes the head skeletal muscle programme instead. The switch from cardiac to skeletal muscle competence is Wnt-independent as Wnt caudalises the head mesoderm and also suppresses Msc-inducing Bmp provided by the prechordal plate, thus suppressing both the cardiac and the head skeletal muscle programmes. Our study for the first time suggests a specific transition state in the embryo when cardiac competence is replaced by skeletal muscle competence. It sets the stage to unravel the cardiac-skeletal muscle antagonism that is known to partially collapse in heart failure.


Asunto(s)
Proteínas Morfogenéticas Óseas , Mesodermo , Animales , Embrión de Pollo , Mesodermo/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Cabeza , Cráneo/metabolismo , Músculo Esquelético/metabolismo , Regulación del Desarrollo de la Expresión Génica
17.
J Oral Biosci ; 65(3): 233-242, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37277025

RESUMEN

OBJECTIVES: This study aimed to compare the extracellular matrix of primary cartilage with the secondary cartilage of chicks using immunohistochemical analyses in order to understand the features of chick secondary chondrogenesis. METHODS: Immunohistochemical analysis was performed on the extracellular matrix of quadrate (primary), squamosal, surangular, and anterior pterygoid secondary cartilages using various antibodies targeting the extracellular matrix of cartilage and bone. RESULTS: The localization of collagen types I, II, and X, versican, aggrecan, hyaluronan, link protein, and tenascin-C was identified in the quadrate cartilage, with variations within and between the regions. Newly formed squamosal and surangular secondary cartilages showed simultaneous immunoreactivity for all molecules investigated. However, collagen type X immunoreactivity was not observed, and there was weak immunoreactivity for versican and aggrecan in the anterior pterygoid secondary cartilage. CONCLUSIONS: The immunohistochemical localization of extracellular matrix in the quadrate (primary) cartilage was comparable to that of long bone (primary) cartilage in mammals. The fibrocartilaginous nature and rapid differentiation into hypertrophic chondrocytes, which are known structural features of secondary cartilage, were confirmed in the extracellular matrix of squamosal and surangular secondary cartilages. Furthermore, these tissues appear to undergo developmental processes similar to those in mammals. However, the anterior pterygoid secondary cartilage exhibited unique features that differed from primary and other secondary cartilages, suggesting it is formed through a distinct developmental process.


Asunto(s)
Cartílago , Versicanos , Animales , Agrecanos/análisis , Agrecanos/metabolismo , Versicanos/análisis , Versicanos/metabolismo , Cartílago/química , Cartílago/metabolismo , Cráneo/metabolismo , Mamíferos
18.
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
19.
Front Endocrinol (Lausanne) ; 14: 1127536, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37378024

RESUMEN

Introduction: Both the calvarial and the cortical bones develop through intramembranous ossification, yet they have very different structures and functions. The calvaria enables the rapid while protected growth of the brain, whereas the cortical bone takes part in locomotion. Both types of bones undergo extensive modeling during embryonic and post-natal growth, while bone remodeling is the most dominant process in adults. Their shared formation mechanism and their highly distinct functions raise the fundamental question of how similar or diverse the molecular pathways that act in each bone type are. Methods: To answer this question, we aimed to compare the transcriptomes of calvaria and cortices from 21-day old mice by bulk RNA-Seq analysis. Results: The results revealed clear differences in expression levels of genes related to bone pathologies, craniosynostosis, mechanical loading and bone-relevant signaling pathways like WNT and IHH, emphasizing the functional differences between these bones. We further discussed the less expected candidate genes and gene sets in the context of bone. Finally, we compared differences between juvenile and mature bone, highlighting commonalities and dissimilarities of gene expression between calvaria and cortices during post-natal bone growth and adult bone remodeling. Discussion: Altogether, this study revealed significant differences between the transcriptome of calvaria and cortical bones in juvenile female mice, highlighting the most important pathway mediators for the development and function of two different bone types that originate both through intramembranous ossification.


Asunto(s)
Osteogénesis , Cráneo , Ratones , Femenino , Animales , Cráneo/metabolismo , Osteogénesis/genética , Desarrollo Óseo/genética , Hueso Cortical , Expresión Génica
20.
Dev Biol ; 501: 81-91, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37355029

RESUMEN

The skull is a fundamental bone that protects the development of brain and consists of several bony elements, such as the frontal and parietal bones. Frontal bone exhibited superior in osteogenic potential and regeneration of cranial defects compared to parietal bone. However, how this regional difference is regulated remains largely unknown. In this study, we identified an Ap-2ß transcriptional factor with a higher expression in frontal bone, but its molecular function in osteoblasts needs to be elucidated. We found that Ap-2ß knockdown in preosteoblasts leads to reduced proliferation, increased cell death and impaired differentiation. Through RNA-seq analysis, we found that Ap-2ß influences multiple signaling pathways including the Wnt pathway, and overexpression of Ap-2ß showed increased nuclear ß-catenin and its target genes expressions in osteoblasts. Pharmacological activation of Wnt/ß-catenin signaling using LiCl treatment cannot rescue the reduced luciferase activities of the ß-catenin/TCF/LEF reporter in Ap-2ß knockdown preosteoblasts. Besides, transient expression of Ap-2ß via the lentivirus system could sufficiently rescue the inferior osteogenic potential in parietal osteoblasts, while Ap-2ß knockdown in frontal osteoblasts resulted in reduced osteoblast activity, reduced active ß-catenin and target genes expressions. Taken together, our data demonstrated that Ap-2ß modulates osteoblast proliferation and differentiation through the regulation of Wnt/ß-catenin signaling pathway and plays an important role in regulating regional osteogenic potential in frontal and parietal bone.


Asunto(s)
Vía de Señalización Wnt , beta Catenina , beta Catenina/metabolismo , Factores de Transcripción/metabolismo , Diferenciación Celular , Cráneo/metabolismo , Osteogénesis/fisiología , Osteoblastos , Células Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...