Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros











Intervalo de año de publicación
1.
Dis Model Mech ; 17(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38967226

RESUMEN

Robinow syndrome is a rare disease caused by variants of seven WNT pathway genes. Craniofacial features include widening of the nasal bridge and jaw hypoplasia. We used the chicken embryo to test whether two missense human FZD2 variants (1301G>T, p.Gly434Val; 425C>T, p.Pro142Lys) were sufficient to change frontonasal mass development. In vivo, the overexpression of retroviruses with wild-type or variant human FZD2 inhibited upper beak ossification. In primary cultures, wild-type and variant human FZD2 significantly inhibited chondrogenesis, with the 425C>T variant significantly decreasing activity of a SOX9 luciferase reporter compared to that for the wild type or 1301G>T. Both variants also increased nuclear shuttling of ß-catenin (CTNNB1) and increased the expression of TWIST1, which are inhibitory to chondrogenesis. In canonical WNT luciferase assays using frontonasal mass cells, the variants had dominant-negative effects on wild-type FZD2. In non-canonical assays, the 425C>T variant failed to activate the reporter above control levels and was unresponsive to exogenous WNT5A. This is the first single amino acid change to selectively alter ligand binding in a FZD receptor. Therefore, FZD2 missense variants are pathogenic and could lead to the altered craniofacial morphogenesis seen in Robinow syndrome.


Asunto(s)
Condrogénesis , Anomalías Craneofaciales , Receptores Frizzled , Animales , Embrión de Pollo , Humanos , Pico , beta Catenina/metabolismo , Núcleo Celular/metabolismo , Condrogénesis/genética , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Enanismo , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Deformidades Congénitas de las Extremidades , Cráneo/patología , Cráneo/embriología , Proteína 1 Relacionada con Twist/metabolismo , Proteína 1 Relacionada con Twist/genética , Anomalías Urogenitales , Vía de Señalización Wnt
2.
Bone ; 186: 117169, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38880170

RESUMEN

Intrauterine growth restriction (IUGR) affects a large proportion of infants, particularly in underdeveloped countries. Among the main causes of IUGR, maternal endocrine-metabolic dysfunction is highlighted, either due to its high incidence or due to the severity of the immediate and mediated changes that these dysfunctions cause in the fetus and the mother. Although the effects of endocrine and metabolic disorders have been widely researched, there are still no reviews that bring together and summarize the effects of these conditions on bone development in cases of IUGR. Therefore, the present literature review was conducted with the aim of discussing bone changes observed in fetuses with IUGR caused by maternal endocrine-metabolic dysfunction. The main endocrine dysfunctions that occur with IUGR include maternal hyperthyroidism, hypothyroidism, and hypoparathyroidism. Diabetes mellitus, hypertensive disorders, and obesity are the most important maternal metabolic dysfunctions that compromise fetal growth. The bone changes reported in the fetus are, for the most part, due to damage to cell proliferation and differentiation, as well as failures in the synthesis and mineralization of the extracellular matrix, which results in shortening and fragility of the bones. Some maternal dysfunctions, such as hyperthyroidism, have been widely studied, whereas conditions such as hypoparathyroidism and gestational hypertensive disorders require further study regarding the mechanisms underlying the development of bone changes. Similarly, there is a gap in the literature regarding changes related to intramembranous ossification, as most published articles only describe changes in endochondral bone formation associated with IUGR. Furthermore, there is a need for more research aimed at elucidating the late postnatal changes that occur in the skeletons of individuals affected by IUGR and their possible relationships with adult diseases, such as osteoarthritis and osteoporosis.


Asunto(s)
Desarrollo Óseo , Retardo del Crecimiento Fetal , Humanos , Retardo del Crecimiento Fetal/fisiopatología , Femenino , Embarazo , Feto , Animales , Enfermedades del Sistema Endocrino
3.
Protein Cell ; 15(8): 559-574, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-38442300

RESUMEN

Tissue-resident stem cells are essential for development and repair, and in the skeleton, this function is fulfilled by recently identified skeletal stem cells (SSCs). However, recent work has identified that SSCs are not monolithic, with long bones, craniofacial sites, and the spine being formed by distinct stem cells. Recent studies have utilized techniques such as fluorescence-activated cell sorting, lineage tracing, and single-cell sequencing to investigate the involvement of SSCs in bone development, homeostasis, and disease. These investigations have allowed researchers to map the lineage commitment trajectory of SSCs in different parts of the body and at different time points. Furthermore, recent studies have shed light on the characteristics of SSCs in both physiological and pathological conditions. This review focuses on discussing the spatiotemporal distribution of SSCs and enhancing our understanding of the diversity and plasticity of SSCs by summarizing recent discoveries.


Asunto(s)
Desarrollo Óseo , Homeostasis , Células Madre , Humanos , Animales , Células Madre/citología , Células Madre/metabolismo , Huesos/citología , Huesos/metabolismo
4.
J Craniomaxillofac Surg ; 52(3): 310-315, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38212164

RESUMEN

To retrospectively review the clinical effect of comprehensive treatment of alveolar cleft (CTAC) using the mandible as the bone source. Patients with alveolar clefts who met the inclusion criteria were subjected to a CTAC protocol that included the following: (1) preoperative orthodontic treatment for creating good soft-tissue conditions; (2) 'area-like grafting' with subperiosteal osteogenic chin bone instead of cartilaginous osteogenic iliac bone; (3) simulation of normal bone anatomy via a sandwich-like bone graft consisting of 'cortical bone + cancellous bone + cortical bone'; and (4) strong internal fixation to ensure initial bone block stability. At 6 months postoperatively, the titanium plate was removed and cone-beam computed tomography was performed to evaluate the surgical results. A total of 54 patients underwent treatment with the CTAC protocol. The average age at the initial operation was 10.3 ± 2.1 years, and the average hospital stay was 2.8 ± 0.6 days. At 6 months postoperatively, 49 patients (90.7%) showed good clinical results. The transplanted bone block formed a 'cortical bone + cancellous bone + cortical bone' structure similar to that of the normal jawbone. A mature bone bridge formed, and the impacted permanent teeth continued to erupt and enter the bone graft area. CTAC is a comprehensive restorative solution for alveolar cleft repair that integrates multiple concepts, including orthodontics, embryology, anatomy, and improvements to surgical methods. The method is easy to perform, causes little surgical trauma, and shows a stable success rate, and is thus worth promoting.


Asunto(s)
Injerto de Hueso Alveolar , Labio Leporino , Fisura del Paladar , Humanos , Estudios Retrospectivos , Labio Leporino/cirugía , Hueso Esponjoso , Resultado del Tratamiento , Fisura del Paladar/diagnóstico por imagen , Fisura del Paladar/cirugía , Trasplante Óseo/métodos , Mandíbula , Injerto de Hueso Alveolar/métodos
5.
Semin Cell Dev Biol ; 155(Pt B): 58-65, 2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-37423854

RESUMEN

Thrombospondins (TSPs) belong to a functional class of ECM proteins called matricellular proteins that are not primarily structural, but instead influence cellular interactions within the local extracellular environment. The 3D arrangement of TSPs allow interactions with other ECM proteins, sequestered growth factors, and cell surface receptors. They are expressed in mesenchymal condensations and limb buds during skeletal development, but they are not required for patterning. Instead, when absent, there are alterations in musculoskeletal connective tissue ECM structure, organization, and function, as well as altered skeletal cell phenotypes. Both functional redundancies and unique contributions to musculoskeletal tissue structure and physiology are revealed in mouse models with compound TSP deletions. Crucial roles of individual TSPs are revealed during musculoskeletal injury and regeneration. The interaction of TSPs with mesenchymal stem cells (MSC), and their influence on cell fate, function, and ultimately, musculoskeletal phenotype, suggest that TSPs play integral, but as yet poorly understood roles in musculoskeletal health. Here, unique and overlapping contributions of trimeric TSP1/2 and pentameric TSP3/4/5 to musculoskeletal cell and matrix physiology are reviewed. Opportunities for new research are also noted.


Asunto(s)
Proteínas de la Matriz Extracelular , Trombospondinas , Ratones , Animales , Trombospondinas/genética , Trombospondinas/metabolismo , Esqueleto/metabolismo , Fenómenos Fisiológicos Celulares
6.
Vet Pathol ; 61(1): 74-87, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37431760

RESUMEN

Recently, the central and third tarsal bones of 23 equine fetuses and foals were examined using micro-computed tomography. Radiological changes, including incomplete ossification and focal ossification defects interpreted as osteochondrosis, were detected in 16 of 23 cases. The geometry of the osteochondrosis defects suggested they were the result of vascular failure, but this requires histological confirmation. The study aim was to examine central and third tarsal bones from the 16 cases and to describe the tissues present, cartilage canals, and lesions, including suspected osteochondrosis lesions. Cases included 9 males and 7 females from 0 to 150 days of age, comprising 11 Icelandic horses, 2 standardbred horses, 2 warmblood riding horses, and 1 coldblooded trotting horse. Until 4 days of age, all aspects of the bones were covered by growth cartilage, but from 105 days, the dorsal and plantar aspects were covered by fibrous tissue undergoing intramembranous ossification. Cartilage canal vessels gradually decreased but were present in most cases up to 122 days and were absent in the next available case at 150 days. Radiological osteochondrosis defects were confirmed in histological sections from 3 cases and consisted of necrotic vessels surrounded by ischemic chondronecrosis (articular osteochondrosis) and areas of retained, morphologically viable hypertrophic chondrocytes (physeal osteochondrosis). The central and third tarsal bones formed by both endochondral and intramembranous ossification. The blood supply to the growth cartilage of the central and third tarsal bones regressed between 122 and 150 days of age. Radiological osteochondrosis defects represented vascular failure, with chondrocyte necrosis and retention, or a combination of articular and physeal osteochondrosis.


Asunto(s)
Enfermedades de los Caballos , Osteocondrosis , Huesos Tarsianos , Masculino , Femenino , Animales , Caballos , Microtomografía por Rayos X , Osteocondrosis/diagnóstico por imagen , Osteocondrosis/veterinaria , Osteocondrosis/patología , Cartílago/patología , Necrosis/veterinaria , Huesos Tarsianos/diagnóstico por imagen , Huesos Tarsianos/patología , Enfermedades de los Caballos/diagnóstico por imagen , Enfermedades de los Caballos/patología
7.
Regen Ther ; 24: 536-546, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37860130

RESUMEN

Vertebrates form their skeletal tissues from three distinct origins (the neural crest, paraxial mesoderm, and lateral plate mesoderm) through two distinct modes of ossification (intramembranous and endochondral ossification). Since the paraxial mesoderm generates both intramembranous and endochondral bones, it is thought to give rise to both osteoprogenitors and osteo-chondroprogenitors. However, it remains unclear what directs the paraxial mesoderm-derived cells toward these different fates in distinct skeletal elements during human skeletal development. To answer this question, we need experimental systems that recapitulate paraxial mesoderm-mediated intramembranous and endochondral ossification processes. In this study, we aimed to develop a human pluripotent stem cell (hPSC)-based system that models the human intramembranous ossification process. We found that spheroid culture of the hPSC-derived paraxial mesoderm derivatives generates osteoprogenitors or osteo-chondroprogenitors depending on stimuli. The former induced intramembranous ossification, and the latter endochondral ossification, in mouse renal capsules. Transcriptional profiling supported the notion that bone signatures were enriched in the intramembranous bone-like tissues. Thus, we developed a system that recapitulates intramembranous ossification, and that enables the induction of two distinct modes of ossification by controlling the cell fate of the hPSC-derived paraxial mesoderm derivatives.

8.
J Orthop Res ; 41(9): 1934-1944, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36850029

RESUMEN

This study defined and compared the course of native, impaired and growth factor-stimulated bone regeneration in a rat femoral defect model. A mid-diaphyseal defect with rigid internal fixation was surgically created in the right femur of male Fischer rats and serially analyzed over 36 weeks. Native bone regeneration was modeled using a sub-critical, 1 mm size defect, which healed uneventfully. Critical size defects of 5 mm were used to analyze impaired bone regeneration. In a third group, the 5 mm defects were filled with 11 µg of recombinant human bone morphogenetic protein 2 (rhBMP2) impregnated onto an absorbable collagen sponge, modeling its clinical use. Native bone regeneration was characterized by endochondral ossification with progressive remodeling to ultimately resemble intact femora. An endochondral response was also observed under conditions of impaired bone regeneration, but by week 8 medullary capping occurred with fibrofatty consolidation of the tissue within the defect, resembling an atrophic non-union. rhBMP2 treatment was associated with prolonged inflammatory cytokine expression and rapid intramembranous bone formation occurring with reduced expression of cartilage-associated collagens. Between weeks 4 and 36, rhBMP2-treated bones demonstrated decreased trabecular number and increased trabecular separation, which resulted in inferior mechanical properties compared with bones that healed naturally. Clinical Significance: Recombinant human bone morphogenetic protein 2 (rhBMP2) is used clinically to promote healing of long bones. Our data suggest that it drives intramembraneous ossification producing an inferior regenerate that deteriorates with time. Clinical outcomes would be improved by technologies favoring endochondral regenerative ossification.


Asunto(s)
Proteína Morfogenética Ósea 2 , Regeneración Ósea , Ratas , Humanos , Masculino , Animales , Proteína Morfogenética Ósea 2/farmacología , Proteína Morfogenética Ósea 2/uso terapéutico , Cicatrización de Heridas , Fémur , Osteogénesis , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico
9.
J Dent Res ; 102(1): 13-20, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36303415

RESUMEN

While formation and regeneration of the skeleton have been studied for a long period of time, significant scientific advances in this field continue to emerge based on an unmet clinical need to improve options to promote bone repair. In this review, we discuss the relationship between mechanisms of bone formation and bone regeneration. Data clearly show that regeneration is not simply a reinduction of the molecular and cellular programs that were used for development. Instead, the mechanical environment exerts a strong influence on the mode of repair, while during development, cell-intrinsic processes drive the mode of skeletal formation. A major advance in the field has shown that cell fate is flexible, rather than terminal, and that chondrocytes are able to differentiate into osteoblasts and other cell types during development and regeneration. This is discussed in a larger context of regeneration in vertebrates as well as the clinical implication that this shift in understanding presents.


Asunto(s)
Huesos , Cartílago , Animales , Osteogénesis , Condrocitos/metabolismo , Regeneración Ósea , Osteoblastos
10.
Wound Repair Regen ; 31(1): 17-27, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36177656

RESUMEN

Humans and mice have the ability to regenerate the distal digit tip, the terminal phalanx (P3) in response to amputation. What distinguishes P3 regeneration from regenerative failure is formation of the blastema, a proliferative structure that undergoes morphogenesis to regenerate the amputated tissues. P3 regeneration is characterised by the phases of inflammation, tissue histolysis and expansive bone degradation with simultaneous blastema formation, wound closure and finally blastemal differentiation to restore the amputated structures. While each regenerating digit faithfully progresses through all phases of regeneration, phase progression has traditionally been delineated by time, that is, days postamputation (DPA), yet there is widespread variability in the timing of the individual phases. To diminish variability between digits during tissue histolysis and blastema formation, we have established an in-vivo method using microcomputed tomography (micro CT) scanning to identify five distinct stages of the early regeneration response based on anatomical changes of the digit stump. We report that categorising the initial phases of digit regeneration by stage rather than time greatly diminishes the variability between digits with respect to changes in bone volume and length. Also, stages correlate with the levels of cell proliferation, osteoclast recruitment and osteoprogenitor cell recruitment. Importantly, micro CT staging provides a means to estimate open versus closed digit wounds. We demonstrate two spatially distinct and stage specific bone repair/regeneration responses that occur during P3 regeneration. Collectively, these studies showcase the utility of micro CT imaging to infer the composition of radiolucent soft tissues during P3 blastema formation. Specifically, the staging system identifies the onset of cell proliferation, osteoclastogenesis, osteoprogenitor recruitment, the spatial initiation of de novo bone formation and epidermal closure.


Asunto(s)
Osteogénesis , Cicatrización de Heridas , Ratones , Animales , Humanos , Microtomografía por Rayos X , Cicatrización de Heridas/fisiología , Osteogénesis/fisiología , Osteoclastos/fisiología , Regeneración Ósea/fisiología
11.
Matrix Biol ; 113: 100-121, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36261075

RESUMEN

The gelatinases, a subgroup of the matrix metalloproteinases (MMPs) superfamily are composed of two members; MMP2 and MMP9. They are known to degrade gelatin among other components of the extracellular matrix. Recently, the two gelatinases were found to be necessary for neural crest cell migration and to compensate for each other loss in these cells. To characterize their involvement in the skeletal system, and to better reveal their individual or common roles, we have generated double knockout (dKO) mice, lacking both MMP2 and MMP9. Comprehensive analysis of the skeleton morphological and mechanical parameters at postnatal day (P) 0, P21, 3 months (M) and 8M of age, revealed an unexpected distinct role for each gelatinase; MMP2 was found to be involved merely in intramembranous ossification which led to a smaller skull and inferior cortical parameters upon its loss, while MMP9 was found to affect only the endochondral ossification process, which led to shorter long-bones in its absence. Importantly, the dKO mice demonstrated a combination of both the skull and long bone phenotypes as found in the single-KOs, and not a severer additive phenotype. Transcriptome analysis on the cortical bone, the growth plate and the skull frontal bone, found many genes that were differentially expressed as a direct or indirect result of MMP-loss, and reinforced the specific and distinct role of each gelatinase in each bone type. Altogether, these results suggest that although both gelatinases share the same substrates and are highly expressed in flat and long bones, they are indispensable and control separately the development of different bones.


Asunto(s)
Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Cráneo , Animales , Ratones , Placa de Crecimiento/crecimiento & desarrollo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Cráneo/crecimiento & desarrollo
12.
Adv Biol (Weinh) ; 6(11): e2200076, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35859256

RESUMEN

Bone apatite crystals grow in clusters, but the microstructure of these clusters is unknown. This study compares the structural and compositional differences between bone apatite clusters formed in intramembranous (IO) and endochondral ossification (EO). Calvaria (IO) and femurs (EO) are isolated from mice at embryonic days (E) 14.5 to 15.5 and post-natal days (P) 6 to 7, respectively. Results show that the initially formed bone apatite clusters in EO (≅1.2 µm2 ) are >10 times larger than those in IO (≅0.1 µm2 ), without significant changes in ion composition. In IO (E14.5 calvarium), early minerals are formed inside matrix vesicles (MVs). In contrast, in EO (P6 femur epiphysis), no MVs are observed, and chondrocyte-derived plasma membrane nanofragments (PMNFs) are the nucleation site for mineralization. Apatite cluster size difference is linked with the different nucleation sites. Moreover, an alkaline pH and slow P supply into a Ca-rich microenvironment are suggested to facilitate apatite cluster growth, as demonstrated in a biomimetic mineralization system. Together, the results reveal for the first time the distinct and exquisite microstructures of bone apatite clusters in IO and EO, and provide insightful inspirations for the design of more efficient materials for bone tissue engineering and repair.


Asunto(s)
Apatitas , Osteogénesis , Ratones , Animales , Apatitas/química , Cráneo , Condrocitos , Ingeniería de Tejidos
13.
Cell Rep ; 40(2): 111045, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35830813

RESUMEN

The mammalian skull vault is essential to shape the head and protect the brain, but the cellular and molecular events underlying its development remain incompletely understood. Single-cell transcriptomic profiling from early to late mouse embryonic stages provides a detailed atlas of cranial lineages. It distinguishes various populations of progenitors and reveals a high expression of SOXC genes (encoding the SOX4, SOX11, and SOX12 transcription factors) early in development in actively proliferating and myofibroblast-like osteodermal progenitors. SOXC inactivation in these cells causes severe skull and skin underdevelopment due to the limited expansion of cell populations before and upon lineage commitment. SOXC genes enhance the expression of gene signatures conferring dynamic cellular and molecular properties, including actin cytoskeleton assembly, chromatin remodeling, and signaling pathway induction and responsiveness. These findings shed light onto craniogenic mechanisms and SOXC functions and suggest that similar mechanisms could decisively control many developmental, adult, pathological, and regenerative processes.


Asunto(s)
Miofibroblastos , Factores de Transcripción SOXC , Animales , Regulación del Desarrollo de la Expresión Génica , Mamíferos/metabolismo , Ratones , Miofibroblastos/metabolismo , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo
14.
Elife ; 112022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35704354

RESUMEN

The cranial endo and dermal skeletons, which comprise the vertebrate skull, evolved independently over 470 million years ago and form separately during embryogenesis. In mammals, much of the cartilaginous chondrocranium is transient, undergoing endochondral ossification or disappearing, so its role in skull morphogenesis is not well studied and it remains an enigmatic structure. We provide complete 3D reconstructions of the laboratory mouse chondrocranium from embryonic day (E) 13.5 through E17.5 using a novel methodology of uncertainty-guided segmentation of phosphotungstic enhanced 3D micro-computed tomography images with sparse annotation. We evaluate the embryonic mouse chondrocranium and dermatocranium in 3D, and delineate the effects of a Fgfr2 variant on embryonic chondrocranial cartilages and on their association with forming dermal bones using the Fgfr2cC342Y/+ Crouzon syndrome mouse. We show that the dermatocranium develops outside of and in shapes that conform to the chondrocranium. Results reveal direct effects of the Fgfr2 variant on embryonic cartilage, on chondrocranium morphology, and on the association between chondrocranium and dermatocranium development. Histologically, we observe a trend of relatively more chondrocytes, larger chondrocytes, and/or more matrix in the Fgfr2cC342Y/+ embryos at all timepoints before the chondrocranium begins to disintegrate at E16.5. The chondrocrania and forming dermatocrania of Fgfr2cC342Y/+ embryos are relatively large, but a contrasting trend begins at E16.5 and continues into early postnatal (P0 and P2) timepoints, with the skulls of older Fgfr2cC342Y/+ mice reduced in most dimensions compared to Fgfr2c+/+ littermates. Our findings have implications for the study and treatment of human craniofacial disease, for understanding the impact of chondrocranial morphology on skull growth, and potentially on the evolution of skull morphology.


Asunto(s)
Disostosis Craneofacial , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Animales , Cartílago , Disostosis Craneofacial/patología , Modelos Animales de Enfermedad , Mamíferos , Ratones , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Cráneo/anatomía & histología , Microtomografía por Rayos X
15.
Anat Rec (Hoboken) ; 305(9): 2166-2174, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35122476

RESUMEN

Identification of skeletal maturity is of interest as a measure of species longevity and for identifying its maximal achievable size/mass. Measurement of age on the basis of growth arrest/accentuation lines and external fundamental system evidences cessation or at least extreme slowing of circumferential bone growth. Such intramembranous (periosteal)-derived growth is distinct from the endochondral ossification responsible for longitudinal growth and therefore achievable organismal size/mass. As subchondral transcortical channels are required for nourishment, their loss should identify cessation of longitudinal growth. Predicated on phylogenetic bracketing/relationship and shared anatomical structures with and without growth plates, birds represent an appropriate model for the study of dinosaur ontogeny. Persistence of transcortical subchondral channels in the long bones of birds are examined at ×100-200 magnification and correlated with bone length. Transcortical channels are present in subchondral articular surfaces, but disappear when terminal longitudinal growth is achieved. Articular vascular channels perforating articular surfaces from within the bone are detected. Loss of penetrating channels is interpreted as evidence of skeletal growth cessation, identifying the longitudinal bone length at which skeletal growth cessation has been achieved. The current study provides evidence that maximal bone length does correlate with endochondral cessation growth. Failure of circumferential growth reduction/cessation to correlate with bone length may be related to lack of synchronicity of periosteal-based circumferential growth with the endochondral process responsible for bone lengthening. Loss/closure of articular vascular channels may be the most reliable measure of a bird's achievement of maximal growth (indicating cessation of appendicular element lengthening).


Asunto(s)
Desarrollo Óseo , Huesos , Animales , Aves , Huesos/diagnóstico por imagen , Osteogénesis , Filogenia
16.
Cureus ; 13(8): e17051, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34522529

RESUMEN

Osteochondroma, often referred to as exostosis, is the most common benign bone tumor characterized by a bony protuberance surrounded by a cartilaginous surface. Most osteochondromas are found on the metaphysis of long bones, with the dorsal aspect of the scapula being a rare site of occurrence for an exostosis. Radiographic imaging, preferably through MRI or CT, assists in the identification of benign growth; however, a definitive diagnosis requires a biopsy. Open surgical resection and arthroscopic excision are the definitive treatment modalities of the nidus. Postoperative care requires immobilization of the limb for two months, with at least four months being the appropriate timeline for complete recovery.

17.
Dev Biol ; 478: 133-143, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34245724

RESUMEN

Vascular endothelial growth factor A (VEGF-A) is expressed by several cell types and is a crucial factor for angiogenic-osteogenic coupling. However, the immunolocalization of VEGF-A during the early stages of the alveolar process formation remains underexplored. Thus, we analyzed the spatio-temporal immunolocalization of VEGF-A and its relationship with Runt-related transcription factor 2 (Runx2) and osterix (Osx) during the early steps of intramembranous ossification of the alveolar process in rat embryos. Embryo heads (E) of 16, 18 and 20-day-old rats were processed for paraffin embedding. Histomorphometry and immunohistochemistry to detect VEGF-A, Runx2, and Osx (osteoblast differentiation markers) were performed. The volume density of bone tissue including bone cells and blood vessels increased significantly in E18 and E20. Cells showing high VEGF-A immunoreactivity were initially observed within a perivascular niche in the ectomesenchyme; afterwards, these cells were diffusely located near bone formation sites. Runx2-and Osx-immunopositive cells were observed in corresponded regions of cells showing strong VEGF-A immunoreactivity. Although these immunostained cells were observed in all specimens, this immunolocalization pattern was more evident in E16 specimens and gradually decreased in E18 and E20 specimens. Double immunofluorescence labelling showed intracellular co-localization of Osx and VEGF-A in cells surrounding the developing alveolar process, indicating a crucial role of VEGF-A in osteoblast differentiation. Our results showed VEGF-A immunoexpression in osteoblasts and its precursors during the maxillary alveolar process formation of rat embryos. Moreover, the VEGF-A-positive cells located within a perivascular niche at the early stages of the alveolar process development suggest a crosstalk between endothelium and ectomesenchymal cells, reinforcing the angiogenic-osteogenic coupling in this process.


Asunto(s)
Proceso Alveolar/embriología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proceso Alveolar/citología , Proceso Alveolar/metabolismo , Animales , Células Endoteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Técnicas para Inmunoenzimas , Mesodermo/citología , Mesodermo/metabolismo , Osteoblastos/citología , Osteoclastos/metabolismo , Ratas , Ratas Wistar
18.
Acta Biomater ; 130: 485-496, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34129957

RESUMEN

Most fractures heal by a combination of endochondral and intramembranous ossification dependent upon strain and vascularity at the fracture site. Many biomaterials-based bone regeneration strategies rely on the use of calcium phosphates such as nano-crystalline hydroxyapatite (nHA) to create bone-like scaffolds. In this study, nHA was dispersed in reactive polymers to form composite scaffolds that were evaluated both in vitro and in vivo. Matrix assays, immunofluorescent staining, and Western blots demonstrated that nHA influenced mineralization and subsequent osteogenesis in a dose-dependent manner in vitro. Furthermore, nHA dispersed in polymeric composites promoted osteogenesis by a similar mechanism as particulated nHA. Scaffolds were implanted into a 2-mm defect in the femoral diaphysis or metaphysis of Sprague-Dawley rats to evaluate new bone formation at 4 and 8 weeks. Two formulations were tested: a poly(thioketal urethane) scaffold without nHA (PTKUR) and a PTKUR scaffold augmented with 22 wt% nHA (22nHA). The scaffolds supported new bone formation in both anatomic sites. In the metaphysis, augmentation of scaffolds with nHA promoted an intramembranous healing response. Within the diaphysis, nHA inhibited endochondral ossification. Immunohistochemistry was performed on cryo-sections of the bone/scaffold interface in which CD146, CD31, Endomucin, CD68, and Myeloperoxidase were evaluated. No significant differences in the infiltrating cell populations were observed. These findings suggest that nHA dispersed in polymeric composites induces osteogenic differentiation of adherent endogenous cells, which has skeletal site-specific effects on fracture healing. STATEMENT OF SIGNIFICANCE: Understanding the mechanism by which synthetic scaffolds promote new bone formation in preclinical models is crucial for bone regeneration applications in the clinic where complex fracture cases are seen. In this study, we found that dispersion of nHA in polymeric scaffolds promoted in vitro osteogenesis in a dose-dependent manner through activation of the PiT1 receptor and subsequent downstream Erk1/2 signaling. While augmentation of polymeric scaffolds with nHA enhanced intramembranous ossification in metaphyseal defects, it inhibited endochondral ossification in diaphyseal defects. Thus, our findings provide new insights into designing synthetic bone grafts that complement the skeletal site-specific fracture healing response.


Asunto(s)
Durapatita , Osteogénesis , Animales , Regeneración Ósea , Cartílago , Poliuretanos , Ratas , Ratas Sprague-Dawley , Ingeniería de Tejidos , Andamios del Tejido
19.
Front Cell Dev Biol ; 9: 645386, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33996804

RESUMEN

Epigenetic and chromatin regulation of craniofacial development remains poorly understood. Ankyrin Repeat Domain 11 (ANKRD11) is a chromatin regulator that has previously been shown to control neural stem cell fates via modulation of histone acetylation. ANKRD11 gene variants, or microdeletions of the 16q24.3 chromosomal region encompassing the ANKRD11 gene, cause KBG syndrome, a rare autosomal dominant congenital disorder with variable neurodevelopmental and craniofacial involvement. Craniofacial abnormalities include a distinct facial gestalt, delayed bone age, tooth abnormalities, delayed fontanelle closure, and frequently cleft or submucosal palate. Despite this, the dramatic phenotype and precise role of ANKRD11 in embryonic craniofacial development remain unexplored. Quantitative analysis of 3D images of KBG syndromic subjects shows an overall reduction in the size of the middle and lower face. Here, we report that mice with heterozygous deletion of Ankrd11 in neural crest cells (Ankrd11nchet) display a mild midfacial hypoplasia including reduced midfacial width and a persistent open fontanelle, both of which mirror KBG syndrome patient facial phenotypes. Mice with a homozygous Ankrd11 deletion in neural crest cells (Ankrd11ncko) die at birth. They show increased severity of several clinical manifestations described for KBG syndrome, such as cleft palate, retrognathia, midfacial hypoplasia, and reduced calvarial growth. At E14.5, Ankrd11 expression in the craniofacial complex is closely associated with developing bony structures, while expression at birth is markedly decreased. Conditional deletion of Ankrd11 leads to a reduction in ossification of midfacial bones, with several ossification centers failing to expand and/or fuse. Intramembranous bones show features of delayed maturation, with bone remodeling severely curtailed at birth. Palatal shelves remain hypoplastic at all developmental stages, with a local reduction in proliferation at E13.5. Our study identifies Ankrd11 as a critical regulator of intramembranous ossification and palate development and suggests that Ankrd11nchet and Ankrd11ncko mice may serve as pre-clinical models for KBG syndrome in humans.

20.
Zool Res ; 42(3): 362-376, 2021 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-33998184

RESUMEN

Intermuscular bones (IBs) are slender linear bones embedded in muscle, which ossify from tendons through a process of intramembranous ossification, and only exist in basal teleosts. IBs are essential for fish swimming, but they present a choking risk during human consumption, especially in children, which can lead to commercial risks that have a negative impact on the aquaculture of these fish. In this review, we discuss the morphogenesis and functions of IBs, including their underlying molecular mechanisms, as well as the advantages and disadvantages of different methods for IB studies and techniques for breeding and generating IB-free fish lines. This review reveals that the many key genes involved in tendon development, osteoblast differentiation, and bone formation, e.g., scxa, msxC, sost, twist, bmps, and osterix, also play roles in IB development. Thus, this paper provides useful information for the breeding of new fish strains without IBs via genome editing and artificial selection.


Asunto(s)
Desarrollo Óseo/fisiología , Peces/crecimiento & desarrollo , Animales , Acuicultura , Dieta , Peces/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA