Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Osteoarthritis Cartilage ; 32(5): 548-560, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38160742

RESUMEN

OBJECTIVE: Cartilage tissue engineering strategies that use autologous chondrocytes require in vitro expansion of cells to obtain enough cells to produce functional engineered tissue. However, chondrocytes dedifferentiate during expansion culture, limiting their ability to produce chondrogenic tissue and their utility for cell-based cartilage repair strategies. The current study identified conditions that favor cartilage production and the mechanobiological mechanisms responsible for these benefits. DESIGN: Chondrocytes were isolated from juvenile bovine knee joints and cultured with (primed) or without (unprimed) a growth factor cocktail. Gene expression, cell morphology, cell adhesion, cytoskeletal protein distribution, and cell mechanics were assessed. Following passage 5, cells were embedded into agarose hydrogels to evaluate functional properties of engineered cartilage. RESULTS: Priming cells during expansion culture altered cell phenotype and chondrogenic tissue production. Unbiased ribonucleic acid-sequencing analysis suggested, and experimental studies confirmed, that growth factor priming delays dedifferentiation associated changes in cell adhesion and cytoskeletal organization. Priming also overrode mechanobiological pathways to prevent chondrocytes from remodeling their cytoskeleton to accommodate the stiff, monolayer microenvironment. Passage 1 primed cells deformed less and had lower yes associated protein 1 activity than unprimed cells. Differences in cell adhesion, morphology, and cell mechanics between primed and unprimed cells were mitigated by passage 5. CONCLUSIONS: Priming suppresses mechanobiologic cytoskeletal remodeling to prevent chondrocyte dedifferentiation, resulting in more cartilage-like tissue-engineered constructs.


Asunto(s)
Cartílago Articular , Condrocitos , Animales , Bovinos , Condrocitos/metabolismo , Células Cultivadas , Cartílago , Ingeniería de Tejidos/métodos , Condrogénesis , Péptidos y Proteínas de Señalización Intercelular/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34161267

RESUMEN

Skeletal fragility in the elderly does not simply result from a loss of bone mass. However, the mechanisms underlying the concurrent decline in bone mass, quality, and mechanosensitivity with age remain unclear. The important role of osteocytes in these processes and the age-related degeneration of the intricate lacunocanalicular network (LCN) in which osteocytes reside point to a primary role for osteocytes in bone aging. Since LCN complexity severely limits experimental dissection of these mechanisms in vivo, we used two in silico approaches to test the hypothesis that LCN degeneration, due to aging or an osteocyte-intrinsic defect in transforming growth factor beta (TGF-ß) signaling (TßRIIocy-/-), is sufficient to compromise essential osteocyte responsibilities of mass transport and exposure to mechanical stimuli. Using reconstructed confocal images of bone with fluorescently labeled osteocytes, we found that osteocytes from aged and TßRIIocy-/- mice had 33 to 45% fewer, and more tortuous, canaliculi. Connectomic network analysis revealed that diminished canalicular density is sufficient to impair diffusion even with intact osteocyte numbers and overall LCN architecture. Computational fluid dynamics predicts that the corresponding drop in shear stress experienced by aged or TßRIIocy-/- osteocytes is highly sensitive to canalicular surface area but not tortuosity. Simulated expansion of the osteocyte pericellular space to mimic osteocyte perilacunar/canalicular remodeling restored predicted shear stress for aged osteocytes to young levels. Overall, these models show how loss of LCN volume through LCN pruning may lead to impaired fluid dynamics and osteocyte exposure to mechanostimulation. Furthermore, osteocytes emerge as targets of age-related therapeutic efforts to restore bone health and function.


Asunto(s)
Envejecimiento/fisiología , Huesos/fisiología , Hidrodinámica , Osteocitos/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Análisis de Elementos Finitos , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Transporte de Proteínas , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Estrés Mecánico
3.
Curr Osteoporos Rep ; 21(6): 637-649, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37831357

RESUMEN

PURPOSE OF REVIEW: The integration of data from multiple genomic assays from humans and non-human model organisms is an effective approach to identify genes involved in skeletal fragility and fracture risk due to osteoporosis and other conditions. This review summarizes genome-wide genetic variation and gene expression data resources relevant to the discovery of genes contributing to skeletal fragility and fracture risk. RECENT FINDINGS: Genome-wide association studies (GWAS) of osteoporosis-related traits are summarized, in addition to gene expression in bone tissues in humans and non-human organisms, with a focus on rodent models related to skeletal fragility and fracture risk. Gene discovery approaches using these genomic data resources are described. We also describe the Musculoskeletal Knowledge Portal (MSKKP) that integrates much of the available genomic data relevant to fracture risk. The available genomic resources provide a wealth of knowledge and can be analyzed to identify genes related to fracture risk. Genomic resources that would fill particular scientific gaps are discussed.


Asunto(s)
Fracturas Óseas , Osteoporosis , Humanos , Densidad Ósea/genética , Estudio de Asociación del Genoma Completo , Predisposición Genética a la Enfermedad , Osteoporosis/genética , Fracturas Óseas/genética , Huesos , Expresión Génica , Biología
4.
FASEB J ; 35(10): e21883, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34569659

RESUMEN

Organism scale mechanical forces elicit cellular scale changes through coordinated regulation of multiple signaling pathways. The mechanisms by which cells integrate signaling to generate a unified biological response remains a major question in mechanobiology. For example, the mechanosensitive response of bone and other tissues requires coordinated signaling by the transforming growth factor beta (TGFß) and Wnt pathways through mechanisms that are not well-defined. Here we report a new microRNA-dependent mechanism that mediates mechanosensitive crosstalk between TGFß and Wnt signaling in osteocytes exposed to fluid shear stress (FSS). From 60 mechanosensitive microRNA (miRs) identified by small-RNAseq, miR100 expression is suppressed by in vivo hindlimb loading in the murine tibia and by cellular scale FSS in OCY454 cells. Though FSS activates both TGFß and Wnt signaling in osteocytes, only TGFß represses miR-100 expression. miR-100, in turn, antagonizes Wnt signaling by targeting and inhibiting expression of Frizzled receptors (FZD5/FZD8). Accordingly, miR-100 inhibition blunts FSS- and TGFß-inducible Wnt signaling. Therefore, our results identify FSS-responsive miRNAs in osteocytes, including one that integrates the mechanosensitive function of two essential signaling pathways in the osteoanabolic response of bone to mechanical load.


Asunto(s)
Mecanotransducción Celular , MicroARNs/metabolismo , Osteocitos/metabolismo , Resistencia al Corte , Factor de Crecimiento Transformador beta/metabolismo , Vía de Señalización Wnt , Animales , Línea Celular , Masculino , Ratones , MicroARNs/genética , Factor de Crecimiento Transformador beta/genética
5.
FASEB J ; 35(3): e21263, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33570811

RESUMEN

Bone is a dynamic tissue that constantly adapts to changing mechanical demands. The transforming growth factor beta (TGFß) signaling pathway plays several important roles in maintaining skeletal homeostasis by both coupling the bone-forming and bone-resorbing activities of osteoblasts and osteoclasts and by playing a causal role in the anabolic response of bone to applied loads. However, the extent to which the TGFß signaling pathway in osteocytes is directly regulated by fluid shear stress (FSS) is unknown, despite work suggesting that fluid flow along canaliculi is a dominant physical cue sensed by osteocytes following bone compression. To investigate the effects of FSS on TGFß signaling in osteocytes, we stimulated osteocytic OCY454 cells cultured within a microfluidic platform with FSS. We find that FSS rapidly upregulates Smad2/3 phosphorylation and TGFß target gene expression, even in the absence of added TGFß. Indeed, relative to treatment with TGFß, FSS induced a larger increase in levels of pSmad2/3 and Serpine1 that persisted even in the presence of a TGFß receptor type I inhibitor. Our results show that FSS stimulation rapidly induces phosphorylation of multiple TGFß family R-Smads by stimulating multimerization and concurrently activating several TGFß and BMP type I receptors, in a manner that requires the activity of the corresponding ligand. While the individual roles of the TGFß and BMP signaling pathways in bone mechanotransduction remain unclear, these results implicate that FSS activates both pathways to generate a downstream response that differs from that achieved by either ligand alone.


Asunto(s)
Osteocitos/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta/fisiología , Receptores de Activinas Tipo II/fisiología , Animales , Células Cultivadas , Dispositivos Laboratorio en un Chip , Ratones , Multimerización de Proteína , Receptor Tipo I de Factor de Crecimiento Transformador beta/química , Análisis de Secuencia de ARN , Transducción de Señal/fisiología , Proteína Smad2/fisiología , Proteína smad3/fisiología , Estrés Mecánico
6.
Curr Rheumatol Rep ; 24(6): 184-197, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35499698

RESUMEN

PURPOSE OF REVIEW: The effect of the transforming growth factor beta (TGFß) signaling pathway on joint homeostasis is tissue-specific, non-linear, and context-dependent, representing a unique complexity in targeting TGFß signaling in joint disease. Here we discuss the variety of mechanisms that TGFß signaling employs in the synovial joint to maintain healthy joint crosstalk and the ways in which aberrant TGFß signaling can result in joint degeneration. RECENT FINDINGS: Osteoarthritis (OA) epitomizes a condition of disordered joint crosstalk in which multiple joint tissues degenerate leading to overall joint deterioration. Synovial joint tissues, such as subchondral bone, articular cartilage, and synovium, as well as mesenchymal stem cells, each demonstrate aberrant TGFß signaling during joint disease, whether by excessive or suppressed signaling, imbalance of canonical and non-canonical signaling, a perturbed mechanical microenvironment, or a distorted response to TGFß signaling during aging. The synovial joint relies upon a sophisticated alliance among each joint tissue to maintain joint homeostasis. The TGFß signaling pathway is a key regulator of the health of individual joint tissues, and the subsequent interaction among these different joint tissues, also known as joint crosstalk. Dissecting the sophisticated function of TGFß signaling in the synovial joint is key to therapeutically interrogating the pathway to optimize overall joint health.


Asunto(s)
Cartílago Articular , Osteoartritis , Cartílago Articular/metabolismo , Humanos , Osteoartritis/metabolismo , Transducción de Señal , Membrana Sinovial/metabolismo , Factor de Crecimiento Transformador beta
7.
Curr Osteoporos Rep ; 17(4): 157-168, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31227998

RESUMEN

PURPOSE OF REVIEW: In perilacunar/canalicular remodeling (PLR), osteocytes dynamically resorb, and then replace, the organic and mineral components of the pericellular extracellular matrix. Given the enormous surface area of the osteocyte lacuna-canalicular network (LCN), PLR is important for maintaining homeostasis of the skeleton. The goal of this review is to examine the motivations and critical considerations for the analysis of PLR, in both in vitro and in vivo systems. RECENT FINDINGS: Morphological approaches alone are insufficient to elucidate the complex mechanisms regulating PLR in the healthy skeleton and in disease. Understanding the role and regulation of PLR will require the incorporation of standardized PLR outcomes as a routine part of skeletal phenotyping, as well as the development of improved molecular and cellular outcomes. Current PLR outcomes assess PLR enzyme expression, the LCN, and bone matrix composition and organization, among others. Here, we discuss current PLR outcomes and how they have been applied to study PLR induction and suppression in vitro and in vivo. Given the role of PLR in skeletal health and disease, integrated analysis of PLR has potential to elucidate new mechanisms by which osteocytes participate in skeletal health and disease.


Asunto(s)
Matriz Ósea/metabolismo , Remodelación Ósea/fisiología , Osteocitos/metabolismo , Matriz Ósea/ultraestructura , Anhidrasas Carbónicas/metabolismo , Catepsina K/metabolismo , Línea Celular , Homeostasis , Humanos , Concentración de Iones de Hidrógeno , Imagenología Tridimensional , Metaloproteinasas de la Matriz/metabolismo , Microscopía Confocal , Microscopía Electrónica de Rastreo , Osteocitos/enzimología , Osteocitos/ultraestructura , ATPasas de Translocación de Protón/metabolismo , Microtomografía por Rayos X
8.
Dev Biol ; 385(2): 380-95, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24262986

RESUMEN

Neural crest mesenchyme (NCM) controls species-specific pattern in the craniofacial skeleton but how this cell population accomplishes such a complex task remains unclear. To elucidate mechanisms through which NCM directs skeletal development and evolution, we made chimeras from quail and duck embryos, which differ markedly in their craniofacial morphology and maturation rates. We show that quail NCM, when transplanted into duck, maintains its faster timetable for development and autonomously executes molecular and cellular programs for the induction, differentiation, and mineralization of bone, including premature expression of osteogenic genes such as Runx2 and Col1a1. In contrast, the duck host systemic environment appears to be relatively permissive and supports osteogenesis independently by providing circulating minerals and a vascular network. Further experiments reveal that NCM establishes the timing of osteogenesis by regulating cell cycle progression in a stage- and species-specific manner. Altering the time-course of D-type cyclin expression mimics chimeras by accelerating expression of Runx2 and Col1a1. We also discover higher endogenous expression of Runx2 in quail coincident with their smaller craniofacial skeletons, and by prematurely over-expressing Runx2 in chick embryos we reduce the overall size of the craniofacial skeleton. Thus, our work indicates that NCM establishes species-specific size in the craniofacial skeleton by controlling cell cycle, Runx2 expression, and the timing of key events during osteogenesis.


Asunto(s)
Ciclo Celular/genética , Evolución Molecular , Cara , Osteogénesis/genética , Cráneo/crecimiento & desarrollo , Animales , Secuencia de Bases , Vasos Sanguíneos/crecimiento & desarrollo , Western Blotting , Coturnix , Cartilla de ADN , Patos , Especificidad de la Especie
9.
NMR Biomed ; 28(5): 523-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25761416

RESUMEN

Osteoarthritis (OA) is a common multifactorial and heterogeneous degenerative joint disease, and biochemical changes in cartilage matrix occur during the early stages of OA before morphological changes occur. Thus, it is desired to measure regional biochemical changes in the joint. High-resolution magic angle spinning (HRMAS) NMR spectroscopy is a powerful method of observing cartilaginous biochemical changes ex vivo, including the concentrations of alanine and N-acetyl, which are markers of collagen and total proteoglycan content, respectively. Previous studies have observed significant changes in chondrocyte metabolism of OA cartilage via the altered gene expression profiles of ACAN, COL2A1 and MMP13, which encode aggrecan, type II collagen and matrix metalloproteinase 13 (a protein crucial in the degradation of type II collagen), respectively. Employing HRMAS, this study aimed to elucidate potential relationships between N-acetyl and/or alanine and ACAN, COL2A1 and/or MMP13 expression profiles in OA cartilage. Thirty samples from the condyles of five subjects undergoing total knee arthroplasty to treat OA were collected. HRMAS spectra were obtained at 11.7 T for each sample. RNA was subsequently extracted to determine gene expression profiles. A significant negative correlation between N-acetyl metabolite and ACAN gene expression levels was observed; this provides further evidence of N-acetyl as a biomarker of cartilage degeneration. The alanine doublet was distinguished in the spectra of 15 of the 30 specimens of this study. Alanine can only be detected with HRMAS NMR spectroscopy when the collagen framework has been degraded such that alanine is sufficiently mobile to form a distinguished peak in the spectrum. Thus, HRMAS NMR spectroscopy may provide unique localized measurements of collagenous degeneration in OA cartilage. The identification of imaging markers that could provide a link between OA pathology and chondrocyte metabolism will facilitate the development of more sensitive diagnostic techniques and will improve methods of monitoring treatment for patients suffering from OA.


Asunto(s)
Cartílago Articular/metabolismo , Colágeno/metabolismo , Perfilación de la Expresión Génica/métodos , Espectroscopía de Resonancia Magnética/métodos , Osteoartritis de la Rodilla/metabolismo , Proteoglicanos/metabolismo , Anciano , Alanina/metabolismo , Biomarcadores/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Osteoartritis de la Rodilla/diagnóstico , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Marcadores de Spin , Estadística como Asunto
10.
Curr Osteoporos Rep ; 12(3): 366-75, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24894149

RESUMEN

The ability of bone to resist fracture is determined by the combination of bone mass and bone quality. Like bone mass, bone quality is carefully regulated. Of the many aspects of bone quality, this review focuses on biological mechanisms that control the material quality of the bone extracellular matrix (ECM). Bone ECM quality depends upon ECM composition and organization. Proteins and signaling pathways that affect the mineral or organic constituents of bone ECM impact bone ECM material properties, such as elastic modulus and hardness. These properties are also sensitive to pathways that regulate bone remodeling by osteoblasts, osteoclasts, and osteocytes. Several extracellular proteins, signaling pathways, intracellular effectors, and transcription regulatory networks have been implicated in the control of bone ECM quality. A molecular understanding of these mechanisms will elucidate the biological control of bone quality and suggest new targets for the development of therapies to prevent bone fragility.


Asunto(s)
Densidad Ósea , Remodelación Ósea , Huesos/metabolismo , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Matriz Ósea/metabolismo , Calcificación Fisiológica , Homeostasis , Humanos , Osteoblastos , Osteoclastos , Osteocitos , Transducción de Señal
11.
Proc Natl Acad Sci U S A ; 108(35): 14416-21, 2011 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-21873221

RESUMEN

The structure of human cortical bone evolves over multiple length scales from its basic constituents of collagen and hydroxyapatite at the nanoscale to osteonal structures at near-millimeter dimensions, which all provide the basis for its mechanical properties. To resist fracture, bone's toughness is derived intrinsically through plasticity (e.g., fibrillar sliding) at structural scales typically below a micrometer and extrinsically (i.e., during crack growth) through mechanisms (e.g., crack deflection/bridging) generated at larger structural scales. Biological factors such as aging lead to a markedly increased fracture risk, which is often associated with an age-related loss in bone mass (bone quantity). However, we find that age-related structural changes can significantly degrade the fracture resistance (bone quality) over multiple length scales. Using in situ small-angle X-ray scattering and wide-angle X-ray diffraction to characterize submicrometer structural changes and synchrotron X-ray computed tomography and in situ fracture-toughness measurements in the scanning electron microscope to characterize effects at micrometer scales, we show how these age-related structural changes at differing size scales degrade both the intrinsic and extrinsic toughness of bone. Specifically, we attribute the loss in toughness to increased nonenzymatic collagen cross-linking, which suppresses plasticity at nanoscale dimensions, and to an increased osteonal density, which limits the potency of crack-bridging mechanisms at micrometer scales. The link between these processes is that the increased stiffness of the cross-linked collagen requires energy to be absorbed by "plastic" deformation at higher structural levels, which occurs by the process of microcracking.


Asunto(s)
Envejecimiento/fisiología , Huesos/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Fenómenos Biomecánicos , Productos Finales de Glicación Avanzada/análisis , Humanos , Persona de Mediana Edad , Tomografía Computarizada por Rayos X
12.
Tissue Eng Part A ; 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38323585

RESUMEN

Tissue engineering strategies show great potential for repairing osteochondral defects in osteoarthritic joints; however, these approaches often rely on passaging cells multiple times to obtain enough cells to produce functional tissue. Unfortunately, monolayer expansion culture causes chondrocyte dedifferentiation, which is accompanied by a phenotypical and morphological shift in chondrocyte properties that leads to a reduction in the quality of de novo cartilage produced. Thus, the objective of this study was to evaluate transcriptional variations during in vitro expansion culture and determine how differences in cell phenotype from monolayer expansion alter development of functional engineered cartilage. We used an unbiased approach to explore genome-wide transcriptional differences in chondrocyte phenotype at passage 1 (P1), P3, and P5, and then seeded cells into hydrogel scaffolds at P3 and P5 to assess cells' abilities to produce cartilaginous extracellular matrix in three dimensional (3D). We identified distinct phenotypic differences, specifically for genes related to extracellular organization and cartilage development. Both P3 and P5 chondrocytes were able to produce chondrogenic tissue in 3D, with P3 cells producing matrix with greater compressive properties and P5 cells secreting matrix with higher glycosaminoglycan/DNA and collagen/DNA ratios. Furthermore, we identified 24 genes that were differentially expressed with passaging and enriched in human osteoarthritis (OA) genome-wide association studies, thereby prioritizing them as functionally relevant targets to improve protocols that recapitulate functional healthy cartilage with cells from adult donors. Specifically, we identified novel genes, such as TMEM190 and RAB11FIP4, which were enriched with human hip OA and may play a role in chondrocyte dedifferentiation. This work lays the foundation for several pathways and genes that could be modulated to enhance the efficacy for chondrocyte culture for tissue regeneration, which could have transformative impacts for cell-based cartilage repair strategies.

13.
Sci Rep ; 14(1): 14655, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918485

RESUMEN

Osteocytes locally remodel their surrounding tissue through perilacunar canalicular remodeling (PLR). During lactation, osteocytes remove minerals to satisfy the metabolic demand, resulting in increased lacunar volume, quantifiable with synchrotron X-ray radiation micro-tomography (SRµCT). Although the effects of lactation on PLR are well-studied, it remains unclear whether PLR occurs uniformly throughout the bone and what mechanisms prevent PLR from undermining bone quality. We used SRµCT imaging to conduct an in-depth spatial analysis of the impact of lactation and osteocyte-intrinsic MMP13 deletion on PLR in murine bone. We found larger lacunae undergoing PLR are located near canals in the mid-cortex or endosteum. We show lactation-induced hypomineralization occurs 14 µm away from lacunar edges, past a hypermineralized barrier. Our findings reveal that osteocyte-intrinsic MMP13 is crucial for lactation-induced PLR near lacunae in the mid-cortex but not for whole-bone resorption. This research highlights the spatial control of PLR on mineral distribution during lactation.


Asunto(s)
Remodelación Ósea , Lactancia , Osteocitos , Microtomografía por Rayos X , Animales , Lactancia/fisiología , Femenino , Osteocitos/metabolismo , Osteocitos/fisiología , Ratones , Remodelación Ósea/fisiología , Metaloproteinasa 13 de la Matriz/metabolismo
14.
Bone Res ; 12(1): 13, 2024 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-38409111

RESUMEN

Poor bone quality is a major factor in skeletal fragility in elderly individuals. The molecular mechanisms that establish and maintain bone quality, independent of bone mass, are unknown but are thought to be primarily determined by osteocytes. We hypothesize that the age-related decline in bone quality results from the suppression of osteocyte perilacunar/canalicular remodeling (PLR), which maintains bone material properties. We examined bones from young and aged mice with osteocyte-intrinsic repression of TGFß signaling (TßRIIocy-/-) that suppresses PLR. The control aged bone displayed decreased TGFß signaling and PLR, but aging did not worsen the existing PLR suppression in male TßRIIocy-/- bone. This relationship impacted the behavior of collagen material at the nanoscale and tissue scale in macromechanical tests. The effects of age on bone mass, density, and mineral material behavior were independent of osteocytic TGFß. We determined that the decline in bone quality with age arises from the loss of osteocyte function and the loss of TGFß-dependent maintenance of collagen integrity.


Asunto(s)
Remodelación Ósea , Osteocitos , Humanos , Anciano , Masculino , Animales , Ratones , Remodelación Ósea/fisiología , Colágeno/farmacología , Envejecimiento , Factor de Crecimiento Transformador beta/farmacología
15.
Biophys J ; 104(8): 1794-804, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23601326

RESUMEN

It was recently demonstrated that mechanical shearing of synovial fluid (SF), induced during joint motion, rapidly activates latent transforming growth factor ß (TGF-ß). This discovery raised the possibility of a physiological process consisting of latent TGF-ß supply to SF, activation via shearing, and transport of TGF-ß into the cartilage matrix. Therefore, the two primary objectives of this investigation were to characterize the secretion rate of latent TGF-ß into SF, and the transport of active TGF-ß across the articular surface and into the cartilage layer. Experiments on tissue explants demonstrate that high levels of latent TGF-ß1 are secreted from both the synovium and all three articular cartilage zones (superficial, middle, and deep), suggesting that these tissues are capable of continuously replenishing latent TGF-ß to SF. Furthermore, upon exposure of cartilage to active TGF-ß1, the peptide accumulates in the superficial zone (SZ) due to the presence of an overwhelming concentration of nonspecific TGF-ß binding sites in the extracellular matrix. Although this response leads to high levels of active TGF-ß in the SZ, the active peptide is unable to penetrate deeper into the middle and deep zones of cartilage. These results provide strong evidence for a sequential physiologic mechanism through which SZ chondrocytes gain access to active TGF-ß: the synovium and articular cartilage secrete latent TGF-ß into the SF and, upon activation, TGF-ß transports back into the cartilage layer, binding exclusively to the SZ.


Asunto(s)
Cartílago Articular/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Sitios de Unión , Bovinos , Condrocitos/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Humanos , Técnicas In Vitro , Líquido Sinovial/metabolismo
16.
Arthritis Rheum ; 64(10): 3278-89, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22674505

RESUMEN

OBJECTIVE: To identify mechanisms by which Smad3 maintains articular cartilage and prevents osteoarthritis. METHODS: A combination of in vivo and in vitro approaches was used to test the hypothesis that Smad3 represses Runx2-inducible gene expression to prevent articular cartilage degeneration. Col2-Cre;Smad3(fl/fl) mice allowed study of the chondrocyte-intrinsic role of Smad3 independently of its role in the perichondrium or other tissues. Primary articular cartilage chondrocytes from Smad3(fl/fl) mice and ATDC5 chondroprogenitor cells were used to evaluate Smad3 and Runx2 regulation of matrix metalloproteinase 13 (MMP-13) messenger RNA (mRNA) and protein expression. RESULTS: Chondrocyte-specific reduction of Smad3 caused progressive articular cartilage degeneration due to imbalanced cartilage matrix synthesis and degradation. In addition to reduced type II collagen mRNA expression, articular cartilage from Col2-Cre;Smad3(fl/fl) mice was severely deficient in type II collagen and aggrecan protein due to excessive MMP-13-mediated proteolysis of these key cartilage matrix constituents. Normally, transforming growth factor ß (TGFß) signals through Smad3 to confer a rapid and dynamic repression of Runx2-inducible MMP-13 expression. However, we found that in the absence of Smad3, TGFß signals through p38 and Runx2 to induce MMP-13 expression. CONCLUSION: Our findings elucidate a mechanism by which Smad3 mutations in humans and mice cause cartilage degeneration and osteoarthritis. Specifically, Smad3 maintains the balance between cartilage matrix synthesis and degradation by inducing type II collagen expression and repressing Runx2-inducible MMP-13 expression. Selective activation of TGFß signaling through Smad3, rather than p38, may help to restore the balance between matrix synthesis and proteolysis that is lost in osteoarthritis.


Asunto(s)
Cartílago Articular/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Osteoartritis/prevención & control , Proteína smad3/metabolismo , Animales , Cartílago Articular/citología , Condrocitos/citología , Condrocitos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Metaloproteinasa 13 de la Matriz/genética , Ratones , Osteoartritis/genética , Osteoartritis/metabolismo , Proteína smad3/genética
17.
Bone ; 175: 116836, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37414200

RESUMEN

Bone derives its ability to resist fracture from bone mass and quality concurrently; however, many questions about the molecular mechanisms controlling bone quality remain unanswered, limiting the development of diagnostics and therapeutics. Despite the increasing evidence on the importance of miR181a/b-1 in bone homeostasis and disease, whether and how osteocyte-intrinsic miR181a/b-1 controls bone quality remains elusive. Osteocyte-intrinsic deletion of miR181a/b-1 in osteocytes in vivo resulted in compromised overall bone mechanical behavior in both sexes, although the parameters affected by miR181a/b-1 varied distinctly based on sex. Furthermore, impaired fracture resistance in both sexes was unexplained by cortical bone morphology, which was altered in female mice and intact in male mice with miR181a/b-1-deficient osteocytes. The role of miR181a/b-1 in the regulation of osteocyte metabolism was apparent in bioenergetic testing of miR181a/b-1-deficient OCY454 osteocyte-like cells and transcriptomic analysis of cortical bone from mice with osteocyte-intrinsic ablation of miR181a/b-1. Altogether, this study demonstrates the control of osteocyte bioenergetics and the sexually dimorphic regulation of cortical bone morphology and mechanical properties by miR181a/b-1, hinting at the role of osteocyte metabolism in the regulation of mechanical behavior.


Asunto(s)
Huesos , Osteocitos , Ratones , Masculino , Animales , Femenino , Osteocitos/metabolismo , Huesos/metabolismo , Hueso Cortical/metabolismo , Densidad Ósea , Metabolismo Energético
18.
Bone Rep ; 18: 101647, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36636109

RESUMEN

Subchondral bone participates in crosstalk with articular cartilage to maintain joint homeostasis, and disruption of either tissue results in overall joint degeneration. Among the subchondral bone changes observed in osteoarthritis (OA), subchondral bone plate (SBP) thickening has a time-dependent relationship with cartilage degeneration and has recently been shown to be regulated by osteocytes. Here, we evaluate the effect of age on SBP thickness and cartilage degeneration in aging mice. We find that SBP thickness significantly increases by 18-months of age, corresponding temporally with increased cartilage degeneration. To identify factors in subchondral bone that may participate in bone cartilage crosstalk or OA, we leveraged mouse transcriptomic data from one joint tissue compartment - osteocyte-enriched bone - to search for enrichment with human OA in UK Biobank and Arthritis Research UK Osteoarthritis Genetics (arcOGEN) GWAS using the mouse2human (M2H, www.mouse2human.org) strategy. Genes differentially expressed in aging mouse bone are significantly enriched for human OA, showing joint site-specific (knee vs. hip) relationships, exhibit temporal associations with age, and unique gene clusters are implicated in each type of OA. Application of M2H identifies genes with known and unknown functions in osteocytes and OA development that are clinically associated with human OA. Altogether, this work prioritizes genes with a potential role in bone/cartilage crosstalk for further mechanistic study based on their association with human OA in GWAS.

19.
J Bone Metab ; 30(1): 1-29, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36950837

RESUMEN

Molecular omics technologies, including proteomics, have enabled the elucidation of key signaling pathways that mediate bidirectional communication between the brain and bone tissues. Here we provide a brief summary of the clinical and molecular evidence of the need to study the bone-brain axis of cross-tissue cellular communication. Clear clinical and molecular evidence suggests biological interactions and similarities between bone and brain cells. Here we review the current mass spectrometric techniques for studying brain and bone diseases with an emphasis on neurodegenerative diseases and osteoarthritis/osteoporosis, respectively. Further study of the bone-brain axis on a molecular level and evaluation of the role of proteins, neuropeptides, osteokines, and hormones in molecular pathways linked to bone and brain diseases is critically needed. The use of mass spectrometry and other omics technologies to analyze these cross-tissue signaling events and interactions will help us better understand disease progression and comorbidities and potentially identify new pathways and targets for therapeutic interventions. Proteomic measurements are particularly favorable for investigating the role of signaling and secreted and circulating analytes and identifying molecular and metabolic pathways implicated in age-related diseases.

20.
J Gerontol A Biol Sci Med Sci ; 78(10): 1733-1739, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37148367

RESUMEN

The National Institute on Aging sponsored a symposium at the Gerontological Society of America (GSA) annual meeting in Indianapolis, Indiana, to discuss recent discoveries related to senescent and inflammatory mechanisms in aging and disease. Consistent with the 2022 Biological Sciences GSA program led by Dr. Rozalyn Anderson, the symposium featured early-stage investigators and a leader in the field of geroscience research. Cell senescence and immune interactions coordinate homeostatic and protective programming throughout the life span. Dysfunctional communication in this exchange eventuates in inflammation-related compositional changes in aged tissues, including propagation of the senescence-associated secretory phenotype and accumulation of senescent and exhausted immune cells. Presentations in this symposium explored senescent and immune-related dysfunction in aging from diverse viewpoints and featured emerging cellular and molecular methods. A central takeaway from the event was that the use of new models and approaches, including single-cell -omics, novel mouse models, and 3D culture systems, is revealing dynamic properties and interactions of senescent and immune cell fates. This knowledge is critical for devising new therapeutic approaches with important translational relevance.


Asunto(s)
Envejecimiento , National Institute on Aging (U.S.) , Animales , Estados Unidos , Ratones , Fenotipo , Senescencia Celular , Inflamación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA