Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.857
Filtrar
1.
J Biomech Eng ; 146(8)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38530647

RESUMO

Articular cartilage (AC) is a load-bearing tissue that covers long bones in synovial joints. The biphasic/poroelastic mechanical properties of AC help it to protect joints by distributing loads, absorbing impact forces, and reducing friction. Unfortunately, alterations in these mechanical properties adversely impact cartilage function and precede joint degeneration in the form of osteoarthritis (OA). Thus, understanding what factors regulate the poroelastic mechanical properties of cartilage is of great scientific and clinical interest. Transgenic mouse models provide a valuable platform to delineate how specific genes contribute to cartilage mechanical properties. However, the poroelastic mechanical properties of murine articular cartilage are challenging to measure due to its small size (thickness ∼ 50 microns). In the current study, our objective was to test whether the poroelastic mechanical properties of murine articular cartilage can be determined based solely on time-dependent cell death measurements under constant loading conditions. We hypothesized that in murine articular cartilage subjected to constant, sub-impact loading from an incongruent surface, cell death area and tissue strain are closely correlated. We further hypothesized that the relationship between cell death area and tissue strain can be used-in combination with inverse finite element modeling-to compute poroelastic mechanical properties. To test these hypotheses, murine cartilage-on-bone explants from different anatomical locations were subjected to constant loading conditions by an incongruent surface in a custom device. Cell death area increased over time and scaled linearly with strain, which rose in magnitude over time due to poroelastic creep. Thus, we were able to infer tissue strain from cell death area measurements. Moreover, using tissue strain values inferred from cell death area measurements, we applied an inverse finite element modeling procedure to compute poroelastic material properties and acquired data consistent with previous studies. Collectively, our findings demonstrate in the key role poroelastic creep plays in mediating cell survival in mechanically loaded cartilage and verify that cell death area can be used as a surrogate measure of tissue strain that enables determination of murine cartilage mechanical properties.


Assuntos
Cartilagem Articular , Osteoartrite , Animais , Camundongos , Condrócitos/fisiologia , Estresse Mecânico , Cartilagem Articular/fisiologia , Morte Celular
2.
Poult Sci ; 103(4): 103534, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38401226

RESUMO

The poultry skeletal system serves multiple functions, not only providing structural integrity but also maintaining the balance of essential minerals such as calcium and phosphorus. However, in recent years, the consideration of skeletal traits has been overlooked in the selective breeding of broilers, resulting in an inadequate adaptation of the skeletal system to cope with the rapid increase in body weight. Consequently, this leads to lameness and bone diseases such as tibial dyschondroplasia (TD), which significantly impact the production performance of broilers. Accumulating evidence has shown that microRNAs (miRNA) play a crucial role in the differentiation, formation, and disease of cartilage. However, the miRNA-mediated molecular mechanism underlying chicken TD formation is still poorly understood. The objective of this study was to investigate the biological function and regulatory mechanism of miRNA in chicken TD formation. Based on transcriptome sequencing of tibial cartilage in the healthy group and TD group, miR-206a-3p was found to be highly expressed in TD cartilage. The function of miR-206a-3p was explored through the transfection test of miR-206a-3p mimics and miR-206a-3p inhibitor. In this study, we utilized qRT-PCR, CCK-8, EdU, western blot, and flow cytometry to detect the proliferation, differentiation, and apoptosis of chondrocytes. The results revealed that miR-206a-3p suppressed the proliferation and differentiation of TD chondrocytes while promoting their programmed cell death. Furthermore, through biosynthesis and dual luciferase assays, it was determined that BMP6 was the direct target gene of miR-206a-3p. This finding was further supported by rescue experiments which confirmed the involvement of BMP6 in the regulatory pathway governed by miR-206a-3p. Our results suggest that miR-206a-3p can inhibits the proliferation and differentiation promote apoptosis through the target gene BMP-6 and suppressing the Smad2/3 signaling pathway in chicken TD chondrocytes.


Assuntos
MicroRNAs , Osteocondrodisplasias , Animais , Condrócitos/fisiologia , Galinhas/genética , Galinhas/metabolismo , Osteocondrodisplasias/genética , Osteocondrodisplasias/veterinária , Proteína Morfogenética Óssea 6/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proliferação de Células , Apoptose
3.
J Orthop Res ; 42(1): 54-65, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37415557

RESUMO

Tissue swelling represents an early sign of osteoarthritis, reflecting osmolarity changes from iso- to hypo-osmotic in the diseased joints. Increased tissue hydration may drive cell swelling. The opposing cartilages in a joint may swell differently, thereby predisposing the more swollen cartilage and cells to mechanical injuries. However, our understanding of the tissue-cell interdependence in osmotically loaded joints is limited as tissue and cell swellings have been studied separately. Here, we measured tissue and cell responses of opposing patellar (PAT) and femoral groove (FG) cartilages in lapine knees exposed to an extreme hypo-osmotic challenge. We found that the tissue matrix and most cells swelled during the hypo-osmotic challenge, but to a different extent (tissue: <3%, cells: 11%-15%). Swelling-induced tissue strains were anisotropic, showing 2%-4% stretch and 1%-2% compression along the first and third principal directions, respectively. These strains were amplified by 5-8 times in the cells. Interestingly, the first principal strains of tissue and cells occurred in different directions (60-61° for tissue vs. 8-13° for cells), suggesting different mechanisms causing volume expansion in the tissue and the cells. Instead of the continuous swelling observed in the tissue matrix, >88% of cells underwent regulatory volume decrease to return to their pre-osmotic challenge volumes. Cell shapes changed in the early phase of swelling but stayed constant thereafter. Kinematic changes to tissue and cells were larger for PAT cartilage than for FG cartilage. We conclude that the swelling-induced deformation of tissue and cells is anisotropic. Cells actively restored volume independent of the surrounding tissues and seemed to prioritize volume restoration over shape restoration. Our findings shed light on tissue-cell interdependence in changing osmotic environments that is crucial for cell mechano-transduction in swollen/diseased tissues.


Assuntos
Cartilagem Articular , Condrócitos , Pressão Osmótica , Condrócitos/fisiologia , Concentração Osmolar , Osmose
4.
Adv Biol (Weinh) ; 8(1): e2300268, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37688354

RESUMO

One of the main components of articular cartilage is the chondrocyte's pericellular matrix (PCM), which is critical for regulating mechanotransduction, biochemical cues, and healthy cartilage development. Here, individual primary human chondrocytes (PHC) are encapsulated and cultured in 50 µm diameter alginate microgels using drop-based microfluidics. This unique culturing method enables PCM formation and manipulation of individual cells. Over ten days, matrix formation is observed using autofluorescence imaging, and the elastic moduli of isolated cells are measured using AFM. Matrix production and elastic modulus increase are observed for the chondrons cultured in microgels. Furthermore, the elastic modulus of cells grown in microgels increases ≈ten-fold over ten days, nearly reaching the elastic modulus of in vivo PCM. The AFM data is further analyzed using a Gaussian mixture model and shows that the population of PHCs grown in microgels exhibit two distinct populations with elastic moduli averaging 9.0 and 38.0 kPa. Overall, this work shows that microgels provide an excellent culture platform for the growth and isolation of PHCs, enabling PCM formation that is mechanically similar to native PCM. The microgel culture platform presented here has the potential to revolutionize cartilage regeneration procedures through the inclusion of in vitro developed PCM.


Assuntos
Cartilagem Articular , Microgéis , Humanos , Condrócitos/fisiologia , Microscopia de Força Atômica , Matriz Extracelular/fisiologia , Mecanotransdução Celular , Cartilagem Articular/fisiologia
5.
Am J Orthod Dentofacial Orthop ; 164(2): 201-214, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36922241

RESUMO

INTRODUCTION: Transdifferentiation of chondrocytes into bone cells explains most of the prenatal and early postnatal condylar growth, but its role during later postnatal growth and the mechanisms regulating transdifferentiation remain unknown. This study aimed to quantify the effects of mechanical loading on chondrocyte-derived osteogenesis during late postnatal condylar growth using a short-term mandibular laterotrusion model. METHODS: Thirty 4-week-old Aggrecan-CreERT2, R26RtdTomato, and 2.3Col1a1-GFP compound mice received tamoxifen injections and were divided into control and experimental groups. Appliances were bonded to shift the mandibles of the experimental mice for 5 days, causing protrusion and retrusion of the right and left condyles, respectively. Radiographic, microcomputed tomographic, and histomorphometric analyses were performed. RESULTS: The experimental and control groups showed substantial transdifferentiation of chondrocytes into bone cells. The experimental mice developed asymmetric mandibles, with the protrusive side significantly longer than the retrusive side. The protrusive condyles showed significantly increased chondrogenesis and greater numbers of chondrocyte-derived osteogenic cells, especially in the posterior third. The opposite effects were seen on the retrusive side. CONCLUSIONS: Transdifferentiation of chondrocytes into bone cells occurs during late postnatal condylar growth. Laterotrusion regulates condylar chondrogenesis and chondrocyte transdifferentiation, which alters the amount and direction of condylar growth. Our study demonstrated that chondrocytes are key players in condylar bone formation and should be the focus of studies to control and further understand condylar growth.


Assuntos
Condrócitos , Côndilo Mandibular , Gravidez , Feminino , Camundongos , Animais , Condrócitos/fisiologia , Côndilo Mandibular/diagnóstico por imagem , Transdiferenciação Celular , Osteogênese , Mandíbula
6.
Comput Methods Programs Biomed ; 231: 107419, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36842346

RESUMO

BACKGROUND AND OBJECTIVE: Osteoarthritis (OA) is a pervasive and debilitating disease, wherein degeneration of cartilage features prominently. Despite extensive research, we do not yet understand the cause or progression of OA. Studies show biochemical, mechanical, and biological factors affect cartilage health. Mechanical loads influence synthesis of biochemical constituents which build and/or break down cartilage, and which in turn affect mechanical loads. OA-associated biochemical profiles activate cellular activity that disrupts homeostasis. To understand the complex interplay among mechanical stimuli, biochemical signaling, and cartilage function requires integrating vast research on experimental mechanics and mechanobiology-a task approachable only with computational models. At present, mechanical models of cartilage generally lack chemo-biological effects, and biochemical models lack coupled mechanics, let alone interactions over time. METHODS: We establish a first-of-its kind virtual cartilage: a modeling framework that considers time-dependent, chemo-mechano-biologically induced turnover of key constituents resulting from biochemical, mechanical, and/or biological activity. We include the "minimally essential" yet complex chemical and mechanobiological mechanisms. Our 3-D framework integrates a constitutive model for the mechanics of cartilage with a novel model of homeostatic adaptation by chondrocytes to pathological mechanical stimuli, and a new application of anisotropic growth (loss) to simulate degradation clinically observed as cartilage thinning. RESULTS: Using a single set of representative parameters, our simulations of immobilizing and overloading successfully captured loss of cartilage quantified experimentally. Simulations of immobilizing, overloading, and injuring cartilage predicted dose-dependent recovery of cartilage when treated with suramin, a proposed therapeutic for OA. The modeling framework prompted us to add growth factors to the suramin treatment, which predicted even better recovery. CONCLUSIONS: Our flexible framework is a first step toward computational investigations of how cartilage and chondrocytes mechanically and biochemically evolve in degeneration of OA and respond to pharmacological therapies. Our framework will enable future studies to link physical activity and resulting mechanical stimuli to progression of OA and loss of cartilage function, facilitating new fundamental understanding of the complex progression of OA and elucidating new perspectives on causes, treatments, and possible preventions.


Assuntos
Cartilagem Articular , Osteoartrite , Humanos , Cartilagem Articular/patologia , Suramina/farmacologia , Modelos Biológicos , Osteoartrite/metabolismo , Osteoartrite/patologia , Condrócitos/patologia , Condrócitos/fisiologia
7.
Elife ; 122023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36656123

RESUMO

Development of the craniofacial skeleton requires interactions between progenitor cells and the collagen-rich extracellular matrix (ECM). The mediators of these interactions are not well-defined. Mutations in the discoidin domain receptor 2 gene (DDR2), which encodes a non-integrin collagen receptor, are associated with human craniofacial abnormalities, such as midface hypoplasia and open fontanels. However, the exact role of this gene in craniofacial morphogenesis is not known. As will be shown, Ddr2-deficient mice exhibit defects in craniofacial bones including impaired calvarial growth and frontal suture formation, cranial base hypoplasia due to aberrant chondrogenesis and delayed ossification at growth plate synchondroses. These defects were associated with abnormal collagen fibril organization, chondrocyte proliferation and polarization. As established by localization and lineage-tracing studies, Ddr2 is expressed in progenitor cell-enriched craniofacial regions including sutures and synchondrosis resting zone cartilage, overlapping with GLI1 + cells, and contributing to chondrogenic and osteogenic lineages during skull growth. Tissue-specific knockouts further established the requirement for Ddr2 in GLI +skeletal progenitors and chondrocytes. These studies establish a cellular basis for regulation of craniofacial morphogenesis by this understudied collagen receptor and suggest that DDR2 is necessary for proper collagen organization, chondrocyte proliferation, and orientation.


We each have unique facial features that are key to our identities. These features are inherited, but the mechanisms are poorly understood. People with the genetic disease spondylo-meta-epiphyseal dysplasia, or SMED, have characteristic facial and skull abnormalities including a flattened face and shortened skull. SMED is associated with mutations that inactivate the gene encoding a protein called discoidin domain receptor 2 (DDR2), which is a receptor for collagen. Collagen is the major structural protein in the human body, supporting the structure of cells and tissues. It also controls cell behaviors including growth, migration and differentiation, and it helps form tissues such as cartilage or bone. At least some of the effects of collagen on cells depend on its interaction with DDR2. Since the facial and skull abnormalities in mice with mutations that stop DDR2 from working correctly resemble those of SMED patients, these mice can be used to understand the cellular basis for this disease, as well as the role of DDR2 in the embryonic development of the face and skull. Therefore, Mohamed et al. set out to understand how loss of DDR2 causes the characteristic facial and skull defects associated with SMED. Mohamed et al. used mice that had been genetically modified so that DDR2 could be inactivated in skeletal progenitor cells, cartilage cells and bone cells (osteoblasts). Examining these mice, they found that the shortened skulls and flat face characteristic of mice lacking DDR2 are due to bones at the skull base failing to elongate correctly due to defects in the growth centers that depend on cartilage. Mohamed et al. also discovered that the cells that normally produce DDR2 are the progenitors of cartilage and bone-forming cells, which partly explains why lacking this protein leads to issues in growth of these tissues. In addition to shedding light on the causes of SMED, Mohamed et al.'s results also provide general insights into the mechanisms controlling the formation of facial and skull bones that depend on interactions between cells and collagen. This information may help explain how other abnormalities in the face and skull emerge, and provide a basis for how the shape of the skull has changed during human evolution. In the future, it may be possible to manipulate the activity of DDR2 to correct skull defects.


Assuntos
Receptor com Domínio Discoidina 2 , Animais , Humanos , Camundongos , Cartilagem , Condrócitos/fisiologia , Colágeno , Receptor com Domínio Discoidina 2/genética , Receptores de Colágeno
10.
Methods Mol Biol ; 2598: 289-300, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36355299

RESUMO

Viral gene transfer, known as transduction, is a powerful research tool for studying the biology of chondrocytes in novel ways and also a technology enabling the use of gene therapy for regenerating cartilage and treating diseases that affect cartilage, such as osteoarthritis. Adenovirus, retrovirus, lentivirus, and adeno-associated virus (AAV) are most commonly used to transduce chondrocytes. Although AAV is able to transduce chondrocytes in situ by intra-articular injection, chondrocytes are most commonly transduced in monolayer culture using the four vectors mentioned above. Protocols for achieving this are described, along with a discussion of the variables that can influence transduction efficiency.


Assuntos
Cartilagem Articular , Condrócitos , Condrócitos/fisiologia , Transdução Genética , Vetores Genéticos/genética , Técnicas de Transferência de Genes , Dependovirus/genética , Terapia Genética/métodos , Genes Virais
11.
Dev Biol ; 493: 80-88, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36368521

RESUMO

Bones and articular cartilage are important load-bearing tissues. The fluid flow inside the bone cells and cell interaction with the extracellular matrix serve as the mechanical cues for bones and joints. Piezo1 is an ion channel found on the cell surface of many cell types, including osteocytes and chondrocytes. It is activated in response to mechanical stimulation, which subsequently mediates a variety of signaling pathways in osteoblasts, osteocytes, and chondrocytes. Piezo1 activation in osteoblastic cells positively regulates osteogenesis, while its activation in joints mediates cartilage degradation. This review focuses on the most recent research on Piezo1 in bone development and regeneration.


Assuntos
Osso e Ossos , Condrócitos , Estresse Mecânico , Condrócitos/fisiologia , Homeostase , Biofísica
12.
Life Sci ; 309: 121043, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36206835

RESUMO

Tissue engineering holds great promise in the generation of cartilage analogues for cartilage injury repair and replacement. However, for a long time, a variety of issues have remained unsolved in articular cartilage tissue engineering concerning immunogenicity, stability, and mechanical strength, among others. One of the most remarkable reasons lies in the lack or insufficiency of recapitulating the chondrocyte biomechanical microenvironment (BME) in the articular cartilage tissue engineering. In recent years, an increasing number of studies have disclosed the crucial role of the BME in chondrocyte phenotype and cartilage functions, which has inspired more precise and individualized research in articular cartilage tissue engineering by engineering the chondrocyte BME. This review first takes an in-depth look into the chondrocyte BME and its crucial effects on chondrocytes and articular cartilage tissues. Then, as the core of this work, the principal strategies and their approaches of engineering the chondrocyte BME towards articular cartilage tissue engineering were comprehensively discussed, from the perspectives of simulating the main characteristics of chondrocyte BME including engineering the heterogeneous matrix and the dynamic mechanical stimulation. The current limitations in this emerging area and potential strategies were also proposed to shed some light on the future directions in this field. Although there are still challenges to obtaining engineered articular cartilages with desired performance, the road ahead is bright under the constant efforts in engineering the chondrocyte BME at higher levels towards articular cartilage tissue engineering.


Assuntos
Cartilagem Articular , Condrócitos , Condrócitos/fisiologia , Cartilagem Articular/fisiologia , Engenharia Tecidual , Fenótipo
13.
J Biomech ; 144: 111336, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36240656

RESUMO

Aging is the most prominent risk factor for osteoarthritis onset, but the etiology of aging-associated cartilage degeneration is not fully understood. Recent studies by Guilak and colleagues have highlighted the crucial roles of cell-matrix interactions in cartilage homeostasis and disease. This study thus quantified aging-associated changes in cartilage biomechanics and chondrocyte intracellular calcium signaling, [Ca2+]i, activities in wild-type mice at 3, 12 and 22 months of age. In aged mice, articular cartilage exhibits reduced staining of sulfated glycosaminoglycans (sGAGs), indicating decreased aggrecan content. On cartilage surface, collagen fibrils undergo significant thickening while retaining their transverse isotropic architecture, and exhibit signs of fibril crimping in the 22-month group. These compositional and structural changes contribute to a significant decrease in cartilage modulus at 22 months of age (0.55 ± 0.25 MPa, mean ± 95 % CI, n = 8) relative to those at 3 and 12 months (1.82 ± 0.48 MPa and 1.45 ± 0.46 MPa, respectively, n ≥ 8). Despite the decreases in sGAG content and tissue modulus, chondrocytes do not exhibit significantly demoted [Ca2+]i activities in situ, in both physiological (isotonic) and osmotically instigated (hypo- and hypertonic) conditions. At 12 months of age, there exists a sub-population of chondrocytes with hyper-active [Ca2+]i responses under hypotonic stimuli, possibly indicating a phenotypic shift of chondrocytes during aging. Together, these results yield new insights into aging-associated biomechanical and mechanobiological changes of murine cartilage, providing a benchmark for elucidating the molecular mechanisms of age-related changes in cell-matrix interactions.


Assuntos
Cartilagem Articular , Condrócitos , Camundongos , Animais , Condrócitos/fisiologia , Fenômenos Biomecânicos , Sinalização do Cálcio , Cartilagem Articular/fisiologia , Envelhecimento
14.
J World Fed Orthod ; 11(5): 146-155, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36153283

RESUMO

BACKGROUND: We and others have reported that low-magnitude high-frequency dynamic loading has an osteogenic effect on alveolar bone. Since chondrocytes and osteoblasts originate from the same progenitor cells, we reasoned that dynamic loading may stimulate a similar response in chondrocytes. A stimulating effect could be beneficial for patients with damaged condylar cartilage or mandibular deficiency. METHODS: Studies were conducted on growing Sprague-Dawley rats divided into three groups: control, static load, and dynamic load. The dynamic load group received a dynamic load on the lower right molars 5 minutes per day with a 0.3 g acceleration and peak strain of 30 µÎµ registered by accelerometer and strain gauge. The static load group received an equivalent magnitude of static force (30 µÎµ). The control group did not receive any treatment. Samples were collected at days 0, 28, and 56 for reverse transcriptase polymerase chain reaction analysis, microcomputed tomography, and histology and fluorescent microscopy analysis. RESULTS: Our experiments showed that dynamic loading had a striking effect on condylar cartilage, increasing the proliferation and differentiation of mesenchymal cells into chondrocytes, and promoting chondrocyte maturation. This effect was accompanied by increased endochondral bone formation resulting in lengthening of the condylar process. CONCLUSIONS: Low-magnitude, high-frequency dynamic loading can have a positive effect on condylar cartilage and endochondral bone formation in vivo. This effect has the potential to be used as a treatment for regenerating condylar cartilage and to enhance the effect of orthopedic appliances on mandibular growth.


Assuntos
Condrócitos , Côndilo Mandibular , Animais , Cartilagem/patologia , Condrócitos/fisiologia , Ratos , Ratos Sprague-Dawley , Microtomografia por Raio-X
15.
Osteoarthritis Cartilage ; 30(12): 1547-1560, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36150678

RESUMO

Articular cartilage (AC) has limited capacity for repair. The first attempt to repair cartilage using tissue engineering was reported in 1977. Since then, cell-based interventions have entered clinical practice in orthopaedics, and several tissue engineering approaches to repair cartilage are in the translational pipeline towards clinical application. Classically, these involve a scaffold, substrate or matrix to provide structure, and cells such as chondrocytes or mesenchymal stromal cells to generate the tissue. We discuss the advantages and drawbacks of the use of various cell types, natural and synthetic scaffolds, multiphasic or gradient-based scaffolds, and self-organizing or self-assembling scaffold-free systems, for the engineering of cartilage constructs. Several challenges persist including achieving zonal tissue organization and integration with the surrounding tissue upon implantation. Approaches to improve cartilage thickness, organization and mechanical properties include mechanical stimulation, culture under hypoxic conditions, and stimulation with growth factors or other macromolecules. In addition, advanced technologies such as bioreactors, biosensors and 3D bioprinting are actively being explored. Understanding the underlying mechanisms of action of cell therapy and tissue engineering approaches will help improve and refine therapy development. Finally, we discuss recent studies of the intrinsic cellular and molecular mechanisms of cartilage repair that have identified novel signals and targets and are inspiring the development of molecular therapies to enhance the recruitment and cartilage reparative activity of joint-resident stem and progenitor cells. A one-fits-all solution is unrealistic, and identifying patients who will respond to a specific targeted treatment will be critical.


Assuntos
Cartilagem Articular , Células-Tronco Mesenquimais , Humanos , Engenharia Tecidual , Condrócitos/fisiologia , Células-Tronco Mesenquimais/metabolismo , Terapia Baseada em Transplante de Células e Tecidos , Tecidos Suporte/química
16.
Methods Mol Biol ; 2579: 227-233, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36045210

RESUMO

Cell proliferation is one of the key events that regulates organism development. In the limb, chondrocytes differentiate into a multi-layered cellular template called the growth plate. Chondrocyte proliferation is essential to provide the necessary cells that allow growth of a bone. Deregulated cell proliferation will lead to truncated bone elements. Immunofluorescence is a biological technique that uses specific antibodies to detect the subcellular localization of a proliferative marker within cellular or tissue context. In this chapter, we illustrate how to perform immunofluorescence to detect the localization of Ki-67 (a marker of actively growing/proliferating chondrocytes) in order to assess the growth fraction of the columnar chondrocytes in the growth plate in paraffin-embedded mouse tissue limb.


Assuntos
Condrócitos , Lâmina de Crescimento , Animais , Diferenciação Celular/fisiologia , Condrócitos/fisiologia , Imunofluorescência , Antígeno Ki-67 , Camundongos , Osteogênese/fisiologia
17.
Ann Biomed Eng ; 50(12): 1911-1922, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35879583

RESUMO

Mechanical stimuli are fundamental in the development of organs and tissues, their growth, regeneration or disease. They influence the biochemical signals produced by the cells, and, consequently, the development and spreading of a disease. Moreover, tumour cells are usually characterized by a decrease in the cell mechanical properties that may be directly linked to their metastatic potential. Thus, recently, the experimental and computational study of cell biomechanics is facing a growing interest. Various experimental approaches have been implemented to describe the passive response of cells; however, cell variability and complex experimental procedures may affect the obtained mechanical properties. For this reason, in-silico computational models have been developed through the years, to overcome such limitations, while proposing valuable tools to understand cell mechanical behaviour. This being the case, we propose a combined continuous-tensegrity finite element (FE) model to analyse the mechanical response of a cell and its subcomponents, observing how every part contributes to the overall mechanical behaviour. We modelled both Atomic Force Microscopy (AFM) indentation and micropipette aspiration techniques, as common mechanical tests for cells and elucidated also the role of cell cytoplasm and cytoskeleton in the global cell mechanical response.


Assuntos
Condrócitos , Citoesqueleto , Fenômenos Biomecânicos , Microscopia de Força Atômica , Condrócitos/fisiologia , Simulação por Computador , Estresse Mecânico , Modelos Biológicos
18.
Am J Physiol Cell Physiol ; 323(1): C236-C247, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35649254

RESUMO

Mechanical stress and the stiffness of the extracellular matrix are key drivers of tissue development and homeostasis. Aberrant mechanosensation is associated with a wide range of pathologies, including osteoarthritis. Matrix (or substrate) stiffness plays a major role in cell spreading, adhesion, proliferation, and differentiation. However, how specific cells sense substrate stiffness still remains unclear. The primary cilium is an essential cellular organelle that senses and integrates mechanical and chemical signals from the extracellular environment. We hypothesized that the primary cilium dynamically alters its length and position to fine-tune cell mechanosignaling based on substrate stiffness alone. We used a hydrogel system of varying substrate stiffness to examine the role of stiffness on cilia frequency, length, and centriole position as well as cell and nuclei area over time. Contrary to other cell types, we show that chondrocyte primary cilia shorten on softer substrates, demonstrating tissue-specific mechanosensing that is aligned with the tissue stiffness the cells originate from. We further show that stiffness determines centriole positioning to either the basal or apical membrane during attachment and spreading, with centrioles positioned toward the basal membrane on stiffer substrates. These phenomena are mediated by force generation actin-myosin stress fibers in a time-dependent manner. Finally, we show on stiff substrates that primary cilia are involved in tension-mediated cell spreading. We propose that substrate stiffness plays a role in cilia positioning, regulating cellular responses to external forces, and maybe a key driver of mechanosignaling-associated diseases.


Assuntos
Centríolos , Osteoartrite , Centríolos/metabolismo , Condrócitos/fisiologia , Cílios/metabolismo , Matriz Extracelular , Humanos , Osteoartrite/metabolismo
19.
Acta Biomater ; 146: 274-283, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35487427

RESUMO

Osteoarthritis (OA) is a joint disease affecting millions of patients worldwide. During OA onset and progression, the articular cartilage is destroyed, but the underlying complex mechanisms remain unclear. Here, we uncover changes in the thickness of collagen fibers and their composition at the onset of OA. For articular cartilage explants from knee joints of OA patients, we find that type I collagen-rich fibrocartilage-like tissue was formed in macroscopically intact cartilage, distant from OA lesions. Importantly, the number of thick fibers (>100 nm) has decreased early in the disease, followed by complete absence of thick fibers in advanced OA. We have obtained these results by a combination of high-resolution atomic force microscopy imaging under near-native conditions, immunofluorescence, scanning electron microscopy and a fluorescence-based classification of the superficial chondrocyte spatial organization. Taken together, our data suggests that the loss of tissue functionality in early OA cartilage is caused by a reduction of thick type II collagen fibers, likely due to the formation of type I collagen-rich fibrocartilage, followed by the development of focal defects in later OA stages. We anticipate that such an integrative characterization will be very beneficial for an in-depth understanding of other native biological tissues and the development of sustainable biomaterials. STATEMENT OF SIGNIFICANCE: In early osteoarthritis (OA) the cartilage appears macroscopically intact. However, this study demonstrates that the collagen network already changes in early OA by collagen fiber thinning and the formation of fibrocartilage-like tissue. Both nanoscopic deficiencies already occur in macroscopically intact regions of the human knee joint and are likely connected to processes that result in a weakened extracellular matrix. This study enhances the understanding of earliest progressive cartilage degeneration in the absence of external damage. The results suggest a determination of the mean collagen fiber thickness as a new target for the detection of early OA and a regulation of type I collagen synthesis as a new path for OA treatment.


Assuntos
Cartilagem Articular , Osteoartrite , Cartilagem Articular/patologia , Condrócitos/fisiologia , Colágeno Tipo I , Colágeno Tipo II , Humanos , Osteoartrite/patologia
20.
PLoS One ; 17(2): e0263430, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35139106

RESUMO

BMP7 is a morphogen capable of counteracting the OA chondrocyte hypertrophic phenotype via NKX3-2. NKX3-2 represses expression of RUNX2, an important transcription factor for chondrocyte hypertrophy. Since RUNX2 has previously been described as an inhibitor for 47S pre-rRNA transcription, we hypothesized that BMP7 positively influences 47S pre-rRNA transcription through NKX3-2, resulting in increased protein translational capacity. Therefor SW1353 cells and human primary chondrocytes were exposed to BMP7 and rRNA (18S, 5.8S, 28S) expression was determined by RT-qPCR. NKX3-2 knockdown was achieved via transfection of a NKX3-2-specific siRNA duplex. Translational capacity was assessed by the SUNsET assay, and 47S pre-rRNA transcription was determined by transfection of a 47S gene promoter-reporter plasmid. BMP7 treatment increased protein translational capacity. This was associated by increased 18S and 5.8S rRNA and NKX3-2 mRNA expression, as well as increased 47S gene promotor activity. Knockdown of NKX3-2 led to increased expression of RUNX2, accompanied by decreased 47S gene promotor activity and rRNA expression, an effect BMP7 was unable to restore. Our data demonstrate that BMP7 positively influences protein translation capacity of SW1353 cells and chondrocytes. This is likely caused by an NKX3-2-dependent activation of 47S gene promotor activity. This finding connects morphogen-mediated changes in cellular differentiation to an aspect of ribosome biogenesis via key transcription factors central to determining the chondrocyte phenotype.


Assuntos
Proteína Morfogenética Óssea 7/fisiologia , Condrócitos/metabolismo , Proteínas de Homeodomínio/fisiologia , Biossíntese de Proteínas/genética , RNA Ribossômico/metabolismo , Fatores de Transcrição/fisiologia , Proteína Morfogenética Óssea 7/farmacologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Humanos , Regiões Promotoras Genéticas/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , RNA Ribossômico/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...