Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Bone Res ; 12(1): 7, 2024 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-38311627

RESUMO

Osteoarthritis (OA) is a debilitating degenerative disease affecting multiple joint tissues, including cartilage, bone, synovium, and adipose tissues. OA presents diverse clinical phenotypes and distinct molecular endotypes, including inflammatory, metabolic, mechanical, genetic, and synovial variants. Consequently, innovative technologies are needed to support the development of effective diagnostic and precision therapeutic approaches. Traditional analysis of bulk OA tissue extracts has limitations due to technical constraints, causing challenges in the differentiation between various physiological and pathological phenotypes in joint tissues. This issue has led to standardization difficulties and hindered the success of clinical trials. Gaining insights into the spatial variations of the cellular and molecular structures in OA tissues, encompassing DNA, RNA, metabolites, and proteins, as well as their chemical properties, elemental composition, and mechanical attributes, can contribute to a more comprehensive understanding of the disease subtypes. Spatially resolved biology enables biologists to investigate cells within the context of their tissue microenvironment, providing a more holistic view of cellular function. Recent advances in innovative spatial biology techniques now allow intact tissue sections to be examined using various -omics lenses, such as genomics, transcriptomics, proteomics, and metabolomics, with spatial data. This fusion of approaches provides researchers with critical insights into the molecular composition and functions of the cells and tissues at precise spatial coordinates. Furthermore, advanced imaging techniques, including high-resolution microscopy, hyperspectral imaging, and mass spectrometry imaging, enable the visualization and analysis of the spatial distribution of biomolecules, cells, and tissues. Linking these molecular imaging outputs to conventional tissue histology can facilitate a more comprehensive characterization of disease phenotypes. This review summarizes the recent advancements in the molecular imaging modalities and methodologies for in-depth spatial analysis. It explores their applications, challenges, and potential opportunities in the field of OA. Additionally, this review provides a perspective on the potential research directions for these contemporary approaches that can meet the requirements of clinical diagnoses and the establishment of therapeutic targets for OA.


Assuntos
Osteoartrite , Humanos , Osteoartrite/diagnóstico , Membrana Sinovial/metabolismo , Metabolômica , Fenótipo , Proteômica
2.
Theranostics ; 13(13): 4636-4649, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37649605

RESUMO

Background: Enzymes are central components of many physiological processes, and changes in enzyme activity are linked to numerous disease states, including osteoarthritis (OA). Assessing changes in enzyme function can be challenging because of difficulties in separating affected tissue areas that result in the homogenisation of healthy and diseased cells. Direct correlation between spatially-resolved enzyme distribution(s) and diseased cells/tissues can thus lead to advances in our understanding of OA pathophysiology. Herein, we present a method that uses mass spectrometry imaging (MSI) to visualise the distribution of lipase enzymes and their downstream lipid products in fresh bone and cartilage tissue sections. Immunohistostaining of adjacent tissue sections was then used to identify OA cells/tissues, which were then statistically correlated with molecular-level images. Methods: MSI was used to image lipase enzymes, their substrates, and their metabolic products to validate enzymatic activity and correlate to OA regions determined by immunohistochemistry (IHC). Based on the modified Mankin score, six non-OA and OA patient-matched osteochondral samples were analysed by matrix-assisted laser desorption/ionisation mass spectrometry imaging (MALDI-MSI). Due to the involvement of phospholipase A2 (PLA2) in inflammatory pathways, explant tissues were treated with IL-1ß to mimic inflammation observed in OA. Bovine explant tissues were then subject to MSI methods to observe the spatial distribution of PLA2. Results: Compared with non-OA samples, OA samples showed an elevated level of multiple arachidonic acid (AA)-containing phospholipids (P < 0.001), in which the elevation in the surface and deep layer cartilage of OA tissues is correlated to elevated PLA2 activity (P < 0.001). Bovine explant tissues treated with IL-1ß to mimic OA pathophysiology validated these results and displayed elevated PLA2 levels in OA mimic samples relative to the controls (P < 0.001). It was established that the PLA2G2A isoform specifically was responsible for PLA2 enzyme activity changes in OA tissues (P < 0.001). Conclusion: Our results present a reliable method for imaging enzyme dynamics in OA cartilage, which sets up the foundation for future spatial enzyme dynamics in the OA field. We demonstrated that OA patients exhibit increased expression of PLA2G2A at the superficial and deep cartilage zone that degrades cartilage differently at the spatial level. A tissue-specific PLA2G2A precision inhibition may be the potential target for OA.


Assuntos
Osteoartrite , Humanos , Animais , Bovinos , Osteoartrite/diagnóstico por imagem , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Inflamação , Lipase , Poliésteres
3.
Sci Rep ; 13(1): 10200, 2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37353503

RESUMO

The osteochondral interface is a thin layer that connects hyaline cartilage to subchondral bone. Subcellular elemental distribution can be visualised using synchrotron X-ray fluorescence microscopy (SR-XFM) (1 µm). This study aims to determine the relationship between elemental distribution and osteoarthritis (OA) progression based on disease severity. Using modified Mankin scores, we collected tibia plates from 9 knee OA patients who underwent knee replacement surgery and graded them as intact cartilage (non-OA) or degraded cartilage (OA). We used a tape-assisted system with a silicon nitride sandwich structure to collect fresh-frozen osteochondral sections, and changes in the osteochondral unit were defined using quantified SR-XFM elemental mapping at the Australian synchrotron's XFM beamline. Non-OA osteochondral samples were found to have significantly different zinc (Zn) and calcium (Ca) compositions than OA samples. The tidemark separating noncalcified and calcified cartilage was rich in zinc. Zn levels in OA samples were lower than in non-OA samples (P = 0.0072). In OA samples, the tidemark had less Ca than the calcified cartilage zone and subchondral bone plate (P < 0.0001). The Zn-strontium (Sr) colocalisation index was higher in OA samples than in non-OA samples. The lead, potassium, phosphate, sulphur, and chloride distributions were not significantly different (P > 0.05). In conclusion, SR-XFM analysis revealed spatial elemental distribution at the subcellular level during OA development.


Assuntos
Cartilagem Articular , Osteoartrite do Joelho , Humanos , Cartilagem Articular/diagnóstico por imagem , Síncrotrons , Raios X , Austrália , Osteoartrite do Joelho/diagnóstico por imagem , Progressão da Doença , Zinco , Microscopia de Fluorescência
4.
Aging Dis ; 14(3): 592-604, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37191424

RESUMO

Osteoarthritis is the most prevalent degenerative joint disease and one of the leading causes of physical impairment in the world's aging population. The human lifespan has significantly increased as a result of scientific and technological advancements. According to estimates, the world's elderly population will increase by 20% by 2050. Aging and age-related changes are discussed in this review in relation to the development of OA. We specifically discussed the cellular and molecular changes that occur in the chondrocytes during aging and how these changes may make synovial joints more susceptible to OA development. These changes include chondrocyte senescence, mitochondrial dysfunction, epigenetic modifications, and decreased growth factor response. The age-associated changes occur not only in the chondrocytes but also in the matrix, subchondral bone, and synovium. This review aims to provide an overview of the interplay between chondrocytes and matrix and how age-related changes affect the normal function of cartilage and contribute to OA development. Understanding the alterations that affect the function of chondrocytes will emerge new possibilities for prospective therapeutic options for the treatment of OA.

5.
Mol Pharm ; 20(6): 2966-2977, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-37216314

RESUMO

Coenzyme Q10 is a potent antioxidant that plays an important role in the maintenance of various biochemical pathways of the body and has a wide range of therapeutic applications. However, it has low aqueous solubility and oral bioavailability. Mesoporous silica nanoparticles (MCM-41 and SBA-15 types) exhibiting varying pore sizes and modified with phosphonate and amino groups were used to study the influence of pore structure and surface chemistry on the solubility, in vitro release profile, and intracellular ROS inhibition activity of coenzyme Q10. The particles were thoroughly characterized to confirm the morphology, size, pore profile, functionalization, and drug loading. Surface modification with phosphonate functional groups was found to have the strongest impact on the solubility enhancement of coenzyme Q10 when compared to that of pristine and amino-modified particles. Phosphonate-modified MCM-41 nanoparticles (i.e., MCM-41-PO3) induced significantly higher coenzyme Q10 solubility than the other particles studied. Furthermore, MCM-41-PO3 led to a twofold decrease in ROS generation in human chondrocyte cells (C28/I2), compared to the free drug in a DMSO/DMEM mixture. The results confirmed the significant contribution of small pore size and negative surface charge of MSNs that enable coenzyme Q10 confinement to allow enhanced drug solubility and antioxidant activity.


Assuntos
Antioxidantes , Nanopartículas , Humanos , Solubilidade , Antioxidantes/farmacologia , Espécies Reativas de Oxigênio , Nanopartículas/química , Dióxido de Silício/química , Porosidade , Portadores de Fármacos/química
6.
Cartilage ; : 19476035231169940, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37086004

RESUMO

OBJECTIVE: Chronic inflammation plays an important role in the osteoarthritis (OA) pathology but how this influence OA disease progression is unclear. Leukotriene B4 (LTB4) is a potent proinflammatory lipid mediator generated from arachidonic acid through the sequential activities of 5-lipoxygenase, 5-lipoxygenase-activating protein, Leukotriene A4 hydrolase (LTA4H) and its downstream product LTB4. The aim of this study is to investigate the involvement and the potential therapeutic target of the LTB4 pathway in OA disease progression. DESIGN: Both clinical human cartilage samples (n = 7) and mice experimental OA models (n = 6) were used. The levels of LTA4H and leukotriene B4 receptor 1 were first examined using immunostaining in human OA/non-OA cartilage and mice experimental OA models. We also determined whether the LTA4H pathway was associated with cartilage degeneration and synovitis inflammation in OA mice models and human articular chondrocytes. RESULTS: We found that both LTA4H and LTB4 receptor (BLT1) were highly expressed in human and mice OA cartilage. Inhibition of LTA4H suppressed cartilage degeneration and synovitis in OA mice model. Furthermore, inhibition of LTA4H promoted cartilage regeneration by upregulating chondrogenic genes expression such as aggrecan (ACAN), collagen 2A1 (COL2A1), and SRY-Box transcription factor 9 (SOX9). CONCLUSIONS: Our results indicate that the LTA4H pathway is a crucial regulator of OA pathogenesis and suggest that LTA4H could be a therapeutic target in combat OA.

7.
Cells ; 12(4)2023 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-36831245

RESUMO

Osteoarthritis (OA) remains a prevalent disease affecting more than 20% of the global population, resulting in morbidity and lower quality of life for patients. The study of OA pathophysiology remains predominantly in animal models due to the complexities of mimicking the physiological environment surrounding the joint tissue. Recent development in microfluidic organ-on-chip (OoC) systems have demonstrated various techniques to mimic and modulate tissue physiological environments. Adaptations of these techniques have demonstrated success in capturing a joint tissue's tissue physiology for studying the mechanism of OA. Adapting these techniques and strategies can help create human-specific in vitro models that recapitulate the cellular processes involved in OA. This review aims to comprehensively summarise various demonstrations of microfluidic platforms in mimicking joint microenvironments for future platform design iterations.


Assuntos
Osteoartrite , Qualidade de Vida , Animais , Humanos , Microfluídica/métodos , Modelos Animais
8.
Mol Med ; 28(1): 102, 2022 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-36058911

RESUMO

BACKGROUND: It is well-known that both macrophages and osteocytes are critical regulators of osteogenesis and osteoclastogenesis, yet there is limited understanding of the macrophage-osteocyte interaction, and how their crosstalk could affect bone homeostasis and mineralization. This research therefore aims to investigate the effects of macrophage polarization on osteocyte maturation and mineralization process. METHODS: A macrophage-derived conditioned medium based osteocyte culture was set up to investigate the impact of macrophages on osteocyte maturation and terminal mineralization. Surgically induced osteoarthritis (OA) rat model was used to further investigate the macrophage-osteocyte interaction in inflammatory bone remodeling, as well as the involvement of the Notch signaling pathway in the mineralization process. RESULTS: Our results identified that osteocytes were confined in an immature stage after the M1 macrophage stimulation, showing a more rounded morphology, higher expression of early osteocyte marker E11, and significantly lower expression of mature osteocyte marker DMP1. Immature osteocytes were also found in inflammatory bone remodeling areas, showing altered morphology and mineralized structures similar to those observed under the stimulation of M1 macrophages in vitro, suggesting that M1 macrophages negatively affect osteocyte maturation, leading to abnormal mineralization. The Notch signaling pathway was found to be down regulated in M1 macrophage-stimulated osteocytes as well as osteocytes in inflammatory bone. Overexpression of the Notch signaling pathway in osteocytes showed a significant circumvention on the negative effects from M1 macrophage. CONCLUSION: Taken together, our findings provide valuable insights into the mechanisms involved in abnormal bone mineralization under inflammatory conditions.


Assuntos
Calcinose , Osteócitos , Animais , Calcificação Fisiológica , Calcinose/metabolismo , Macrófagos , Osteócitos/metabolismo , Osteogênese , Ratos , Transdução de Sinais
9.
Histochem Cell Biol ; 158(5): 463-469, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35809120

RESUMO

The anatomy of the osteochondral junction is complex because several tissue components exist as a unit, including uncalcified cartilage (with superficial, middle, and deep layers), calcified cartilage, and subchondral bone. Furthermore, it is difficult to study because this region is made up of a variety of cell types and extracellular matrix compositions. Using X-ray fluorescence microscopy, we present a protocol for simultaneous elemental detection on fresh frozen samples. We transferred the osteochondral sample using a tape-assisted system and successfully tested it in synchrotron X-ray fluorescence. This protocol elucidates the distinct distribution of elements at the human knee's osteochondral junction, making it a useful tool for analyzing the co-distribution of various elements in both healthy and diseased states.


Assuntos
Cartilagem Articular , Humanos , Cartilagem Articular/metabolismo , Secções Congeladas , Osso e Ossos
10.
Front Immunol ; 13: 907750, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35860250

RESUMO

Obesity remains the most important risk factor for the incidence and progression of osteoarthritis (OA). The leading cause of OA was believed to be overloading the joints due to excess weight which in turn leads to the destruction of articular cartilage. However, recent studies have proved otherwise, various other factors like adipose deposition, insulin resistance, and especially the improper coordination of innate and adaptive immune responses may lead to the initiation and progression of obesity-associated OA. It is becoming increasingly evident that multiple inflammatory cells are recruited into the synovial joint that serves an important role in pathological changes in the synovial joint. Polarization of macrophages and macrophage-produced mediators are extensively studied and linked to the inflammatory and destructive responses in the OA synovium and cartilage. However, the role of other major innate immune cells such as neutrophils, eosinophils, and dendritic cells in the pathogenesis of OA has not been fully evaluated. Although cells of the adaptive immune system contribute to the pathogenesis of obesity-induced OA is still under exploration, a quantity of literature indicates OA synovium has an enriched population of T cells and B cells compared with healthy control. The interplay between a variety of immune cells and other cells that reside in the articular joints may constitute a vicious cycle, leading to pathological changes of the articular joint in obese individuals. This review addresses obesity and the role of all the immune cells that are involved in OA and summarised animal studies and human trials and knowledge gaps between the studies have been highlighted. The review also touches base on the interventions currently in clinical trials, different stages of the testing, and their shortcomings are also discussed to understand the future direction which could help in understanding the multifactorial aspects of OA where inflammation has a significant function.


Assuntos
Osteoartrite , Animais , Humanos , Inflamação/patologia , Macrófagos/patologia , Obesidade , Osteoartrite/etiologia , Osteoartrite/patologia , Membrana Sinovial
11.
Aging Dis ; 13(4): 1166-1182, 2022 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-35855332

RESUMO

Articular cartilage function depends on the temporal and zonal distribution of coordinated metabolic regulation in chondrocytes. Emerging evidence shows the importance of cellular metabolism in the molecular control of the cartilage and its dysregulation in degenerative diseases like osteoarthritis (OA). Compared to most other tissues, chondrocytes are sparsely located in the extracellular matrix, lacking the typical proximity of neural, vascular, and lymphatic tissue. Making up under 5% of the total tissue weight of cartilage, chondrocytes have a relative deficiency of access to nutrients and oxygen, as well as limited pathways for metabolite removal. This makes cartilage a unique tissue with hypocellularity, prolonged metabolic rate, and tissue turnover. Studies in the past decade have shown that several pathways of central carbon metabolism are essential for cartilage homeostasis. Here, we summarised the literature findings on the role of cellular metabolism in determining the chondrocyte function and how this metabolic dysregulation led to cartilage aging in OA and provided an outlook on how the field may evolve in the coming years. Although the various energy metabolism pathways are inextricably linked with one another, for the purpose of this review, we initially endeavoured to examine them individually and in relative isolation. Subsequently, we comment on what is known regarding the integration and linked signalling pathways between these systems and the therapeutic opportunities for targeting OA metabolism.

12.
FASEB J ; 36(2): e22142, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35032407

RESUMO

The calcified cartilage zone (CCZ) is a thin interlayer between the hyaline articular cartilage and the subchondral bone and plays an important role in maintaining the joint homeostasis by providing biological and mechanical support from unmineralized cartilage to the underlying mineralized subchondral bone. The hallmark of CCZ characteristics in osteoarthritis (OA) is less well known. The aim of our study is to evaluate the structural, molecular, and biochemical composition of CCZ in tissues affected by primary knee OA and its relationship with disease severity. We collected osteochondral tissue samples stratified according to disease severity, from 16 knee OA patients who underwent knee replacement surgery. We also used meniscectomy-induced rat samples to confirm the pathophysiologic changes of human samples. We defined the characteristics of the calcified cartilage layer using a combination of morphological, biochemical, proteomic analyses on laser micro-dissected tissue. Our results demonstrated that the Calcium/Phosphate ratio is unchanged during the OA progression, but the calcium-binding protein and cadherin binding protein, as well as carbohydrate metabolism-related proteins, undergo significant changes. These changes were further accompanied by thinning of the CCZ, loss of collagen and proteoglycan content, the occurrence of the endochondral ossification, neovasculature, loss of the elastic module, loss of the collagen direction, and increase of the tortuosity indicating an altered structural and mechanical properties of the CCZ in OA. In conclusion, our results suggest that the calcified cartilage changes can reflect the disease progression.


Assuntos
Cartilagem Articular/metabolismo , Osteoartrite/metabolismo , Animais , Osso e Ossos/metabolismo , Calcificação Fisiológica/fisiologia , Progressão da Doença , Feminino , Humanos , Articulação do Joelho/metabolismo , Osteogênese/fisiologia , Proteoglicanas/metabolismo , Proteômica/métodos , Ratos
13.
Mater Sci Eng C Mater Biol Appl ; 128: 112297, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34474848

RESUMO

INTRODUCTION: Peri-implantitis is a bacterially induced inflammatory disease which affects the hard and soft tissues around a dental implant. Microbial biofilm formation is an important causative factor in peri-implantitis. The aim of this study is to develop an effective multifunctional surface coating for antimicrobial property and to counteract oral biofilm-associated infections via a single polydopamine copper coating (PDAM@Cu) on titanium implant surface to regulate endogenous nitric oxide (NO) generation. METHODS: PDAM@Cu coatings were made with different concentrations of CuCl2 on titanium surfaces with a simple dip coating technique. Coatings were characterised to evaluate Cu concentrations as well as NO release rates from the coatings. Further, salivary biofilms were made on the coatings using Brain Heart Infusion (BHI) media in an anaerobic chamber. Biofilms were prepared with three different mixtures, one of which was saliva only, the second had an addition of sheep's blood, and the third was prepared with NO donors S-nitrosoglutathione (GSNO) and L-glutathione (GSH) in the mixture of saliva and blood to evaluate the effects of endogenously produced NO on biofilms. The effectiveness of coated surfaces on biofilms were assessed using four different methods, namely, crystal violet assay, scanning electron microscopy imaging, 2,3-bis (2-methoxy-4-nitro-5-sulfophenyl)-5-[(phenylamino) carbonyl]-2H-tetrazolium hydroxide (XTT) metabolic assay, and live/dead staining. RESULTS: NO release rates could be controlled with different Cu concentration in PDAM@Cu coatings. NO generated from the PDAM@Cu coatings effectively induced dispersal of biofilms shown by the reduction in biofilm biomass as well as reduced biofilm attachment in samples prepared with blood and NO donors. Cu ions released from the PDAM@Cu coatings resulted in killing of the dispersed bacteria, which was evidenced by the live/dead cell staining and reduced metabolic activity noted from the XTT assay. In contrast, samples prepared with saliva showed no significant reduction in biofilms, indicating the important effect of endogenously generated NO on biofilm dispersal. CONCLUSION: In conclusion, PDAM@Cu coatings with NO generating surfaces have a dual anti-biofilm function, with a synergistic effect on biofilm dispersal from regulated NO generation and bactericidal effects from Cu ions from the coatings.


Assuntos
Cobre , Óxido Nítrico , Animais , Biofilmes , Materiais Revestidos Biocompatíveis/farmacologia , Cobre/farmacologia , Indóis , Polímeros , Ovinos , Propriedades de Superfície , Titânio/farmacologia
14.
Front Cell Dev Biol ; 9: 659654, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34041240

RESUMO

Osteoarthritis (OA) is a long-term condition that causes joint pain and reduced movement. Notably, the same pathways governing cell growth, death, and differentiation during the growth and development of the body are also common drivers of OA. The osteochondral interface is a vital structure located between hyaline cartilage and subchondral bone. It plays a critical role in maintaining the physical and biological function, conveying joint mechanical stress, maintaining chondral microenvironment, as well as crosstalk and substance exchange through the osteochondral unit. In this review, we summarized the progress in research concerning the area of osteochondral junction, including its pathophysiological changes, molecular interactions, and signaling pathways that are related to the ultrastructure change. Multiple potential treatment options were also discussed in this review. A thorough understanding of these biological changes and molecular mechanisms in the pathologic process will advance our understanding of OA progression, and inform the development of effective therapeutics targeting OA.

15.
Front Med (Lausanne) ; 8: 628843, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33829022

RESUMO

Obesogenic diets contribute to the pathology of osteoarthritis (OA) by altering systemic and local metabolic inflammation. Yet, it remains unclear how quickly and reproducibly the body responds to weight loss strategies and improve OA. In this study we tested whether switching obese diet to a normal chow diet can mitigate the detrimental effects of inflammatory pathways that contribute to OA pathology. Male C57BL/6 mice were first fed with obesogenic diet (high fat diet) and switched to normal chow diet (obese diet → normal diet) or continued obese diet or normal diet throughout the experiment. A mouse model of OA was induced by surgical destabilization of the medial meniscus (DMM) model into the knee joint. Outcome measures included changes in metabolic factors such as glucose, insulin, lipid, and serum cytokines levels. Inflammation in synovial biopsies was scored and inflammation was determined using FACs sorted macrophages. Cartilage degeneration was monitored using histopathology. Our results indicate, dietary switching (obese diet → normal diet) reduced body weight and restored metabolic parameters and showed less synovial tissue inflammation. Systemic blood concentrations of pro-inflammatory cytokines IL-1α, IL-6, IL-12p40, and IL-17 were decreased, and anti-inflammatory cytokines IL-4 and IL-13 were increased in dietary switch group compared to mice that were fed with obesogenic diet continuously. Although obese diet worsens the cartilage degeneration in DMM OA model, weight loss induced by dietary switch does not promote the histopathological changes of OA during this study period. Collectively, these data demonstrate that switching obesogenic diet to normal improved metabolic syndrome symptoms and can modulate both systemic and synovium inflammation levels.

16.
Cells ; 10(2)2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33525381

RESUMO

Altered subchondral bone and articular cartilage interactions have been implicated in the pathogenesis of osteoarthritis (OA); however, the mechanisms remain unknown. Exosomes are membrane-derived vesicles that have recently been recognized as important mediators of intercellular communication. Herein, we investigated if OA subchondral bone derived exosomes alter transcriptional and bioenergetic signatures of chondrocytes. Exosomes were isolated and purified from osteoblasts of nonsclerotic or sclerotic zones of human OA subchondral bone and their role on the articular cartilage chondrocytes was evaluated by measuring the extent of extracellular matrix production, cellular bioenergetics, and the expression of chondrocyte activity associated marker genes. Exosomal microRNAs were analyzed using RNA sequencing and validated by quantitative real-time PCR and loss-of-function. In coculture studies, chondrocytes internalized OA sclerotic subchondral bone osteoblast derived exosomes and triggered catabolic gene expression and reduced chondrocyte-specific marker expression a phenomenon that is often observed in OA cartilage. RNA sequencing and miRNA profiling have identified miR-210-5p, which is highly enriched in OA sclerotic subchondral bone osteoblast exosomes, triggered the catabolic gene expression in articular cartilage chondrocytes. Importantly, we demonstrate that miR-210-5p suppresses the oxygen consumption rate of chondrocytes, altering their bioenergetic state that is often observed in OA conditions. These effects were markedly inhibited by the addition of a miR-210-5p inhibitor. Our study indicates that exosomes released by OA sclerotic subchondral bone osteoblasts plays a critical role in progression of cartilage degeneration and might be a potential target for therapeutic intervention in OA.


Assuntos
Osso e Ossos/patologia , Cartilagem Articular/patologia , Condrócitos/patologia , Exossomos/metabolismo , Osteoartrite/patologia , Aerobiose , Idoso de 80 Anos ou mais , Comunicação Celular/genética , Respiração Celular/genética , Condrócitos/metabolismo , Exossomos/ultraestrutura , Feminino , Regulação da Expressão Gênica , Humanos , Hipertrofia , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Osteoartrite/genética , Osteoblastos/patologia , Fenótipo
17.
ACS Appl Bio Mater ; 4(3): 2490-2501, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35014367

RESUMO

Bone and bone-related diseases are the major cause of mobility hindrance and mortality in humans and there is no effective and safe treatment for most of them, especially, for bone and bone metastatic cancers. Bisphosphonates (BPs) are a group of small-molecule drugs for treating osteoporosis and bone cancers but have a very short half-life in circulation, requiring high doses and long-term repeat use that can cause severe side effects. Previous attempts of using nanoparticles to deliver BPs have issues of drug loading capacity and endosome escape/drug release. The present study reports the direct synthesis of BP nanoparticles by precipitating bone-favorable calcium ions and a third-generation BP, risedronate (Ca-RISNPs), to achieve high drug loading, endosomal release, and strong bone-targeting properties. The Ca-RISNPs are monodispersed with high stability at physiological pH but readily dissociate at endosomal pH conditions. They demonstrate strong penetration ability and uniform distribution in human bone and cartilage tissues and the superior drug and DNA (plasmid and oligo double strand DNA) delivery capacity in bone cells. These NPs also exhibit high specificity in killing tumor-associated macrophages (TAMs) and inhibit TAM-induced tumor cell migration. Collectively, our data indicate that this BP nanodrug platform has a great potential in managing bone-related diseases and cancers as a prolonged BP nanodrug and simultaneously as the bone-targeted drug delivery system.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Materiais Biocompatíveis/química , Doenças Ósseas/tratamento farmacológico , Neoplasias Ósseas/tratamento farmacológico , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos , Animais , Antibióticos Antineoplásicos/química , Doenças Ósseas/patologia , Neoplasias Ósseas/patologia , Cálcio/química , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Difosfonatos/química , Doxorrubicina/química , Portadores de Fármacos/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Teste de Materiais , Camundongos , Estrutura Molecular , Nanopartículas/química , Tamanho da Partícula , Células RAW 264.7
18.
J Drug Target ; 29(6): 609-616, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33356642

RESUMO

Osteoarthritis (OA)is a common orthopaedic disease in middle-aged and aged people. To date, no disease-modifying drug is available to prevent the progression of OA. Surgical treatment of OA has complications such as pain and high costs with increased risk of post-operative infections. An intra-articular drug delivery is a conservative treatment method to apply therapeutic composites directly into the OA joint cavity. This method has an advantage to improve the bioavailability of therapeutics and hence is a widely preferred choice to test novel disease-modifying drug targets for OA. Herein, we summarised and discussed the current status of intra-articular therapy for OA treatment as well as outlined drug delivery of small molecular, protein and gene delivery for OA therapy. Currently, new targeted nano-based drug delivery systems, including nanoparticles, exosomes and hydrogel formulations under investigation for OA treatment via intra-articular injection are also addressed. The emerging trend demonstrates that intra-articular drug delivery has vast prospects for the clinical selective treatment of OA. The rational application of intra-articular injection of drugs and biological agents will be of great significance for alleviating the patients with OA, improving their quality of life, delaying surgery, and reducing the disease burden of OA.


Assuntos
Sistemas de Liberação de Medicamentos , Osteoartrite/tratamento farmacológico , Idoso , Animais , Disponibilidade Biológica , Exossomos , Terapia Genética/métodos , Humanos , Hidrogéis , Injeções Intra-Articulares , Pessoa de Meia-Idade , Nanopartículas , Osteoartrite/fisiopatologia , Qualidade de Vida
19.
J Orthop Translat ; 21: 73-80, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32099807

RESUMO

Osteoarthritis (OA) is a prevalent whole joint disease characterised by cartilage degradation, subchondral bone sclerosis and bone remodelling, and synovium inflammation, leading to pain, deformity, and cartilage dysfunction. Currently, there is no appropriate therapy for OA, and available treatments simply aim to reduce pain and swelling. Exosomes are membrane-bound extracellular vesicles secreted by almost all cells, receiving increasing interest because of their effect in cell-to-cell communication. Increasing evidence suggests that exosomes play an important role in cartilage physiological and pathological effects. This article reviews the potential role of exosomes in OA regenerative medicine. Special attention is given to mesenchymal stem cells-derived exosomes due to the extensive research on their cartilage repair property and their function as miRNA cargo. More investigations are needed for the effects of exosomes from synovial fluid and chondrocytes in joints. A better understanding of the mechanisms will contribute to a novel and promising therapy for OA patients. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: A better understanding of the role of extracellular vesicles in regenerative medicine will contribute to a novel and promising therapy for OA patients.

20.
Nutrients ; 12(2)2020 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-32085385

RESUMO

Osteoarthritis (OA) is a degenerative condition of joints, causing pain and swelling, and can be caused or worsened by trauma and obesity. The objectives of this study were to determine whether pain behaviour and progression of OA were increased in rats with trauma-induced OA fed dietary saturated fatty acids (SFA). Male Wistar rats were fed either a corn starch diet (C) or high-carbohydrate high-fat diet (H) with either 20% beef tallow or SFA (lauric (HLA), myristic (HMA), palmitic (HPA) or stearic (HSA) acids) for 16 weeks prior to and 8 weeks after excision of the medial meniscus of right knee joint to initiate OA when pain behaviour, glial activity, progression of knee OA, inflammatory mediators and signs of metabolic syndrome were assessed. Rats fed beef tallow, palmitic or stearic acids showed increased pain symptoms characterised by decreased hind paw/limb withdrawal thresholds and grip strengths and increased spinal astrogliosis and microgliosis compared to rats fed lauric or myristic acids. However, the severity of OA joint damage was unchanged by these dietary manipulations. We conclude that pain symptoms of trauma-induced OA in rats worsen with increased dietary beef tallow or palmitic or stearic acids, but improve with lauric or myristic acids, despite unchanged OA cartilage damage.


Assuntos
Gorduras na Dieta/efeitos adversos , Gorduras/efeitos adversos , Ácidos Graxos/efeitos adversos , Traumatismos do Joelho/complicações , Osteoartrite do Joelho/etiologia , Dor/dietoterapia , Dor/etiologia , Animais , Dieta Hiperlipídica , Carboidratos da Dieta , Progressão da Doença , Ácidos Graxos/administração & dosagem , Ácidos Láuricos/administração & dosagem , Masculino , Ácido Mirístico/administração & dosagem , Ácido Palmítico/efeitos adversos , Ratos Wistar , Ácidos Esteáricos/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA