Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bioact Mater ; 39: 255-272, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38832304

RESUMO

Osteoarthritis (OA) is a major clinical challenge, and effective disease-modifying drugs for OA are still lacking due to the complicated pathology and scattered treatment targets. Effective early treatments are urgently needed to prevent OA progression. The excessive amount of transforming growth factor ß (TGFß) is one of the major causes of synovial fibrosis and subchondral bone sclerosis, and such pathogenic changes in early OA precede cartilage damage. Herein we report a novel strategy of intra-articular sustained-release of pirfenidone (PFD), a clinically-approved TGFß inhibitor, to achieve disease-modifying effects on early OA joints. We found that PFD effectively restored the mineralization in the presence of excessive amount of TGFß1 (as those levels found in patients' synovial fluid). A monthly injection strategy was then designed of using poly lactic-co-glycolic acid (PLGA) microparticles and hyaluronic acid (HA) solution to enable a sustained release of PFD (the "PLGA-PFD + HA" strategy). This strategy effectively regulated OA progression in destabilization of the medial meniscus (DMM)- induced OA mice model, including preventing subchondral bone loss in early OA and subchondral bone sclerosis in late OA, and reduced synovitis and pain with cartilage preservation effects. This finding suggests the promising clinical application of PFD as a novel disease-modifying OA drug.

2.
Sci Bull (Beijing) ; 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38760248

RESUMO

Mechanical loading is required for bone homeostasis, but the underlying mechanism is still unclear. Our previous studies revealed that the mechanical protein polycystin-1 (PC1, encoded by Pkd1) is critical for bone formation. However, the role of PC1 in bone resorption is unknown. Here, we found that PC1 directly regulates osteoclastogenesis and bone resorption. The conditional deletion of Pkd1 in the osteoclast lineage resulted in a reduced number of osteoclasts, decreased bone resorption, and increased bone mass. A cohort study of 32,500 patients further revealed that autosomal dominant polycystic kidney disease, which is mainly caused by loss-of-function mutation of the PKD1 gene, is associated with a lower risk of hip fracture than those with other chronic kidney diseases. Moreover, mice with osteoclast-specific knockout of Pkd1 showed complete resistance to unloading-induced bone loss. A mechanistic study revealed that PC1 facilitated TAZ nuclear translocation via the C-terminal tail-TAZ complex and that conditional deletion of Taz in the osteoclast lineage resulted in reduced osteoclastogenesis and increased bone mass. Pharmacological regulation of the PC1-TAZ axis alleviated unloading- and estrogen deficiency- induced bone loss. Thus, the PC1-TAZ axis may be a potential therapeutic target for osteoclast-related osteoporosis.

3.
Bone Res ; 12(1): 6, 2024 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-38267422

RESUMO

Skeletal stem/progenitor cell (SSPC) senescence is a major cause of decreased bone regenerative potential with aging, but the causes of SSPC senescence remain unclear. In this study, we revealed that macrophages in calluses secrete prosenescent factors, including grancalcin (GCA), during aging, which triggers SSPC senescence and impairs fracture healing. Local injection of human rGCA in young mice induced SSPC senescence and delayed fracture repair. Genetic deletion of Gca in monocytes/macrophages was sufficient to rejuvenate fracture repair in aged mice and alleviate SSPC senescence. Mechanistically, GCA binds to the plexin-B2 receptor and activates Arg2-mediated mitochondrial dysfunction, resulting in cellular senescence. Depletion of Plxnb2 in SSPCs impaired fracture healing. Administration of GCA-neutralizing antibody enhanced fracture healing in aged mice. Thus, our study revealed that senescent macrophages within calluses secrete GCA to trigger SSPC secondary senescence, and GCA neutralization represents a promising therapy for nonunion or delayed union in elderly individuals.


Assuntos
Calosidades , Fraturas Ósseas , Idoso , Humanos , Animais , Camundongos , Consolidação da Fratura , Senescência Celular , Envelhecimento , Macrófagos , Células-Tronco
4.
PLoS One ; 18(10): e0286626, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37797053

RESUMO

Quantification of the histological staining images gives important insights in biomedical research. In wet lab, it is common to have some stains off the target to become unwanted noisy stains during the generation of histological staining images. The current tools designed for quantification of histological staining images do not consider such situations; instead, the stained region is identified based on assumptions that the background is pure and clean. The goal of this study is to develop a light software named Staining Quantification (SQ) tool which could handle the image quantification job with features for removing a large amount of unwanted stains blended or overlaid with Region of Interest (ROI) in complex scenarios. The core algorithm was based on the method of higher order statistics transformation, and local density filtering. Compared with two state-of-art thresholding methods (i.e. Otsu's method and Triclass thresholding method), the SQ tool outperformed in situations such as (1) images with weak positive signals and experimental caused dirty stains; (2) images with experimental counterstaining by multiple colors; (3) complicated histological structure of target tissues. The algorithm was developed in R4.0.2 with over a thousand in-house histological images containing Alizarin Red (AR) and Von Kossa (VK) staining, and was validated using external images. For the measurements of area and intensity in total and stained region, the average mean of difference in percentage between SQ and ImageJ were all less than 0.05. Using this as a criterion of successful image recognition, the success rate for all measurements in AR, VK and external validation batch were above 0.8. The test of Pearson's coefficient, difference between SQ and ImageJ, and difference of proportions between SQ and ImageJ were all significant at level of 0.05. Our results indicated that the SQ tool is well established for automatic histological staining image quantification.


Assuntos
Processamento de Imagem Assistida por Computador , Software , Processamento de Imagem Assistida por Computador/métodos , Algoritmos , Corantes , Coloração pela Prata
5.
Stem Cell Res Ther ; 14(1): 160, 2023 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-37316923

RESUMO

BACKGROUND: Autologous chondrocyte implantation (ACI) has been used to treat articular cartilage defects for over two decades. Adult stem cells have been proposed as a solution to inadequate donor cell numbers often encountered in ACI. Multipotent stem/progenitor cells isolated from adipose, bone marrow, and cartilage are the most promising cell therapy candidates. However, different essential growth factors are required to induce these tissue-specific stem cells to initiate chondrogenic differentiation and subsequent deposition of extracellular matrix (ECM) to form cartilage-like tissue. Upon transplantation into cartilage defects in vivo, the levels of growth factors in the host tissue are likely to be inadequate to support chondrogenesis of these cells in situ. The contribution of stem/progenitor cells to cartilage repair and the quality of ECM produced by the implanted cells required for cartilage repair remain largely unknown. Here, we evaluated the bioactivity and chondrogenic induction ability of the ECM produced by different adult stem cells. METHODS: Adult stem/progenitor cells were isolated from human adipose (hADSCs), bone marrow (hBMSCs), and articular cartilage (hCDPCs) and cultured for 14 days in monolayer in mesenchymal stromal cell (MSC)-ECM induction medium to allow matrix deposition and cell sheet formation. The cell sheets were then decellularized, and the protein composition of the decellularized ECM (dECM) was analyzed by BCA assay, SDS-PAGE, and immunoblotting for fibronectin (FN), collagen types I (COL1) and III (COL3). The chondrogenic induction ability of the dECM was examined by seeding undifferentiated hBMSCs onto the respective freeze-dried solid dECM followed by culturing in serum-free medium for 7 days. The expression levels of chondrogenic genes SOX9, COL2, AGN, and CD44 were analyzed by q-PCR. RESULTS: hADSCs, hBMSCs, and hCDPCs generated different ECM protein profiles and exhibited significantly different chondrogenic effects. hADSCs produced 20-60% more proteins than hBMSCs and hCDPCs and showed a fibrillar-like ECM pattern (FNhigh, COL1high). hCDPCs produced more COL3 and deposited less FN and COL1 than the other cell types. The dECM derived from hBMSCs and hCDPCs induced spontaneous chondrogenic gene expression in hBMSCs. CONCLUSIONS: These findings provide new insights on application of adult stem cells and stem cell-derived ECM to enhance cartilage regeneration.


Assuntos
Células-Tronco Adultas , Condrogênese , Adulto , Humanos , Matriz Extracelular , Células-Tronco , Colágeno Tipo I
6.
Bioact Mater ; 25: 256-272, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36825224

RESUMO

Meniscus, the cushion in knee joint, is a load-bearing tissue that transfers mechanical forces to extracellular matrix (ECM) and tissue resident cells. The mechanoresponse of human tissue resident stem/progenitor cells in meniscus (hMeSPCs) is significant to tissue homeostasis and regeneration but is not well understood. This study reports that a mild cyclic tensile loading regimen of ∼1800 loads/day on hMeSPCs seeded in 3-dimensional (3D) photocrosslinked gelatin methacryloyl (GelMA) hydrogel is critical in maintaining cellular homeostasis. Experimentally, a "slow walk" biomimetic cyclic loading regimen (10% tensile strain, 0.5 Hz, 1 h/day, up to 15 days) is applied to hMeSPCs encapsulated in GelMA hydrogel with a magnetic force-controlled loading actuator. The loading significantly increases cell differentiation and fibrocartilage-like ECM deposition without affecting cell viability. Transcriptomic analysis reveals 332 mechanoresponsive genes, clustered into cell senescence, mechanical sensitivity, and ECM dynamics, associated with interleukins, integrins, and collagens/matrix metalloproteinase pathways. The cell-GelMA constructs show active ECM remodeling, traced using a green fluorescence tagged (GFT)-GelMA hydrogel. Loading enhances nascent pericellular matrix production by the encapsulated hMeSPCs, which gradually compensates for the hydrogel loss in the cultures. These findings demonstrate the strong tissue-forming ability of hMeSPCs, and the importance of mechanical factors in maintaining meniscus homeostasis.

7.
Am J Sports Med ; 51(2): 461-475, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36645043

RESUMO

BACKGROUND: Doxycycline (Doxy) has been shown to facilitate tendon healing by reducing on-site matrix metalloproteinase (MMP) activity, but its effect on graft healing after anterior cruciate ligament reconstruction (ACLR) has not been investigated, and the therapeutic effect of Doxy in preventing ACLR-induced posttraumatic osteoarthritis (PTOA) is unclear. HYPOTHESIS: Doxy promotes graft healing and alleviates the progression of PTOA after ACLR. STUDY DESIGN: Controlled laboratory study. METHODS: Sprague Dawley rats (n = 74; age, 12-13 weeks; male) that underwent ACLR were divided into untreated control and Doxy treatment (50 mg/kg/d orally until sacrifice) groups. At 2 and 6 weeks after surgery, graft healing was assessed by biomechanical testing, histology, immunohistochemical staining, and micro-computed tomography (µCT). The progression of PTOA was evaluated at 6 weeks by histology, the Mankin score, and immunofluorescence staining of the tibial plateau, and osteophyte formation was evaluated by µCT. Hindlimb weight distribution was evaluated at 6 weeks, and gait patterns were evaluated at 2 and 6 weeks. Intra-articular MMP activity was evaluated at 6 weeks in vivo using an MMP-activatable near-infrared fluorescent probe. RESULTS: Graft healing was enhanced by Doxy treatment, and the ultimate failure load (P = .002) and stiffness of the graft (P = .007) were significantly higher in the Doxy group at week 2. Bone mineral density and bone volume/total volume for both the tibial and the femoral tunnels at week 6 in the Doxy group were significantly higher compared with in the control group (P < .05). The overall graft healing scores were significantly higher in the Doxy group. Doxy treatment enhanced graft integration, intratunnel graft integrity, and collagen birefringence; more collagen types 1 and 10 and less MMP-13 were found at the graft-bone interface. At week 6, the Doxy group had a lower modified Mankin score (P = .033) and showed fewer MMP 13-positive chondrocytes at the articular cartilage surface (P = .002), indicating moderate joint cartilage damage. µCT revealed less osteophyte formation, and gait analysis revealed more symmetric weightbearing and gait patterns, after Doxy treatment at week 6 (P < .05). In vivo imaging with the near-infrared fluorescent probe identified significantly lower intra-articular MMP activity in the Doxy group at week 6 (P = .016). CONCLUSION: The oral administration of Doxy was able to synchronously promote graft healing and attenuate PTOA in an ACLR rat model. CLINICAL RELEVANCE: Our results indicated that Doxy, a widely used drug, is potentially beneficial to patients after ACLR.


Assuntos
Lesões do Ligamento Cruzado Anterior , Reconstrução do Ligamento Cruzado Anterior , Osteoartrite , Osteófito , Ratos , Masculino , Animais , Doxiciclina , Ratos Sprague-Dawley , Microtomografia por Raio-X , Corantes Fluorescentes , Osteoartrite/cirurgia , Colágeno , Reconstrução do Ligamento Cruzado Anterior/métodos , Lesões do Ligamento Cruzado Anterior/cirurgia
9.
Cell Death Dis ; 13(5): 494, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35610206

RESUMO

A specific bone capillary subtype, namely type H vessels, with high expression of CD31 and endomucin, was shown to couple angiogenesis and osteogenesis recently. The number of type H vessels in bone tissue declines with age, and the underlying mechanism for this reduction is unclear. Here, we report that microRNA-188-3p (miR-188-3p) involves this process. miRNA-188-3p expression is upregulated in skeletal endothelium and negatively regulates the formation of type H vessels during ageing. Mice with depletion of miR-188 showed an alleviated age-related decline in type H vessels. In contrast, endothelial-specific overexpression of miR-188-3p reduced the number of type H vessels, leading to decreased bone mass and delayed bone regeneration. Mechanistically, we found that miR-188 inhibits type H vessel formation by directly targeting integrin ß3 in endothelial cells. Our findings indicate that miR-188-3p is a key regulator of type H vessel formation and may be a potential therapeutic target for preventing bone loss and accelerating bone regeneration.


Assuntos
MicroRNAs , Osteogênese , Envelhecimento/genética , Animais , Células Endoteliais/metabolismo , Endotélio , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Neovascularização Patológica , Osteogênese/genética
10.
J Cell Physiol ; 237(4): 2258-2270, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35147979

RESUMO

Immune regulation of osteochondral defect regeneration has not yet been rigorously characterized. Although macrophages have been demonstrated to regulate the regeneration process in various tissues, their direct contribution to cartilage regeneration remains to be investigated, particularly the functions of polarized macrophage subpopulations. In this study, we investigated the origins and functions of macrophages during healing of osteochondral injury in the murine model. Upon osteochondral injury, joint macrophages are predominantly derived from circulating monocytes. Macrophages are essential for spontaneous cartilage regeneration in juvenile C57BL/6 mice, by modulating proliferation and apoptosis around the injury site. Exogeneous macrophages also exhibit therapeutic potential in promoting cartilage regeneration in adult mice with poor regenerative capacity, possibly via regulation of PDGFRα+  stem cells, with this process being influenced by initial phenotype and administration timing. Only M2c macrophages are able to promote regeneration of both cartilage tissues and subchondral bone. Overall, we reveal the direct link between macrophages and osteochondral regeneration and highlight the key roles of relevant immunological niches in successful regeneration.


Assuntos
Cartilagem Articular , Macrófagos/fisiologia , Cicatrização , Animais , Cartilagem Articular/citologia , Cartilagem Articular/lesões , Cartilagem Articular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
11.
Sci China Life Sci ; 65(5): 927-939, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34586575

RESUMO

Mesenchymal stem cells (MSCs) are important cell sources in cartilage tissue development and homeostasis, and multiple strategies have been developed to improve MSCs chondrogenic differentiation with an aim of promoting cartilage regeneration. Here we report the effects of combining nanosecond pulsed electric fields (nsPEFs) followed by treatment with ghrelin (a hormone that stimulates release of growth hormone) to regulate chondrogenesis of MSCs. nsPEFs and ghrelin were observed to separately enhance the chondrogenesis of MSCs, and the effects were significantly enhanced when the bioelectric stimulation and hormone were combined, which in turn improved osteochondral tissue repair of these cells within Sprague Dawley rats. We further found that nsPEFs can prime MSCs to be more receptive to subsequent stimuli of differentiation by upregulated Oct4/Nanog and activated JNK signaling pathway. Ghrelin initiated chondrogenic differentiation by activation of ERK1/2 signaling pathway, and RNA-seq results indicated 243 genes were regulated, and JAK-STAT signaling pathway was involved. Interestingly, the sequential order of applying these two stimuli is critical, with nsPEFs pretreatment followed by ghrelin enhanced chondrogenesis of MSCs in vitro and subsequent cartilage regeneration in vivo, but not vice versa. This synergistic prochondrogenic effects provide us new insights and strategies for future cell-based therapies.


Assuntos
Condrogênese , Células-Tronco Mesenquimais , Animais , Diferenciação Celular , Células Cultivadas , Grelina/metabolismo , Grelina/farmacologia , Ratos , Ratos Sprague-Dawley
12.
Arthritis Res Ther ; 23(1): 110, 2021 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-33838669

RESUMO

OBJECTIVE: Osteoarthritis (OA) has long been regarded as a disease of cartilage degeneration, whereas mounting evidence implies that low-grade inflammation contributes to OA. Among inflammatory cells involved, macrophages play a crucial role and are mediated by the local microenvironment to exhibit different phenotypes and polarization states. Therefore, we conducted a systematic review to uncover the phenotypic alterations of macrophages during OA and summarized the potential therapeutic interventions via modulating macrophages. METHODS: A systematic review of multiple databases (PubMed, Web of Science, ScienceDirect, Medline) was performed up to February 29, 2020. Included articles were discussed and evaluated by two independent reviewers. Relevant information was analyzed with a standardized and well-designed template. RESULTS: A total of 28 studies were included. Results were subcategorized into two sections depending on sources from human tissue/cell-based studies (12 studies) and animal experiments (16 studies). The overall observation indicated that M1 macrophages elevated in both synovium and circulation during OA development, along with lower numbers of M2 macrophages. The detailed alterations of macrophages in both synovium and circulation were listed and analyzed. Furthermore, interventions against OA via regulating macrophages in animal models were highlighted. CONCLUSION: This study emphasized the importance of the phenotypic alterations of macrophages in OA development. The classical phenotypic subcategory of M1 and M2 macrophages was questionable due to controversial and conflicting results. Therefore, further efforts are needed to categorize macrophages in an exhaustive manner and to use advanced technologies to identify the individual roles of each subtype of macrophages in OA.


Assuntos
Osteoartrite , Animais , Humanos , Inflamação , Macrófagos , Fenótipo , Membrana Sinovial
14.
Stem Cell Res Ther ; 11(1): 308, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32698858

RESUMO

BACKGROUND: Multiple strategies have been proposed to promote the differentiation potential of mesenchymal stem cells (MSCs), which is the fundamental property in tissue formation and regeneration. However, these strategies are relatively inefficient that limit the application. In this study, we reported a novel and efficient strategy, nanosecond pulsed electric fields (nsPEFs) stimulation, which can enhance the trilineage differentiation potential of MSCs, and further explained the mechanism behind. METHODS: We used histological staining to screen out the nsPEFs parameters that promoted the trilineage differentiation potential of MSCs, and further proved the effect of nsPEFs by detecting the functional genes. In order to explore the corresponding mechanism, we examined the expression of pluripotency genes and the methylation status of their promoters. Finally, we targeted the DNA methyltransferase which was affected by nsPEFs. RESULTS: The trilineage differentiation of bone marrow-derived MSCs was significantly enhanced in vitro by simply pre-treating with 5 pulses of nsPEFs stimulation (energy levels as 10 ns, 20 kV/cm; 100 ns, 10 kV/cm), due to that the nsPEFs demethylated the promoters of stem cell pluripotency genes OCT4 and NANOG through instantaneous downregulation of DNA methylation transferase 1 (DNMT1), thereby increasing the expression of OCT4 and NANOG for up to 3 days, and created a treatment window period of stem cells. CONCLUSIONS: In summary, nsPEFs can enhance MSCs differentiation via the epigenetic regulation and could be a safe and effective strategy for future stem cell application.


Assuntos
Células-Tronco Mesenquimais , Diferenciação Celular , Metilação de DNA , Epigênese Genética , Expressão Gênica , Transferases
15.
J Tissue Eng Regen Med ; 14(8): 1136-1148, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32592324

RESUMO

Mesenchymal stem cells (MSCs) gradually lose multipotency when cultured for prolonged durations in vitro, which significantly hinders subsequent clinical applications. Nanosecond pulsed electric fields (nsPEFs) have been recently investigated to overcome this problem in our lab; however, the differentiation potency of MSCs could only be partially and transiently recovered because the nsPEFs can only be delivered to suspended cells once. Here, we develop a new strategy to apply multiple nsPEFs to adherent MSCs with conductive films to mitigate the decreasing multipotency of prolonged cultured MSCs. The poly(l-lactic acid)/graphitized-carboxylated functionalized carbon nanotubes (PLLA/CNT) films were fabricated as conductive cell culture platforms. Both single and multiple nsPEFs stimulation could significantly increase the differentiation potential of MSCs, as evidenced by upregulated expression of chondrogenic, osteogenic, and adipogenic-related gene (SOX9, RUNX2, and PPAR-γ), as well as increased production of proteoglycans, mineralized calcium, and triglycerides. Multiple nsPEFs stimulation demonstrated significant efficacy in upregulating expression of pluripotency genes of OCT4A (3.5- to 4.5-folds), NANOG (3.5- to 4.0-folds), and SOX2 (1.5- to 2.0-folds) and stably maintaining high expression of these genes for nearly 23 days. Notably, nsPEFs stimulation did not significantly shorten telomere length. In conclusion, multiple nsPEFs stimulation could effectively mitigate decreasing multipotency of MSCs during prolonged in vitro culture.


Assuntos
Eletricidade , Membranas Artificiais , Células-Tronco Mesenquimais/metabolismo , Nanotubos de Carbono/química , Poliésteres/química , Animais , Técnicas de Cultura de Células , Células Cultivadas , Masculino , Células-Tronco Mesenquimais/citologia , Suínos
16.
Int J Biol Sci ; 16(11): 1811-1820, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32398951

RESUMO

Articular cartilage injury is a common clinical problem, which can lead to joint dysfunction, significant pain, and secondary osteoarthritis (OA) in which major surgical procedures are mandatory for treatment. Exosomes, as endosome-derived membrane-bound vesicles, participating in intercellular communications in both physiological and pathophysiological conditions, have been attached great importance in many fields. Recently, the significance of exosomes in the development of OA has been gradually concerned, while the therapeutic value of exosomes in cartilage repair and OA treatment has also been gradually revealed. The functional difference of different types and derivations of exosomes are determined by their specific contents. Herein, we provide comprehensive understanding on exosome and OA, including how exosomes participating in OA, the therapeutic value of exosomes for cartilage injury/OA, and related bioengineering strategies for future therapeutic design.


Assuntos
Cartilagem Articular/lesões , Exossomos , Osteoartrite/patologia , Osteoartrite/terapia , Animais , Técnicas de Cultura de Células , Humanos , Células-Tronco Mesenquimais
17.
Front Cell Dev Biol ; 8: 607764, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33553146

RESUMO

There is emerging awareness that subchondral bone remodeling plays an important role in the development of osteoarthritis (OA). This review presents recent investigations on the cellular and molecular mechanism of subchondral bone remodeling, and summarizes the current interventions and potential therapeutic targets related to OA subchondral bone remodeling. The first part of this review covers key cells and molecular mediators involved in subchondral bone remodeling (osteoclasts, osteoblasts, osteocytes, bone extracellular matrix, vascularization, nerve innervation, and related signaling pathways). The second part of this review describes candidate treatments for OA subchondral bone remodeling, including the use of bone-acting reagents and the application of regenerative therapies. Currently available clinical OA therapies and known responses in subchondral bone remodeling are summarized as a basis for the investigation of potential therapeutic mediators.

18.
Artigo em Inglês | MEDLINE | ID: mdl-31850331

RESUMO

Induced pluripotent stem cells (iPSCs) are reprogrammed somatic cells by defined factors, and have great application potentials in tissue regeneration and disease modeling. Biomaterials have been widely used in stem cell-based studies, and are involved in human iPSCs based studies, but they were not enough emphasized and recognized. Biomaterials can mimic the extracellular matrix and microenvironment, and act as powerful tools to promote iPSCs proliferation, differentiation, maturation, and migration. Many classic and advanced biofabrication technologies, such as cell-sheet approach, electrospinning, and 3D-bioprinting, are used to provide physical cues in macro-/micro-patterning, and in combination with other biological factors to support iPSCs applications. In this review, we highlight the biomaterials and fabrication technologies used in human iPSC-based tissue engineering to model neuromyopathic diseases, particularly those with genetic mutations, such as Duchenne Muscular Dystrophy (DMD), Congenital Heart Diseases (CHD) and Alzheimer's disease (AD).

19.
J Orthop Translat ; 18: 142-153, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31508317

RESUMO

BACKGROUND: Heterotopic ossification (HO) is a pathological condition of abnormal bone formation in soft tissue, which causes pain and restricted range of motion in patients. There are two broad categories of HO, hereditary and acquired. Although different types of HO do not use identical mechanistic pathways of pathogenesis, muscle injury appears to be a unifying feature for all types of HO. However, little is known about the mechanisms by which muscle injury facilitates HO formation. OBJECTIVE AND METHOD: This study aimed to explore the cellular and molecular mechanisms linking muscle injury to HO by using cardiotoxin to induce muscle injury in a bone morphogenetic protein-2 (BMP-2)-induced HO mouse model. RESULTS: We found that muscle injury augmented HO formation and that this effect was correlated with BMP signalling activation and upregulation of BMP-7 expression at the early phase of HO progression. We further demonstrated that inhibition of BMP-7 activity in vitro suppressed the osteogenesis-promoting effect of conditioned medium derived from injured muscle tissue and in vivo reduced the volume of HO formation. We also showed that antiinflammatory drug treatment reduced the volume of HO with concomitant reduction in BMP-7 production. CONCLUSION: In summary, our study has identified BMP-7 as a key osteoinductive factor in injured muscle that facilitates HO formation. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: Our results provide a candidate mechanistic rationale for the use of antiinflammatory drugs in the prevention of HO.

20.
FASEB J ; 33(9): 10231-10239, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31238006

RESUMO

Nerve growth factor (NGF) is a key regulator of chronic osteoarthritic pain, but the exact targets of NGF action on human articular cartilage is unknown. This study aimed to test the hypothesis that the NGF-tropomyosin receptor kinase A (TrkA) (high-affinity NGF receptor) pathway plays a role in the calcification process of human articular chondrocytes (hACs). A 14-aa small peptide of NGF (Nsp) previously shown to activate NGF signaling in rat PC12 cells was used as an NGF signaling agonist, and recombinant NGF and the pan-Trk inhibitor GNF-5837 were employed as signaling modulating agents. The functional consequences of NGF-TrkA signaling were examined in human healthy articular chondrocytes maintained under conditions supportive of osteogenesis in vitro. The NGF-mimetic bioactivity of Nsp was first confirmed on the basis of maintenance of neurite outgrowth in PC12 cells. Primary human chondrocytes responded to Nsp in vitro. Perturbation of NGF signaling with NGF, Nsp, and GNF-5837 resulted in a strong induction of chondrocyte calcification, and gene expression data suggested that the Indian Hedgehog-parathyroid hormone-related protein signaling axis was involved. These findings suggest functional involvement of NGF signaling in calcification of hACs and the importance of NGF signaling in articular cartilage homeostasis.-Jiang, Y., Tuan, R. S. Role of NGF-TrkA signaling in calcification of articular chondrocytes.


Assuntos
Calcificação Fisiológica , Cartilagem Articular/patologia , Condrócitos/patologia , Hipertrofia/patologia , Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Calcificação Vascular/patologia , Animais , Cartilagem Articular/metabolismo , Diferenciação Celular , Condrócitos/metabolismo , Humanos , Hipertrofia/metabolismo , Células PC12 , Ratos , Transdução de Sinais , Calcificação Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...