Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 32(19): 2872-2886, 2023 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-37427980

RESUMO

Mandibuloacral dysplasia type A (MADA) is a rare genetic progeroid syndrome associated with lamin A/C (LMNA) mutations. Pathogenic mutations of LMNA result in nuclear structural abnormalities, mesenchymal tissue damage and progeria phenotypes. However, it remains elusive how LMNA mutations cause mesenchymal-derived cell senescence and disease development. Here, we established an in vitro senescence model using induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs) from MADA patients with homozygous LMNA p.R527C mutation. When expanded to passage 13 in vitro, R527C iMSCs exhibited marked senescence and attenuation of stemness potential, accompanied by immunophenotypic changes. Transcriptome and proteome analysis revealed that cell cycle, DNA replication, cell adhesion and inflammation might play important roles in senescence. In-depth evaluation of changes in extracellular vesicle (EV) derived iMSCs during senescence revealed that R527C iMSC-EVs could promote surrounding cell senescence by carrying pro-senescence microRNAs (miRNAs), including a novel miRNA called miR-311, which can serve as a new indicator for detecting chronic and acute mesenchymal stem cell (MSC) senescence and play a role in promoting senescence. Overall, this study advanced our understanding of the impact of LMNA mutations on MSC senescence and provided novel insights into MADA therapy as well as the link between chronic inflammation and aging development.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Mesenquimais , MicroRNAs , Humanos , Multiômica , Biomarcadores , MicroRNAs/genética , Lamina Tipo A/genética
2.
BMC Biol ; 22(1): 1, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-38167069

RESUMO

BACKGROUND: Cell senescence is a sign of aging and plays a significant role in the pathogenesis of age-related disorders. For cell therapy, senescence may compromise the quality and efficacy of cells, posing potential safety risks. Mesenchymal stem cells (MSCs) are currently undergoing extensive research for cell therapy, thus necessitating the development of effective methods to evaluate senescence. Senescent MSCs exhibit distinctive morphology that can be used for detection. However, morphological assessment during MSC production is often subjective and uncertain. New tools are required for the reliable evaluation of senescent single cells on a large scale in live imaging of MSCs. RESULTS: We have developed a successful morphology-based Cascade region-based convolution neural network (Cascade R-CNN) system for detecting senescent MSCs, which can automatically locate single cells of different sizes and shapes in multicellular images and assess their senescence state. Additionally, we tested the applicability of the Cascade R-CNN system for MSC senescence and examined the correlation between morphological changes with other senescence indicators. CONCLUSIONS: This deep learning has been applied for the first time to detect senescent MSCs, showing promising performance in both chronic and acute MSC senescence. The system can be a labor-saving and cost-effective option for screening MSC culture conditions and anti-aging drugs, as well as providing a powerful tool for non-invasive and real-time morphological image analysis integrated into cell production.


Assuntos
Aprendizado Profundo , Células-Tronco Mesenquimais , Proliferação de Células , Senescência Celular , Células Cultivadas
3.
Biochem Biophys Res Commun ; 663: 61-70, 2023 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-37119767

RESUMO

Articular cartilage (AC) is most susceptible to degeneration in knee osteoarthritis (OA); however, the existing treatments for OA do not target the core link of the pathogenesis-"decreased tissue cell function activity and extracellular matrix (ECM) metabolic disorders" for effective intervention. iMSC hold lower heterogeneity and great promise in biological research and clinical applications. Rps6ka2 may play an important role in the iMSC to treat OA. In this study, the CRISPR/Cas9 gene editing Rps6ka2-/- iMSC were obtained. Effect of Rps6ka2 on iMSC proliferation and chondrogenic differentiation was evaluated in vitro. An OA model was constructed in mice by surgical destabilization of medial meniscus (DMM). The Rps6ka2-/- iMSC and iMSC were injected into the articular cavity twice-weekly for 8 weeks. In vitro experiments showed that Rps6ka2 could promote iMSC proliferation and chondrogenic differentiation. In vivo results further confirmed that Rps6ka2 could improve iMSC viability to promote ECM production to attenuate OA in mice.


Assuntos
Cartilagem Articular , Osteoartrite do Joelho , Camundongos , Animais , Osteoartrite do Joelho/genética , Osteoartrite do Joelho/terapia , Osteoartrite do Joelho/metabolismo , Cartilagem Articular/metabolismo , Diferenciação Celular/genética , Matriz Extracelular , Condrócitos/metabolismo , Modelos Animais de Doenças
4.
Molecules ; 28(23)2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-38067435

RESUMO

Due to the narrow therapeutic window and high mortality of ischemic stroke, it is of great significance to investigate its diagnosis and therapy. We employed weighted gene coexpression network analysis (WGCNA) to ascertain gene modules related to stroke and used the maSigPro R package to seek the time-dependent genes in the progression of stroke. Three machine learning algorithms were further employed to identify the feature genes of stroke. A nomogram model was built and applied to evaluate the stroke patients. We analyzed single-cell RNA sequencing (scRNA-seq) data to discern microglia subclusters in ischemic stroke. The RNA velocity, pseudo time, and gene set enrichment analysis (GSEA) were performed to investigate the relationship of microglia subclusters. Connectivity map (CMap) analysis and molecule docking were used to screen a therapeutic agent for stroke. A nomogram model based on the feature genes showed a clinical net benefit and enabled an accurate evaluation of stroke patients. The RNA velocity and pseudo time analysis showed that microglia subcluster 0 would develop toward subcluster 2 within 24 h from stroke onset. The GSEA showed that the function of microglia subcluster 0 was opposite to that of subcluster 2. AZ_628, which screened from CMap analysis, was found to have lower binding energy with Mmp12, Lgals3, Fam20c, Capg, Pkm2, Sdc4, and Itga5 in microglia subcluster 2 and maybe a therapeutic agent for the poor development of microglia subcluster 2 after stroke. Our study presents a nomogram model for stroke diagnosis and provides a potential molecule agent for stroke therapy.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Humanos , AVC Isquêmico/diagnóstico , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/genética , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/genética , Algoritmos , Aprendizado de Máquina , RNA
5.
Biochem Biophys Res Commun ; 570: 199-205, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34298323

RESUMO

Osteoarthritis (OA) is the most common joint disease worldwide; however, disease-modifying treatments are lacking because of the complicated pathological mechanisms. As a breakthrough, aberrant activation of transforming growth factor-ß 1 (TGF-ß1)in subchondral bone has been confirmed as an essential pathomechanism for OA progression, and has become a potential therapeutic target. In addition to R&D on neutralizing antibodies, small-molecule antagonists and chemical medicines, native antagonists of TGF-ß1 could be exploited as another promising approach. Noggin (NOG) is an antagonist of bone morphogenetic proteins (BMPs) and was reported to effectively attenuate OA by protecting cartilage and preventing pathological subchondral bone remodeling. However, the underlying mechanisms have not been fully clarified. We first detected the distribution of NOG in knee joints of an OA mouse model, which showed upregulation at early stage of OA but downregulation later in the subchondral bone and no significant change in the articular cartilage. Furthermore, the interaction between NOG and TGF-ß1 was verified, which in turn suppressed the downstream SMAD2/3 activity of TGF-ß1. Moreover, the proliferation and chondrogenesis of mesenchymal stem cells (MSCs) were not significantly influenced by NOG. Taken together, the results showed that NOG antagonized TGF-ß1 but did not repress MSC proliferation and chondrogenesis; thus, it seems promising for OA treatment.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Osteoartrite/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Cartilagem Articular/patologia , Proliferação de Células , Condrogênese , Modelos Animais de Doenças , Progressão da Doença , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Modelos Biológicos , Osteoartrite/patologia , Ligação Proteica , Fator de Crescimento Transformador beta1/metabolismo
6.
Biochem Biophys Res Commun ; 554: 33-40, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-33774277

RESUMO

Glucocorticoid-induced osteoporosis (GIOP) has emerged as a challenge after long-term glucocorticoid administration during the clinical therapy of diverse diseases. Although some candidates for GIOP treatment have been explored, there is still a lack of reliable drugs for GIOP prevention. In this study, rat bone marrow stem cells (rBMSCs) were utilized to investigate the feasibility of applying strontium gluconate (GluSr), which displays mild activity, easy absorption and good biocompatibility, for GIOP prevention. Thirty-two SD rats were divided into 4 groups to explore the effects of GluSr on osteoporosis rescue in vivo. Our results suggested that GluSr markedly alleviated dexamethasone (DEX)-induced apoptosis of osteoblast precursor cells and rBMSCs and enhanced rBMSC osteogenesis differentiation in vitro. GluSr also effectively promoted osteoblast survival, inhibited osteoclast differentiation and restored bone formation in GIOP rat models. Microarray analysis of the femora from GIOP rats treated with GluSr revealed that the signalling pathways of the glucocorticoid receptor (GR), oestrogen receptor gene (ESR) and vitamin D receptor (VDR) were involved in bone restoration by GluSr. In summary, our study proved that GluSr enhanced osteoblast differentiation and suppressed osteoclast activity both in vitro and in vivo. GluSr might function as a novel strontium reagent for GIOP prevention.


Assuntos
Glucocorticoides/efeitos adversos , Gluconatos/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Células-Tronco/efeitos dos fármacos , Estrôncio/farmacologia , Animais , Medula Óssea/metabolismo , Células Cultivadas , Dexametasona/efeitos adversos , Modelos Animais de Doenças , Masculino , Camundongos , Osteoblastos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Osteoporose/induzido quimicamente , Osteoporose/metabolismo , Osteoporose/patologia , Ratos , Ratos Sprague-Dawley , Células-Tronco/citologia , Células-Tronco/metabolismo
7.
Biochem Biophys Res Commun ; 532(3): 385-392, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-32888652

RESUMO

A decrease in the number of endogenous stem cells in cartilage is regarded as the cause of cartilage degeneration. Kartogenin (KGN) is known to induce chondrogenesis of cartilage stem/progenitor cells (CSPCs). Using CSPCs isolated from rat cartilage, we analysed changes in the transcriptome after treatment with KGN in vitro. An animal model of destabilization of the medial meniscus (DMM) was then used to identify the effect of intra-articular (IA) KGN injection on CSPC proliferation in vivo. Here, we demonstrated that KGN promoted the proliferation of CSPCs isolated from cartilage. The percentage of G2-M phase cells in the KGN-treated group reached over 10%, nearly twice that in the control group. Transcriptomic profiling of rat CSPCs revealed significant changes in KGN-treated samples compared to control samples. The gene expression levels of IL-6 and its coreceptor Gp130 were much higher in the KGN-treated group than in the control group. Phosphorylation of the IL-6 downstream molecule Stat3 was enhanced via KGN stimulation. The DMM animal model showed increased articular cartilage thickness after IA KGN injection. IHC staining also demonstrated upregulation of Stat3 phosphorylation and enhanced distribution of CD44+/CD105+ cells in cartilage following IA KGN injection. Thus, our data suggested that KGN promoted cartilage regeneration at least partially by stimulating IL-6/Stat3-dependent proliferation.


Assuntos
Anilidas/farmacologia , Cartilagem Articular/efeitos dos fármacos , Interleucina-6/metabolismo , Ácidos Ftálicos/farmacologia , Regeneração/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Animais , Cartilagem Articular/citologia , Cartilagem Articular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Proliferação de Células/fisiologia , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Condrogênese/fisiologia , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Interleucina-6/genética , Masculino , Osteoartrite/tratamento farmacológico , Osteoartrite/patologia , Osteoartrite/fisiopatologia , Ratos , Regeneração/genética , Regeneração/fisiologia , Fator de Transcrição STAT3/genética , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
8.
J Cell Mol Med ; 23(3): 2093-2102, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30609248

RESUMO

Various neuropeptides related to the energy equilibrium affect bone growth in humans and animals. Neuropeptides W (NPW) are identical in the internal ligands of the two G-protein receptors (GPRs) included in subtypes 7 and 8. Neuropeptides W inhibits proliferation in the cultivated rat calvarial osteoblast-like (ROB) cells. This study examines the expression of NPW and GPR7 in murine chondrocyte and their function. An immunohistochemical analysis showed that NPW and GPR7 were expressed in the proliferative chondrocytes of the growth plates in the hind limbs of mice. The NPW mRNA quickly elevated in the early differentiation (7-14 days) of ATDC5 cells, while NPW and GPR7 mRNA were reduced during the late stage (14-21 days) of differentiation. Neuropeptide W-23 (NPW-23) promoted the proliferation of ATDC5 cells, which was attenuated by inhibiting the GPR7, protein kinase A (PKA), protein kinase C (PKC) and ERK1/2 pathways. Neuropeptide W-23 enhanced the early cell differentiation, as evaluated by collagen type II and the aggrecan gene expression, which was unaffected by inhibiting the ERK1/2 pathway, but significantly decreased by inhibiting the PKA, PKC and p38 MAPK pathways. In contrast, NPW-23 was not involved in the terminal differentiation of the chondrocytes, as evaluated by the mineralization of the chondrocytes and the activity of the alkaline phosphatase. Neuropeptides W stimulated the PKA, PKC, p38 MAPK and ERK1/2 activities in a dose- and time-dependent manner in the ATDC5 cells. These results show that NPW promotes the proliferation and early differentiation of murine chondrocyte via GPR7 activation, as well as PKA and PKC-dependent signalling cascades, which may be involved in endochondral bone formation.


Assuntos
Diferenciação Celular/genética , Proliferação de Células/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Neuropeptídeos/genética , Proteína Quinase C/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropeptídeos/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
J Cell Physiol ; 234(4): 3436-3444, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30387127

RESUMO

Runt-related transcription factor-2 (Runx2) is essential for chondrocyte maturation during cartilage development and embryonic mandibular condylar development. The process that chondrocytes, especially a subgroup of hypertrophic chondrocytes (HC), could transform into bone cells in mandibular condyle growth makes chondrocytes crucially important for normal endochondral bone formation. To determine whether Runx2 regulates postnatal condylar cartilage growth and tissue homeostasis, we deleted Runx2 in chondrocytes in postnatal mice and assessed the consequences on temporomandibular joint (TMJ) cartilage growth and remodeling. The cell lineage tracing data provide information demonstrating the role of chondrocytes in subchondral bone remodeling. The histologic and immunohistochemical data showed that Runx2 deficiency caused condylar tissue disorganization, including loss of HC and reduced hypertrophic zone, reduced proliferative chondrocytes, and decreased cartilage matrix production. Expression of Col10a1, Mmp13, Col2a1, Aggrecan, and Ihh was significantly reduced in Runx2 knockout mice. The findings of this study demonstrate that Runx2 is required for chondrocyte proliferation and hypertrophy in TMJ cartilage and postnatal TMJ cartilage growth and homeostasis, and that Runx2 may play an important role in regulation of chondrocyte-derived subchondral bone remodeling.


Assuntos
Proliferação de Células , Condrócitos/metabolismo , Condrogênese , Subunidade alfa 1 de Fator de Ligação ao Core/deficiência , Côndilo Mandibular/metabolismo , Articulação Temporomandibular/metabolismo , Animais , Remodelação Óssea , Linhagem da Célula , Condrócitos/patologia , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Homeostase , Hipertrofia , Côndilo Mandibular/patologia , Camundongos Knockout , Fenótipo , Articulação Temporomandibular/patologia
10.
Int J Mol Sci ; 19(12)2018 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-30487470

RESUMO

Chondrocyte dysfunction occurs during the development of osteoarthritis (OA), typically resulting from a deleterious increase in oxidative stress. Accordingly, strategies for arresting oxidative stress-induced chondrocyte dysfunction may lead to new potential therapeutic targets for OA treatment. Forkhead box O (FoxO) transcription factors have recently been shown to play a protective role in chondrocyte dysfunction through the regulation of inflammation, autophagy, aging, and oxidative stress. They also regulate growth, maturation, and matrix synthesis in chondrocytes. In this review, we discuss the recent progress made in the field of oxidative stress-induced chondrocyte dysfunction. We also discuss the protective role of FoxO transcription factors as potential molecular targets for the treatment of OA. Understanding the function of FoxO transcription factors in the OA pathology may provide new insights that will facilitate the development of next-generation therapies to prevent OA development and to slow OA progression.


Assuntos
Condrócitos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Osteoartrite/metabolismo , Animais , Autofagia/fisiologia , Fatores de Transcrição Forkhead/genética , Humanos , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia
11.
Connect Tissue Res ; 58(6): 573-585, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28102712

RESUMO

Bone morphogenetic proteins (BMPs) play roles in promoting cell anabolism, especially in extracellular matrix production. The difference between BMP members in their capacity to modulate intervertebral disc cell activity is yet to be defined. BMP-7/OP-1 has been shown to retard disc degeneration. We compared the activity of BMP-7 with that of BMP-2 on nucleus pulposus (NP) cell phenotype and function, and investigated how they differentially affect the gene expression profiles of signaling cascade components in human NP cells under degenerative states. We found that while both BMP-2 and BMP-7 enhanced matrix production of bovine NP cells, BMP-7 is more potent than BMP-2 at various dosages (50-800 ng/ml). BMP-7 exerted a relatively stronger stimulation on sulfated glycosaminoglycan production and proliferation in human NP cells. Degenerated NP cells showed an overall weaker response to the BMPs than non-degenerated cells, and were more sensitive to BMP-7 than BMP-2 stimulation. Compared to BMP-2, BMP-7 not only induced the gene expression of canonical BMP components, but also evoked changes in MAPKs as well as CREB1 and EP300 gene expression in degenerated NP cells, suggesting potential activation of the cAMP dependent protein kinase related pathways. In contrast to BMP-2, BMP-7 concomitantly inhibited the expression of profibrotic genes. We propose that BMP-2 and BMP-7, and likely other BMPs, may operate multifaceted but discrete molecular machineries that give rise to their different capacity in regulating NP cell phenotype. Further investigations into such differential capacity may possibly derive alternative cues important for IVD repair or engineering.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 7/metabolismo , Núcleo Pulposo/metabolismo , Células Cultivadas , Matriz Extracelular/metabolismo , Humanos , Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
12.
BMC Biotechnol ; 16(1): 78, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27829414

RESUMO

BACKGROUND: The incapacity of articular cartilage (AC) for self-repair after damage ultimately leads to the development of osteoarthritis. Stem cell-based therapy has been proposed for the treatment of osteoarthritis (OA) and induced pluripotent stem cells (iPSCs) are becoming a promising stem cell source. RESULTS: Three steps were developed to differentiate human iPSCs into chondrocytes which were transplanted into rat OA models induced by monosodium iodoacetate (MIA). After 6 days embryonic body (EB) formation and 2 weeks differentiation, the gene and protein expression of Col2A1, GAG and Sox9 has significantly increased compare to undifferentiated hiPSCs. After 15 weeks transplantation, no immune responses were observed, micro-CT showed gradual engraftment and the improvement of subchondrol plate integrity, and histological examinations demonstrated articular cartilage matrix production. CONCLUSIONS: hiPSC could be an efficient and clinically translatable approach for cartilage tissue regeneration in OA cartilages.


Assuntos
Cartilagem Articular/crescimento & desenvolvimento , Condrócitos/citologia , Condrócitos/transplante , Células-Tronco Pluripotentes Induzidas/citologia , Osteoartrite/patologia , Osteoartrite/terapia , Animais , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Diferenciação Celular/imunologia , Células Cultivadas , Condrócitos/imunologia , Condrogênese/imunologia , Osteoartrite/imunologia , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
13.
Ageing Res Rev ; 96: 102273, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38492810

RESUMO

Cardiovascular disease is currently the largest cause of mortality and disability globally, surpassing communicable diseases, and atherosclerosis is the main contributor to this epidemic. Aging is intimately linked to atherosclerosis development and progression, however, the mechanism of aging in atherosclerosis is not well known. To emphasize the significant research on the involvement of senescent cells in atherosclerosis, we begin by outlining compelling evidence that indicates various types of senescent cells and SASP factors linked to atherosclerotic phenotypes. We subsequently provide a comprehensive summary of the existing knowledge, shedding light on the intricate mechanisms through which cellular senescence contributes to the pathogenesis of atherosclerosis. Further, we cover that senescence can be identified by both structural changes and several senescence-associated biomarkers. Finally, we discuss that preventing accelerated cellular senescence represents an important therapeutic potential, as permanent changes may occur in advanced atherosclerosis. Together, the review summarizes the relationship between cellular senescence and atherosclerosis, and inspects the molecular knowledge, and potential clinical significance of senescent cells in developing senescent-based therapy, thus providing crucial insights into their biology and potential therapeutic exploration.


Assuntos
Aterosclerose , Senescência Celular , Humanos , Envelhecimento , Biomarcadores , Fenótipo , Aterosclerose/terapia
14.
J Dent ; 144: 104957, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38527517

RESUMO

INTRODUCTION: Osteoclasts (OCs) play a crucial role in maintaining bone health. Changes in OC activity are linked to different bone diseases, making them an intriguing focus for research. However, most studies on OCs have relied on 2D cultures, limiting our understanding of their behavior. Yet, there's a lack of knowledge regarding platforms that effectively support osteoclast formation in 3D cultures. METHODS: In our investigation, we explored the capacity of collagen and GelMA hydrogels to facilitate osteoclast development in 3D culture settings. We assessed the osteoclast development by using different hydrogels and cell seeding strategies and optimizing cell seeding density and cytokine concentration. The osteoclast development in 3D cultures was further validated by biochemical assays and immunochemical staining. RESULTS: Our findings revealed that 0.3 % (w/v) collagen was conducive to osteoclast formation in both 2D and 3D cultures, demonstrated by increased multinucleation and higher TRAP activity compared to 0.6 % collagen and 5 % to 10 % (w/v) GelMA hydrogels. Additionally, we devised a "sandwich" technique using collagen substrates and augmented the initial macrophage seeding density and doubling cytokine concentrations, significantly enhancing the efficiency of OC culture in 3D conditions. Notably, we validated osteoclasts derived from macrophages in our 3D cultures express key osteoclast markers like cathepsin K and TRAP. CONCLUSIONS: To conclude, our study contributes to establishing an effective method for cultivating osteoclasts in 3D environments in vitro. This innovative approach not only promises a more physiologically relevant platform to study osteoclast behavior during bone remodeling but also holds potential for applications in bone tissue engineering. CLINICAL SIGNIFICANCE: This study introduces an efficient method for cultivating osteoclasts in 3D environments in vitro. It offers a more physiologically relevant platform to investigate osteoclast behavior and holds promise to advance research in bone biology and regenerative dentistry.


Assuntos
Técnicas de Cultura de Células , Hidrogéis , Osteoclastos , Osteoclastos/citologia , Animais , Diferenciação Celular , Colágeno , Camundongos , Técnicas de Cultura de Células em Três Dimensões/métodos , Macrófagos/citologia , Catepsina K , Citocinas/metabolismo , Células Cultivadas
15.
Sci Total Environ ; 916: 170300, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38272090

RESUMO

Reservoirs are regarded as potential collection sites for microplastics (MPs), and ample water resources in plateau regions provide favorable natural conditions for hydroelectric power generation. However, research on the impact of cascade reservoir construction in the plateau region on the fate of MPs within the watershed is limited. In this study, the Yalong River, an alpine canyon river in the eastern Qinghai-Tibet Plateau, was selected as the research area. This study explored the distribution of MPs at various depths in water, sediment, and riverbank soil as well as the formation of "MP communities" within the river-cascade reservoir system. Furthermore, the effects of dam construction on MPs' migration in different environments were analyzed. The results revealed that the abundance of MPs in the water and sediment within the cascade reservoir area (CRA) was significantly higher than that in the river area (RA) (P < 0.001). Additionally, the trend of increasing MPs in water with decreasing altitude was notably slower in CRA. Regarding shape, the proportion of fibers in the water within the CRA was significantly lower than that in the RA, with a smaller vertical migration rate in the water than in the sediment. The proportion of MPs < 500 µm in the water within the CRA was significantly higher than that in the RA. High-density MPs were notably deposited in the reservoir sediments. The analysis of the MP communities revealed that the construction of cascade dams led to relative geographical isolation between different sampling sites, reducing the similarity of MP communities in the CRA. This study established a theoretical foundation for understanding the impact of cascade dam construction on the fate characteristics of MPs and their potential risks in plateau areas.

16.
Biomaterials ; 307: 122526, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38513434

RESUMO

Stem cell therapies have shown great potential for treating myocardial infarction (MI) but are limited by low cell survival and compromised functionality due to the harsh microenvironment at the disease site. Here, we presented a Mesenchymal stem cell (MSC) spheroid-based strategy for MI treatment by introducing a protein/polyphenol self-assembling armor coating on the surface of cell spheroids, which showed significantly enhanced therapeutic efficacy by actively manipulating the hostile pathological MI microenvironment and enabling versatile functionality, including protecting the donor cells from host immune clearance, remodeling the ROS microenvironment and stimulating MSC's pro-healing paracrine secretion. The underlying mechanism was elucidated, wherein the armor protected to prolong MSCs residence at MI site, and triggered paracrine stimulation of MSCs towards immunoregulation and angiogenesis through inducing hypoxia to provoke glycolysis in stem cells. Furthermore, local delivery of coated MSC spheroids in MI rat significantly alleviated local inflammation and subsequent fibrosis via mediation macrophage polarization towards pro-healing M2 phenotype and improved cardiac function. In general, this study provided critical insight into the enhanced therapeutic efficacy of stem cell spheroids coated with a multifunctional armor. It potentially opens up a new avenue for designing immunomodulatory treatment for MI via stem cell therapy empowered by functional biomaterials.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Infarto do Miocárdio , Ratos , Animais , Infarto do Miocárdio/patologia , Células-Tronco/patologia , Esferoides Celulares/patologia , Cicatrização
17.
Med Biol Eng Comput ; 61(7): 1675-1686, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36853396

RESUMO

Accurate continuous estimation of multi-DOF movement is crucial for simultaneous control of advanced myoelectric prosthetic. The decoupling of multi-DOF is a challenge for continuous estimation. In this paper, we propose a model combined non-negative matrix factorization (NMF) with Hadamard product and L2 regulation to suppress the non-active DOF and achieve the multi-DOF movement continuous estimation. The L2 regulation of non-active DOF activation coefficient was added to the object function of NMF with the benefit of Hadamard product. The angles were estimated by a linear combination of the activation coefficients. We performed a set of continuous estimation experiments for single-DOF and multi-DOF movements of wrist flexion/extend and hand open/close. The results illustrated that the novel model could suppress non-active DOF in single-DOF movement better than other methods based on muscle synergy theory. Moreover, we investigated the robustness of suppression effect and the similarity of synergy matrices at different speeds for NMF-based methods, and the results showed that the proposed method had a superior performance.


Assuntos
Músculo Esquelético , Extremidade Superior , Eletromiografia/métodos , Músculo Esquelético/fisiologia , Punho/fisiologia , Movimento/fisiologia
18.
Br J Pharmacol ; 180(13): 1748-1765, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36721985

RESUMO

BACKGROUND AND PURPOSE: Isoxazole 9 (ISX9) is a neurogenesis-promoting small molecule compound that can up-regulate the expression of NeuroD1 and induce differentiation of neuronal, cardiac and islet endocrine progenitors. So far, the molecular mechanisms underlying the action of ISX9 still remain elusive. EXPERIMENTAL APPROACH: To identify a novel agonist of the Wnt/ß-catenin, a cell-based SuperTOPFlash reporter system was used to screen known-compound libraries. An activation effect of ISX9 on the Wnt/ß-catenin pathway was analysed with the SuperTOPFlash or SuperFOPFlash reporter system. Effects of ISX9 on Axin1/LRP6 interaction were examined using a mammalian two-hybrid system, co-immunoprecipitation, microscale thermophoresis, emission spectra and mass spectrometry assays. The expression of Wnt target and stemmness marker genes were evaluated with real-time PCR and immunoblotting. In vivo hair regeneration abilities of ISX9 were analysed by immunohistochemical staining, real-time PCR and immunoblotting in hair regrowth model using C57BL/6J mice. KEY RESULTS: In this study, ISX9 was identified as a novel agonist of the Wnt/ß-catenin pathway. ISX9 targeted Axin1 by covalently binding to its N-terminal region and potentiated the LRP6-Axin1 interaction, thereby resulting in the stabilization of ß-catenin and up-regulation of Wnt target genes and stemmness marker genes. Moreover, the topical application of ISX9 markedly promoted hair regrowth in C57BL/6J mice and induced hair follicle transition from telogen to anagen via enhancing Wnt/ß-catenin pathway. CONCLUSIONS AND IMPLICATIONS: Taken together, our study unravelled that ISX9 could activate Wnt/ß-catenin signalling by potentiating the association between LRP6 and Axin1, and may be a promising therapeutic agent for alopecia treatment.


Assuntos
Via de Sinalização Wnt , beta Catenina , Camundongos , Animais , Proteína Axina/genética , Proteína Axina/metabolismo , Proteína Axina/farmacologia , beta Catenina/metabolismo , Camundongos Endogâmicos C57BL , Cabelo , Mamíferos/metabolismo
19.
iScience ; 26(1): 105808, 2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36619982

RESUMO

The early postnatal limb developmental progression bridges embryonic and mature stages and mirrors the pathological remodeling of articular cartilage. However, compared with multitudinous research on embryonic limb development, the early postnatal stage seems relatively unnoticed. Here, a systematic work to portray the postnatal limb developmental landscape was carried out by characterization of 19,952 single cells from murine hindlimbs at 4 postnatal stages using single-cell RNA sequencing technique. By delineation of cell heterogeneity, the candidate progenitor sub-clusters marked by Cd34 and Ly6e were discovered in articular cartilage and enthesis, and three cellular developmental branches marked by Col10a1, Spp1, and Tnni2 were reflected in growth plate. The representative transcriptomes and developmental patterns were intensively explored, and the key regulation mechanisms as well as evolvement in osteoarthritis were discussed. Above all, these results expand horizons of postnatal limb developmental biology and reach the interconnections between limb development, remodeling, and regeneration.

20.
Heliyon ; 9(1): e12683, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36647346

RESUMO

Mesenchymal stem/stromal cells (MSCs) show tremendous potential for regenerative medicine due to their self-renewal, multi-differentiation and immunomodulatory capabilities. Largely studies had indicated conventional tissue-derived MSCs have considerable limited expandability and donor variability which hinders further application. Induced pluripotent stem cell (iPSCs)-derived MSCs (iMSCs) have created exciting source for standardized cellular therapy. However, the cellular and molecular differences between iMSCs and the cognate tissue-derived MSCs remains poorly explored. In this study, we first successfully reprogrammed human umbilical cords-derived mesenchymal stem/stromal cells (UMSCs) into iPSCs by using the cocktails of mRNA. Subsequently, iPSCs were further differentiated into iMSCs in xeno-free induction medium. Then, iMSCs were compared with the donor matched UMSCs by assessing proliferative state, differentiation capability, immunomodulatory potential through immunohistochemical analysis, flow cytometric analysis, transcriptome sequencing analysis, and combine with coculture with immune cell population. The results showed that iMSCs exhibited high expression of MSCs positive-makers CD73, CD90, CD105 and lack expression of negative-maker cocktails CD34, CD45, CD11b, CD19, HLA-DR; also successfully differentiated into osteocytes, chondrocytes and adipocytes. Further, the iMSCs were similar with their parental UMSCs in cell proliferative state detected by the CCK-8 assay, and in cell rejuvenation state assessed by ß-Galactosidase staining and telomerase activity related mRNA and protein analysis. However, iMSCs exhibited similarity to resident MSCs in Homeobox (Hox) genes expression profile and presented better neural differentiation potential by activation of NESTIN related pathway. Moreover, iMSCs owned enhanced immunosuppression capacity through downregulation pools of pro-inflammatory factors, including IL6, IL1B etc. and upregulation anti-inflammatory factors NOS1, TGFB etc. signals. In summary, our study provides an attractive cell source for basic research and offers fundamental biological insight of iMSCs-based therapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA