Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Nanobiotechnology ; 22(1): 242, 2024 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-38735936

RESUMO

BACKGROUND: Two-dimensional ultrathin Ti3C2 (MXene) nanosheets have gained significant attention in various biomedical applications. Although previous studies have described the accumulation and associated damage of Ti3C2 nanosheets in the testes and placenta. However, it is currently unclear whether Ti3C2 nanosheets can be translocated to the ovaries and cause ovarian damage, thereby impairing ovarian functions. RESULTS: We established a mouse model with different doses (1.25, 2.5, and 5 mg/kg bw/d) of Ti3C2 nanosheets injected intravenously for three days. We demonstrated that Ti3C2 nanosheets can enter the ovaries and were internalized by granulosa cells, leading to a decrease in the number of primary, secondary and antral follicles. Furthermore, the decrease in follicles is closely associated with higher levels of FSH and LH, as well as increased level of E2 and P4, and decreased level of T in mouse ovary. In further studies, we found that exposure toTi3C2 nanosheets increased the levels of Beclin1, ATG5, and the ratio of LC3II/Ι, leading to autophagy activation. Additionally, the level of P62 increased, resulting in autophagic flux blockade. Ti3C2 nanosheets can activate autophagy through the PI3K/AKT/mTOR signaling pathway, with oxidative stress playing an important role in this process. Therefore, we chose the ovarian granulosa cell line (KGN cells) for in vitro validation of the impact of autophagy on the hormone secretion capability. The inhibition of autophagy initiation by 3-Methyladenine (3-MA) promoted smooth autophagic flow, thereby partially reduced the secretion of estradiol and progesterone by KGN cells; Whereas blocking autophagic flux by Rapamycin (RAPA) further exacerbated the secretion of estradiol and progesterone in cells. CONCLUSION: Ti3C2 nanosheet-induced increased secretion of hormones in the ovary is mediated through the activation of autophagy and impairment of autophagic flux, which disrupts normal follicular development. These results imply that autophagy dysfunction may be one of the underlying mechanisms of Ti3C2-induced damage to ovarian granulosa cells. Our findings further reveal the mechanism of female reproductive toxicity induced by Ti3C2 nanosheets.


Assuntos
Autofagia , Células da Granulosa , Nanoestruturas , Ovário , Titânio , Animais , Feminino , Autofagia/efeitos dos fármacos , Titânio/toxicidade , Titânio/química , Titânio/farmacologia , Camundongos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Nanoestruturas/química , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo
2.
J Cell Physiol ; 236(7): 5278-5292, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33452687

RESUMO

Osteoarthritis (OA) is the most common joint disease. The surface of joint cartilage is a defensive and first affected structure of articular cartilage (AC) during the pathogenesis of OA. Alk5 signaling is critical for maintaining AC homeostasis, however, the role and underlying mechanism for the involvement of Alk5 signaling in the phenotypes of articular cartilage stem cells (ACSCs) at the surface of AC is still unclear. The role of Alk5 in OA development was explored using an ACSCs-specific Alk5-deficient (cKO) mouse model. Alterations in cartilage structure were evaluated histologically. Senescence was detected by SA-ß-gal, while reactive oxygen species (ROS), MitoTracker, and LysoTracker staining were used to detect changes related to senescence. In addition, mice were injected intra-articularly with ganciclovir to limit the detrimental roles of senescent cells (SnCs). Alk5 cKO mice showed a decreased number of the slow-cell cycle cells and less lubricant secretion at the surface accompanied with drastically accelerated cartilage degeneration under ageing and surgically induced OA conditions. Further studies showed that Alk5 deficient ACSCs exhibited senescence-like manifestations including decreased proliferation and differentiation, more SA-ß-gal-positive cells and ROS production, as well as significantly swollen mitochondria and lysosome breakdown. We further found that local limitation of the detrimental roles of SnCs can attenuate the development of posttraumatic OA. Taken together, our findings suggest that Alk5 signaling acts as an important regulator of the SnCs in the superficial layer during AC maintenance and OA initiation.


Assuntos
Cartilagem Articular/metabolismo , Senescência Celular/fisiologia , Osteoartrite/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Células-Tronco/metabolismo , Animais , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Cartilagem Articular/patologia , Camundongos , Camundongos Knockout , Osteoartrite/patologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
3.
Ann Rheum Dis ; 79(1): 112-122, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31662319

RESUMO

OBJECTIVES: This study aims to investigate the role and mechanism of FGFR3 in macrophages and their biological effects on the pathology of arthritis. METHODS: Mice with conditional knockout of FGFR3 in myeloid cells (R3cKO) were generated. Gait behaviours of the mice were monitored at different ages. Spontaneous synovial joint destruction was evaluated by digital radiographic imaging and µCT analysis; changes of articular cartilage and synovitis were determined by histological analysis. The recruitment of macrophages in the synovium was examined by immunostaining and monocyte trafficking assay. RNA-seq analysis, Western blotting and chemotaxis experiment were performed on control and FGFR3-deficient macrophages. The peripheral blood from non-osteoarthritis (OA) donors and patients with OA were analysed. Mice were treated with neutralising antibody against CXCR7 to investigate the role of CXCR7 in arthritis. RESULTS: R3cKO mice but not control mice developed spontaneous cartilage destruction in multiple synovial joints at the age of 13 months. Moreover, the synovitis and macrophage accumulation were observed in the joints of 9-month-old R3cKO mice when the articular cartilage was not grossly destructed. FGFR3 deficiency in myeloid cells also aggravated joint destruction in DMM mouse model. Mechanically, FGFR3 deficiency promoted macrophage chemotaxis partly through activation of NF-κB/CXCR7 pathway. Inhibition of CXCR7 could significantly reverse FGFR3-deficiency-enhanced macrophage chemotaxis and the arthritic phenotype in R3cKO mice. CONCLUSIONS: Our study identifies the role of FGFR3 in synovial macrophage recruitment and synovitis, which provides a new insight into the pathological mechanisms of inflammation-related arthritis.


Assuntos
Cartilagem Articular/patologia , Quimiocina CXCL12/metabolismo , Macrófagos/metabolismo , Osteoartrite/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptores CXCR/genética , Sinovite/genética , Animais , Quimiotaxia/genética , Marcha , Regulação da Expressão Gênica , Humanos , Articulações/metabolismo , Articulações/patologia , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Células Mieloides , NF-kappa B/metabolismo , Osteoartrite/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores CXCR/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Sinovite/patologia
4.
J Biol Chem ; 293(23): 8761-8774, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29691281

RESUMO

Temporomandibular joint osteoarthritis (TMJ OA) is a common degenerative disease with few effective disease-modifying treatments in the clinic. Fibroblast growth factor (FGF) signaling is implicated in articular cartilage homeostasis, but the functional roles of FGFR1 in TMJ OA remain largely unknown. In this study, we report that deletion of Fgfr1 in TMJ chondrocytes delayed TMJ OA progression in the age-associated spontaneous OA model and the abnormal dental occlusion OA model. Immunohistochemical staining revealed that Fgfr1 deficiency decreased the expressions of MMP13 (matrix metalloproteinase-13), ADAMTS5 (a disintegrin and metalloproteinase with thrombospondin motifs 5), and COL10A1 but increased aggrecan expression level in two TMJ OA models. Furthermore, our data show that inactivation of FGFR1 signaling may promote autophagic activity in TMJ. FGFR1 inhibitor decreased the expressions of Mmp13, Adamts5, and Runx2 in IL-1ß-stimulated condylar chondrocytes, whereas autophagy inhibitors abrogated the protective effects of the FGFR1 inhibitor. Thus, our study indicates inactivated FGFR1 signaling ameliorates TMJ OA progression partially by promoting autophagic activity. Manipulation of this signaling may be a potential therapeutic approach to modify TMJ OA.


Assuntos
Autofagia , Condrócitos/patologia , Deleção de Genes , Osteoartrite/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Articulação Temporomandibular/patologia , Animais , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Condrócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoartrite/patologia , Articulação Temporomandibular/metabolismo
5.
J Cell Physiol ; 234(1): 595-605, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-30078186

RESUMO

Activation of transforming growth factor-ß (TGF-ß) signaling has been used to enhance healing of meniscal degeneration in several models. However, the exact role and molecular mechanism of TGF-ß signaling in meniscus maintenance and degeneration are still not understood due to the absence of in vivo evidence. In this study, we found that the expression of activin receptor-like kinases 5 (ALK5) in the meniscus was decreased with the progression of age and/or osteoarthritis induced meniscal degeneration. Col2α1 positive cells were found to be specifically distributed in the superficial and inner zones of the anterior horn, as well as the inner zone of the posterior horn in mice, indicating that Col2α1-CreERT2 mice can be a used for studying gene function in menisci. Furthermore, we deleted Alk5 in Col2α1 positive cells in meniscus by administering tamoxifen. Alterations in the menisci structure were evaluated histologically. The expression levels of genes and proteins associated with meniscus homeostasis and TGF-ß signaling were analyzed by quantitative real-time PCR analysis (qRT-PCR) and immunohistochemistry (IHC). Our results revealed severe and progressive meniscal degeneration phenotype in 3- and 6-month-old Alk5 cKO mice compared with Cre-negative control, including aberrantly increased hypertrophic meniscal cells, severe fibrillation, and structure disruption of meniscus. qRT-PCR and IHC results showed that disruption of anabolic and catabolic homeostasis of chondrocytes may contribute to the meniscal degeneration phenotype observed in Alk5 cKO mice. Thus, TGF-ß/ALK5 signaling plays a chondro-protective role in menisci homeostasis, in part, by inhibiting matrix degradation and maintaining extracellular matrix proteins levels in meniscal tissues.


Assuntos
Colágeno Tipo II/genética , Menisco/fisiopatologia , Osteoartrite/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Animais , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Condrócitos/metabolismo , Condrócitos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Menisco/metabolismo , Camundongos , Camundongos Knockout , Osteoartrite/fisiopatologia , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética
6.
J Cell Mol Med ; 21(12): 3231-3243, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28643466

RESUMO

The osteoarthritis (OA) progression is now considered to be related to inflammation. Anemonin (ANE) is a small natural molecule extracted from various kinds of Chinese traditional herbs and has been shown to inhibiting inflammation response. In this study, we examined whether ANE could attenuate the progression of OA via suppression of IL-1ß/NF-κB pathway activation. Destabilization of the medial meniscus (DMM) was performed in 10-week-old male C57BL/6J mice. ANE was then intra-articularly injected into joint capsule for 8 and 12 weeks. Human articular chondrocytes and cartilage explants challenged with interleukin-1ß (IL-1ß) were treated with ANE. We found that ANE delayed articular cartilage degeneration in vitro and in vivo. In particular, proteoglycan loss and chondrocyte hypertrophy were significantly decreased in ANE -treated mice compared with vehicle-treated mice. ANE decreased the expressions of matrix metalloproteinase-13 (MMP13), A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5), collagen X (Col X) while increasing Aggrecan level in murine with DMM surgery. ANE treatment also attenuated proteoglycan loss in human cartilage explants treated with IL-1ß ex vivo. ANE is a potent protective molecule for OA; it delays OA progression by suppressing ECM loss and chondrocyte hypertrophy partially by suppressing IL-1ß/NF-κB pathway activation.


Assuntos
Anti-Inflamatórios/farmacologia , Cartilagem Articular/efeitos dos fármacos , Furanos/farmacologia , Interleucina-1beta/genética , NF-kappa B/genética , Osteoartrite/tratamento farmacológico , Proteína ADAMTS5/antagonistas & inibidores , Proteína ADAMTS5/genética , Proteína ADAMTS5/metabolismo , Agrecanas/agonistas , Agrecanas/genética , Agrecanas/metabolismo , Animais , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Injeções Intra-Articulares , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/metabolismo , Cápsula Articular/efeitos dos fármacos , Cápsula Articular/metabolismo , Cápsula Articular/patologia , Masculino , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/patologia , Cultura Primária de Células , Transdução de Sinais , Técnicas de Cultura de Tecidos
7.
Hum Mol Genet ; 21(26): 5443-55, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23014564

RESUMO

Gain-of-function mutations in fibroblast growth factor receptor-3 (FGFR3) lead to several types of human skeletal dysplasia syndromes including achondroplasia, hypochondroplasia and thanatophoric dysplasia (TD). Currently, there are no effective treatments for these skeletal dysplasia diseases. In this study, we screened, using FGFR3 as a bait, a random 12-peptide phage library and obtained 23 positive clones that share identical amino acid sequences (VSPPLTLGQLLS), named as peptide P3. This peptide had high binding specificity to the extracellular domain of FGFR3. P3 inhibited tyrosine kinase activity of FGFR3 and its typical downstream molecules, extracellular signal-regulated kinase/mitogen-activated protein kinase. P3 also promoted proliferation and chondrogenic differentiation of cultured ATDC5 chondrogenic cells. In addition, P3 alleviated the bone growth retardation in bone rudiments from mice mimicking human thanatophoric dysplasia type II (TDII). Finally, P3 reversed the neonatal lethality of TDII mice. Thus, this study identifies a novel inhibitory peptide for FGFR3 signaling, which may serve as a potential therapeutic agent for the treatment of FGFR3-related skeletal dysplasia.


Assuntos
Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Displasia Tanatofórica/genética , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Clonagem Molecular , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Biblioteca de Peptídeos , Peptídeos/metabolismo , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Análise de Sequência de DNA , Crânio/anormalidades , Crânio/metabolismo , Crânio/patologia , Displasia Tanatofórica/metabolismo , Displasia Tanatofórica/patologia
8.
J Mol Med (Berl) ; 102(5): 693-707, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38492027

RESUMO

Physical therapy is extensively employed in clinical settings. Nevertheless, the absence of suitable animal models has resulted in an incomplete understanding of the in vivo mechanisms and cellular distribution that respond to physical stimuli. The objective of this research was to create a mouse model capable of indicating the cells affected by physical stimuli. In this study, we successfully established a mouse line based on the heat shock protein 70 (Hsp70) promoter, wherein the expression of CreERT2 can be induced by physical stimuli. Following stimulation of the mouse tail, ear, or cultured calvarias with heat shock (generated by heating, ultrasound, or laser), a distinct Cre-mediated excision was observed in cells stimulated by these physical factors with minimal occurrence of leaky reporter expression. The application of heat shock to Hsp70-CreERT2; FGFR2-P253R double transgenic mice or Hsp70-CreERT2 mice infected with AAV-BMP4 at calvarias induced the activation of Cre-dependent mutant FGFR2-P253R or BMP4 respectively, thereby facilitating the premature closure of cranial sutures or the repair of calvarial defects. This novel mouse line holds significant potential for investigating the underlying mechanisms of physical therapy, tissue repair and regeneration, lineage tracing, and targeted modulation of gene expression of cells in local tissue stimulated by physical factor at the interested time points. KEY MESSAGES: In the study, an Hsp70-CreERT2 transgenic mouse was generated for heat shock-induced gene modulation. Heat shock, ultrasound, and laser stimulation effectively activated Cre expression in Hsp70-CreERT2; reporter mice, which leads to deletion of floxed DNA sequence in the tail, ear, and cultured calvaria tissues of mice. Local laser stimuli on cultured calvarias effectively induce Fgfr2-P253R expression in Hsp70-mTmG-Fgfr2-P253R mice and result in accelerated premature closure of cranial suture. Heat shock activated AAV9-FLEX-BMP4 expression and subsequently promoted the repair of calvarial defect of Hsp70-CreERT2; Rosa26-mTmG mice.


Assuntos
Proteína Morfogenética Óssea 4 , Proteínas de Choque Térmico HSP70 , Camundongos Transgênicos , Regiões Promotoras Genéticas , Animais , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Camundongos , Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 4/genética , Resposta ao Choque Térmico/genética , Crânio/metabolismo , Regulação da Expressão Gênica , Integrases/metabolismo , Integrases/genética
9.
Adv Sci (Weinh) ; 11(7): e2306143, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38083984

RESUMO

Macrophages are heterogenic phagocytic cells that play distinct roles in physiological and pathological processes. Targeting different types of macrophages has shown potent therapeutic effects in many diseases. Although many approaches are developed to target anti-inflammatory macrophages, there are few researches on targeting pro-inflammatory macrophages, which is partially attributed to their non-s pecificity phagocytosis of extracellular substances. In this study, a novel recombinant protein is constructed that can be anchored on an exosome membrane with the purpose of targeting pro-inflammatory macrophages via antigen recognition, which is named AnCar-ExoLaIMTS . The data indicate that the phagocytosis efficiencies of pro-inflammatory macrophages for different AnCar-ExoLaIMTS show obvious differences. The AnCar-ExoLaIMTS3 has the best targeting ability for pro-inflammatory macrophages in vitro and in vivo. Mechanically, AnCar-ExoLaIMTS3 can specifically recognize the leucine-rich repeat domain of the TLR4 receptor, and then enter into pro-inflammatory macrophages via the TLR4-mediated receptor endocytosis pathway. Moreover, AnCar-ExoLaIMTS3 can efficiently deliver therapeutic cargo to pro-inflammatory macrophages and inhibit the synovial inflammatory response via downregulation of HIF-1α level, thus ameliorating the severity of arthritis in vivo. Collectively, the work established a novel gene/drug delivery system that can specifically target pro-inflammatory macrophages, which may be beneficial for the treatments of arthritis and other inflammatory diseases.


Assuntos
Artrite , Macrófagos , Humanos , Macrófagos/metabolismo , Artrite/tratamento farmacológico , Fagocitose , Anti-Inflamatórios/uso terapêutico , Comunicação Celular
10.
Exp Mol Med ; 55(11): 2376-2389, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37907740

RESUMO

Osteoarthritis (OA) is a full-joint, multifactorial, degenerative and inflammatory disease that seriously affects the quality of life of patients due to its disabling and pain-causing properties. ER stress has been reported to be closely related to the progression of OA. The inositol-requiring enzyme 1α/X-box-binding protein-1 spliced (IRE1α/XBP1s) pathway, which is highly expressed in the chondrocytes of OA patients, promotes the degradation and refolding of abnormal proteins during ER stress and maintains the stability of the ER environment of chondrocytes, but its function and the underlying mechanisms of how it contributes to the progression of OA remain unclear. This study investigates the role of IRE1α/ERN1 in OA. Specific deficiency of ERN1 in chondrocytes spontaneously resulted in OA-like cartilage destruction and accelerated OA progression in a surgically induced arthritis model. Local delivery of AdERN1 relieved degradation of the cartilage matrix and prevented OA development in an ACLT-mediated model. Mechanistically, progranulin (PGRN), an intracellular chaperone, binds to IRE1α, promoting its phosphorylation and splicing of XBP1u to generate XBP1s. XBP1s protects articular cartilage through TNF-α/ERK1/2 signaling and further maintains collagen homeostasis by regulating type II collagen expression. The chondroprotective effect of IRE1α/ERN1 is dependent on PGRN and XBP1s splicing. ERN1 deficiency accelerated cartilage degeneration in OA by reducing PGRN expression and XBP1s splicing, subsequently decreasing collagen II expression and triggering collagen structural abnormalities and an imbalance in collagen homeostasis. This study provides new insights into OA pathogenesis and the UPR and suggests that IRE1α/ERN1 may serve as a potential target for the treatment of joint degenerative diseases, including OA.


Assuntos
Cartilagem Articular , Osteoartrite , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Progranulinas/metabolismo , Endorribonucleases/genética , Endorribonucleases/metabolismo , Qualidade de Vida , Osteoartrite/metabolismo , Condrócitos/metabolismo , Cartilagem Articular/metabolismo , Colágeno/metabolismo , Homeostase , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo
11.
Int J Biol Sci ; 18(1): 315-330, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34975335

RESUMO

X-box binding protein 1(XBP1) is a critical component for unfolded protein response (UPR) in ER stress. According to previous studies performed with different XBP1-deficient mice, the XBP1 gene affects mouse cartilage development and causes other related diseases. However, how the complete transcriptome, including mRNA and ncRNAs, affects the function of cartilage and other tissues when XBP1 is deficient in chondrocytes is unclear. In this study, we aimed to screen the differentially expressed (DE) mRNAs, circRNAs, lncRNAs and miRNAs in XBP1 cartilage-specific knockout (CKO) mice using high throughput sequencing and construct the circRNA-miRNA-mRNA and lncRNA-miRNA-mRNA regulatory networks. DE LncRNAs (DE-LncRNAs), circRNAs (DE-circRNAs), miRNAs (DE-miRNAs), and mRNAs [differentially expressed genes (DEGs)] between the cartilage tissue of XBP1 CKO mice and controls were identified, including 441 DE-LncRNAs, 15 DE-circRNAs, 6 DE-miRNAs, and 477 DEGs. Further, 253,235 lncRNA-miRNA-mRNA networks and 1,822 circRNA-miRNA-mRNA networks were constructed based on the correlation between lncRNAs/circRNAs, miRNAs, mRNAs. The whole transcriptome analysis revealed that XBP1 deficiency in cartilage affects the function of cartilage and other different tissues, as well as associated diseases. Overall, our findings may provide potential biomarkers and mechanisms for the diagnosis and treatment of cartilage and other related diseases.


Assuntos
Cartilagem/metabolismo , MicroRNAs/metabolismo , RNA Circular/metabolismo , RNA Longo não Codificante/metabolismo , RNA Mensageiro/metabolismo , Proteína 1 de Ligação a X-Box/deficiência , Animais , Perfilação da Expressão Gênica , Camundongos
12.
Bone Res ; 10(1): 2, 2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-34983922

RESUMO

The intervertebral disc (IVD) is the largest avascular tissue. Hypoxia-inducible factors (HIFs) play essential roles in regulating cellular adaptation in the IVD under physiological conditions. Disc degeneration disease (DDD) is one of the leading causes of disability, and current therapies are ineffective. This study sought to explore the role of HIFs in DDD pathogenesis in mice. The findings of this study showed that among HIF family members, Hif1α was significantly upregulated in cartilaginous endplate (EP) and annulus fibrosus (AF) tissues from human DDD patients and two mouse models of DDD compared with controls. Conditional deletion of the E3 ubiquitin ligase Vhl in EP and AF tissues of adult mice resulted in upregulated Hif1α expression and age-dependent IVD degeneration. Aberrant Hif1α activation enhanced glycolytic metabolism and suppressed mitochondrial function. On the other hand, genetic ablation of the Hif1α gene delayed DDD pathogenesis in Vhl-deficient mice. Administration of 2-methoxyestradiol (2ME2), a selective Hif1α inhibitor, attenuated experimental IVD degeneration in mice. The findings of this study show that aberrant Hif1α activation in EP and AF tissues induces pathological changes in DDD, implying that inhibition of aberrant Hif1α activity is a potential therapeutic strategy for DDD.

13.
Stem Cell Res Ther ; 13(1): 227, 2022 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-35659742

RESUMO

BACKGROUND: Intervertebral disc degeneration (IVDD) can cause low back pain, a major public health concern. IVDD is characterized with loss of cells especially those in nucleus pulposus (NP), due to the limited proliferative potential and regenerative ability. Few studies, however, have been carried out to investigate the in vivo proliferation events of NP cells and the cellular contribution of a specific subpopulation of NP during postnatal growth or regeneration. METHODS: We generated FGFR3-3*Flag-IRES-GFP mice and crossed FGFR3-CreERT2 mice with Rosa26-mTmG, Rosa26-DTA and Rosa26-Confetti mice, respectively, to perform inducible genetic tracing studies. RESULTS: Expression of FGFR3 was found in the outer region of NP with co-localized expressions of proliferating markers. By fate mapping studies, FGFR3-positive (FGFR3+) NP cells were found proliferate from outer region to inner region of NP during postnatal growth. Clonal lineage tracing by Confetti mice and ablation of FGFR3·+ NP cells by DTA mice further revealed that the expansion of the FGFR3+ cells was required for the morphogenesis and homeostasis of postnatal NP. Moreover, in degeneration and regeneration model of mouse intervertebral disc, FGFR3+ NP cells underwent extensive expansion during the recovery stage. CONCLUSION: Our present work demonstrates that FGFR3+ NP cells are novel subpopulation of postnatal NP with long-existing proliferative capacity shaping the adult NP structure and participating in the homeostasis maintenance and intrinsic repair of NP. These findings may facilitate the development of new therapeutic approaches for IVD regeneration.


Assuntos
Degeneração do Disco Intervertebral , Disco Intervertebral , Dor Lombar , Núcleo Pulposo , Animais , Células Cultivadas , Degeneração do Disco Intervertebral/terapia , Camundongos , Núcleo Pulposo/metabolismo
14.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 28(6): 1251-6, 2011 Dec.
Artigo em Zh | MEDLINE | ID: mdl-22295724

RESUMO

Tumor cells exhibit two main different migration strategies when invading in 3D environment, i. e. mesenchymal migration and amoeboid migration. This review summarizes the internal reasons and characteristics on various modes of migration adaptation to the microenvironment, and the molecular mechanisms in particular environment where they are mutually interchangeable. A study of the mechanisms that may possibly trigger mesenchymal-amoeboid transition/amoeboid-mesenchymal transition help us to understand the change and the plasticity in the migration strategies of tumor cells. These are important for the development of a cancer treatment, which would efficiently suppress tumor cell invasiveness.


Assuntos
Movimento Celular/fisiologia , Invasividade Neoplásica , Metástase Neoplásica/fisiopatologia , Matriz Extracelular/patologia , Humanos , Integrinas/fisiologia
15.
J Orthop Translat ; 30: 41-50, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34611513

RESUMO

OBJECTIVE: Synovial fibrosis is a characteristic symptom of osteoarthritis (OA), which is closely associated with joint pain and stiffness. Previous studies have reported that low-intensity pulsed ultrasound (LIPUS) can alleviate cartilage degradation in OA. However, the functions and mechanisms of LIPUS in OA synovial fibrosis are still unknown. METHODS: The destabilization of the medial meniscus (DMM) mouse model of OA was established in C57 male mice and fibroblast-like synoviocytes (FLS) were isolated from synovial tissue of OA patients. The knee joint diameter, Masson's trichrome (MT) and Hematoxylin-eosin (HE) staining were used to evaluate synovial fibrosis and hyperplasia. The Immunohistochemistry (IHC) staining was performed to detected the expression of synovial fibrosis makers and the activation of Wnt/ß-catenin signaling in vivo. FLS were treated with TGF-ß1 to serve as an in vitro model of synovial fibrosis, Wnt3a was used to activate the Wnt/ß-catenin signaling in cells. Cell proliferation was detected by using EdU assay, cell viability was performed by CCK8 assay. The protein levels of α-SMA, CTGF, Col Ⅰ, ß-catenin, active ß-catenin, c-Myc and cyclin D1 were examined by western blot and immunofluorescence staining. RESULTS: Two weeks after the LIPUS treatment, the synovial fibrosis, synovial hyperplasia and synoviocyte proliferation in the DMM model were significantly decreased. In vitro, LIPUS directly inhibited the TGF-ß1-induced fibrotic response and proliferation of FLS. Meanwhile, LIPUS suppressed Wnt/ß-catenin signaling in the synovium of DMM mice and cultured FLS. More importantly, we found that the synovial fibrosis makers, Wnt/ß-catenin pathway downstream proteins and FLS proliferation were significantly decreased in Wnt3a-stimulated FLS following LIPUS treatment. CONCLUSIONS: Our results present a novel role of LIPUS in OA-related synovial fibrosis, which is associated with its ability to repress Wnt/ß-catenin signaling in FLS. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: This study provides new insight into the clinical application of LIPUS as a therapeutic option to manage synovial fibrosis in OA.

16.
Int J Biol Sci ; 17(15): 4140-4153, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34803488

RESUMO

Systemic application of glucocorticoids is an essential anti-inflammatory and immune-modulating therapy for severe inflammatory or autoimmunity conditions. However, its long-term effects on articular cartilage of patients' health need to be further investigated. In this study, we studied the effects of dexamethasone (Dex) on the homeostasis of articular cartilage and the progress of destabilization of medial meniscus (DMM)-induced osteoarthritis (OA) in adult mice. Long-term administration of Dex aggravates the proteoglycan loss of articular cartilage and drastically accelerates cartilage degeneration under surgically induced OA conditions. In addition, Dex increases calcium content in calcified cartilage layer of mice and the samples from OA patients with a history of long-term Dex treatment. Moreover, long term usage of Dex results in decrease subchondral bone mass and bone density. Further studies showed that Dex leads to calcification of extracellular matrix of chondrocytes partially through activation of AKT, as well as promotes apoptosis of chondrocytes in calcified cartilage layer. Besides, Dex weakens the stress-response autophagy with the passage of time. Taken together, our data indicate that long-term application of Dex may predispose patients to OA and or even accelerate the OA disease progression development of OA patients.


Assuntos
Apoptose/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Dexametasona/efeitos adversos , Matriz Extracelular/efeitos dos fármacos , Osteoartrite/etiologia , Animais , Calcinose , Dexametasona/administração & dosagem , Esquema de Medicação , Glucocorticoides/administração & dosagem , Glucocorticoides/efeitos adversos , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite/patologia
17.
ACS Nano ; 15(10): 15874-15891, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34586802

RESUMO

The clinical application of small interfering RNA (siRNA) drugs provides promising opportunities to develop treatment strategies for autoimmune inflammatory diseases. In this study, siRNAs targeting the endoplasmic reticulum to nucleus signaling 1 (ERN1) gene (siERN1) were screened. Two cationic polymers, polyethylenimine (PEI) and poly(ß-amino amine) (PBAA), which can improve the efficiency of the siRNA transfection, were used as siERN1 delivery carriers. They were implemented to construct a nanodrug delivery system with macrophage-targeting ability and dual responsiveness for the treatment of autoimmune inflammatory diseases. In terms of the mechanism, siERN1 can regulate the intracellular calcium ion concentration by interfering with the function of inositol 1,4,5-trisphosphate receptor 1/3 (IP3R1/3) and thus inducing M2 polarization of macrophages. Furthermore, siERN1-nanoprodrug [FA (folic acid)-PEG-R(RKKRRQRRR)-NPs(ss-PBAA-PEI)@siERN1] acts as a conductor of macrophage polarization by controlling the calcium ion concentration and is an inhibitor of MyD88-dependent Toll-like receptor signaling. The results revealed that the FA-PEG-R-NPs@siERN1 has universal biocompatibility, long-term drug release responsiveness, superior targeting properties, and therapeutic effects in mouse collagen-induced arthritis and inflammatory bowel disease models. In conclusion, this study reveals a potential strategy to treat autoimmune inflammatory disorders.


Assuntos
Polietilenoimina , Receptores Toll-Like , Animais , Macrófagos , Camundongos , RNA Interferente Pequeno , Transfecção
18.
Nat Commun ; 12(1): 4391, 2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34282140

RESUMO

Acquired heterotopic ossification (HO) is the extraskeletal bone formation after trauma. Various mesenchymal progenitors are reported to participate in ectopic bone formation. Here we induce acquired HO in mice by Achilles tenotomy and observe that conditional knockout (cKO) of fibroblast growth factor receptor 3 (FGFR3) in Col2+ cells promote acquired HO development. Lineage tracing studies reveal that Col2+ cells adopt fate of lymphatic endothelial cells (LECs) instead of chondrocytes or osteoblasts during HO development. FGFR3 cKO in Prox1+ LECs causes even more aggravated HO formation. We further demonstrate that FGFR3 deficiency in LECs leads to decreased local lymphatic formation in a BMPR1a-pSmad1/5-dependent manner, which exacerbates inflammatory levels in the repaired tendon. Local administration of FGF9 in Matrigel inhibits heterotopic bone formation, which is dependent on FGFR3 expression in LECs. Here we uncover Col2+ lineage cells as an origin of lymphatic endothelium, which regulates local inflammatory microenvironment after trauma and thus influences HO development via FGFR3-BMPR1a pathway. Activation of FGFR3 in LECs may be a therapeutic strategy to inhibit acquired HO formation via increasing local lymphangiogenesis.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Vasos Linfáticos/metabolismo , Ossificação Heterotópica/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Tendão do Calcâneo , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Endotélio Linfático/metabolismo , Técnicas de Silenciamento de Genes , Linfangiogênese , Masculino , Células-Tronco Mesenquimais , Camundongos , Tenotomia
19.
FEBS Open Bio ; 10(3): 434-443, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31975545

RESUMO

Low-intensity pulsed ultrasound (LIPUS), a noninvasive physical therapy, was recently demonstrated to be an effective treatment for osteoarthritis (OA). Vascular endothelium growth factor A (VEGFA) has been found to be upregulated in the articular cartilage, synovium and subchondral bone of OA patients, leading to cartilage degeneration, synovitis and osteophyte formation. However, the functions and mechanisms of LIPUS in regulating chondrocyte-derived VEGFA expression are still unclear. In this study, we investigated whether LIPUS attenuated OA progression by (a) decreasing the percentage of VEGFA-positive cells in mouse articular cartilage destabilised through medial meniscus surgery and (b) relieving interleukin-1ß-induced VEGFA expression in mouse primary chondrocytes. However, this function was negated by a p38 mitogen-activated protein kinase (p38 MAPK) inhibitor. In addition, we found that LIPUS ameliorated VEGFA-mediated disorders in cartilage extracellular matrix metabolism and chondrocyte hypertrophy during OA development. In conclusion, our data indicate a novel effect of LIPUS in regulating the expression of osteoarthritic chondrocyte-derived VEGFA through the suppression of p38 MAPK activity.


Assuntos
Cartilagem/metabolismo , Condrócitos/metabolismo , Terapia por Ultrassom/métodos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Cartilagem/fisiologia , Doenças das Cartilagens/metabolismo , Cartilagem Articular/metabolismo , Matriz Extracelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite/metabolismo , Osteoartrite/fisiopatologia , Osteófito/metabolismo , Substâncias Protetoras/metabolismo , Substâncias Protetoras/farmacologia , Sinovite/metabolismo , Ondas Ultrassônicas , Fator A de Crescimento do Endotélio Vascular/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/farmacologia
20.
Autophagy ; 16(7): 1262-1278, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31500508

RESUMO

Synovitis is implicated in the pathology of osteoarthritis (OA) and significantly contributes to the development of OA. As a noninvasive physical therapy, low-intensity pulsed ultrasound (LIPUS) has been reported to possess anti-inflammatory effect in recent years. However, the role of LIPUS on synovitis of OA and the underlying mechanisms are little known. The present study showed that LIPUS ameliorated synovial inflammation in destabilization of the medial meniscus (DMM) mouse model and air pouch model, and alleviated pain gait patterns of DMM mouse. LIPUS dramatically inhibited the production of mature IL1B/IL-1ß (interleukin 1 beta) in vitro and in vivo. In addition, LIPUS upregulated the macroautophagy/autophagy level as well as accelerated the formation of an SQSTM1 (sequestosome1)-PKM (pyruvate kinase, muscle) complex in the lipopolysaccharide (LPS)-adenosine triphosphate (ATP)-treated macrophages. Besides, LIPUS downregulated the level of PKM2 in LPS-ATP-treated macrophages, which could be reversed by SQSTM1 knockdown. In brief, the present study for the first time demonstrates that LIPUS inhibits the production of mature IL1B partially via SQSTM1-dependent autophagic degradation of PKM2 in LPS-ATP-treated macrophages, which may further ameliorate the synovial inflammation and gait patterns in animal models. Our data provide new clues for the treatments of synovitis and other inflammatory diseases using LIPUS. ABBREVIATIONS: 3-MA: 3-methyladenene; ATG7: autophagy-related 7; ATP: adenosine triphosphate; BafA1: bafilomycin A1; BMDMs: bone marrow derived macrophages; CHX: cycloheximide; DMM: destabilization of the medial meniscus; ELISA: enzyme-linked immunosorbent assay; GFP: green fluorescent protein; IL1B/IL-1ß: interleukin 1 beta; LIPUS: low-intensity pulsed ultrasound; LIR: LC3-interacting region; LPS: lipopolysaccharide; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MDP: muramyl dipeptide; NFKB/NF-κB: nuclear factor kappa B; NLRP3: NLR family, pyrin domain containing 3; OA: osteoarthritis; PKM/PKM2: pyruvate kinase M1/2; PMA: phorbol-12-myristate-13-acetate; PYCARD/ASC; PYD and CARD domain containing; RFP: red fluorescent protein; siRNAs: small interfering RNAs; SQSTM1: sequestosome 1; TEM: transmission electron microscopy.


Assuntos
Anti-Inflamatórios/farmacologia , Autofagia , Interleucina-1beta/metabolismo , Proteólise , Piruvato Quinase/metabolismo , Proteína Sequestossoma-1/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Autofagia/efeitos dos fármacos , Modelos Animais de Doenças , Marcha/efeitos dos fármacos , Humanos , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Meniscos Tibiais/patologia , Meniscos Tibiais/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Dor/patologia , Dor/fisiopatologia , Proteólise/efeitos dos fármacos , Células RAW 264.7 , Membrana Sinovial/patologia , Células THP-1 , Ondas Ultrassônicas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA