Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
Chin J Traumatol ; 27(3): 168-172, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38262890

RESUMO

PURPOSE: To identify the risk factors for training-related lower extremity muscle injuries in young males by a non-invasive method of body composition analysis. METHODS: A total of 282 healthy young male volunteers aged 18 - 20 years participated in this cohort study. Injury location, degree, and injury rate were adjusted by a questionnaire based on the overuse injury assessment methods used in epidemiological studies of sports injuries. The occurrence of training injuries is monitored and diagnosed by physicians and treated accordingly. The body composition was measured using the BodyStat QuadScan 4000 multifrequency Bio-impedance system at 5, 50, 100 and 200 kHz to obtain 4 impedance values. The Shapiro-Wilk test was used to check whether the data conformed to a normal distribution. Data of normal distribution were shown as mean ± SD and analyzed by t-test, while those of non-normal distribution were shown as median (Q1, Q3) and analyzed by Wilcoxon rank sum test. The receiver operator characteristic curve and logistic regression analysis were performed to investigate risk factors for developing training-related lower extremity injuries and accuracy. RESULTS: Among the 282 subjects, 78 (27.7%) developed training injuries. Lower extremity training injuries revealed the highest incidence, accounting for 23.4% (66 cases). These patients showed higher percentages of lean body mass (p = 0.001), total body water (TBW, p = 0.006), extracellular water (p = 0.020) and intracellular water (p = 0.010) as well as a larger ratio of basal metabolic rate/total weight (p = 0.006), compared with those without lower extremity muscle injuries. On the contrary, the percentage of body fat (p = 0.001) and body fat mass index (p = 0.002) were lower. Logistic regression analysis showed that TBW percentage > 65.35% (p = 0.050, odds ratio = 3.114) and 3rd space water > 0.95% (p = 0.045, odds ratio = 2.342) were independent risk factors for lower extremity muscle injuries. CONCLUSION: TBW percentage and 3rd space water measured with bio-impedance method are potential risk factors for predicting the incidence of lower extremity muscle injuries in young males following training.


Assuntos
Água Corporal , Extremidade Inferior , Músculo Esquelético , Humanos , Masculino , Fatores de Risco , Adulto Jovem , Adolescente , Extremidade Inferior/lesões , Músculo Esquelético/lesões , Traumatismos em Atletas/epidemiologia , Composição Corporal , Estudos de Coortes
3.
BMC Ophthalmol ; 24(1): 32, 2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38254055

RESUMO

PURPOSE: To investigate the changes in aqueous humor (AH) protein profiles before and after intravitreal aflibercept (IVA) treatment in patients with proliferative diabetic retinopathy (PDR). METHODS: 5 PDR patients provided 10 samples of AH before and after IVA treatment (pre-group vs. post-group). Proteins were identified using liquid chromatography-tandem mass spectrometry. Then, bioinformatics was employed to investigate the functional significance of differentially expressed proteins (DEPs) and hub proteins. RESULTS: A total of 16 DEPs were identified, consisting of 8 downregulated proteins and 8 upregulated proteins. Bioinformatics analysis indicated that the most significantly enriched biological process was "blood coagulation, intrinsic pathway." The most significantly enriched signaling pathway was "complement and coagulation cascades." HBB, HPX, VEGFA, and CA1 were identified as hub proteins for IVA treatment. CONCLUSIONS: Together with the downregulation of the intravitreal vascular endothelial growth factor level, IVA may also change the AH protein composition in PDR patients, with DEPs involved in the blood coagulation, intrinsic pathway, complement, and coagulation cascades. IVA treatment may protect against PDR by regulating HBB, HPX, VEGFA, and CA1 expression.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Proteínas Recombinantes de Fusão , Humanos , Humor Aquoso , Retinopatia Diabética/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico
4.
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
5.
Heliyon ; 9(11): e21531, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38027717

RESUMO

Background: Developing effective therapeutic strategies to delay the progression of chronic kidney disease (CKD) remains a significant challenge. Low-intensity pulsed ultrasound (LIPUS) has demonstrated potential for treating CKD, but the underlying molecular mechanisms are still elusive. This study aimed to evaluate the therapeutic efficacy of LIPUS and to elucidate the involved genes and signaling pathways. Methods: The CKD model was established in rats using Adriamycin (ADR). The bilateral kidneys of CKD rats were continuously stimulated with LIPUS for a period of four weeks. The therapeutic efficacy was defined by renal function and histopathological evaluation. RNA sequencing was employed to profile the transcriptome of rat kidneys in each group. Cluster analysis was utilized to identify differentially expressed genes (DEGs), followed by enrichment analysis of their associated pathways using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. Results: LIPUS treatment improved ADR-induced renal dysfunction in the CKD group. Renal fibrosis and pathological damages were also alleviated in the ADR + LIPUS group compared to the ADR group. Cluster analysis identified 844 DEGs. GO enrichment analysis revealed enrichment in inflammatory response terms, while KEGG enrichment analysis highlighted the nuclear factor kappa B (NF-κB) signaling and ferroptosis-related pathways. Conclusion: Continuous LIPUS treatment improved ADR-induced renal fibrosis and dysfunction. The therapeutic effect of LIPUS was primarily due to its ability to suppress the CKD-related inflammation, which was associated with the modulation of the NF-κB and ferroptosis signaling pathways. These findings provide a new insight into the potential molecular mechanisms of LIPUS in treating CKD. Further research is necessary to confirm these findings and to identify potential therapeutic targets within these pathways.

6.
Front Pediatr ; 11: 1183345, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37435167

RESUMO

Persistent fifth aortic arch (PFAA) is an extremely rare congenital cardiovascular anomaly resulting from the failure of the fifth aortic arch to degenerate during embryonic development; it is often associated with various other cardiovascular anomalies. Despite being first reported by Van Praagh in 1969, there have been only a few individual case reports. Owing to its rarity and lack of comprehensive understanding, PFAA is often misdiagnosed or missed diagnosed during clinical. Thus, this review aimed to summarise the embryonic development, pathological classification, imaging diagnosis, and clinical treatment of PFAA to improve its overall understanding, ultimately helping in accurate diagnosis and treatment.

7.
J Proteomics ; 276: 104838, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36764651

RESUMO

To investigate the changes in aqueous humor (AH) protein profiles before and after intravitreal conbercept (IVC) treatment in proliferative diabetic retinopathy (PDR) patients. Ten PDR patients provided 20 samples of AH before (pre group) and after (post group) IVC treatment. Liquid chromatography-tandem mass spectrometry was performed to identify proteins. Bioinformatics analysis was used to explore the functional relevance of differentially expressed proteins (DEPs) and hub proteins. Parallel reaction monitoring (PRM) method was used to verify the hub proteins in another 8 samples of AH before and after IVC treatment in 4 PDR patients. A total of 30 DEPs were identified, consisting of 14 downregulated proteins and 16 upregulated proteins. Bioinformatics analysis indicated that DEPs mostly involved in neutrophil degranulation, antioxidant activity, secretory granule lumen, cytoplasmic vesicle lumen, vesicle lumen, and fluid shear stress. HP, VEGFA, CTSD, and LYZ were identified as hub proteins, among which HP and CTSD were verified by PRM. In addition to decreasing the intravitreal vascular endothelial growth factor level, IVC may alter the AH protein profile in PDR patients, especially HP and CTSD, with the DEPs involved in neutrophil degranulation, antioxidant activity, secretory granule lumen, cytoplasmic vesicle lumen, vesicle lumen, and fluid shear stress. SIGNIFICANCE: Patients with proliferative diabetic retinopathy (PDR) regularly receive intravitreal conbercept treatment these days. The effect of this treatment has been determined by previous studies. However, the mechanism of IVC in PDR is not eventually determined. No studies have compared the aqueous humor (AH) protein profile before and after IVC treatment in the same patient. This is a topic deserving of further exploration. A proteomic method based on liquid chromatography-tandem mass spectrometry (LC-MS/MS) was utilized in this study to analyze and assess the AH samples to explore the mechanism underlying the effects of IVC treatment on PDR.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Humanos , Retinopatia Diabética/metabolismo , Cromatografia Líquida/métodos , Fator A de Crescimento do Endotélio Vascular , Humor Aquoso/metabolismo , Proteômica/métodos , Antioxidantes , Espectrometria de Massas em Tandem/métodos
8.
Food Chem ; 397: 133833, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-35933751

RESUMO

Reports on meat quality and flavor evaluation of Nanjing water boiled salted duck (NWSD) produced by different Muscovy duck (Cairina moschata) ingredients are limited. To select a suitable Muscovy duck ingredient for the NWSD processing, six kinds of NWSD products were produced using female (65, 70, and 75 days) and male (75, 80, and 85 days) Muscovy duck ingredients. The meat quality, volatile organic compounds (VOCs), smell and taste were investigated by using colorimeter, texture analyzer, headspace-gas chromatography-ion mobility spectroscopy (HS-GC-IMS), electronic nose (E-nose), electronic tongue (E-tongue), etc. Results exhibited that 32 iconic VOCs were obtained by using partial least squares discrimination analysis (PLS-DA), principal component analysis (PCA), and variable importance projection (VIP) methods. 80-day-old male Muscovy duck showed moderate moisture and protein content, good meat texture and bright color, diverse iconic VOCs and clear differentiation, making it the preferred ingredient for NWSD processing.


Assuntos
Patos , Compostos Orgânicos Voláteis , Animais , Patos/metabolismo , Nariz Eletrônico , Feminino , Masculino , Carne , Olfato , Compostos Orgânicos Voláteis/metabolismo , Água/metabolismo
9.
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
10.
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.

11.
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
12.
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
13.
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
14.
Theranostics ; 10(16): 7111-7130, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32641982

RESUMO

CATSHL syndrome, characterized by camptodactyly, tall stature and hearing loss, is caused by loss-of-function mutations of fibroblast growth factor receptors 3 (FGFR3) gene. Most manifestations of patients with CATSHL syndrome start to develop in the embryonic stage, such as skeletal overgrowth, craniofacial abnormalities, however, the pathogenesis of these phenotypes especially the early maldevelopment remains incompletely understood. Furthermore, there are no effective therapeutic targets for this skeleton dysplasia. Methods: We generated fgfr3 knockout zebrafish by CRISPR/Cas9 technology to study the developmental mechanisms and therapeutic targets of CATSHL syndrome. Several zebrafish transgenic lines labeling osteoblasts and chondrocytes, and live Alizarin red staining were used to analyze the dynamical skeleton development in fgfr3 mutants. Western blotting, whole mount in situ hybridization, Edu labeling based cell proliferation assay and Wnt/ß-catenin signaling antagonist were used to explore the potential mechanisms and therapeutic targets. Results: We found that fgfr3 mutant zebrafish, staring from early development stage, showed craniofacial bone malformation with microcephaly and delayed closure of cranial sutures, chondroma-like lesion and abnormal development of auditory sensory organs, partially resembling the clinical manifestations of patients with CATSHL syndrome. Further studies showed that fgfr3 regulates the patterning and shaping of pharyngeal arches and the timely ossification of craniofacial skeleton. The abnormal development of pharyngeal arch cartilage is related to the augmented hypertrophy and disordered arrangement of chondrocytes, while decreased proliferation, differentiation and mineralization of osteoblasts may be involved in the delayed maturation of skull bones. Furthermore, we revealed that deficiency of fgfr3 leads to enhanced IHH signaling and up-regulated canonical Wnt/ß-catenin signaling, and pharmacological inhibition of Wnt/ß-catenin could partially alleviate the phenotypes of fgfr3 mutants. Conclusions: Our study further reveals some novel phenotypes and underlying developmental mechanism of CATSHL syndrome, which deepens our understanding of the pathogenesis of CATSHL and the role of fgfr3 in skeleton development. Our findings provide evidence that modulation of Wnt/ß-catenin activity could be a potential therapy for CATSHL syndrome and related skeleton diseases.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Condrócitos/patologia , Condrogênese/genética , Deformidades Congênitas da Mão/genética , Perda Auditiva/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Crânio/embriologia , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Doenças do Desenvolvimento Ósseo/patologia , Sistemas CRISPR-Cas/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Embrião não Mamífero , Técnicas de Inativação de Genes , Deformidades Congênitas da Mão/patologia , Perda Auditiva/patologia , Proteínas Hedgehog/metabolismo , Humanos , Mutação , Via de Sinalização Wnt/genética , Peixe-Zebra
15.
Bone Res ; 8: 25, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32596023

RESUMO

Exosomes participate in many physiological and pathological processes by regulating cell-cell communication, which are involved in numerous diseases, including osteoarthritis (OA). Exosomes are detectable in the human articular cavity and were observed to change with OA progression. Several joint cells, including chondrocytes, synovial fibroblasts, osteoblasts, and tenocytes, can produce and secrete exosomes that influence the biological effects of targeted cells. In addition, exosomes from stem cells can protect the OA joint from damage by promoting cartilage repair, inhibiting synovitis, and mediating subchondral bone remodeling. This review summarizes the roles and therapeutic potential of exosomes in OA and discusses the perspectives and challenges related to exosome-based treatment for OA patients in the future.

16.
Biomed Res Int ; 2020: 4307385, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32596310

RESUMO

Synovitis plays an important role in the pathogenesis of arthritis, which is closely related to the joint swell and pain of patients. The purpose of this study was to investigate the anti-inflammatory effects of pulsed electromagnetic fields (PEMF) on synovitis and its underlying mechanisms. Destabilization of the medial meniscus (DMM) model and air pouch inflammation model were established to induce synovitis in C57BL/6 mice. The mice were then treated by PEMF (pulse waveform, 1.5 mT, 75 Hz, 10% duty cycle). The synovitis scores as well as the levels of IL-1ß and TNF-α suggested that PEMF reduced the severity of synovitis in vivo. Moreover, the proportion of neutrophils in the synovial-like layer was decreased, while the proportion of macrophages increased after PEMF treatment. In addition, the phagocytosis of apoptotic neutrophils by macrophages (efferocytosis) was enhanced by PEMF. Furthermore, the data from western blot assay showed that the phosphorylation of P38 was inhibited by PEMF. In conclusion, our current data show that PEMF noninvasively exhibits the anti-inflammatory effect on synovitis via upregulation of the efferocytosis in macrophages, which may be involved in the phosphorylation of P38.


Assuntos
Campos Eletromagnéticos , Macrófagos/efeitos da radiação , Fagocitose/efeitos da radiação , Sinovite/radioterapia , Animais , Apoptose/efeitos da radiação , Masculino , Camundongos Endogâmicos C57BL
17.
Nat Commun ; 11(1): 479, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980602

RESUMO

Congenital scoliosis (CS) is a complex genetic disorder characterized by vertebral malformations. The precise etiology of CS is not fully defined. Here, we identify that mutation in dual serine/threonine and tyrosine protein kinase (dstyk) lead to CS-like vertebral malformations in zebrafish. We demonstrate that the scoliosis in dstyk mutants is related to the wavy and malformed notochord sheath formation and abnormal axial skeleton segmentation due to dysregulated biogenesis of notochord vacuoles and notochord function. Further studies show that DSTYK is located in late endosomal/lysosomal compartments and is involved in the lysosome biogenesis in mammalian cells. Dstyk knockdown inhibits notochord vacuole and lysosome biogenesis through mTORC1-dependent repression of TFEB nuclear translocation. Inhibition of mTORC1 activity can rescue the defect in notochord vacuole biogenesis and scoliosis in dstyk mutants. Together, our findings reveal a key role of DSTYK in notochord vacuole biogenesis, notochord morphogenesis and spine development through mTORC1/TFEB pathway.


Assuntos
Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Escoliose/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/anormalidades , Peixe-Zebra/genética , Transporte Ativo do Núcleo Celular , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Modelos Biológicos , Mutação , Notocorda/anormalidades , Notocorda/metabolismo , Notocorda/ultraestrutura , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Escoliose/congênito , Escoliose/metabolismo , Transdução de Sinais , Coluna Vertebral/anormalidades , Coluna Vertebral/metabolismo , Fatores de Transcrição/metabolismo , Vacúolos/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/metabolismo
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...