Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Aging Dis ; 15(1): 153-168, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37307832

RESUMEN

Reactive astrocytes (RAs) produce chondroitin sulfate proteoglycans (CSPGs) in large quantities after spinal cord injury (SCI) and inhibit axon regeneration through the Rho-associated protein kinase (ROCK) pathway. However, the mechanism of producing CSPGs by RAs and their roles in other aspects are often overlooked. In recent years, novel generation mechanisms and functions of CSPGs have gradually emerged. Extracellular traps (ETs), a new recently discovered phenomenon in SCI, can promote secondary injury. ETs are released by neutrophils and microglia, which activate astrocytes to produce CSPGs after SCI. CSPGs inhibit axon regeneration and play an important role in regulating inflammation as well as cell migration and differentiation; some of these regulations are beneficial. The current review summarized the process of ET-activated RAs to generate CSPGs at the cellular signaling pathway level. Moreover, the roles of CSPGs in inhibiting axon regeneration, regulating inflammation, and regulating cell migration and differentiation were discussed. Finally, based on the above process, novel potential therapeutic targets were proposed to eliminate the adverse effects of CSPGs.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato , Traumatismos de la Médula Espinal , Humanos , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Axones/metabolismo , Regeneración Nerviosa , Traumatismos de la Médula Espinal/tratamiento farmacológico , Inflamación
2.
Int Immunopharmacol ; 122: 110561, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37451018

RESUMEN

Aseptic inflammation is a major cause of late failure in total joint arthroplasty, and the primary factor contributing to the development and perpetuation of aseptic inflammation is classical macrophage activation (M1 phenotype polarization) induced by wear particles. CD73 (ecto-5'-nucleotidase) is an immunosuppressive factor that establishes an adenosine-induced anti-inflammatory environment. Although CD73 has been shown to suppress inflammation by promoting alternate macrophage activation (M2 phenotype polarization), its role in wear particle-induced aseptic inflammation is currently unknown. Our experiments were based on metabolomic assay results in a mouse model of aseptic loosening, and studied the function of CD73 in vivo and in vitro using a mouse aseptic loosening model and a mouse bone marrow derived macrophage (BMDM) inflammation model. Results show that aseptic loosening (AL) reduces the purine metabolic pathway and decreases the native expression of the metabolite adenosine. In vivo, CD73 expression was low in the bone tissue surrounding the titanium nail and synovial-like interface tissue, while in vitro experiments demonstrated that CD73 knockdown promoted titanium particles-induced aseptic inflammation. CD73 overexpression mitigated the titanium particle-mediated enhancement of LPS-induced M1 polarization while promoting the titanium particle-mediated attenuation of IL-4-induced M2 polarization. In BMDM exposed to titanium particles, CD73 promotes M2 polarization via the p38 pathway. Meanwhile, local injection of recombinant mouse CD73 protein slightly alleviated the progression of AL. Collectively, our data suggest that CD73 alleviates the process of AL, and this function is achieved by promoting alternate activation of macrophages.


Asunto(s)
Osteólisis , Titanio , Humanos , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Macrófagos/metabolismo , Inflamación/metabolismo , Adenosina/metabolismo , Osteólisis/metabolismo
3.
EMBO J ; 42(14): e113168, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37248947

RESUMEN

Enhanced expression of the cold-shock protein RNA binding motif 3 (RBM3) is highly neuroprotective both in vitro and in vivo. Whilst upstream signalling pathways leading to RBM3 expression have been described, the precise molecular mechanism of RBM3 cold induction remains elusive. To identify temperature-dependent modulators of RBM3, we performed a genome-wide CRISPR-Cas9 knockout screen using RBM3-reporter human iPSC-derived neurons. We found that RBM3 mRNA and protein levels are robustly regulated by several splicing factors, with heterogeneous nuclear ribonucleoprotein H1 (HNRNPH1) being the strongest positive regulator. Splicing analysis revealed that moderate hypothermia significantly represses the inclusion of a poison exon, which, when retained, targets the mRNA for nonsense-mediated decay. Importantly, we show that HNRNPH1 mediates this cold-dependent exon skipping via its thermosensitive interaction with a G-rich motif within the poison exon. Our study provides novel mechanistic insights into the regulation of RBM3 and provides further targets for neuroprotective therapeutic strategies.


Asunto(s)
Venenos , Humanos , Proteínas y Péptidos de Choque por Frío/metabolismo , Frío , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
4.
Biochem Pharmacol ; 211: 115516, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36966936

RESUMEN

Osteoporosis is a common inflammaging-related condition, where long-term accumulation of pro-inflammatory cytokines causes massive bone loss. Periplocin, a cardiotonic steroid isolated from Periploca forrestii, has been proved to reduce inflammation in several inflammatory diseases, such as rheumatoid arthritis. However, its effect and mechanism of inflammation in osteoporosis, in which pro-inflammatory factors accelerate bone loss, has not been well demonstrated. In this study, periplocin attenuated receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation of bone marrow-derived macrophages (BMMs) and RAW264.7 cells in vitro. It reduced osteoclast numbers and bone resorption in a concentration- and time-dependent manner. Further, periplocin treatment resulted in reduced bone loss on mice with ovariectomy-induced osteoporosis in vivo. By transcriptome sequencing, periplocin was indicated to function through inhibition of the mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signaling pathways and attenuating interactions between NF-κB and nuclear factor of activated T-cells 1 (NFATc1). It was further detected to bind low density lipoprotein receptor-related protein 4 (LRP4) in osteoclasts to exert anti-inflammatory and anti-osteoclastic effects. Overall, the findings have highlighted a better understanding for the anti-inflammatory and anti-osteoclastic role of periplocin in osteoporosis and its mechanism, bringing new possibilities for osteoporosis treatment.


Asunto(s)
Resorción Ósea , Osteoporosis , Animales , Femenino , Ratones , Antiinflamatorios/farmacología , Resorción Ósea/prevención & control , Resorción Ósea/metabolismo , Diferenciación Celular , Inflamación/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoclastos , Osteogénesis , Osteoporosis/tratamiento farmacológico , Osteoporosis/prevención & control , Ligando RANK/farmacología , Receptores de LDL/metabolismo
5.
J Bone Miner Res ; 38(5): 775-791, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36907987

RESUMEN

Osteoporosis is a common bone disease caused by an imbalance of bone resorption and formation that results in a loss of total bone density. SMAD2/3 signal transduction is known to play a crucial role in osteogenic differentiation through transforming growth factor-beta (TGF-ß). By screening a library of small-molecule compounds, the current study identifies higenamine (HG) as an active osteogenic agent that could be a therapeutic candidate for osteoporosis. In vitro data demonstrated that HG effectively induced expressions of osteogenic markers in mouse bone marrow stromal cell (BMSCs) and preosteoblastic cell cultures. Further, HG treatment resulted in enhanced bone formation and prevented accelerated bone loss on two animal models that mimic spontaneous senile osteoporosis and postmenopausal osteoporosis. IQ motif-containing GTPase-activating protein 1 (IQGAP1) was confirmed as a novel target of HG, where HG appears to bind to the Glu-1019 site of IQGAP1 to exert its osteogenic effects. Data subsequently suggested that HG promoted phosphorylation of SMAD2/3 and regulated the SMAD2/3 pathway by inhibiting SMAD4 ubiquitination. Overall, the findings highlight HG as a new small-molecule drug to promote bone formation through SMAD2/3 pathway in osteoporosis. © 2023 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Osteogénesis , Osteoporosis , Ratones , Animales , Transducción de Señal , Diferenciación Celular , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Estrógenos
6.
Neural Regen Res ; 18(5): 1076-1083, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36254996

RESUMEN

Studies have shown that gut microbiota metabolites can enter the central nervous system via the blood-spinal cord barrier and cause neuroinflammation, thus constituting secondary injury after spinal cord injury. To investigate the correlation between gut microbiota and metabolites and the possible mechanism underlying the effects of gut microbiota on secondary injury after spinal cord injury, in this study, we established mouse models of T8-T10 traumatic spinal cord injury. We used 16S rRNA gene amplicon sequencing and metabolomics to reveal the changes in gut microbiota and metabolites in fecal samples from the mouse model. Results showed a severe gut microbiota disturbance after spinal cord injury, which included marked increases in pro-inflammatory bacteria, such as Shigella, Bacteroides, Rikenella, Staphylococcus, and Mucispirillum and decreases in anti-inflammatory bacteria, such as Lactobacillus, Allobaculum, and Sutterella. Meanwhile, we identified 27 metabolites that decreased and 320 metabolites that increased in the injured spinal cord. Combined with pathway enrichment analysis, five markedly differential amino acids (L-leucine, L-methionine, L-phenylalanine, L-isoleucine and L-valine) were screened out, which play a pivotal role in activating oxidative stress and inflammatory responses following spinal cord injury. Integrated correlation analysis indicated that the alteration of gut microbiota was related to the differences in amino acids, which suggests that disturbances in gut microbiota might participate in the secondary injury through the accumulation of partial metabolites that activate oxidative stress and inflammatory responses. Findings from this study provide a new theoretical basis for improving the secondary injury after spinal cord injury through fecal microbial transplantation.

7.
Biomed Pharmacother ; 153: 113500, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36076590

RESUMEN

Spinal cord injury (SCI) triggers complex inflammatory and pathological processes that ultimately lead to scar formation, thus resulting in the severe and irreversible loss of function. The composition of spinal cord scars in SCI can be divided into cellular components and non-cellular components such as the extracellular matrix (ECM). Studies targeting the ECM, such as chondroitin sulfate proteoglycans (CSPGs), have achieved promising therapeutic results. On the other hand, with the application of new techniques in the field of SCI, an increasing body of research has been targeted to the complex mechanisms that occur in the cellular components during scar formation. Here, we meticulously describe the spatial distribution and dynamic changes of the cellular components involved in the formation of scars following SCI, including microglia, astrocytes, fibroblasts, and macrophages, paying particular attention to the recent application of new technologies. In addition, we outline the therapeutic approaches that target the cellular components of scar formation and explore potential therapeutic measures to improve functional recovery after SCI.


Asunto(s)
Gliosis , Traumatismos de la Médula Espinal , Astrocitos/patología , Proteoglicanos Tipo Condroitín Sulfato/uso terapéutico , Gliosis/metabolismo , Gliosis/patología , Humanos , Médula Espinal/patología , Traumatismos de la Médula Espinal/patología
8.
CNS Neurosci Ther ; 28(11): 1883-1894, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35906830

RESUMEN

AIM: To investigate the effect of apigenin on fibrous scar formation after mouse spinal cord injury (SCI). METHODS: The pneumatic impactor strike method was used to establish an SCI model. Mice were intraperitoneally injected with 5 mg/kg or 20 mg/kg apigenin daily for 28 days after SCI. The Basso Mouse Scale (BMS) score, hematoxylin-eosin staining, and immunohistochemical staining were used to assess the effect of apigenin on scar formation and motor function recovery. Western blotting and qRT-PCR were used to detect the expression of fibrosis-related parameters in spinal cord tissue homogenates. NIH-3 T3 cells and mouse primary spinal cord fibroblasts, α-Smooth muscle actin (α-SMA), collagen 1, and fibronectin were used to evaluate apigenin's effect in vitro. Western blotting and immunofluorescence techniques were used to study the effect of apigenin on TGFß/SMADs signaling. RESULTS: Apigenin inhibited fibrous scar formation in the mouse spinal cord and promoted the recovery of motor function. It reduced the expression of fibroblast-related parameters and increased the content of nerve growth factor in vivo, decreasing myofibroblast activation and collagen fiber formation by inhibiting TGFß-induced SMAD2/3 phosphorylation and nuclear translocation in vitro. CONCLUSION: Apigenin inhibits fibrous scar formation after SCI by decreasing fibrosis-related factor expression through TGFß/SMADs signaling.


Asunto(s)
Cicatriz , Traumatismos de la Médula Espinal , Actinas/metabolismo , Animales , Apigenina/farmacología , Apigenina/uso terapéutico , Cicatriz/tratamiento farmacológico , Cicatriz/etiología , Cicatriz/metabolismo , Colágeno/metabolismo , Colágeno/farmacología , Eosina Amarillenta-(YS)/metabolismo , Eosina Amarillenta-(YS)/farmacología , Fibronectinas/metabolismo , Fibronectinas/farmacología , Hematoxilina/metabolismo , Hematoxilina/farmacología , Ratones , Factores de Crecimiento Nervioso/metabolismo , Recuperación de la Función , Transducción de Señal , Médula Espinal/patología , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
9.
FASEB J ; 35(12): e22014, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34751973

RESUMEN

Spinal cord injury (SCI) is a complex pathological change that includes primary SCI and gradually evolves into secondary SCI. Accumulating evidence demonstrates that circular RNAs (circRNAs) are involved in the pathology of a variety of neurological diseases and injuries. However, the characteristics and function of circRNAs in SCI have yet to be elucidated. Although previous research demonstrated that circPrkcsh induces astrocytes to produce inflammatory factors and chemokines, the precise function and mechanism of circPrkcsh in microglia after SCI remains unknown. In this study, we constructed a mouse model of SCI by applying a SCI impactor. Quantitative Real-time PCR and Fluorescence in situ hybridization analysis revealed that circPrkcsh was upregulated in the microglia of SCI mice when compared to sham-operated mice. Gain- or loss-of-function experiments and in vivo assays further indicated that circPrkcsh promotes microglia M1 polarization both in vivo and in vitro. Furthermore, bioinformatics analysis, dual-luciferase assays, and RNA immunoprecipitation assays, confirmed that circPrkcsh serves as a competing endogenous RNA (ceRNA) to promote the expression of MEKK1 mRNA by sponging miR-488. Double knockout rescue experiments further showed that circPrkcsh regulates the MEKK1/JNK/p38 MAPK pathway via miR-488. Our research provides a better understanding of the mechanism of circPrkcsh in SCI and demonstrates that the circPrkcsh/miR-488/Mekk1 axis is a promising regulatory method for the treatment of SCI.


Asunto(s)
Glucosidasas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Sistema de Señalización de MAP Quinasas , Macrófagos/inmunología , MicroARNs/genética , ARN Circular/genética , Traumatismos de la Médula Espinal/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Transducción de Señal , Traumatismos de la Médula Espinal/etiología , Traumatismos de la Médula Espinal/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
10.
Int J Biol Sci ; 17(11): 2795-2810, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34345208

RESUMEN

Reactive astrocytes are implicated in traumatic spinal cord injury (TSCI). Interestingly, naïve astrocytes can easily transform into neurotoxic reactive astrocytes (A1s) with inflammatory stimulation. Previous studies demonstrated that microRNA(miR)-21a-5p was up-regulated in spinal cord tissue after TSCI; however, it is not clear whether this affected reactive astrocyte polarization. Here, we aim to detect the effects of miR-21a-5p on the induction of A1 formation and the underlying mechanisms. Our study found that the expression of miR-21a-5p was significantly increased while that of Cntfr α was decreased, since naïve astrocytes transformed into A1s 3 days post-TSCI; the binding site between miR-21a-5p and Cntfr α was further confirmed in astrocytes. After treatment with CNTF, the levels of A1 markers decreased while that of A2 increased. The expression of A1 markers significantly decreased with the downregulation of miR-21a-5p, while Cntfr α siRNA treatment caused the opposite both in vitro and in vivo. To summarize, miR-21a-5p/Cntfr α promotes A1 induction and might enhance the inflammatory process of TSCI; furthermore, we identified, for the first time, the effect and potential mechanism by which CNTF inhibits naïve astrocytes transformation into A1s. Collectively, our findings demonstrate that targeting miR-21a-5p represents a prospective therapy for promoting the recovery of TSCI.


Asunto(s)
Astrocitos/metabolismo , Subunidad alfa del Receptor del Factor Neurotrófico Ciliar/metabolismo , Factor Neurotrófico Ciliar/metabolismo , MicroARNs/metabolismo , Factor de Transcripción STAT3/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Astrocitos/citología , Modelos Animales de Enfermedad , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Traumatismos de la Médula Espinal/patología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA