Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell Signal ; 121: 111275, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38942343

RESUMO

Keloid formation, characterized by aberrant fibroproliferation and immune dysregulation, remains a challenging clinical concern. This study aims to elucidate the neuroimmune mechanisms underlying keloid pathogenesis and explores the efficacy of a combined treatment approach involving modulation of the α7 nicotinic acetylcholine receptor (α7nAchR), a key player in neural transmission, and programmed death ligand 1 (PD-L1), an immune checkpoint molecule, for keloid intervention. A key innovation lies in the identification of signal peptide-CUB-EGF-like domain-containing protein 3 (SCUBE3) as a potential target gene influenced by this dual treatment. We elucidate the underlying mechanism, wherein the hypoxic keloid microenvironment fosters an upsurge in SCUBE3 secretion. Subsequently, SCUBE3 forms complexes with TGF-ß, initiating the activation of the PI3K/AKT/NF-κB signaling pathway. Notably, SCUBE3 is secreted in the form of exosomes, thereby exerting a profound influence on the differentiation of T cells and macrophages within the keloid milieu. This research not only provides a comprehensive understanding of the molecular mechanisms involved but also offers a promising avenue for the development of targeted therapies to address keloid-associated fibrosis and immune dysregulation. In conclusion, the combined inhibition of α7nAchR and PD-L1 represents a promising therapeutic strategy with SCUBE3 as a pivotal molecular target in the complex landscape of keloid pathophysiology.


Assuntos
Antígeno B7-H1 , Queloide , Receptor Nicotínico de Acetilcolina alfa7 , Humanos , Antígeno B7-H1/metabolismo , Queloide/metabolismo , Queloide/patologia , Queloide/imunologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Transdução de Sinais , Fosfatidilinositol 3-Quinases/metabolismo
2.
Cell Prolif ; 55(12): e13318, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35932176

RESUMO

OBJECTIVES: Diabetes mellitus is associated with refractory wound healing, yet current therapies are insufficient to accelerate the process of healing. Recent studies have indicated chemically modified mRNA (modRNA) as a promising therapeutic intervention. The present study aimed to explore the efficacy of small skin engineered to express modified mRNAs encoding the stromal cell-derived factor-1α (SDF-1α) facilitating wound healing in a full-thickness skin defect rat model. This study, devised therapeutic strategies for diabetic wounds by pre-treating small skin with SDF-1α modRNA. MATERIALS AND METHODS: The in vitro transfection efficiency was evaluated using fluorescence microscopy and the content of SDF-1α in the medium was determined using ELISA after the transfection of SDF-1α into the small skin. To evaluate the effect of SDF-1α modRNA and transplantation of the small skin cells on wound healing, an in vivo full-thickness skin defect rat model was assessed. RESULTS: The results revealed that a modRNA carrying SDF-1α provided potent wound healing in the small skin lesions reducing reduced scar thickness and greater angiogenesis (CD31) in the subcutaneous layer. The SDF-1α cytokines were significantly secreted by the small skin after transfection in vitro. CONCLUSIONS: This study demonstrated the benefits of employing small skin combined with SDF-1α modRNA in enhancing wound healing in diabetic rats having full-thickness skin defects.


Assuntos
Quimiocina CXCL12 , Diabetes Mellitus Experimental , Ratos , Animais , Quimiocina CXCL12/genética , Quimiocina CXCL12/farmacologia , RNA Mensageiro/genética , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/patologia , Neovascularização Fisiológica , Cicatrização , Pele/patologia
3.
J Control Release ; 347: 237-255, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35489544

RESUMO

Osteoarthritis (OA) is a chronic disease caused by joint inflammation. Its occurrence and development depend on a continuous inflammation environment. The activated M1 macrophages play a critical role in the inflammatory response of OA. Regulating the pro-inflammatory M1 to anti-inflammatory M2 macrophages in the OA articular cavity could be a rational strategy for OA treatment. It has been acknowledged that activated macrophages could proactively capture opsonized nanoparticles in the bloodstream and then accumulate into the reticuloendothelial system (RES) organs. Based on this fact, a trapping strategy is proposed, which transforms a normal nanoparticle into an opsonized attractant to target and regulate macrophage polarization. In this study, the opsonized nanoparticle (IgG/Bb@BRPL) had several key features, including an immunoglobulin IgG (the opsonized layer), an anti-inflammatory agent berberine (Bb), and an oxidative stress-responsive bilirubin grafted polylysine biomaterial (BR-PLL) for drug loading (the inner nanocore). In vitro studies confirmed that IgG/Bb@BRPL prefer to be phagocytosed by M1 macrophage, not M0. And the internalized IgG/Bb@BRPL effectively promoted macrophage polarization toward the M2 phenotype and protected nearby chondrocytes. In vivo studies suggested that IgG/Bb@BRPL significantly enhanced therapeutic outcomes by suppressing inflammation and promoting cartilage repair while not prolonging the retention period compared to non-opsonized counterparts. This proof-of-concept study provided a novel opsonization trapping strategy for OA drug delivery and treatment.


Assuntos
Nanopartículas , Osteoartrite , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Humanos , Imunoglobulina G/farmacologia , Inflamação , Macrófagos , Osteoartrite/tratamento farmacológico
5.
Front Cell Dev Biol ; 9: 623959, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33614652

RESUMO

Random skin flaps are frequently applied in plastic and reconstructive surgery for patients suffering from soft tissue defects caused by congenital deformities, trauma and tumor resection. However, ischemia and necrosis in distal parts of random skin flaps remains a common challenge that limits the clinical application of this procedure. Recently, chemically modified mRNA (modRNA) was found to have great therapeutic potential. Here, we explored the potential of fibroblasts engineered to express modified mRNAs encoding the stromal cell-derived factor-1α (SDF-1α) to improve vascularization and survival of therapeutic random skin flaps. Our study showed that fibroblasts pre-treated with SDF-1α modRNA have the potential to salvage ischemic skin flaps. Through a detailed analysis, we revealed that a fibroblast SDF-1α modRNA combinatorial treatment dramatically reduced tissue necrosis and significantly promoted neovascularization in random skin flaps compared to that in the control and vehicle groups. Moreover, SDF-1α modRNA transcription in fibroblasts promoted activation of the SDF-1α/CXCR4 pathway, with concomitant inactivation of the MEK/ERK, PI3K/AKT, and JAK2/STAT3 signaling pathways, indicating a possible correlation with cell proliferation and migration. Therefore, fibroblast-mediated SDF-1α modRNA expression represents a promising strategy for random skin flap regeneration.

6.
Front Pharmacol ; 11: 761, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32581781

RESUMO

Spinal cord injury (SCI) is a clinical tough neurological problem without efficient cure currently. Blood-spinal cord barrier (BSCB) interruption is not only a crucial pathological feature for SCI process but is a possible target for future SCI treatments; however, few treatments have been developed to intervene BSCB. In the present study, we intravenously injected CO-releasing molecule3 (CORM-3), a classical exogenous CO donor, to the rats experiencing SCI and assessed its protection on BSCB integrity in rats. Our results demonstrated that the exogenous increasing of CO by CORM-3 blocked the tight junction (TJ) protein degeneration and neutrophils infiltration, subsequently suppressed the BSCB damage and improved the motor recovery after SCI. And we certified that the CO-induced down-regulation of MMP-9 expression and activity in neutrophil might be associated with the NF-κB signaling. Taken together, our study indicates that CO-releasing molecule (CORM)-3 ameliorates BSCB after spinal cord injury.

7.
Front Pharmacol ; 11: 107, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32161544

RESUMO

Osteoarthritis (OA) is a chronic degenerative disease wherein the articular cartilage exhibits inflammation and degradation. Scutellarin (SCU) is a flavonoid glycoside with a range of pharmacological activities, as shown in previous studies demonstrating its anti-inflammatory activity. How SCU impacts the progression of OA, however, has not been explored to date. Herein, we assessed the impact of SCU on murine chondrocytes in an OA model system. In in vitro assays, we measured chondrocyte expression of key OA-associated factors such as matrix metalloproteinase 13 (MMP-13), a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS-5), cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) via qRT-PCR and Western blotting, the expression of interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), and prostaglandin E2 (PGE2) were detected by qRT-PCR. Our results showed that the downregulation of MMP-13, ADAMTS-5, COX-2, and iNOS expression by SCU and the overproduction of IL-6, TNF-α, and PGE2 induced by IL-1ß were all inhibited by SCU in a concentration-dependent manner. Moreover, SCU was able to reverse aggrecan and collagen II degradation and nuclear factor-κB (NF-κB) and nuclear factor erythroid-derived 2-like 2 (Nrf2) signaling pathway activation both in vivo and in vitro. We further used a destabilization of the medial meniscus (DMM) murine model of OA to explore the therapeutic benefits of SCU in vivo. Together, our findings suggest SCU to be a potentially valuable therapeutic agent useful for treating OA.

8.
Front Pharmacol ; 10: 393, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31040782

RESUMO

Osteoarthritis (OA), defined as a long-term progressive joint disease, is characterized by cartilage impairment and erosion. In recent decades, magnolol, as a type of lignin extracted from Magnolia officinalis, has been proved to play a potent anti-inflammatory role in various diseases. The current research sought to examine the latent mechanism of magnolol and its protective role in alleviating the progress of OA in vivo as well as in vitro experimentations. In vitro, the over-production of Nitric oxide (NO), prostaglandin E2 (PGE2), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6), induced by interleukin-1 beta (IL-1ß), were all inhibited notably by magnolol in a concentration-dependent manner. Moreover, magnolol could also downregulate the expression of metalloproteinase 13 (MMP13) and thrombospondin motifs 5 (ADAMTS5). All these changes ultimately led to the deterioration of the extracellular matrix (ECM) induced by IL-1ß. Mechanistically, magnolol suppressed the activation of PI3K/Akt/NF-κB pathway. Furthermore, a powerful binding capacity between magnolol and PI3K was also revealed in our molecular docking research. In addition, magnolol-induced protective effects in OA development were also detected in a mouse model. In summary, this research suggested that magnolol possessed a new therapeutic potential for the development of OA.

9.
Am J Transl Res ; 11(3): 1417-1427, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30972171

RESUMO

Cartilage lesion repair is difficult due to the limited self-repair capability of cartilage and its lack of vascularization. Our previous study established a sandwich model for engineering cartilage with acellular cartilage sheets (ACSs) and chondrocytes. However, there is still debate over which agent achieves the optimal decellularization of cartilage sheets. In addition, changes in the extracellular matrix after decellularization are worth studying. We aimed to determine the optimal decellularization reagents and decellularization time for preparing cartilage sheets. This study compared the effects of 2 extraction chemicals [t-octylphenoxypolyethoxyethanol (Triton X-100) and sodium dodecyl sulfate (SDS)] on cartilage sheets. The sheets were soaked in various concentrations (0.1-2%) of the extraction solutions for various time periods (24-72 h). After the decellularization process with the various treatments, we examined the cell removal and preservation of the matrix components and microstructure to determine which method was the most efficient while inducing minimal damage to the perichondrium. Both protocols achieved decellularization within an acceptable time. DNA analysis showed that the reagent removed nearly all of the DNA from the cartilage sheets. The growth factor contents in the Triton X-100 samples were higher than those in the SDS samples, quantified by enzyme-linked immunosorbent assay (ELISA). Furthermore, Triton X-100 decreased the glycosaminoglycan (GAG) and increased the chondromodulin-I contents compared with SDS. The results of a Cell Counting Kit-8 (CCK-8) assay revealed that the ACSs were not cytotoxic. In conclusion, our results demonstrate that cartilage sheets decellularized by 1% SDS for 24 h or by 2% Triton X-100 for 48 h may be suitable candidate scaffolds for cartilage tissue engineering.

10.
Food Funct ; 10(4): 2198-2208, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30942801

RESUMO

Osteoarthritis (OA) is a common arthrosis characterized by degeneration and inflammation of articular cartilage. In recent decades, peiminine (Pm) has been identified as one of the active ingredients of Fritillaria plants. According to reports, Pm has a potent anti-inflammatory effect in various diseases. However, the effectiveness of Pm as an anti-inflammatory in OA has not previously been reported. This research aims to evaluate the anti-inflammatory effect of Pm on interleukin (IL)-1ß-induced mice chondrocytes and its chondroprotective effect in a mouse OA model with surgical destabilization of the medial meniscus. IL-1ß-induced expression of nitric oxide (NO), prostaglandin E2 (PGE2), cyclooxygenase-2 (COX-2), tumor necrosis factor alpha (TNF-α), inducible nitric oxide synthase (iNOS) and interleukin-6 (IL-6) were all inhibited significantly by Pm pretreatment in vitro. In addition, Pm also inhibited the expression of thrombospondin motifs 5 (ADAMTS-5) and matrix metalloproteinase-13 (MMP-13), which are responsible for the degradation of the extracellular matrix (ECM). Additionally, the degradation of aggrecan and collagen II was reversed by Pm. Furthermore, Pm inhibited Akt phosphorylation and the nuclear transfer of nuclear factor-κB (NF-κB) and activated Nrf2/HO-1 signaling pathways both in vitro and in vivo. These findings suggested that Pm alleviated inflammatory effects in the IL-1ß-induced chondrocytes. Therefore, Pm might be a potential therapeutic agent for OA.


Assuntos
Cartilagem Articular/efeitos dos fármacos , Cevanas/administração & dosagem , Condrócitos/efeitos dos fármacos , Interleucina-1beta/imunologia , Osteoartrite/tratamento farmacológico , Animais , Cartilagem Articular/citologia , Cartilagem Articular/imunologia , Células Cultivadas , Condrócitos/imunologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Dinoprostona/imunologia , Modelos Animais de Doenças , Humanos , Interleucina-1beta/genética , Interleucina-6/genética , Interleucina-6/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/imunologia , Óxido Nítrico/imunologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Osteoartrite/genética , Osteoartrite/imunologia
11.
Cell Death Dis ; 10(2): 134, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755595

RESUMO

Bone marrow derived stem cells (BMSCs) transplantation are viewed as a promising therapeutic candidate for spinal cord injury (SCI). However, the inflammatory microenvironment in the spinal cord following SCI limits the survival and efficacy of transplanted BMSCs. In this study, we investigate whether injured neuronal cells derived exosomes would influence the survival of transplanted BMSCs after SCI. In order to mimic the microenvironment in SCI that the neuronal cells or transplanted BMSCs suffer in vivo, PC12 cells conditioned medium and PC12 cell's exosomes collected from H2O2-treated PC12 cell's culture medium were cultured with BMSCs under oxidative stress in vitro. PC12 cells conditioned medium and PC12 cell's exosomes significantly accelerated the apoptosis of BMSCs induced by H2O2. Moreover, the cleaved caspase-3, cytochrome (Cyt) C, lactate dehydrogenase (LDH) releases, and apoptotic percentage were increased, and the ratio of Bcl-2/Bax and cell viability were decreased. Inhibition of exosome secretion via Rab27a small interfering RNA prevented BMSCs apoptosis in vitro. In addition, hypoxia-preconditioned promoted the survival of BMSCs under oxidative stress both in vivo after SCI and in vitro. Our results also indicate that HIF-1α plays a central role in the survival of BMSCs in hypoxia pretreatment under oxidative stress conditions. siRNA-HIF-1α increased apoptosis of BMSCs; in contrast, HIF-1α inducer FG-4592 attenuated apoptosis of BMSCs. Taken together, we found that the injured PC12 cells derived exosomes accelerate BMSCs apoptosis after SCI and in vitro, hypoxia pretreatment or activating expression of HIF-1α to be important in the survival of BMSCs after transplantation, which provides a foundation for application of BMSCs in therapeutic potential for SCI.


Assuntos
Hipóxia Celular/efeitos dos fármacos , Exossomos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Neurônios/metabolismo , Traumatismos da Medula Espinal/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados , Modelos Animais de Doenças , Peróxido de Hidrogênio/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Células PC12 , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/terapia , Proteínas rab27 de Ligação ao GTP/genética
12.
EBioMedicine ; 40: 643-654, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30612943

RESUMO

BACKGROUND: Genetic overexpression or pharmacological activation of heme oxygenase (HO) are identified as potential therapeutic target for spinal cord injury (SCI); however, the role of carbon monoxide (CO), which is a major product of haem degenerated by HO, in SCI remains unknown. Applying hemin or chemicals which may regulate HO expression or activity to increase CO production are inadequate to elaborate the direct role of CO. Here, we assessed the effect of CO releasing molecule-3 (CORM-3), the classical donor of CO, in SCI and explained its possible protective mechanism. METHODS: Rat SCI model was performed with a vascular clip (30 g) compressing at T9 vertebral level for 1 min and CO was delivered immediately after SCI by CORM-3. The neurological deficits and neuron survival were assessed. Inflammasome and inositol-requiring enzyme 1 (IRE1) pathway were measured by western blot and immunofluorescence. For in vitro study, oxygen glucose deprivation (OGD) simulated the SCI-inflammasome change in cultured the primary neurons. FINDINGS: CORM-3 suppressed inflammasome signaling and pyroptosis occurrence, which consequently alleviated neuron death and improved motor functional recovery following SCI. As a pivotal sensor involving in endoplasmic reticulum stress-medicated inflammasome signaling, IRE1 and its downstream X-box binding protein 1 (XBP1) were activated in SCI tissues as well as in OGD neurons; while inhibition of IRE1 by STF-083010 in SCI rats or by si-RNA in OGD neurons suppressed inflammasome signaling and pyroptosis. Interestingly, the SCI/OGD-stimulated IRE1 activation was attenuated by CORM-3 treatment. INTERPRETATIONS: CO may alleviate neuron death and improve motor functional recovery in SCI through IRE1 regulation, and administration of CO could be a promising therapeutic strategy for SCI.


Assuntos
Inflamassomos/metabolismo , Neurônios/metabolismo , Compostos Organometálicos/metabolismo , Traumatismos da Medula Espinal/metabolismo , Animais , Biomarcadores , Monóxido de Carbono/metabolismo , Contagem de Células , Morte Celular , Modelos Animais de Doenças , Feminino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Compostos Organometálicos/farmacologia , Fosforilação , Interferência de RNA , Ratos , Traumatismos da Medula Espinal/etiologia , Traumatismos da Medula Espinal/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA