Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Invest Dermatol ; 143(4): 648-660.e6, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36309321

RESUMO

The transition from the proinflammatory phase to the prohealing phase in wound healing is essential for effective skin wound repair, which involves the balance of M1 and M2 polarization of wound-infiltrating macrophages. P311 plays an essential role in promoting wound closure by enhancing the biological function of epidermal stem cells, endothelial cells, and fibroblasts. Nevertheless, whether and how P311 regulates macrophage polarization remains unclear. In this study, we showed that P311 deficiency reduced the M2 polarization of macrophages, thereby attenuating the secretion of M2-like cytokines. The P311 deficiency prolonged the transition from the proinflammatory phase to the prohealing phase, accompanied by weakened angiogenesis and retarded granulation tissue formation, both of which coordinately hinder the healing of skin wounds. Mechanistically, P311 deficiency downregulated the expression of IL-4 receptor on macrophages, followed by less activation of the IL-4 receptor‒signal transducer and activator of transcription 6 signaling pathway, resulting in impaired M2 macrophage polarization. We further revealed that the mTOR signaling pathway was associated with the regulation of P311 on the expression of IL-4 receptor in macrophages. Thus, our study has highlighted the pivotal role of P311 in promoting the M2 polarization of macrophages for effective skin wound healing.


Assuntos
Células Endoteliais , Pele , Pele/lesões , Macrófagos/metabolismo , Cicatrização , Transdução de Sinais
2.
Angew Chem Int Ed Engl ; 61(17): e202200480, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35143118

RESUMO

Fenton reactions have been recently applied in tumor catalytic therapy, whose efficacy suffers from the unsatisfactory reaction kinetics of Fe3+ to Fe2+ conversion. Here we introduce a co-catalytic concept in tumor catalytic therapy by using a two-dimensional molybdenum disulfide (MoS2 ) nanosheet atomically dispersed with Fe species. The single-atom Fe species act as active sites for triggering Fenton reactions, while the abundant sulfur vacancies generated on the nanosheet favor electron capture by hydrogen peroxide for promoting hydroxyl radical production. Moreover, the 2D MoS2 support also acts as a co-catalyst to accelerate the conversion of Fe3+ to Fe2+ by the oxidation of active Mo4+ sites to Mo6+ , thereby promoting the whole catalytic process. The 2D nanocatalyst exhibits a desirable catalytic performance, as well as a significantly enhanced anticancer efficacy both in vitro and in vivo, which indicates the feasibility for applying such a co-catalytic concept in tumor therapy.


Assuntos
Molibdênio , Neoplasias , Catálise , Humanos , Peróxido de Hidrogênio/química , Neoplasias/tratamento farmacológico , Oxirredução
3.
Front Immunol ; 13: 821932, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154140

RESUMO

As a potential clinical therapeutic cell for injured tissue repair, mesenchymal stem cells (MSCs) have attracted increasing attention. Enhancing the pro-healing function of MSCs has gradually become an essential topic in improving the clinical efficacy of MSCs. Recently, studies have shown that neuronal protein 3.1 (P311) plays a crucial role in promoting skin wound healing, suggesting P311 gene modification may improve the pro-healing function of MSCs. In this study, we demonstrated that increasing the in vivo expression of P311 could significantly enhance the ability of MSCs to lessen the number of inflammatory cells, increase the expression of IL10, reduce the levels of TNF-α and IFN-γ, increase collagen deposition, promote angiogenesis, and ultimately accelerate skin wound closure and improve the quality of wound healing. Importantly, we uncovered that P311 enhanced the pro-angiogenesis function of MSCs by increasing the production of vascular endothelial growth factor (VEGF) in vitro and in vivo. Mechanistically, we revealed that the mTOR signalling pathway was closely related to the regulation of P311 on VEGF production in MSCs. Together, our data displayed that P311 gene modification in MSCs augments their capabilities to promote skin wound closure, which might bring the dawn for its clinical application in the future.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Pele/patologia , Fatores de Crescimento do Endotélio Vascular/metabolismo , Cicatrização/fisiologia , Indutores da Angiogênese , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética
4.
Nat Commun ; 12(1): 3393, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099712

RESUMO

The iron gall ink-triggered chemical corrosion of hand-written documents is a big threat to Western cultural heritages, which was demonstrated to result from the iron gall (GA-Fe) chelate-promoted reactive oxygen species generation. Such a phenomenon has inspired us to apply the pro-oxidative mechanism of GA-Fe to anticancer therapy. In this work, we construct a composite cancer nanomedicine by loading gallate into a Fe-engineered mesoporous silica nanocarrier, which can degrade in acidic tumor to release the doped Fe3+ and the loaded gallate, forming GA-Fe nanocomplex in situ. The nanocomplex with a highly reductive ligand field can promote oxygen reduction reactions generating hydrogen peroxide. Moreover, the resultant two-electron oxidation form of GA-Fe is an excellent Fenton-like agent that can catalyze hydrogen peroxide decomposition into hydroxyl radical, finally triggering severe oxidative damage to tumors. Such a therapeutic approach by intratumoral synthesis of GA-Fe nano-metalchelate may be instructive to future anticancer researches.


Assuntos
Antineoplásicos/administração & dosagem , Ácido Gálico/administração & dosagem , Ferro/administração & dosagem , Nanopartículas Metálicas/administração & dosagem , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Catálise , Complexos de Coordenação/administração & dosagem , Complexos de Coordenação/química , Complexos de Coordenação/metabolismo , Portadores de Fármacos/química , Feminino , Ácido Gálico/química , Ácido Gálico/metabolismo , Células HeLa , Células Endoteliais da Veia Umbilical Humana , Humanos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Concentração de Íons de Hidrogênio , Radical Hidroxila/química , Radical Hidroxila/metabolismo , Injeções Intravenosas , Ferro/química , Ferro/metabolismo , Ligantes , Nanopartículas Metálicas/química , Camundongos , Neoplasias/patologia , Oxirredução , Oxigênio/metabolismo , Dióxido de Silício/química , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Sci Bull (Beijing) ; 66(5): 464-477, 2021 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-36654184

RESUMO

Ferroptosis and autophagy, playing significant roles in tumor treatment, are two typical forms of the programmed cell death. However, the rational combination of ferroptosis and autophagy for synergistic tumor therapy is still highly challenging. Herein, we report on an intriguing nanomedicine strategy for achieving autophagy-enhanced ferroptosis on efficiently combating cancer, which was based on the construction of trehalose-loaded mSiO2@MnOx-mPEG (TreMMM) nanoparticles with satisfactory biocompatibility. The nanoparticles are endowed with high glutathione (GSH) consumption efficiency, thereby inducing cancer-cell ferroptosis via inactivating glutathione peroxidases 4 (GPX4). Subsequently, the TreMMM degradation due to the GSH depletion and pH sensitivity contributed to the trehalose release for inducing autophagy, promoting/enhancing ferroptosis by NCOA4-mediated degradation of ferritin. A substantial in vitro and in vivo antitumor effect was achieved by such an intriguing autophagy-enhanced ferroptosis. Therefore, the rational combination of GSH-consumption-induced ferroptosis and trehalose-induced autophagy by nanomedicine design provides an alternative but effective strategy for tumor treatment.

6.
Stem Cell Res Ther ; 11(1): 220, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32513275

RESUMO

BACKGROUND: Restenosis is a serious problem in patients who have undergone percutaneous transluminal angioplasty. Endothelial injury resulting from surgery can lead to endothelial dysfunction and neointimal formation by inducing aberrant proliferation and migration of vascular smooth muscle cells. Exosomes secreted by mesenchymal stem cells have been a hot topic in cardioprotective research. However, to date, exosomes derived from mesenchymal stem cells (MSC-Exo) have rarely been reported in association with restenosis after artery injury. The aim of this study was to investigate whether MSC-Exo inhibit neointimal hyperplasia in a rat model of carotid artery balloon-induced injury and, if so, to explore the underlying mechanisms. METHODS: Characterization of MSC-Exo immunophenotypes was performed by electron microscopy, nanoparticle tracking analysis and western blot assays. To investigate whether MSC-Exo inhibited neointimal hyperplasia, rats were intravenously injected with normal saline or MSC-Exo after carotid artery balloon-induced injury. Haematoxylin-eosin staining was performed to examine the intimal and media areas. Evans blue dye staining was performed to examine re-endothelialization. Moreover, immunohistochemistry and immunofluorescence were performed to examine the expression of CD31, vWF and α-SMA. To further investigate the involvement of MSC-Exo-induced re-endothelialization, the underlying mechanisms were studied by cell counting kit-8, cell scratch, immunofluorescence and western blot assays. RESULTS: Our data showed that MSC-Exo were ingested by endothelial cells and that systemic injection of MSC-Exo suppressed neointimal hyperplasia after artery injury. The Evans blue staining results showed that MSC-Exo could accelerate re-endothelialization compared to the saline group. The immunofluorescence and immunohistochemistry results showed that MSC-Exo upregulated the expression of CD31 and vWF but downregulated the expression of α-SMA. Furthermore, MSC-Exo mechanistically facilitated proliferation and migration by activating the Erk1/2 signalling pathway. The western blot results showed that MSC-Exo upregulated the expression of PCNA, Cyclin D1, Vimentin, MMP2 and MMP9 compared to that in the control group. Interestingly, an Erk1/2 inhibitor reversed the expression of the above proteins. CONCLUSION: Our data suggest that MSC-Exo can inhibit neointimal hyperplasia after carotid artery injury by accelerating re-endothelialization, which is accompanied by activation of the Erk1/2 signalling pathway. Importantly, our study provides a novel cell-free approach for the treatment of restenosis diseases after intervention.


Assuntos
Lesões das Artérias Carótidas , Exossomos , Células-Tronco Mesenquimais , Animais , Proliferação de Células , Células Endoteliais , Humanos , Hiperplasia , Ratos
7.
Med Sci Monit ; 26: e918881, 2020 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-32417848

RESUMO

BACKGROUND This retrospective study aimed to investigate the epidemiology of burns to the hand, including the causes, demographic data, management, and outcome in a single center in Southwest China between 2012 and 2017. MATERIAL AND METHODS A retrospective study included 470 patients with hand burns who were treated at a single hospital in Southwest China between 2012 and 2017. Demographic, injury-related, and clinical data were obtained from the clinical electronic data collection system. RESULTS In 470 patients, men were more commonly admitted to hospital with hand burns (73.62%). Children under 10 years (29.57%) were the main patient group. Hospital admissions occurred in the coldest months, from December to March (55.11%). In 60.21% of cases, hand burns occurred outside the workplace. Fire (40.42%), electricity (30.85%), and hot liquids (20.21%) were the main causes of hand burns. Data from 428 patients showed that burns with a larger total body surface area and deeper burns were associated with surgery and amputation. Burn depth was a risk factor for skin grafting, and lack of burn cooling before hospital admission increased the risk of amputation. Data from 117 patients with localized burns showed that full-thickness burns and lack of cooling before admission were associated with an increased hospital stay. CONCLUSIONS The findings suggest that in Southwest China, prevention programs for children aged 0-9 years, injuries occurring in winter and non-workplace sites, and fire burns were imperative.


Assuntos
Unidades de Queimados/estatística & dados numéricos , Queimaduras/fisiopatologia , Traumatismos da Mão/epidemiologia , Prevenção de Acidentes/métodos , Distribuição por Idade , Queimaduras/epidemiologia , China/epidemiologia , Feminino , Mãos , Hospitalização , Humanos , Tempo de Internação/estatística & dados numéricos , Masculino , Estudos Retrospectivos , Fatores de Risco , Transplante de Pele/métodos , Resultado do Tratamento
8.
Int J Nanomedicine ; 14: 3345-3360, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31190796

RESUMO

Background: Designing a wound dressing that effectively prevents multi-drug-resistant bacterial infection and promotes angiogenesis and re-epithelialization is of great significance for wound management. Methods and results: In this study, a biocompatible composite membrane comprising biomimetic polydopamine-modified eggshell membrane nano/microfibres coated with KR-12 antimicrobial peptide and hyaluronic acid (HA) was developed in an eco-friendly manner. The physicochemical properties of the composite membrane were thoroughly characterized, and the results showed that the surface hydrophilicity and water absorption ability of the composite membrane were improved after the successive conjugation of the HA and the KR-12 peptide. Furthermore, the in vitrobiological results revealed that the composite membrane had excellent antibacterial activity against Gram-positive Staphylococcus aureus, methicillin-resistant Staphylococcus aureus (MRSA) and Gram-negative Escherichia coli, and it could prevent MRSA biofilm formation on its surface. Additionally, it promoted the proliferation of keratinocytes and human umbilical vein endothelial cells and increased the secretion of VEGF. Finally, an in vivo animal study indicated that the composite membrane could promote wound healing via accelerating angiogenesis and re-epithelialization, which were demonstrated by the enhanced expression of angiogenetic markers (CD31 and VEGF) and keratinocyte proliferation marker (PCNA), respectively. Conclusion: These results indicated that the composite membrane is a potential candidate of wound dressings.


Assuntos
Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Casca de Ovo/química , Ácido Hialurônico/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Peptídeos/farmacologia , Reepitelização/efeitos dos fármacos , Animais , Antibacterianos/farmacologia , Galinhas , Escherichia coli/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Peptídeos/química , Porosidade , Staphylococcus aureus/efeitos dos fármacos
9.
Burns Trauma ; 7: 37, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31890716

RESUMO

BACKGROUND: The intestinal barrier integrity can be disrupted due to burn injury, which is responsible for local and systemic inflammatory responses. Anti-inflammation strategy is one of the proposed therapeutic approaches to control inflammatory cascade at an early stage. Interleukin-17A (IL-17A) plays a critical role in inflammatory diseases. However, the role of IL-17A in the progression of burn-induced intestinal inflammation is poorly understood. In this study, we aimed to investigate the effect of IL-17A and associated pro-inflammatory cytokines that were deeply involved in the pathogenesis of burn-induced intestinal inflammatory injury, and furthermore, we sought to determine the early source of IL-17A in the intestine. METHODS: Mouse burn model was successfully established with infliction of 30% total body surface area scald burn. The histopathological manifestation, intestinal permeability, zonula occludens-1 expression, pro-inflammatory cytokines were determined with or without IL-17A-neutralization. Flow cytometry was used to detect the major source of IL-17A+ cells in the intestine. RESULTS: Burn caused intestinal barrier damage, increase of intestinal permeability, alteration of zonula occludens-1 expressions, elevation of IL-17A, IL-6, IL-1ß and tumor necrosis factor-α (TNF-α), whereas IL-17A neutralization dramatically alleviated burn-induced intestinal barrier disruption, maintained zonula occludens-1 expression, and noticeably, inhibited pro-inflammatory cytokines elevation. In addition, we observed that the proportion of intestinal IL-17A+Vγ4+ T subtype cells (but not IL-17A+Vγ1+ T subtype cells) were increased in burn group, and neutralization of IL-17A suppressed this increase. CONCLUSIONS: The main original findings of this study are intestinal mucosa barrier is disrupted after burn through affecting the expression of pro-inflammatory cytokines, and a protective role of IL-17A neutralization for intestinal mucosa barrier is determined. Furthermore, Vγ4+ T cells are identified as the major early producers of IL-17A that orchestrate an inflammatory response in the burn model. These data suggest that IL-17A blockage may provide a unique target for therapeutic intervention to treat intestinal insult after burn.

10.
Biomed Mater ; 11(1): 015008, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26836957

RESUMO

Recently, cell-based therapies have attracted attention as promising treatments for acute liver failure (ALF). Bone marrow-derived mesenchymal stem cells (MSCs) are potential candidates for co-culture with hepatocytes in poly(lactic acid-glycolic acid) (PLGA) scaffolds to support hepatocellular function. However, the mechanism of culturing protocol using PLGA scaffolds for MSC differentiation into hepatocyte-like cells as well as the therapeutic effect of cell seeded PLGA scaffolds on ALF remain unsatisfactory in clinical application. Here, MSCs and hepatocytes were co-cultured at ratios of 1:2.5 (MSCs: Hep), 1:5 and 1:10, respectively. The proliferation abilities of these co-cultured cells were detected by CCK8, MTT, EdU and by scanning electron microscopy (SEM), and the ability of MSCs to differentiate into hepatocytes was detected by PCR, western blot and immunofluorescence staining. Therapeutic trials of cell seeded PLGA scaffolds were conducted through mouse abdominal cavity transplantation. Results showed that the 1:5 group showed significantly higher cellular proliferation than the 1:2.5 and 1:10 groups, supernatant albumin and urea nitrogen levels were also significantly higher in the 1:5 group than in other two groups. Similarly, the 1:5 group demonstrated better DNA transcription and liver-specific protein (albumin, CK18 and P450) production. Meanwhile, the GalN-stimulated levels of ALT, AST and TBil in mouse serum were down-regulated significantly more by (MSC + Hep)-PLGA scaffold treatment than MSC-PLGA or Hep-PLGA scaffold treatments. Furthermore, the (MSC + Hep)-PLGA scaffold-treated ALF mice showed a lower immunogenic response level than the other two groups. These data suggested that the ratio of 1:5 (MSC:Hep) co-cultures was the optimal ratio for MSCs to support hepatocellular metabolism and function in PLGA scaffolds in vitro, the (MSC + Hep)-PLGA scaffold treatment could perform better restoration for damaged liver function and could give ALF mice a greater survival rate than the monocell seeded PLGA scaffold treatment.


Assuntos
Técnicas de Cocultura/instrumentação , Hepatócitos/transplante , Ácido Láctico/química , Falência Hepática Aguda/terapia , Transplante de Células-Tronco Mesenquimais/instrumentação , Ácido Poliglicólico/química , Alicerces Teciduais , Animais , Materiais Biocompatíveis/síntese química , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Feminino , Hepatócitos/citologia , Falência Hepática Aguda/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Camundongos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Sprague-Dawley , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos , Resultado do Tratamento
11.
PLoS One ; 8(4): e61026, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23613775

RESUMO

Autophagy is involved in many human diseases, such as cancer, cardiovascular disease and virus infection, including human immunodeficiency virus (HIV), hepatitis C virus (HCV), influenza A virus (IAV) and coxsackievirus B3/B4 (CVB3/B4), so a drug screening model targeting autophagy may be very useful for the therapy of these diseases. In our study, we established a drug screening model based on the inhibition of the dissociation of Beclin1-Bcl2 heterodimer, an important negative regulator of autophagy, using bimolecular fluorescence complementation (BiFC) technique for developing novel autophagy inhibitors and anti-IAV agents. From 86 examples of traditional Chinese medicines, we found Syzygium aromaticum L. had the best activity. We then determined the anti-autophagy and anti-IAV activity of eugenol, the major active compound of Syzygium aromaticum L., and explored its mechanism of action. Eugenol could inhibit autophagy and IAV replication, inhibited the activation of ERK, p38MAPK and IKK/NF-κB signal pathways and antagonized the effects of the activators of these pathways. Eugenol also ameliorated the oxidative stress and inhibited the expressions of autophagic genes. We speculated that the mechanism underlying might be that eugenol inhibited the oxidative stress and the activation of ERK1/2, p38MAPK and IKK/NF-κB pathways, subsequently inhibited the dissociation of Beclin1-Bcl2 heterodimer and autophagy, and finally impaired IAV replication. These results might conversely display the reasonableness of the design of our screening model. In conclusion, we have established a drug screening model for developing novel autophagy inhibitor, and find eugenol as a promising inhibitor for autophagy and IAV infection.


Assuntos
Antivirais/farmacologia , Autofagia/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Eugenol/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Linhagem Celular , Medicamentos de Ervas Chinesas/farmacologia , Humanos , Syzygium/química
12.
PLoS One ; 7(8): e42706, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22900043

RESUMO

In this research, we have established a drug screening method based on the autophagy signal pathway using the bimolecular fluorescence complementation-fluorescence resonance energy transfer (BiFC-FRET) technique to develop novel anti-influenza A virus (IAV) drugs. We selected Evodia rutaecarpa Benth out of 83 examples of traditional Chinese medicine and explored the mechanisms of evodiamine, the major active component of Evodia rutaecarpa Benth, on anti-IAV activity. Our results showed that evodiamine could significantly inhibit IAV replication, as determined by a plaque inhibition assay, an IAV vRNA promoter luciferase reporter assay and the Sulforhodamine B method using cytopathic effect (CPE) reduction. Additionally, evodiamine could significantly inhibit the accumulation of LC3-II and p62, and the dot-like aggregation of EGFP-LC3. This compound also inhibited the formation of the Atg5-Atg12/Atg16 heterotrimer, the expressions of Atg5, Atg7 and Atg12, and the cytokine release of TNF-α, IL-1ß, IL-6 and IL-8 after IAV infection. Evodiamine inhibited IAV-induced autophagy was also dependent on its action on the AMPK/TSC2/mTOR signal pathway. In conclusion, we have established a new drug screening method, and selected evodiamine as a promising anti-IAV compound.


Assuntos
Antivirais/farmacologia , Autofagia/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Vírus da Influenza A/efeitos dos fármacos , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Adenilato Quinase/metabolismo , Animais , Autofagia/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Citocinas/biossíntese , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA