Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
World J Psychiatry ; 14(7): 1053-1061, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39050200

RESUMO

BACKGROUND: Having a gynecological tumor or undergoing treatment can be a traumatic experience for women, as it affects their self-image and sexual relationships and can lead to psychological reactions. Psychological adjustment following cancer occurrence remains a key issue among the survivors. AIM: To examine the current status of quality of life (QoL), anxiety, and depression in patients with gynecological cancer and to analyze the factors associated with it. METHODS: Data for 160 patients with gynecological malignancies treated at Shanxi Bethune Hospital from June 2020 to June 2023 were collected and analyzed retrospectively. Patients' QoL was assessed using the European Organization for Research on Treatment of Cancer Quality of Life Questionnaire Core 30 and the Functional Assessment of Cancer Therapy-General Questionnaire. Their emotional status was evaluated using the Self-Rating Anxiety/Depression Scale. The associated factors of anxiety and depression were analyzed. RESULTS: The overall QoL score of the patients 6 months after surgery was 76.39 ± 3.63 points. This included low levels of social and emotional function and severe fatigue and pain. The scores for physiological, functional, emotional, social, and family well-being exhibited an upward trend following surgery compared with those before surgery. One month after surgery, some patients experienced anxiety and depression, with an incidence of 18.75% and 18.13%, respectively. Logistic analysis revealed that good sleep was a protective factor against anxiety and depression in patients with gynecological tumors, whereas physical pain was a risk factor. CONCLUSION: Patients with gynecological malignancies often experience anxiety and depression. By analyzing the factors that affect patients' QoL, effective nursing measures can be administered.

2.
Adv Sci (Weinh) ; 11(28): e2401327, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38725147

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal interstitial lung disease, with limited therapeutic options available. Impaired autophagy resulting from aberrant TRB3/p62 protein-protein interactions (PPIs) contributes to the progression of IPF. Restoration of autophagy by modulating the TRB3/p62 PPIs has rarely been reported for the treatment of IPF. Herein, peptide nanofibers are developed that specifically bind to TRB3 protein and explored their potential as a therapeutic approach for IPF. By conjugating with the self-assembling fragment (Ac-GFFY), a TRB3-binding peptide motif A2 allows for the formation of nanofibers with a stable α-helix secondary structure. The resulting peptide (Ac-GFFY-A2) nanofibers exhibit specific high-affinity binding to TRB3 protein in saline buffer and better capacity of cellular uptake to A2 peptide. Furthermore, the TRB3-targeting peptide nanofibers efficiently interfere with the aberrant TRB3/p62 PPIs in activated fibroblasts and fibrotic lung tissue of mice, thereby restoring autophagy dysfunction. The TRB3-targeting peptide nanofibers inhibit myofibroblast differentiation, collagen production, and fibroblast migration in vitro is demonstrated, as well as bleomycin-induced pulmonary fibrosis in vivo. This study provides a supramolecular method to modulate PPIs and highlights a promising strategy for treating IPF diseases by restoring autophagy.


Assuntos
Autofagia , Bleomicina , Modelos Animais de Doenças , Nanofibras , Fibrose Pulmonar , Nanofibras/química , Animais , Autofagia/efeitos dos fármacos , Camundongos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/tratamento farmacológico , Camundongos Endogâmicos C57BL , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/induzido quimicamente , Peptídeos/farmacologia
3.
Front Oncol ; 13: 1100045, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36756161

RESUMO

Follistatin-like 1 (FSTL1) is a cancer-related matricellular secretory protein with contradictory organ-specific roles. Its contribution to the pathogenesis of cervical carcinoma is still not clear. Meanwhile, it is necessary to identify novel candidate genes to understand cervical carcinoma's pathogenesis further and find potential therapeutic targets. We collected cervical carcinoma samples and matched adjacent tissues from patients with the locally-advanced disease and used cervical carcinoma cell lines HeLa and C33A to evaluate the effects of FSTL1 on CC cells. The mRNA transcription and protein expression of FSTL1 in cervical carcinoma tumor biopsy tissues were lower than those of matched adjacent tissues. Patients with a lower ratio of FSTL1 mRNA between the tumor and its matched adjacent tissues showed a correlation with the advanced cervical carcinoma FIGO stages. High expression of FSTL1 markedly inhibited the proliferation, motility, and invasion of HeLa and C33A. Regarding mechanism, FSTL1 plays its role by negatively regulating the BMP4/Smad1/5/9 signaling. Our study has demonstrated the tumor suppressor effect of FSTL1, and these findings suggested a potential therapeutic target and biomarker for cervical carcinoma.

4.
Sleep Breath ; 27(3): 1165-1173, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36115873

RESUMO

PURPOSE: Intermittent hypoxia (IH) mimicking obstructive sleep apnea (OSA) has been confirmed to induce tumor lung metastasis via oxidative stress and inflammation responses. Follistatin-like 1 (Fstl1), as a matricellular protein, plays critical roles in inflammatory diseases and cancer. This study aimed to investigate the effect and mechanism of Fstl1 on OSA-IH-induced tumor lung metastasis. METHODS: Fstl1+/+ or Fstl1+/- mice inoculated with B16F10 melanoma cells were exposed to OSA-IH. The number and area of mouse lung metastatic colonies were assessed. Markers for tumor metastasis, oxidative stress, and inflammation in lung melanoma tissue or B16F10 melanoma cells were quantified by western blotting, qRT-PCR, and immunohistochemistry. The migration of B16F10 cells was examined by wound healing assay. RESULTS: Fstl1 levels are decreased in lung tissues from OSA-IH injured mice inoculated with melanoma cells. Fstl1-deficient mice were highly susceptible to the OSA-IH model of melanoma lung metastasis, as assessed by increased number and area of lung metastatic colonies, and by the elevated levels of HIF-1α, Vegf, N-cadherin, and E-cadherin. Lung melanoma tissue in Fstl1+/- mice provided evidence of increased oxidative stress, as determined by increased levels of NRF2 and P22phox and decreased level of Sod2, as well as increased inflammatory response, as determined by elevated levels of NF-κB P65, Tnf-α and Il-6. Conversely, stable overexpression of Fstl1 in B16F10 cells under OSA-IH exposure attenuated the migration of B16F10 cells and levels of tumor-related markers, as well as decreased oxidative stress and inflammatory responses. CONCLUSION: These results suggest that Fstl1 may protect against OSA-IH-induced tumor lung metastasis through oxidative stress and inflammatory responses. Fstl1 may serve as a promising target for OSA-related cancer.


Assuntos
Proteínas Relacionadas à Folistatina , Neoplasias Pulmonares , Melanoma , Apneia Obstrutiva do Sono , Animais , Camundongos , Folistatina , Proteínas Relacionadas à Folistatina/genética , Hipóxia/metabolismo , Inflamação/metabolismo , Neoplasias Pulmonares/patologia , Apneia Obstrutiva do Sono/metabolismo
5.
Stem Cell Res Ther ; 13(1): 403, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35932064

RESUMO

BACKGROUND: Mesenchymal stem cell (MSC) therapy has been shown to be a promising option for liver fibrosis treatment. However, critical factors affecting the efficacy of MSC therapy for liver fibrosis remain unknown. Follistatin-like 1 (FSTL1), a TGF-ß-induced matricellular protein, is documented as an intrinsic regulator of proliferation and differentiation in MSCs. In the present study, we characterized the potential role of FSTL1 in MSC-based anti-fibrotic therapy and further elucidated the mechanisms underlying its action. METHODS: Human umbilical cord-derived MSCs were characterized by flow cytometry. FSTL1low MSCs were achieved by FSTL1 siRNA. Migration capacity was evaluated by wound-healing and transwell assay. A murine liver fibrotic model was created by carbon tetrachloride (CCl4) injection, while control MSCs or FSTL1low MSC were transplanted via intravenous injection 12 weeks post CCl4 injection. Histopathology, liver function, fibrosis degree, and inflammation were analysed thereafter. Inflammatory cell infiltration was evaluated by flow cytometry after hepatic nonparenchymal cell isolation. An MSC-macrophage co-culture system was constructed to further confirm the role of FSTL1 in the immunosuppressive capacity of MSCs. RNA sequencing was used to screen target genes of FSTL1. RESULTS: FSTL1low MSCs had comparable gene expression for surface markers to wildtype but limited differentiation and migration capacity. FSTL1low MSCs failed to alleviate CCl4-induced hepatic fibrosis in a mouse model. Our data indicated that FSTL1 is essential for the immunosuppressive action of MSCs on inflammatory macrophages during liver fibrotic therapy. FSTL1 silencing attenuated this capacity by inhibiting the downstream JAK/STAT1/IDO pathway. CONCLUSIONS: Our data suggest that FSTL1 facilitates the immunosuppression of MSCs on macrophages and that guarantee the anti-fibrotic effect of MSCs in liver fibrosis.


Assuntos
Proteínas Relacionadas à Folistatina , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Folistatina/efeitos adversos , Folistatina/metabolismo , Proteínas Relacionadas à Folistatina/genética , Humanos , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/genética , Cirrose Hepática/terapia , Células-Tronco Mesenquimais/metabolismo , Camundongos
6.
Tissue Eng Regen Med ; 19(5): 961-968, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35809187

RESUMO

Senescence is an inevitable natural life process that involves structural and functional degeneration of tissues and organs. Recently, the process of skin aging has attracted much attention. Determining a means to delay or even reverse skin aging has become a research hotspot in medical cosmetology and anti-aging. Dysfunction in the epidermis and fibroblasts and changes in the composition and content of the extracellular matrix are common pathophysiological manifestations of skin aging. Reactive oxygen species and matrix metalloproteinases play essential roles in this process. Stem cells are pluripotent cells that possess self-replication abilities and can differentiate into multiple functional cells under certain conditions. These cells also possess a strong ability to facilitate tissue repair and regeneration. Stem cell transplantation has the potential for application in anti-aging therapy. Increasing studies have demonstrated that stem cells perform functions through paracrine processes, particularly those involving exosomes. Exosomes are nano-vesicular substances secreted by stem cells that participate in cell-to-cell communication by transporting their contents into target cells. In this chapter, the biological characteristics of exosomes were reviewed, including their effects on extracellular matrix formation, epidermal cell function, fibroblast function and antioxidation. Exosomes derived from stem cells may provide a new means to reverse skin aging.


Assuntos
Exossomos , Envelhecimento da Pele , Fibroblastos , Espécies Reativas de Oxigênio , Células-Tronco
7.
Expert Opin Drug Deliv ; 19(7): 815-832, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35738312

RESUMO

INTRODUCTION: Triple-negative breast cancer (TNBC) is a subtype of severely aggressive breast cancer that lacks the expression of oestrogen receptor (ER), progesterone receptor and human epidermal growth factor receptor 2 (HER2) and is highly metastatic and related to a poor prognosis. Current standard treatments are still limited to systemic chemotherapy, radiotherapy, and surgical resection. More effective treatments are urgently needed. AREAS COVERED: The immunogenicity of TNBC has provided opportunities for the development of targeted immunotherapy. In this review, we focus on the recent development in antibody-based drug modalities, including angiogenesis inhibitors, immune checkpoint inhibitors, antibody-drug conjugates, immunoconjugates, T cell-redirecting bispecific antibodies and CAR-T cells, and their mechanisms of action in TNBC. EXPERT OPINION: At present, the treatment of TNBC is still a major challenge that needs to be addressed. Novel immunotherapies are promising opportunities for improving the management of this aggressive disease.


Assuntos
Imunoconjugados , Neoplasias de Mama Triplo Negativas , Inibidores da Angiogênese , Humanos , Imunoconjugados/uso terapêutico , Imunoterapia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo
8.
Cancer Res ; 81(22): 5692-5705, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34551961

RESUMO

The tumor microenvironment plays a critical role in maintaining the immature phenotype of tumor-initiating cells (TIC) to promote cancer. Hepatocellular carcinoma (HCC) is a unique disease in that it develops in the setting of fibrosis and cirrhosis. This pathologic state commonly shows an enrichment of stromal myofibroblasts, which constitute the bulk of the tumor microenvironment and contribute to disease progression. Follistatin-like 1 (FSTL1) has been widely reported as a proinflammatory mediator in different fibrosis-related and inflammatory diseases. Here we show FSTL1 expression to be closely correlated with activated fibroblasts and to be elevated in regenerative, fibrotic, and disease liver states in various mouse models. Consistently, FSTL1 lineage cells gave rise to myofibroblasts in a CCL4-induced hepatic fibrosis mouse model. Clinically, high FSTL1 in fibroblast activation protein-positive (FAP+) fibroblasts were significantly correlated with more advanced tumors in patients with HCC. Although FSTL1 was expressed in primary fibroblasts derived from patients with HCC, it was barely detectable in HCC cell lines. Functional investigations revealed that treatment of HCC cells and patient-derived 3D organoids with recombinant FSTL1 or with conditioned medium collected from hepatic stellate cells or from cells overexpressing FSTL1 could promote HCC growth and metastasis. FSTL1 bound to TLR4 receptor, resulting in activation of AKT/mTOR/4EBP1 signaling. In a preclinical mouse model, blockade of FSTL1 mitigated HCC malignancy and metastasis, sensitized HCC tumors to sorafenib, prolonged survival, and eradicated the TIC subset. Collectively, these data suggest that FSTL1 may serve as an important novel diagnostic/prognostic biomarker and therapeutic target in HCC. SIGNIFICANCE: This study shows that FSTL1 secreted by activated fibroblasts in the liver microenvironment augments hepatocellular carcinoma malignancy, providing a potential new strategy to improve treatment of this aggressive disease.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/secundário , Fibroblastos/patologia , Proteínas Relacionadas à Folistatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/patologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Fibroblastos/metabolismo , Proteínas Relacionadas à Folistatina/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Biomolecules ; 11(8)2021 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-34439906

RESUMO

Skin fibrotic diseases, such as keloids, are mainly caused by pathologic scarring of wounds during healing and characterized by benign cutaneous overgrowths of dermal fibroblasts. Current surgical and therapeutic modalities of skin fibrosis are unsatisfactory. Pinocembrin, a natural flavonoid, has been shown to possess a vast range of pharmacological activities including antimicrobial, antioxidant, anti-inflammatory, and anti-tumor activities. In this study we explored the potential effect and mechanisms of pinocembrin on skin fibrosis in vitro and in vivo. In vitro studies indicated that pinocembrin dose-dependently suppressed proliferation, migration, and invasion of keloid fibroblasts and mouse primary dermal fibroblasts. The in vivo studies showed that pinocembrin could effectively alleviate bleomycin (BLM)-induced skin fibrosis and reduce the gross weight and fibrosis-related protein expression of keloid tissues in xenograft mice. Further mechanism studies indicated that pinocembrin could suppress TGF-ß1/Smad signaling and attenuate TGF-ß1-induced activation of skin fibroblasts. In conclusion, our results demonstrate the therapeutic potential of pinocembrin for skin fibrosis.


Assuntos
Fibrose/patologia , Flavanonas/farmacologia , Pele/efeitos dos fármacos , Pele/patologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Animais Recém-Nascidos , Movimento Celular , Proliferação de Células , Feminino , Fibroblastos/metabolismo , Flavonoides/metabolismo , Humanos , Técnicas In Vitro , Queloide/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Cicatrização
10.
Mol Ther ; 29(1): 347-364, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33007201

RESUMO

Follistatin-like 1 (FSTL1) is a matricellular protein that is upregulated during development and disease, including idiopathic pulmonary fibrosis (IPF), keloid, and arthritis. The profibrotic and pro-inflammatory roles of FSTL1 have been intensively studied during the last several years, as well as in this report. We screened and identified epitope-specific monoclonal neutralizing antibodies (nAbs) to functionally block FSTL1. FSTL1 nAbs attenuated bleomycin-induced pulmonary and dermal fibrosis in vivo and transforming growth factor (TGF)-ß1-induced dermal fibrosis ex vivo in human skin. In addition, FSTL1 nAbs significantly reduced existing lung fibrosis and skin fibrosis in experimental models. FSTL1 nAbs exerted their potent antifibrotic effects via reduced TGF-ß1 responsiveness and subsequent myofibroblast activation and extracellular matrix production. We also observed that FSTL1 nAbs attenuated the severity of collagen-induced arthritis in mice, which was accompanied by reduced inflammatory responses in vitro. Our findings suggest that FSTL1 nAbs are a promising new therapeutic strategy for the treatment of multiple organ fibrosis and systemic autoimmune diseases.


Assuntos
Doenças Autoimunes/etiologia , Doenças Autoimunes/metabolismo , Biomarcadores , Suscetibilidade a Doenças , Proteínas Relacionadas à Folistatina/metabolismo , Fibrose Pulmonar Idiopática/etiologia , Fibrose Pulmonar Idiopática/metabolismo , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/patologia , Modelos Animais de Doenças , Descoberta de Drogas , Fibrose , Proteínas Relacionadas à Folistatina/antagonistas & inibidores , Proteínas Relacionadas à Folistatina/genética , Expressão Gênica , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/patologia , Camundongos , Terapia de Alvo Molecular , Fator de Crescimento Transformador beta1/metabolismo
11.
J Breath Res ; 15(2)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33339013

RESUMO

The coexistence of chronic obstructive pulmonary disease (COPD) and obstructive sleep apnea (OSA) can cause multiple system damage, and the main physiological mechanisms are continuous hypoxia and intermittent hypoxia (IH). Airway mucus hypersecretion is an important clinical feature of COPD, which can cause a progressive decline of lung function, acute COPD aggravation, and disease progression. The purpose of our study is to determine the influence of the coexistence of mild OSA on airway mucus hypersecretion. Clinical data and airway epithelial samples of 36 subjects were collected. The average fluorescence intensity of MUC5AC and the number of goblet cells were measured through immunofluorescence staining. MUC5AC expression was measured in human bronchial epithelial (HBE) cells exposed to normoxia, IH, particulate matter (PM), and PM + IH using real-time quantitative polymerase chain reaction and western blotting. FEV1% pred and FEV1/FVC were higher in patients with COPD-OSA overlap syndrome (OS) than in patients with COPD alone. Patients with OS had less sputum volume than patients with COPD alone. MUC5AC expression and the number of goblet cells in the airway epithelium in the COPD alone group were significantly higher than those in the OS groups. The PM + IH group had lower MUC5AC mRNA and protein expression in HBE cells than the PM group. The coexistence of mild OSA may reduce goblet cell proliferation and MUC5AC expression in the airway epithelium of patients with COPD. Mild IH inhibited PM-induced up-regulation of MUC5AC expression in the mRNA and protein levels in HBE cells.


Assuntos
Doença Pulmonar Obstrutiva Crônica , Apneia Obstrutiva do Sono , Testes Respiratórios , Células Epiteliais , Humanos , Muco , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/metabolismo , Apneia Obstrutiva do Sono/complicações
12.
Phytomedicine ; 81: 153428, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33341025

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a fatal and progressive fibrotic lung disease lacking a validated and effective therapy. Aberrant activation of the Wnt/ß-catenin signaling cascade plays the key role in the pathogenesis of IPF. Betulinic acid is a natural pentacyclic triterpenoid molecule that has excellent antitumor and antiviral activities. HYPOTHESIS: We hypothesized that BA has an anti-pulmonary fibrosis effect mediated by the suppression of the Wnt/ß-catenin pathway. Study design Pulmonary fibrosis markers were detected in vitro and in vivo to confirm the antifibrotic effect of BA. The Wnt/ß-catenin pathway-related proteins were overexpressed to determine the effect of BA on Wnt signaling. METHODS AND RESULTS: BA dose-dependently inhibited Wnt3a-induced fibroblast activation in vitro. Moreover, BA decreased Wnt3a- and LiCl-induced transcriptional activity, as assessed by the TOPFlash assay in fibroblasts, and repressed the expression of the Wnt target genes cyclin D1, axin 2, and S100A4. Further investigation indicated that BA restrained the nuclear accumulation of ß-catenin, mainly by increasing the phospho-ß-catenin ratio (S33/S37/T41 and S45), inhibited the phosphorylation of DVL2 and LRP, and decreased the levels of Wnt3a and LRP6. In agreement with the results of the in vitro assays, the in vivo experiments indicated that BA significantly decreased bleomycin-induced pulmonary fibrosis in mice and suppressed myofibroblast activation by inhibiting Wnt/ß-catenin signaling. CONCLUSION: BA may directly interfere with the Wnt/ß-catenin pathway to subsequently repress myofibroblast activation and pulmonary fibrosis.


Assuntos
Fibrose Pulmonar Idiopática/tratamento farmacológico , Triterpenos Pentacíclicos/farmacologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Bleomicina/toxicidade , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Masculino , Camundongos Endogâmicos C57BL , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Ácido Betulínico
13.
Int Immunopharmacol ; 90: 107230, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33290968

RESUMO

Inflammation is a defense response of the body to stimuli. Lung injury caused by external stimuli can stimulate inflammatory cells to accumulate at the site of injury and secrete cytokines. Pinocembrin is a flavonoid with anti-inflammatory effects. Based on previous studies, we further explored the anti-inflammatory mechanisms of pinocembrin in vitro and in vivo. In vitro studies indicated that pinocembrin inhibited lipopolysaccharide (LPS)-stimulated inflammatory response in macrophages. In vivo studies also showed that pinocembrin could reduce LPS and bleomycin (BLM) induced lung inflammatory response in mice. Further mechanistic studies indicated that pinocembrin could regulate the TLR4-NF-κB signaling pathway and suppressed the activation and assembly of NLRP3 inflammasomes. In summary, pinocembrin could relieve pulmonary inflammatory response induced by LPS and BLM mainly via inhibiting TLR4-NF-κB-NLRP3 inflammasome axis. These results contribute to the understanding of the anti-inflammatory mechanisms of pinocembrin and serve as reference for future research on pinocembrin.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Flavanonas/uso terapêutico , Inflamassomos/efeitos dos fármacos , Pneumonia/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Bleomicina , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B , Proteína 3 que Contém Domínio de Pirina da Família NLR , Óxido Nítrico/metabolismo , Pneumonia/induzido quimicamente , Células RAW 264.7 , Receptor 4 Toll-Like
14.
J Cell Mol Med ; 24(15): 8623-8635, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32643868

RESUMO

Liver fibrosis is a disease caused by long-term damage that is related to a number of factors. The current research on the treatment of liver fibrosis mainly focuses on the activation of hepatic stellate cell, in addition to protecting liver cells. byakangelicin has certain anti-inflammatory ability, but its effect on liver fibrosis is unclear. This study aims to explore whether byakangelicin plays a role in the development of liver fibrosis and to explore its mechanism. We determined that byakangelicin has a certain ability to resist fibrosis and reduce liver cell damage in a model of carbon tetrachloride-induced liver fibrosis in mice. Thereafter, we performed further verification in vitro. The signalling pathways of two important pro-fibrotic cytokines, transforming growth factor-ß and platelet-derived growth factor, were studied. Results showed that byakangelicin can inhibit related pathways. According to the hepatoprotective effect of byakangelicin observed in animal experiments, we studied the effect of byakangelicin on 4-HNE-induced hepatocyte (HepG2) apoptosis and explored its related pathways. The results showed that byakangelicin could attenuate 4-HNE-induced hepatocyte apoptosis via inhibiting ASK-1/JNK signalling. In conclusion, byakangelicin could improve carbon tetrachloride-induced liver fibrosis and liver injury by inhibiting hepatic stellate cell proliferation and activation and suppressing hepatocyte apoptosis.


Assuntos
Tetracloreto de Carbono/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Furocumarinas/farmacologia , Cirrose Hepática/etiologia , Cirrose Hepática/patologia , Substâncias Protetoras/farmacologia , Animais , Apoptose/efeitos dos fármacos , Biópsia , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Citocinas/metabolismo , Modelos Animais de Doenças , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/prevenção & controle , Masculino , Camundongos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo
15.
Mol Cell Biol ; 40(13)2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32312884

RESUMO

RNA helicase DHX33 was found to regulate the transcription of multiple genes involved in cancer development. But the underlying molecular mechanism remains unclear. Here, we found DHX33 associated extensively with gene promoters at CG-rich region. Its deficiency reduced the loading of active RNA polymerase II at gene promoters. Furthermore, we observed a functional interaction between DHX33, AP-2ß, and DNA demethylation protein Gadd45a (growth arrest and DNA damage inductile protein 45a) at specific gene promoters. DHX33 is required to recruit GADD45a, thereby causing local DNA demethylation through further recruiting ten-eleven-translocation (Tet) methylcytosine dioxygenase enzyme, as manifested by reduced 5-hydroxymethyl cytosine levels for a subset of genes after DHX33 deficiency. This process might involve R-loop formation in GC skew as a guidance signal at promoter sites. Our report provides for the first time, to our knowledge, original evidence that DHX33 alters epigenetic marks and regulates specific gene transcription through interaction with Gadd45a.


Assuntos
Proteínas de Ciclo Celular/metabolismo , RNA Helicases DEAD-box/metabolismo , Desmetilação do DNA , Neoplasias/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/genética , Regiões Promotoras Genéticas , Mapas de Interação de Proteínas , Ativação Transcricional
16.
ORL J Otorhinolaryngol Relat Spec ; 81(5-6): 317-326, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31639804

RESUMO

BACKGROUND: This work aimed to explore the predictors of lymph node metastasis (LNM) and analyze the prognosis of patients with clinically node-negative (cN0) T1-T2 supraglottic laryngeal carcinoma (SGLC). METHODS: Data for 130 patients with cN0 T1-T2 SGLC who initially underwent surgery were retrospectively reviewed. Occult LNM incidence, relevant factors, and prognosis were analyzed. RESULTS: Of the 130 patients with cN0 T1-T2 SGLC, 21 (16.2%) had occult LNM. Based on univariate and multivariable regression analyses, male sex and poor tumor differentiation predicted the incidence of occult LNM. The incidence of occult LNM was 20.9% in males and 5.1% in females (p = 0.035). Patients with poorly differentiated tumors had a higher incidence of occult LNM (42.9%) than patients with well-differentiated (10.3%) and moderately differentiated tumors (14.3%; p < 0.05). Thirteen patients (10%) had cervical recurrence, and all had T2 tumors (p = 0.02). The 5-year disease-specific survival rates were 70 and 90% for patients with and without LNM, respectively (p = 0.000). CONCLUSIONS: Sex and tumor differentiation are potential predictors of occult nodal disease. Female patients with cN0 T1-T2 SGLC are less likely than male patients to have neck metastasis. Poorly differentiated tumors are associated with the frequency of neck metastasis, and selective neck dissection is strongly recommended for these tumors.


Assuntos
Neoplasias Laríngeas/patologia , Neoplasias Laríngeas/cirurgia , Laringectomia/métodos , Metástase Linfática , Adulto , Idoso , Feminino , Humanos , Excisão de Linfonodo , Masculino , Pessoa de Meia-Idade , Esvaziamento Cervical , Gradação de Tumores , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Prognóstico , Estudos Retrospectivos , Fatores Sexuais
17.
Protein Sci ; 28(10): 1819-1829, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31351024

RESUMO

Fstl1 is a TGF-ß superfamily binding protein which involved in many pathological processes. The function of Fstl1 has been widely elucidated, but its structural characterization has not been explored. Here we solved the high-resolution crystal structure of FK domain of murine Fstl1, analyzed its unique characteristics, and investigated its contribution to the function of full-length Fstl1. We found that Fstl1-FK forms a stable dimer in both solution and crystal, which suggest that this protein may function as a dimer during its interaction with TGF-ß, a molecule known to form dimer during activation process. We also found this FK domain is indispensable for the proper function of Fstl1 during the transduction of TGF-ß signaling. These observations provide important insights into the understanding of Fstl1 and may facilitate the exploration of this molecule in clinical study.


Assuntos
Proteínas Relacionadas à Folistatina/química , Proteínas Relacionadas à Folistatina/metabolismo , Fibrose Pulmonar/metabolismo , Células A549 , Animais , Bleomicina/administração & dosagem , Clonagem Molecular , Cristalografia por Raios X , Proteínas Relacionadas à Folistatina/genética , Humanos , Injeção Intratimpânica , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Domínios Proteicos , Fibrose Pulmonar/induzido quimicamente , Células Tumorais Cultivadas
18.
Int J Mol Med ; 44(1): 273-280, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31115510

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by progressive lung scarring and excessive extracellular matrix depositon. When stimulated, alveolar epithelial cells (AECs) are aberrantly activated, the expression of profibrotic molecules is enhanced, and lung fibrosis is promoted, but the mechanism for this is unclear. It has been reported that a downregulation of the Na,K­ATPase ß1 subunit in renal epithelial cells is involved in renal fibrosis development, but the role of this protein in lung fibrosis remains unknown. In the present study, the expression of the Na,K­ATPase ß1 subunit was revealed to be markedly decreased in AECs of patients with IPF and a bleomycin­induced pulmonary fibrosis mouse model. Treatment with transforming growth factor ß­1 led to significantly downregulation of the Na,K­ATPase ß1 subunit in lung adenocarcioma A549 cells. Furthermore, the knockdown of the Na,K­ATPase ß1 subunit in A549 cells resulted in the upregulation of profibrotic molecules, activation of the neurogenic locus notch homolog protein 1 and extracellular signal­regulated kinase 1/2 signaling pathways and induction of endoplasmic reticulum stress. These findings reveal that the downregulation of the Na,K­ATPase ß1 subunit enhances the expression of profibrotic molecules in AECs and may contribute to IPF pathogenesis.


Assuntos
Células Epiteliais Alveolares/enzimologia , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica , Fibrose Pulmonar Idiopática/enzimologia , Sistema de Sinalização das MAP Quinases , ATPase Trocadora de Sódio-Potássio/biossíntese , Células A549 , Adulto , Células Epiteliais Alveolares/patologia , Animais , Estresse do Retículo Endoplasmático , Humanos , Fibrose Pulmonar Idiopática/patologia , Masculino , Camundongos , Pessoa de Meia-Idade
19.
Int J Radiat Oncol Biol Phys ; 103(1): 208-216, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30171878

RESUMO

PURPOSE: Radiation-induced pulmonary fibrosis (RIPF) is a severe and life-threatening complication of radiation therapy in patients with thoracic cancer; however, the exact molecular mechanisms remain unknown, and there is no effective treatment method in clinic. Here, we assessed the role of follistatin-like 1 (Fstl1) in RIPF. METHODS AND MATERIALS: Protein and messenger RNA levels of Fstl1 in lung tissues from symptomatic RIPF patients, Rhesus macaques, and mice were assessed. Fibrotic and inflammatory responses to radiation-induced lung injury and accumulation of myofibroblasts in Fstl1 haplodeficient (Fstl1+/-) mice were determined. Finally, radiation-induced differentiation and activation of fibroblasts in primary Fstl1+/- lung fibroblasts were evaluated. RESULTS: FSTL1 amounts were significantly increased in serum and/or radiation-injured lung specimens from symptomatic RIPF patients, Rhesus macaques, and mice. Haplodeletion of Fstl1 in Fstl1+/- mice was protective against x-ray-induced lung injury in mice in vivo, as well as myofibroblast activation in vitro. CONCLUSIONS: These findings suggest that Fstl1 plays an important role in lung fibrosis and may offer a potential approach to attenuate RIPF in radiation therapy of patients with thoracic cancer.


Assuntos
Proteínas Relacionadas à Folistatina/fisiologia , Fibrose Pulmonar/prevenção & controle , Pneumonite por Radiação/prevenção & controle , Animais , Diferenciação Celular/efeitos da radiação , Proteínas Relacionadas à Folistatina/sangue , Proteínas Relacionadas à Folistatina/genética , Deleção de Genes , Humanos , Macaca mulatta , Masculino , Camundongos , Miofibroblastos/efeitos da radiação , Fibrose Pulmonar/etiologia
20.
Transl Oncol ; 12(1): 162-169, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30342294

RESUMO

Radiation-induced pulmonary fibrosis (RIPF) is a common complication in patients with lung cancer and breast cancer after receiving thoracic radiotherapy. The average incidence of RIPF is 16%-28% after radiotherapy. RIPF includes a heterogeneous group of lung disorders characterized by progressive and irreversible destruction of lung architecture and disruption of gas exchange. The clinical signs of RIPF include increasing dyspnea, deteriorating lung function, and accumulation of interstitial fluid, eventually leading to respiratory failure. No medical therapy for RIPF has been approved for routine clinical use despite the apparent need for an effective treatment. Numerous signaling pathways are involved in the initiation and progression of RIPF. Also, various approaches for RIPF treatments have focused on several aspects of the current understanding of the molecular pathology of RIPF. This review used the mechanistic categories of associated cell signaling pathways, epithelial cell dysfunction and senescence, abnormal lung remodeling, and aberrant innate and adaptive immunity to review the published literature on RIPF to date and then to identify potential areas for the effective treatment of RIPF.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA