Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Inflammation ; 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39017810

RESUMO

Psoriasis is a chronic skin inflammatory disorder characterized by the hyper-activation of the immune system and the over-proliferation of epidermal keratinocytes. This study aimed to investigate the anti-psoriatic activity of Biochanin A (BCA), a phytomolecule with known anti-inflammatory and anti-cancer properties, using the IMQ-induced psoriasis-like mouse model. Network pharmacology analysis was performed to investigate the targetability of Biochanin A (BCA) against psoriasis. Psoriasis-like skin inflammation was established using BALB/c mice by topical application of IMQ (5%). BCA cream (0.3%, 1%, 3%) was applied on the skin regions every day for 6 days. The skin phenotypes-erythema and scaling were scored every day. On the 7th day, skin tissues were collected for gene expression analysis, histopathological analysis, cytokine levels determination, and western blot analysis for signaling mechanisms. The network pharmacology analysis has identified 57 common targets between psoriasis and BCA. The topical application of IMQ induced a typical psoriasis-like skin phenotype including redness, skin thickening, and plaque formation. Upon BCA treatment, the psoriasis-like symptoms were significantly reduced in a dose-dependent manner. The targets identified by the network pharmacology (MMP9, EGFR, and PTGS2) and the pro-inflammatory cytokine gene expression were found to be significantly elevated in IMQ controls, and upon BCA treatment they were found significantly reduced. The release of cytokines linked to psoriasis (IL-17A and IL-23) were significantly reduced upon BCA treatment. Furthermore, our findings demonstrated that BCA treatment alleviated the psoriasis-like symptoms via modulating NF-κB and MAPK signaling pathways. Our results demonstrate the therapeutic potential of BCA against IMQ-induced psoriasis-like skin inflammation.

2.
Life Sci ; 346: 122626, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38614295

RESUMO

AIM: Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive condition with unknown aetiology that causes the lung parenchyma to scar incessantly, lowering the quality of life and hastening death. In this investigation, we studied the anti-fibrotic activity of Geneticin (a derivative of gentamycin) using in vitro and in vivo models. MAIN METHODS: The TGF-ß-mediated differentiation model was adopted to investigate (fibrotic marker's levels/expression) the anti-fibrotic activity of geneticin (GNC) in in-vitro scenarios (LL29 and DHLF cells). In vivo, the bleomycin (BLM)-induced pulmonary fibrosis model was employed by administering BLM intratracheally. Post 14 days of BLM administration, animals were treated with geneticin (6.25, 12.5, and 25 mg·kg-1) for another 14 days, and their therapeutic effect was investigated using a spectrum of techniques. KEY FINDINGS: RTqPCR and western-blot results revealed that geneticin treatment significantly attenuated the TGF-ß/BLM mediated fibrotic cascade of markers in both in-vitro and in-vivo models respectively. Further, the BLM-induced pulmonary fibrosis model revealed, that geneticin dose-dependently reduced the BLM-induced inflammatory cell infiltrations, and thickness of the alveoli walls, improved the structural distortion of the lung, and aided in improving the survival rate of the rats. Picrosirus and Masson's trichrome staining indicated that geneticin therapy reduced collagen deposition and, as a result, lung functional characteristics were improved as assessed by flexivent. Mechanistic studies have shown that geneticin reduced fibrosis by attenuating the TGF-ß/Smad through modulating the AMPK/SIRT1 signaling. SIGNIFICANCE: These findings suggest that geneticin may be a promising therapeutic agent for the treatment of pulmonary fibrosis in clinical settings.


Assuntos
Proteínas Quinases Ativadas por AMP , Bleomicina , Fibrose Pulmonar , Transdução de Sinais , Sirtuína 1 , Fator de Crescimento Transformador beta , Animais , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/patologia , Fibrose Pulmonar/induzido quimicamente , Ratos , Sirtuína 1/metabolismo , Sirtuína 1/genética , Masculino , Bleomicina/toxicidade , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Smad/metabolismo , Ratos Sprague-Dawley , Modelos Animais de Doenças
3.
ACS Appl Bio Mater ; 6(11): 4814-4827, 2023 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-37886889

RESUMO

Bacterial infections and persistent inflammation can impede the intrinsic healing process of wounds. To combat this issue, researchers have delved into the potential use of carbon dots (CDs) in the regulation of inflammation and counteract infections. These CDs were synthesized using a microwave-assisted hydrothermal process and have demonstrated outstanding antibacterial and antibiofilm properties against Gram-positive and Gram-negative bacteria. Additionally, CDs displayed biocompatibility at therapeutic concentrations and the ability to specifically target mitochondria. CD treatment effectively nullified lipopolysaccharide-triggered reactive oxygen species production by macrophages, while simultaneously promoting macrophage polarization toward an anti-inflammatory phenotype (M2), leading to a reduction in inflammation and an acceleration in wound healing. In vitro scratch assays also revealed that CDs facilitated the tissue-repairing process by stimulating epithelial cell migration during reepithelialization. In vivo studies using CDs topically applied to lipopolysaccharide (LPS)-stimulated wounds in C57/BL6 mice demonstrated significant improvements in wound healing due to enhanced fibroblast proliferation, angiogenesis, and collagen deposition. Crucially, histological investigations showed no indications of systemic toxicity in vital organs. Collectively, the application of CDs has shown immense potential in speeding up the wound-healing process by regulating inflammation, preventing bacterial infections, and promoting tissue repair. These results suggest that further clinical translation of CDs should be considered.


Assuntos
Antibacterianos , Infecções Bacterianas , Camundongos , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Lipopolissacarídeos/farmacologia , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Cicatrização , Macrófagos , Inflamação
4.
Life Sci ; 330: 122027, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37597767

RESUMO

AIMS: Acute lung inflammation, particularly acute respiratory distress syndrome (ARDS), is caused by a variety of pathogens including bacteria and viruses. ß-Glucans have been reported to possess both anti-inflammatory and immunomodulatory properties. The current study evaluated the therapeutic effect of ß-glucans on polyinosinic:polycytidylic acid (Poly(I:C)) induced lung inflammation in both hamster and mice models. MAIN METHODS: Poly(I:C)-induced ALI/inflammation models were developed in hamsters (2.5 mg/kg) and mice (2 mg/kg) by delivering the Poly(I:C) intratracheally, and followed with and without ß-glucan administration. After treatment, lung mechanics were assessed and lung tissues were isolated and analyzed for mRNA/protein expression, and histopathological examinations. KEY FINDINGS: Poly(I:C) administration, caused a significant elevation of inflammatory marker's expression in lung tissues and showed abnormal lung mechanics in mice and hamsters. Interestingly, treatment with ß-glucan significantly (p < 0.001) reversed the Poly(I:C)-induced inflammatory events and inflammatory markers expression in both mRNA (IL-6, IL-1ß, TNF-α, CCL2 and CCL7) and protein levels (TNF-α, CD68, myeloperoxidase, neutrophil elastase, MUC-5Ac and iNOS). Lung functional assays revealed that ß-glucan treatment significantly improved lung mechanics. Histopathological analysis showed that ß-glucan treatment significantly attenuated the Poly(I:C) induced inflammatory cell infiltration, injury and goblet cell population in lung tissues. Consistent with these findings, ß-glucan treatment markedly reduced the number of neutrophils and macrophages in lung tissues. Our findings further demonstrated that ß-glucan could reduce inflammation by suppressing the MAPK pathway. SIGNIFICANCE: These results suggested that ß-glucan may attenuate the pathogenic effects of Poly(I:C)-induced ALI/ARDS via modulating the MAPK pathway, indicating ß-glucan as a possible therapeutic agent for the treatment of viral-pulmonary inflammation/injury.


Assuntos
Lesão Pulmonar Aguda , Pneumonia , Síndrome do Desconforto Respiratório , Viroses , Cricetinae , Animais , Camundongos , Fator de Necrose Tumoral alfa , Pneumonia/induzido quimicamente , Pneumonia/tratamento farmacológico , Inflamação/tratamento farmacológico , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Células Caliciformes
5.
J Nutr Biochem ; 116: 109294, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36948431

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive disorder that severely impairs lung function, by increasing lung stiffness. Sesamol, a phenolic Phyto-molecule isolated from sesame seeds, possess a rich source of protein and is known to have extensive nutritional and health effects. Here we investigated the effect of sesamol on TGF-ß/periostin-induced fibroblast differentiation in in vitro and bleomycin-induced pulmonary fibrosis in an in vivo model. Our results demonstrated that activation of (DHLF, LL29, NHLF and A549) cells with TGF-ß, elevates the epithelial to mesenchymal transition, extracellular matrix, and collagen deposition and periostin signaling marker's expression, further treatment with sesamol attenuated these markers significantly. In addition, sesamol treatment improved the TGF-ß-induced contraction and migration of cells. Mechanistic studies showed that activation of IPF cells with periostin increased the TGF-ß signaling and treatment with sesamol significantly abrogated the periostin-induced TGF-ß activation and its downstream fibrotic marker's expression. In in vivo, sesamol treatment attenuated the lung inflammation, infiltration of cells, wall thickening and the formation of fibrous bands significantly in BLM-induced fibrosis rats. Molecular studies revealed that sesamol treatment reduced the bleomycin-induced fibrotic, inflammatory, apoptotic marker's expression by modulating the TGF-ß/periostin crosstalk signaling in a dose-dependent manner. Further, treatment with sesamol dramatically improved lung function and decreased mortality. Our study first time reports the sesamol's inhibitory effects on periostin signalling. Collectively, our study demonstrated that periostin and TGF-ß seem to work in a positive-feedback loop, inducing the other, therefore, targeting TGF-ß/periostin signaling may provide a better therapeutic approach against IPF and other fibrotic disorders.


Assuntos
Fibrose Pulmonar , Animais , Ratos , Bleomicina/toxicidade , Transição Epitelial-Mesenquimal , Fibroblastos/metabolismo , Pulmão , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Fator de Crescimento Transformador beta/metabolismo
6.
Inflammation ; 46(3): 787-807, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36622573

RESUMO

Ulcerative colitis (UC) is an intestinal inflammatory disease characterised by the loss of intestinal crypts, edema, mucosal ulceration, and infiltration of inflammatory cells in the mucosa. The current study aimed to investigate the protective and therapeutic effects of sinigrin and underlying mechanisms in a dextran sulfate sodium (DSS)-induced mouse model of ulcerative colitis. DSS-induced colitis models were used to demonstrate sinigrin's therapeutic/protective action. Mice were orally administered with sinigrin (15 mg/kg or 30 mg/kg) for a period of 12 days in both prophylactic and therapeutic models. Animal weights, stool consistency, and bleeding parameters were measured throughout the experimental period. After the experimental period, colon lengths were measured, and colon tissues were harvested to determine the levels of oxidative stress-inducing factors (nitrates and MDA levels) and anti-oxidant components (GSH, SOD, and catalase). Furthermore, gene expression analysis, IL-17 levels, and inflammatory marker expressions were measured using RT-qPCR, ELISA, and immunohistochemical methods respectively. Furthermore, histopathological observations and elucidation of the mechanism of action were determined using H&E analysis and Western blot analysis. Sinigrin treatment (in both prophylactic and therapeutic models) significantly mitigated the DSS-induced body weight loss, attenuated the colon length shrinkage, and improved the disease index score (p < 0.001). Further results revealed that sinigrin's protective/therapeutic effect is associated with a significant attenuation of pro­inflammatory cytokine production (p < 0.001), reversing the anti-oxidant enzyme levels (p < 0.001) and substantial improvement (2 folds) of the disruption of the colonic morphology in colon tissues compared to DSS control. Immunohistochemical analysis showed that sinigrin treatment remarkably reduced the DSS-induced myeloperoxidase, neutrophil elastase, and CD68 expression in colon tissues. Additionally, sinigrin successfully abrogated the DSS-induced IL-17 levels (p < 0.001) and improved the colonic barrier in colon tissues. Overall, these results demonstrated that sinigrin exerts protective and therapeutic effects on DSS­induced colitis, by enhancing the anti-oxidant enzymes and suppressing the intestinal inflammatory cascade of markers by regulating the MAPK pathway.


Assuntos
Colite Ulcerativa , Colite , Animais , Camundongos , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/patologia , Sulfato de Dextrana/toxicidade , Interleucina-17 , Antioxidantes/uso terapêutico , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
7.
Inflammopharmacology ; 31(2): 823-844, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36662401

RESUMO

Acute respiratory distress syndrome (ARDS) is one of the major causes of mortality in COVID-19 patients, due to limited therapeutic options. This prompted us to explore natural sources to mitigate this condition. Gymnema Sylvestre (GS) is an ancient medicinal plant known to have various therapeutic effects. This investigation examined the therapeutic effect of hydroalcoholic extract of Gymnema Sylvestre (HAEGS) against lipopolysaccharide (LPS)-induced lung injury and ARDS in in vitro and in vivo models. UHPLC-HRMS/GC-MS was employed for characterizing the HAEGS and identified several active derivatives including gymnemic acid, gymnemasaponins, gymnemoside, gymnemasin, quercetin, and long fatty acids. Gene expression by RT-qPCR and DCFDA analysis by flow cytometry revealed that several inflammatory cytokine/chemokine, cell injury markers, and reactive oxygen species (ROS) levels were highly upregulated in LPS control and were significantly reduced upon HAEGS treatment. Consistent with the in vitro studies, we found that in LPS-induced ARDS model, pre-treatment with HAEGS significantly suppressed the LPS-induced elevation of inflammatory cell infiltrations, cytokine/chemokine marker expression, ROS levels, and lung injury in a dose-dependent manner. Further mechanistic studies demonstrated that HAEGS suppressed oxidative stress by modulating the NRF2 pathway and ameliorated the ARDS through the NF-κB/MAPK signalling pathway. Additional fractionation results revealed that fraction 6 which has the exclusive composition of gymnemic acid derivatives showed better anti-inflammatory effects (inhibition of IL-6 and IL-1ß) at lower concentrations compared to HAEGS. Overall, HAEGS significantly mitigated LPS-induced lung injury and ARDS by targeting the NF-κB/MAPK signalling pathway. Thus, our work unravels the protective role of HAEGS for the first time in managing ARDS.


Assuntos
COVID-19 , Gymnema sylvestre , Lesão Pulmonar , Síndrome do Desconforto Respiratório , Ratos , Animais , NF-kappa B/metabolismo , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Gymnema sylvestre/metabolismo , Espécies Reativas de Oxigênio , Lesão Pulmonar/tratamento farmacológico , Lipopolissacarídeos/farmacologia , Síndrome do Desconforto Respiratório/tratamento farmacológico , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Citocinas
8.
Phytomedicine ; 92: 153729, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34517257

RESUMO

BACKGROUND: Inflammation-mediated lung injury is a major cause of health problems in many countries and has been the leading cause of morbidity/mortality in intensive care units. In the current COVID-19 pandemic, the majority of the patients experienced serious pneumonia resulting from inflammation (Acute respiratory distress syndrome/ARDS). Pathogenic infections cause cytokine release syndrome (CRS) by hyperactivation of immune cells, which in turn release excessive cytokines causing ARDS. Currently, there are no standard therapies for viral, bacterial or pathogen-mediated CRS. PURPOSE: This study aimed to investigate and validate the protective effects of Dehydrozingerone (DHZ) against LPS induced lung cell injury by in-vitro and in-vivo models and to gain insights into the molecular mechanisms that mediate these therapeutic effects. METHODS: The therapeutic activity of DHZ was determined in in-vitro models by pre-treating the cells with DHZ and exposed to LPS to stimulate the inflammatory cascade of events. We analysed the effect of DHZ on LPS induced inflammatory cytokines, chemokines and cell damage markers expression/levels using various cell lines. We performed gene expression, ELISA, and western blot analysis to elucidate the effect of DHZ on inflammation and its modulation of MAPK and NF-κB pathways. Further, the prophylactic and therapeutic effect of DHZ was evaluated against the LPS induced ARDS model in rats. RESULTS: DHZ significantly (p < 0.01) attenuated the LPS induced ROS, inflammatory cytokine, chemokine gene expression and protein release in macrophages. Similarly, DHZ treatment protected the lung epithelial and endothelial cells by mitigating the LPS induced inflammatory events in a dose-dependent manner. In vivo analysis showed that DHZ treatment significantly (p < 0.001) mitigated the LPS induced ARDS pathophysiology of increase in the inflammatory cells in BALF, inflammatory cytokine and chemokines in lung tissues. LPS stimulated neutrophil-mediated events, apoptosis, alveolar wall thickening and alveolar inflammation were profoundly reduced by DHZ treatment in a rat model. CONCLUSION: This study demonstrates for the first time that DHZ has the potential to ameliorate LPS induced ARDS by inhibiting cytokine storm and oxidative through modulating the MAPK and NF-κB pathways. This data provides pre-clinical support to develop DHZ as a potential therapeutic agent against ARDS.


Assuntos
COVID-19 , Síndrome do Desconforto Respiratório , Animais , Síndrome da Liberação de Citocina , Células Endoteliais/metabolismo , Humanos , Lipopolissacarídeos , Pulmão/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo , Pandemias , Ratos , Síndrome do Desconforto Respiratório/tratamento farmacológico , SARS-CoV-2 , Estirenos
9.
Phytomedicine ; 78: 153298, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32781391

RESUMO

BACKGROUND: Idiopathic Pulmonary Fibrosis (IPF) is a progressive inflammatory disorder driven by a fibrotic cascade of events such as epithelial to mesenchymal transition, extracellular matrix production and collagen formation in the lungs in a sequential manner. IPF incidences were raising rapidly across the world. FDA approved pirfenidone and nintedanib (tyrosine kinase inhibitors) are being used as a first-line treatment drugs for IPF, however, neither the quality of life nor survival rates have been improved because of patient noncompliance due to multiple side effects. Thus, the development of novel therapeutic approaches targeting TGF-ß mediated cascade of fibrotic events is urgently needed to improve the survival of the patients suffering from devastating disease. PURPOSE: The aim of this study was to investigate and validate the anti-fibrotic properties of Biochanin-A (isoflavone) against TGF-ß mediated fibrosis in in vitro, ex vivo, in vivo models and to determine the molecular mechanisms that mediate these anti-fibrotic effects. METHODS: The therapeutic activity of BCA was determined in in vitro/ex vivo models. Cells were pre-treated with BCA and incubated in presence or absence of recombinant-TGF-ß to stimulate the fibrotic cascade of events. Pulmonary fibrosis was developed by intratracheal administration of bleomycin in rats. BCA treatment was given for 14 days from post bleomycin instillation and then various investigations (collagen content, fibrosis gene/protein expression and histopathological changes) were performed to assess the anti-fibrotic activity of BCA. RESULTS: In vitro/ex vivo (Primary normal, IPF cell line and primary IPF cells/ Precision cut mouse lung slices) experiments revealed that, BCA treatment significantly (p < 0.001) reduced the expression of TGF-ß modulated fibrotic genes/protein expressions (including their functions) which are involved in the cascade of fibrotic events. BCA treatment significantly (p < 0.01) reduced the bleomycin-induced inflammatory cell-infiltration, inflammatory markers expression, collagen deposition and expression of fibrotic markers in lung tissues equivalent or better than pirfenidone treatment. In addition, BCA treatment significantly (p < 0.001) attenuated the TGF-ß1/BLM-mediated increase of TGF-ß/Smad2/3 phosphorylation and resulted in the reduction of pathological abnormalities in lung tissues determined by histopathology observations. CONCLUSION: Collectively, BCA treatment demonstrated the remarkable therapeutic effects on TGF-ß/BLM mediated pulmonary fibrosis using IPF cells and rodent models. This current study may offer a novel treatment approach to halt and may be even rescue the devastating lung scarring of IPF.


Assuntos
Colágeno/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Genisteína/farmacologia , Miofibroblastos/efeitos dos fármacos , Fibrose Pulmonar/tratamento farmacológico , Animais , Bleomicina/toxicidade , Diferenciação Celular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Humanos , Camundongos Endogâmicos C57BL , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Ratos Wistar , Reprodutibilidade dos Testes , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...