Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Inflammation ; 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39017810

ABSTRACT

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.
Inflamm Res ; 73(7): 1223-1237, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38789791

ABSTRACT

INTRODUCTION: Inflammation and oxidative stress are key factors in the development of pulmonary fibrosis (PF) by promoting the differentiation of fibroblasts through modulating various pathways including Wnt/ß-catenin, TGF-ß and mTOR signalling. OBJECTIVE AND METHODS: This study aimed to evaluate the effects and elucidate the mechanisms of vistusertib (VSB) in treating pulmonary inflammation/fibrosis, specifically by targeting the mTOR pathway using various in vitro and in vivo models. RESULTS: Lipopolysaccharide (LPS)-induced inflammation model in macrophages (RAW 264.7), epithelial (BEAS-2B) and endothelial (HMVEC-L) cells revealed that treatment with VSB significantly reduced the IL-6, TNF-α, CCL2, and CCL7 expression. TGF-ß induced differentiation was also significantly reduced upon VSB treatment in fibrotic cells (LL29 and DHLF). Further, bleomycin-induced inflammation and fibrosis models demonstrated that treatment with VSB significantly ameliorated the severe inflammation, and lung architectural distortion, by reducing the inflammatory markers expression/levels, inflammatory cells and oxidative stress indicators. Further, fibrosis model results exhibited that, VSB treatment significantly reduced the α-SMA, collagen and TGF-ß expressions, improved the lung architecture and restored lung functions. CONCLUSION: Overall, this study uncovers the anti-inflammatory/anti-fibrotic effects of VSB by modulating the mTOR activation. Although VSB was tested for lung fibrosis, it can be tested for other fibrotic disorders to improve the patient's survival and quality of life.


Subject(s)
Bleomycin , Lung , Oxidative Stress , Pneumonia , Pulmonary Fibrosis , Signal Transduction , TOR Serine-Threonine Kinases , Animals , TOR Serine-Threonine Kinases/metabolism , Oxidative Stress/drug effects , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Humans , Signal Transduction/drug effects , Pneumonia/drug therapy , Pneumonia/metabolism , Pneumonia/chemically induced , Pneumonia/pathology , Lung/pathology , Lung/drug effects , Lung/metabolism , Mice, Inbred C57BL , Male , Lipopolysaccharides , Cytokines/metabolism , RAW 264.7 Cells , Cell Line , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology
3.
Inflammation ; 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38466531

ABSTRACT

Acute myocarditis, also known as myocardial inflammation, is a self-limited condition caused by systemic infection with cardiotropic pathogens, primarily viruses, bacteria, or fungi. Despite significant research, inflammatory cardiomyopathy exacerbated by heart failure, arrhythmia, or left ventricular dysfunction and it has a dismal prognosis. In this study, we aimed to evaluate the therapeutic effect of yohimbine against lipopolysaccharide (LPS) induced myocarditis in rat model. The anti-inflammatory activity of yohimbine was assessed in in-vitro using RAW 264.7 and H9C2 cells. Myocarditis was induced in rats by injecting LPS (10 mg/kg), following the rats were treated with dexamethasone (2 mg/kg) or yohimbine (2.5, 5, and 10 mg/kg) for 12 h and their therapeutic activity was examined using various techniques. Yohimbine treatment significantly attenuated the LPS-mediated inflammatory markers expression in the in-vitro model. In-vivo studies proved that yohimbine treatment significantly reduced the LPS-induced increase of cardiac-specific markers, inflammatory cell counts, and pro-inflammatory markers expression compared to LPS-control samples. LPS administration considerably affected the ECG, RR, PR, QRS, QT, ST intervals, and hemodynamic parameters, and caused abnormal pathological parameters, in contrast, yohimbine treatment substantially improved the cardiac parameters, mitigated the apoptosis in myocardial cells and ameliorated the histopathological abnormalities that resulted in an improved survival rate. LPS-induced elevation of cardiac troponin-I, myeloperoxidase, CD-68, and neutrophil elastase levels were significantly attenuated upon yohimbine treatment. Further investigation showed that yohimbine exerts an anti-inflammatory effect partly by modulating the MAPK pathway. This study emphasizes yohimbine's therapeutic benefit against LPS-induced myocarditis and associated inflammatory markers response by regulating the MAPK pathway.

4.
Phytomedicine ; 123: 155182, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37952411

ABSTRACT

BACKGROUND AND PURPOSE: Chronic liver injury, caused by various aetiologies, causes recurrent tissue damage, culminating in decreased liver regenerative ability and resulting in fibrosis followed by cirrhosis. In this study, the anti-fibrotic activity of Yohimbine hydrochloride (YHC) was investigated using various in vitro models and in vivo models. METHODS: To assess the anti-inflammatory, antioxidant, and anti-fibrotic effects of YHC, lipopolysaccharide or TGF-ß induced differentiation or lipid-induced oxidative-stress models were employed using HLECs, HSC-LX2, and HepG2 cells. Further, thioacetamide (TAA) induced hepatic inflammation/fibrosis models were utilized to validate the YHC's anti-fibrotic activity in rats. RESULTS: Inflammation/differentiation experiments in HLECs and HSC-LX2 revealed that YHC treatment significantly (p < 0.001) mitigated the lipopolysaccharide or TGF-ß induced upregulation of inflammatory and fibrotic markers expression respectively. In addition, YHC dose-dependently reduced the TGF-ß induced migration and palmitic acid-induced oxidative stress in HepG2 cells. Further, TAA administration (5 weeks) in vivo rat model showed increased inflammatory marker levels/expression, oxidative stress, and pathological abnormalities. Additionally, TAA administration (9 weeks) elevated the fibrotic marker expression, collagen deposition in liver tissues, and shortened longevity in rats. Treatment with YHC dose-dependently mitigated the TAA-induced abnormalities in both inflammation and fibrosis models and improved the survival of the rats. Further mechanistic approaches revealed that TAA administration elevated the JNK, Wnt components and ß-catenin expression in hepatic stellate cells and animal tissues. Further treatment with YHC significantly modulated the JNK/Wnt/ß-catenin signaling. Moreover, the ß-catenin nuclear translocation results showed that ß-catenin levels were significantly elevated in the nuclear fraction of TAA control samples and reduced in YHC-treated samples. CONCLUSION: Yohimbine treatment significantly improved inflammation and fibrosis by inhibiting differentiation, oxidative stress, and collagen deposition by partly modulating the JNK/Wnt/ß-catenin pathway. These results might serve as a foundation for proposing yohimbine as a potential lead compound for liver fibrosis.


Subject(s)
Lipopolysaccharides , beta Catenin , Rats , Animals , beta Catenin/metabolism , Yohimbine/pharmacology , Yohimbine/metabolism , Yohimbine/therapeutic use , Lipopolysaccharides/pharmacology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver , Oxidative Stress , Collagen/metabolism , Hepatic Stellate Cells , Inflammation/metabolism , Transforming Growth Factor beta/metabolism , Thioacetamide
5.
Int J Pharm ; 649: 123644, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38040396

ABSTRACT

Nintedanib (NIN) and pirfenidone are the only approved drugs for the treatment of Idiopathic Pulmonary Fibrosis (IPF). However, NIN and pirfenidone have low oral bioavailability and limited therapeutic potential, requiring higher dosages to increase their efficacy, which causes significant liver and gastrointestinal toxicities. In this study, we aimed to develop nintedanib-loaded solid lipid nanoparticles (NIN-SLN) to improve the oral bioavailability and therapeutic potential against TGF-ß-induced differentiation in IPF fibroblasts and bleomycin (BLM)-induced lung fibrosis in rat models. NIN-SLN was prepared using a double-emulsification method and characterization studies (Particle size, zeta potential, entrapment efficiency and other parameters) were performed using various techniques. NIN-SLN treatment significantly (p < 0.001) downregulated α-SMA and COL3A1 expression in TGF-ß stimulated DHLF and LL29 cells. NIN-SLN showed a 2.87-fold increase in the bioavailability of NIN and also improved the NIN levels in lung tissues compared to NIN alone. Pharmacodynamic investigation revealed that NIN-SLN (50 mg/Kg) treatment significantly attenuated BLM-induced lung fibrosis by inhibiting epithelial-to-mesenchymal-transition (EMT), extracellular matrix remodelling, and collagen deposition compared to free NIN. Additionally, in the BLM model of fibrosis, NIN-SLN greatly improved the BLM-caused pathological changes, attenuated the NIN-induced gastrointestinal abnormalities, and significantly improved the lung functional indices compared to free NIN. Collectively, NIN-SLN could be a promising nanoformulation for the management of pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung , Rats , Animals , Biological Availability , Lung/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/therapeutic use , Bleomycin
6.
Bioorg Med Chem Lett ; 97: 129549, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37952597

ABSTRACT

Therapeutically active lipids in drug delivery systems offer customization for enhanced pharmaceutical and biological effects, improving safety and efficacy. Biologically active N, N-didodecyl-3,4-dimethoxy-N-methylbenzenaminium lipid (Q) was synthesized and employed to create a liposome formulation (FQ) encapsulating melphalan (M) through a thin film hydration method. Synthesized cationic lipids and their liposomal formulation underwent characterization and assessment for additive anti-cancer effects on myeloma and melanoma cancer cell lines. These effects were evaluated through various studies, including cytotoxicity assessments, cell cycle arrest analysis, apoptosis measurements, mitochondrial membrane potential depolarization, DNA fragmentation, and a significant reduction in tumorigenic potential, as evidenced by a decrease in both the number and percentage area of cancer spheroids.


Subject(s)
Antineoplastic Agents , Liposomes , Humans , Cell Line , Drug Delivery Systems , Lipids , Melphalan/pharmacology , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
7.
Life Sci ; 330: 122027, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37597767

ABSTRACT

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.


Subject(s)
Acute Lung Injury , Pneumonia , Respiratory Distress Syndrome , Virus Diseases , Cricetinae , Animals , Mice , Tumor Necrosis Factor-alpha , Pneumonia/chemically induced , Pneumonia/drug therapy , Inflammation/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Goblet Cells
8.
NanoImpact ; 31: 100479, 2023 07.
Article in English | MEDLINE | ID: mdl-37572937

ABSTRACT

Our earlier reports established that zinc oxide nanoflowers (ZONF) show significant pro-angiogenic properties, where reactive oxygen species, nitric oxide and MAPK-AKT-eNOS cell signaling axis play an essential task. Considering the significance of angiogenesis in healthcare, our research group has recently demonstrated the in vivo therapeutic application of ZONF (10 mg/kg b.w.) for treating peripheral artery disease. Moreover, based on the angio-neural crosstalk between vascular and neuronal systems, we have further demonstrated the neuritogenic and neuroprotective characteristics of pro-angiogenic nanoflowers (10 mg/kg b.w.) for the treatment of cerebral ischemia. However, it is crucial for a therapeutic material to be non-toxic for its practical clinical applications and therefore assessment of its in vivo toxicity and adverse effect is highly important. Herein, for the first time, we investigate a detailed nanotoxicology of therapeutically active ZONF in Swiss albino mice to evaluate their safety profile and comprehend their aspects for future clinical applications. The maximum tolerated dose (MTD) of ZONF was found to be 512.5 mg/kg b.w. which was employed for acute exposure (2 weeks), showing slight toxicity. However, sub-chronic (4 weeks) and long term chronic (8-12 weeks) studies of nanoflowers exhibited their non-toxic nature particularly at lower therapeutic doses (1-10 mg/kg b.w.). Additionally, in depth genotoxicity study revealed that lower therapeutic dose of ZONF (10 mg/kg b.w.) did not exhibit significant toxicity even in genetic level. Overall, the present nanotoxicology of ZONF suggests their high biocompatible nature at therapeutic dose, offering the basis of their future clinical applications in ischemic and other vascular diseases.


Subject(s)
Zinc Oxide , Mice , Animals , Zinc Oxide/toxicity , Reactive Oxygen Species
9.
Eur J Pharmacol ; 953: 175820, 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37245857

ABSTRACT

In idiopathic pulmonary fibrosis (IPF), excessive collagen deposition predisposes to irreversible lung function decline, respiratory failure, and ultimately death. Due to the limited therapeutic efficacy of FDA-approved medications, novel drugs are warranted for better treatment outcomes. Dehydrozingerone (DHZ) is an analogue of curcumin that has been investigated against pulmonary fibrosis using a bleomycin-induced pulmonary fibrosis model in rats. In in vitro, TGF-ß-induced differentiation models (using NHLF, LL29, DHLF and A549 cells) were adopted to assess fibrotic markers expression and explored the mechanism of action. DHZ administration attenuated the bleomycin-induced elevation of lung index, inflammatory cell infiltrations, and hydroxyproline levels in lung tissues. Furthermore, treatment with DHZ mitigated the bleomycin-mediated elevation of extracellular matrix (ECM), epithelial-to-mesenchymal-transition (EMT), and collagen deposition markers and improved lung mechanics. In addition, treatment with DHZ significantly suppressed the BLM-induced apoptosis and rescued the BLM-induced pathological abnormalities in lung tissues. In vitro assays revealed that DHZ suppressed the expression of TGF-ß-elevated collagen deposition, EMT and ECM markers in both mRNA/protein levels. Our findings showed that DHZ has anti-fibrotic effect against pulmonary fibrosis by modulating Wnt/ß-catenin signaling, suggesting that DHZ may serve as a potential treatment option for IPF.


Subject(s)
Epithelial-Mesenchymal Transition , Idiopathic Pulmonary Fibrosis , Rats , Animals , beta Catenin/metabolism , Lung , Idiopathic Pulmonary Fibrosis/pathology , Transforming Growth Factor beta/metabolism , Inflammation/drug therapy , Inflammation/pathology , Collagen/metabolism , Bleomycin/pharmacology , Transforming Growth Factor beta1/metabolism
10.
J Nutr Biochem ; 116: 109294, 2023 06.
Article in English | MEDLINE | ID: mdl-36948431

ABSTRACT

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.


Subject(s)
Pulmonary Fibrosis , Animals , Rats , Bleomycin/toxicity , Epithelial-Mesenchymal Transition , Fibroblasts/metabolism , Lung , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/metabolism
11.
Inflammation ; 46(3): 787-807, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36622573

ABSTRACT

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.


Subject(s)
Colitis, Ulcerative , Colitis , Animals , Mice , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Dextran Sulfate/toxicity , Interleukin-17 , Antioxidants/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Disease Models, Animal , Mice, Inbred C57BL
12.
Eur J Pharmacol ; 937: 175366, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36375494

ABSTRACT

Hepatic fibrosis is a progressive consequence of injury to the liver cells. Liver fibrosis causes hepatic dysfunction and also plays a key role in the pathogenesis of other chronic ailments. Dehydrozingerone (DHZ) is a half-structural analogue of curcumin and is known to have several therapeutic benefits. However, the impact of DHZ on liver fibrosis was not investigated. The current investigation attempted to determine the anti-fibrotic effect of DHZ against thioacetamide-induced liver fibrosis in rats and TGF-ß-induced differentiation in human HSC-LX2 cells and to uncover the possible mechanisms. In in-vivo, DHZ significantly reduced the TAA-induced liver index and ameliorated the liver functional parameters. TAA elevated the fibrotic marker's expression in TAA control, on the other hand, DHZ treatment significantly mitigated the same in mRNA and protein levels. Additionally, these findings were supported by histological investigations and immunohistochemistry studies of the fibrotic marker's expressions. DHZ treatment effectively reduced oxidative stress by increasing catalase activity and decreased the expression of inflammatory markers (myeloperoxidase and neutrophil-elastase) in liver tissues. Additionally, collagen staining and histological findings confirmed that DHZ administration significantly reduced TAA induced pathological deformities and elevated collagen levels. In-vitro results showed that TGF-ß-induced differentiation was suppressed by DHZ treatment in a dose-dependent manner. Mechanistic approaches in HSC-LX2 and liver tissues revealed that DHZ treatment mitigated fibrosis by modulating the MAPK-pathway. Overall, these results show that DHZ exhibited anti-fibrotic action by reducing fibrotic markers and their activities through regulation of the MAPK-pathway, suggesting that DHZ may be a promising therapeutic molecule for liver fibrosis.


Subject(s)
Hepatic Stellate Cells , Thioacetamide , Rats , Humans , Animals , Thioacetamide/pharmacology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Biomarkers/metabolism , Collagen/metabolism , Transforming Growth Factor beta/metabolism
13.
Drug Dev Ind Pharm ; 48(8): 384-396, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36047536

ABSTRACT

OBJECTIVE: The main objective is to formulate solid lipid nanoparticles conjugated with cyclic RGDfk peptide encapsulated with gemcitabine hydrochloride drug for targeting breast cancer. SIGNIFICANCE: The hydrophilic nature of gemcitabine hampers passive transport by cell membrane permeation that may lead to drug resistance as it has to enter the cells via nucleoside transporters. The art of encapsulating the drug in a nanovesicle and then anchoring it with a targeting ligand is one of the present areas of research in cancer chemotherapy. METHODS: In this study, solid lipid nanoparticles were prepared by double emulsification and solvent evaporation method. Cyclic RGDfk and gemcitabine hydrochloride were used as targeting ligands and chemotherapeutic drugs, respectively, for targeting breast cancer. The prepared nanoparticles were evaluated for in vitro and in vivo performance to showcase the targeting efficiency and therapeutic benefits of the gemcitabine-loaded ligand conjugated nanoparticles. RESULTS: When compared with gemcitabine (GEM) and GEM loaded nanoparticles (GSLN), the ligand conjugated GEM nanoparticles (cGSLN) showed superior cytotoxicity, apoptosis, and inhibition of 3D multicellular spheroids in human breast cancer cells (MDA MB 231). The in vivo tumor regression studies in orthotopic breast cancer induced Balb/C mice showed that cGSLN displayed superior tumor suppression and also the targeting potential of the cGSLN toward induced breast cancer. CONCLUSION: Prepared nanoformulations showed enhanced anticancer activity in both 2D and 3D cell culture models along with antitumor efficacy in orthotopic breast cancer mouse models.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Nanoparticles , Humans , Mice , Animals , Female , Integrin beta3/therapeutic use , Integrin alphaV , Ligands , Cell Line, Tumor , Breast Neoplasms/pathology , Mice, Inbred BALB C , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Gemcitabine
14.
Apoptosis ; 27(11-12): 825-839, 2022 12.
Article in English | MEDLINE | ID: mdl-35829938

ABSTRACT

Polyphenols are naturally occurring organic compounds with varying structures represented by four major groups: flavonoids, phenolic acids, lignans and stilbenes. Several studies suggested that these secondary metabolites have health benefits due to its anti-tumorigenic effect. Therefore, substantial effort has been put forward to isolate and characterize these natural compounds and synthesize analogues that may serve as potential anti-cancer therapeutics. This present study is aimed at designing and synthesis of azaflavanone derivative and in understanding its mechanism of action in vitro and in vivo. Molecular docking studies predicted that the compound can potentially bind strongly to the Cyclin E1-Cdk2 complex which is a key mediator of the cell cycle progression indicating a biological interference in aggressive prostate cancer. Further downstream studies to understand its cytotoxicity and mechanism of action showed this azaflavanone derivative markedly inhibits viability of prostate cancer cells (DU145) showing an IC50 value of 0.4 µM compared to other cancer cells. The pharmacological ROS insult using the azaflavanone derivative increases the oxidative damage leading to high expression of apoptotic markers with increasing concentration. On compound treatment, the cells lose the metabolic flexibility accompanied by mitochondrial dysfunction leading to cell cycle arrest and apoptosis. Further, no compound mediated toxicity was observed in xenograft mouse model of prostate cancer at a concentration as high as 5 mg/kg. The tumor burden was reduced to 60% rendering the azaflavanone derivative a potential candidate in cancer therapeutics. Collectively, the compound triggers cell cycle arrest and ROS mediated oxidative stress sensitizing the cancerous cells towards apoptosis.


Subject(s)
Apoptosis , Prostatic Neoplasms , Male , Humans , Mice , Animals , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Molecular Docking Simulation , Prostatic Neoplasms/pathology , Cell Proliferation
15.
Molecules ; 27(4)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35208958

ABSTRACT

Chikungunya is an infectious disease caused by mosquito-transmitted chikungunya virus (CHIKV). It was reported that NS1 and E2 siRNAs administration demonstrated CHIKV inhibition in in vitro as well as in vivo systems. Cationic lipids are promising for designing safe non-viral vectors and are beneficial in treating chikungunya. In this study, nanodelivery systems (hybrid polymeric/solid lipid nanoparticles) using cationic lipids (stearylamine, C9 lipid, and dioctadecylamine) and polymers (branched PEI-g-PEG -PEG) were prepared, characterized, and complexed with siRNA. The four developed delivery systems (F1, F2, F3, and F4) were assessed for stability and potential toxicities against CHIKV. In comparison to the other nanodelivery systems, F4 containing stearylamine (Octadecylamine; ODA), with an induced optimum cationic charge of 45.7 mV in the range of 152.1 nm, allowed maximum siRNA complexation, better stability, and higher transfection, with strong inhibition against the E2 and NS1 genes of CHIKV. The study concludes that cationic lipid-like ODA with ease of synthesis and characterization showed maximum complexation by structural condensation of siRNA owing to high transfection alone. Synergistic inhibition of CHIKV along with siRNA was demonstrated in both in vitro and in vivo models. Therefore, ODA-based cationic lipid nanoparticles can be explored as safe, potent, and efficient nonviral vectors overcoming siRNA in vivo complexities against chikungunya.


Subject(s)
Amines , Chikungunya Fever , Chikungunya virus/growth & development , Liposomes , Nanoparticles , RNA, Small Interfering , Amines/chemistry , Amines/pharmacology , Animals , Chikungunya Fever/drug therapy , Chikungunya Fever/metabolism , Chlorocebus aethiops , Liposomes/chemistry , Liposomes/pharmacology , Mice , Nanoparticles/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology , Vero Cells
16.
Colloids Surf B Biointerfaces ; 211: 112305, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34998178

ABSTRACT

Erlotinib-based EGFR targeted therapy has proven significant clinical improvement against non-small cell lung cancer (NSCLC). However, the anticancer activity of Erlotinib (Ertb) is limited by the development of Ertb resistance and possess a challenge to clinicians and patients. To explore a better therapeutic strategy, we evaluated Ertb in combinations with different natural products. We identified that Ertb and Quercetin (Quer) combination is more synergistic against A549 and NCI H460 cells compared to Ertb with Fisetin/Carnosic acid/Luteolin. To further improve the efficacy and overcome the limitation of free therapeutics, Ertb and Quer loaded solid lipid nanoparticles (EQNPs) were prepared using Chitosan-MA-TPGS polymer by hot homogenization method. The drug-loaded nanoparticles (NPs) have shown high encapsulation efficiency (77% Ertb and 71.4% Quer) as well as small particle size of 87.3 ± 0.78 nm and positive zeta potential + 13.4 ± 1.12 mV. At pH 5.5, Ertb and Quer were released at their highest levels. We found that, EQNPs decreased the expression of P-glycoprotein (P-gp) and nuclear epidermal growth factor receptor (nEGFR). EQNPs increased the uptake of Ertb and Quer, and apoptosis induction in Ertb resistant A549/ER cells. Further, in vivo EQNPs formulation have shown increased uptake of nanoparticles in the lung tissue and significantly reduced the expression of nEGFR. Thus, EQNPs may be developed as a targeted medicine with minimum side effects for treatment of NSCLC to improve the quality of life and survival of NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Nanoparticles , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Liposomes , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Nanoparticles/therapeutic use , Phosphatidylinositol 3-Kinases/pharmacology , Phosphatidylinositol 3-Kinases/therapeutic use , Proto-Oncogene Proteins c-akt , Quality of Life , Quercetin/pharmacology
18.
Phytomedicine ; 92: 153729, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34517257

ABSTRACT

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.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , Animals , Cytokine Release Syndrome , Endothelial Cells/metabolism , Humans , Lipopolysaccharides , Lung/metabolism , NF-kappa B/metabolism , Oxidative Stress , Pandemics , Rats , Respiratory Distress Syndrome/drug therapy , SARS-CoV-2 , Styrenes
19.
Bioorg Med Chem Lett ; 49: 128307, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34363936

ABSTRACT

We report herein, the design, synthesis and study of anticancer properties of sulfenylated 2-phenylimidazo[1,2-a]pyridines and their analogues. A set of twenty sulfenylated imidazo[1, 2-a]pyridine derivatives were synthesized. Whereby elusive amendments to the imidazo[1,2-a]pyridine motif confer dramatic changes in functional affinity of a novel action to modulate anticancer activity in seven different human cancer cell lines i.e.: MDA MB 231 (breast), HepG2 (liver), Hela (cervical), A549 (lung), U87MG (glioblastoma), SKMEL-28 (skin melanoma) and DU-145 (prostate) by employing MTT assay. Among the series, compounds 4e (naphthalene), 4f (styrene) and 4h (thiomethyl) showed potent activity towards human liver cancer cells HepG2. Cell cycle analysis results revealed that these compounds arrested the cell cycle at G2/M phase and induced apoptosis in human liver cancer cells HepG2. It was further confirmed by Hoechst staining, Measurement of mitochondrial membrane potential (ΔΨm) and Annexin V-FITC assay.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Sulfides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , HEK293 Cells , Humans , Imidazoles/chemical synthesis , Mice , Sulfides/chemical synthesis
20.
Int Immunopharmacol ; 96: 107754, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34162135

ABSTRACT

Psoriasis is a most common chronic autoimmune-arbitrated cutaneous inflammatory skin disorder by unclear pathogenesis. In this current study we demonstrated the effect of galangin (GAL) on imiquimod (IMQ)-induced psoriasis-like skin inflammation and decipher its possible protective mechanism which has not been investigated. The in vivo results revealed that GAL at 1% w/w and 2% w/w for six consecutive days markedly reduced IMQ-induced PASI scoring, skin, ear thickness, hematological markers, levels of nitrites, TBARS, MPO, histopathological, as well modulated the protein levels of pro-inflammatory mediators of COX-2, iNOS, NF-κB pathway and pro-inflammatory cytokines IL-17, IL-23, IL-1ß in the skin and also IL-6, TNF-α in both skin and serum. Besides, GAL restored the levels of antioxidants markers such as SOD, CAT, GST, GSH, GR and Vit-C, anti-inflammatory cytokine of IL-10, and the protein levels of Nrf2/HO-1 in the skin compared to the IMQ group. Finally, our study demonstrates that GAL exerted its protective effect by up-regulating the anti-inflammatory and the antioxidant markers against psoriasis pre-clinical models indicating its potency for treating psoriasis in humans.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dermatitis/drug therapy , Dermatitis/metabolism , Flavonoids/pharmacology , NF-E2-Related Factor 2/agonists , NF-kappa B/genetics , Psoriasis/drug therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/metabolism , Body Weight/drug effects , Cytokines/blood , Dermatitis/etiology , Dermatitis/pathology , Disease Models, Animal , Down-Regulation/drug effects , Flavonoids/therapeutic use , Heme Oxygenase-1/metabolism , Imiquimod/toxicity , Male , Membrane Proteins/metabolism , Mice, Inbred BALB C , NF-kappa B/metabolism , Oxidative Stress/drug effects , Peroxidase/metabolism , Psoriasis/blood , Psoriasis/chemically induced , Signal Transduction/drug effects , Spleen/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...