Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Toxicol In Vitro ; 89: 105579, 2023 Jun.
Article En | MEDLINE | ID: mdl-36870549

Malignant melanoma is considered a deadly aggressive form of skin cancer that frequently metastasizes to various distal organs, which harbors mutations of the BRAF or NRAS which occur in 30 to 50% of melanoma patients. The growth factors secreted by melanoma cells contribute to tumor angiogenesis with the acquisition of metastatic potential by epithelial-mesenchymal transition (EMT) and drive melanoma growth toward a more aggressive form. Niclosamide (NCL) is an FDA-approved anthelmintic drug and is reported to have strong anti-cancer properties against various solid and liquid tumors. Its role in BRAF or NRAS mutated cells is unknown. In this context, we uncovered the role of NCL in impeding malignant metastatic melanoma in vitro in SK-MEL-2 and SK-MEL-28 cell lines. We found that NCL induces significant ROS generation and apoptosis through a series of molecular mechanisms, such as depolarization of mitochondrial membrane potential, arresting the cell cycle at the sub G1 phase with a significant increase in the DNA cleavage via topoisomerase II in both cell lines. We also found that NCL potently inhibited metastasis, which was examined by scratch wound assay, Additionally, we found that NCL inhibits the most important markers involved in the EMT signaling cascade that are stimulated by TGF-ß such as N-cadherin, Snail, Slug, Vimentin, α-SMA and p-Smad 2/3. This work provides useful insights into the mechanism of NCL in BRAF/NRAF mutant melanoma cells via inhibition of molecular signaling events involved in EMT signaling, and apoptosis induction.


Melanoma , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Niclosamide/pharmacology , Epithelial-Mesenchymal Transition , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Apoptosis , Cell Line, Tumor , Mutation , Membrane Proteins/genetics , GTP Phosphohydrolases/genetics
2.
Int Immunopharmacol ; 115: 109701, 2023 Feb.
Article En | MEDLINE | ID: mdl-36641892

Acute respiratory distress syndrome (ARDS) is associated with severe lung inflammation, edema, hypoxia, and high vascular permeability. The COVID-19-associated pandemic ARDS caused by SARS-CoV-2 has created dire global conditions and has been highly contagious. Chronic inflammatory disease enhances cancer cell proliferation, progression, and invasion. We investigated how acute lung inflammation activates the tumor microenvironment and enhances lung metastasis in LPS induced in vitro and in vivo models. Respiratory illness is mainly caused by cytokine storm, which further influences oxidative and nitrosative stress. The LPS-induced inflammatory cytokines made the conditions suitable for the tumor microenvironment in the lungs. In the present study, we observed that LPS induced the cytokine storm and promoted lung inflammation via BRD4, which further caused the nuclear translocation of p65 NF-κB and STAT3. The transcriptional activation additionally triggers the tumor microenvironment and lung metastasis. Thus, BRD4-regulated p65 and STAT3 transcriptional activity in ARDS enhances lung tumor metastasis. Moreover, LPS-induced ARDS might promote the tumor microenvironment and increase cancer metastasis into the lungs. Collectively, BRD4 plays a vital role in inflammation-mediated tumor metastasis and is found to be a diagnostic and molecular target in inflammation-associated cancers.


COVID-19 , Lung Neoplasms , Pneumonia , Respiratory Distress Syndrome , Humans , Nuclear Proteins/genetics , Lipopolysaccharides/pharmacology , Tumor Microenvironment , Cytokine Release Syndrome , SARS-CoV-2 , Transcription Factors/genetics , Lung/pathology , Respiratory Distress Syndrome/chemically induced , Pneumonia/chemically induced , Inflammation , Cell Cycle Proteins/genetics
3.
J Nanobiotechnology ; 19(1): 101, 2021 Apr 13.
Article En | MEDLINE | ID: mdl-33849555

BACKGROUND: Psoriasis is a chronic autoimmune skin disease characterized by hyperproliferation of keratinocytes. Wide treatment options used to treat psoriasis is associated with various adverse effects. To overcome this nanoformulation is prepared. Selenium is an essential trace element and plays major role in oxidation reduction system. Toxicity and stability limits the applications of selenium. Toxicity can be reduced and stabilized upon preparation into nanoparticles. RESULTS: Selenium nanoparticles (SeNPs) exhibit potent apoptosis through the generation of reactive oxygen species (ROS) with cell cycle arrest. SeNPs topical gel application produced significant attenuation of psoriatic severity with the abrogation of acanthosis and splenomegaly. SeNPs reduced the phosphorylation and expressions of MAPKs, STAT3, GSK-3ß, Akt along with PCNA, Ki67, and cyclin-D1. CONCLUSION: SeNPs inhibit various inflammation and proliferation mediated pathways and could be an ideal candidate for psoriasis therapy. MATERIALS AND METHODS: SeNPs were characterized and various techniques were used to determine apoptosis and other molecular mechanisms. In vivo studies were performed by inducing psoriasis with imiquimod (IMQ). SeNPs were administered via topical route.


Inflammation/drug therapy , Nanoparticles/chemistry , Psoriasis/drug therapy , Selenium/chemistry , Selenium/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints , Cell Line , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Imiquimod , Keratinocytes , Mice , Mice, Inbred BALB C , Nanoparticles/therapeutic use , Oxidation-Reduction , Phosphorylation , Reactive Oxygen Species , STAT3 Transcription Factor/metabolism
4.
Int J Pharm ; 601: 120536, 2021 May 15.
Article En | MEDLINE | ID: mdl-33781885

Acute respiratory distress syndrome (ARDS) is a life threatening respiratory disease associated with pulmonary edema, alveolar dysfunction, hypoxia, and inflammatory cell accumulation. The most contagious form of COVID-19 associated with ARDS caused by SARS-CoV-2. SARS-CoV-2 majorly produces the cytokine storm and severe lung inflammation and ultimately leads to respiratory failure. ARDS is a complex disease and there is no proper therapeutics for effective therapy. Still, there is a huge scope to identify novel targets to combat respiratory illness. In the current study, we have identified the epigenetic regulating protein BRD4 and developed siRNA based nanomedicine to treat the ARDS. The liposomes were prepared by thin-film hydration method, where BRD4 siRNA complexed with cationic lipid and exhibited 96.24 ± 18.01 nm size and stable even in the presence of RNase. BRD4 siRNA lipoplexes (BRD4-siRNA-LP) inhibited inflammatory cells in lungs and suppressed the lipopolysaccharide (LPS) induced the neutrophil infiltration and mast cell accumulation. Also, BRD4 siRNA based nanomedicine significantly reduced the LPS induced cytokine storm followed by inflammatory signaling pathways. Interestingly, BRD4-siRNA-LP suppressed the LPS-induced p65 and STAT3 nuclear translocation and ameliorated the lung inflammation. Thus, BRD4-siRNA-LP could be a plausible therapeutic option for treating ARDS and might be useful for combating the COVID-19 associated respiratory illness.


COVID-19 , Respiratory Distress Syndrome , Cell Cycle Proteins , Humans , Lipopolysaccharides , Nuclear Proteins , Respiratory Distress Syndrome/drug therapy , SARS-CoV-2 , Transcription Factors/genetics
5.
Nanomedicine ; 33: 102351, 2021 04.
Article En | MEDLINE | ID: mdl-33418136

Acute respiratory distress syndrome (ARDS) is a deadly respiratory illness associated with refractory hypoxemia and pulmonary edema. The recent pandemic outbreak of COVID-19 is associated with severe pneumonia and inflammatory cytokine storm in the lungs. The anti-inflammatory phytomedicine nimbolide (NIM) may not be feasible for clinical translation due to poor pharmacokinetic properties and lack of suitable delivery systems. To overcome these barriers, we have developed nimbolide liposomes conjugated with iRGD peptide (iRGD-NIMLip) for targeting lung inflammation. It was observed that iRGD-NIMLip treatment significantly inhibited oxidative stress and cytokine storm compared to nimbolide free-drug (f-NIM), nimbolide liposomes (NIMLip), and exhibited superior activity compared to dexamethasone (DEX). iRGD-NIMLip abrogated the LPS induced p65 NF-κB, Akt, MAPK, Integrin ß3 and ß5, STAT3, and DNMT1 expression. Collectively, our results demonstrate that iRGD-NIMLip could be a promising novel drug delivery system to target severe pathological consequences observed in ARDS and COVID-19 associated cytokine storm.


Anti-Inflammatory Agents/administration & dosage , Limonins/administration & dosage , Liposomes/chemistry , Oligopeptides/chemistry , Respiratory Distress Syndrome/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Cell Line , Drug Delivery Systems , Endotoxins , Humans , Limonins/chemistry , Limonins/therapeutic use , Lung/drug effects , Lung/pathology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/pathology
6.
Bioorg Chem ; 101: 103983, 2020 08.
Article En | MEDLINE | ID: mdl-32683136

A series of new ß-carboline linked aryl sulfonyl piperazine congeners have been synthesized by coupling various ß-carboline acids with substituted aryl sulfonyl piperazines. Evaluation of their anticancer activity against a panel of human cancer cell lines such as colon (HT-29), breast (MDA-MB-231), bone osteosarcoma (MG-63), brain (U87 MG), prostate (PC- 3) and normal monkey kidney (Vero) cell line has been done. Among the series, compound 8ec and 8ed has shown most potent cytotoxicity with an IC50 values of 2.80 ± 0.10 µM and 0.59 ± 0.28 µM respectively against MG-63 cell line and also potent on other cell lines tested. Compounds 8ec and 8ed was found to inhibit Topo II that is confirmed by specific Topo II inhibition assay. DNA binding studies, cell cycle analysis, Annexin V study indicate that these compounds has potential anticancer activity. Molecular docking studies for compound 8ec and 8ed are incorporated to understand the nature of interaction with topoisomerase IIα and dsDNA.


Carbolines/chemistry , Carbolines/chemical synthesis , Molecular Docking Simulation/methods , Topoisomerase II Inhibitors/therapeutic use , Apoptosis , Humans , Molecular Structure , Structure-Activity Relationship , Topoisomerase II Inhibitors/pharmacology
7.
Life Sci ; 254: 117765, 2020 Aug 01.
Article En | MEDLINE | ID: mdl-32437797

The ongoing wreaking global outbreak of the novel human beta coronavirus (CoV) pathogen was presumed to be from a seafood wholesale market in Wuhan, China, belongs to the Coronaviridae family in the Nidovirales order. The virus is highly contagious with potential human-human transmission which was named as the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has spread across six continents and emerged as a global pandemic in short span with alarming levels of spread and severity. This virus associated symptoms and infectious respiratory illness is designated as coronavirus disease 19 (COVID-19). The SARS-CoV-2 possesses enveloped club-like spike protein projections with positive-sense large RNA genome and has a unique replication strategy. This virus was believed to have zoonotic origin with genetical identity to bat and pangolin CoV. In the current review, we introduce a general overview about the human CoVs and the associated diseases, the origin, structure, replication and key clinical events that occur in the COVID-19 pathogenicity. Furthermore, we focused on possible therapeutic options such as repurposing drugs including antimalarials, antivirals, antiparasitic drugs, and anti-HIV drugs, as well as monoclonal antibodies, vaccines as potential treatment options. Also we have summarized the latest research progress on the usage of stem cell therapy, human convalescent serum, interferon's, in the treatment of COVID-19.


Coronavirus Infections/epidemiology , Coronavirus Infections/therapy , Pneumonia, Viral/epidemiology , Pneumonia, Viral/therapy , Animals , Betacoronavirus/isolation & purification , COVID-19 , Coronavirus Infections/virology , Pandemics/prevention & control , Pneumonia, Viral/virology , SARS-CoV-2 , Zoonoses/epidemiology , Zoonoses/therapy , Zoonoses/virology
8.
Cell Death Dis ; 11(1): 21, 2020 01 10.
Article En | MEDLINE | ID: mdl-31924750

Psoriasis is an autoimmune skin disease, where chronic immune responses due to exaggerated cytokine signaling, abnormal differentiation, and evasion of keratinocytes apoptosis plays a crucial role in mediating abnormal keratinocytes hyperproliferation. From the therapeutic perspective, the molecules with strong anti-proliferative and anti-inflammatory properties could have tremendous relevance. In this study, we demonstrated that piperlongumine (PPL) treatment effectively abrogated the hyperproliferation and differentiation of keratinocytes by inducing ROS-mediated late apoptosis with loss of mitochondrial membrane potential. Besides, the arrest of cell cycle was found at Sub-G1 phase as a result of DNA fragmentation. Molecularly, inhibition of STAT3 and Akt signaling was observed with a decrease in proliferative markers such as PCNA, ki67, and Cyclin D1 along with anti-apoptotic Bcl-2 protein expression. Keratin 17 is a critical regulator of keratinocyte differentiation, and it was found to be downregulated with PPL significantly. Furthermore, prominent anti-inflammatory effects were observed by inhibition of lipopolysaccharide (LPS)/Imiquimod (IMQ)-induced p65 NF-κB signaling cascade and strongly inhibited the production of cytokine storm involved in psoriasis-like skin inflammation, thus led to the restoration of normal epidermal architecture with reduction of epidermal hyperplasia and splenomegaly. In addition, PPL epigenetically inhibited histone-modifying enzymes, which include histone deacetylases (HDACs) of class I (HDAC1-4) and class II (HDAC6) evaluated by immunoblotting and HDAC enzyme assay kit. In addition, our results show that PPL effectively inhibits the nuclear translocation of p65 and a histone modulator HDAC3, thus sequestered in the cytoplasm of macrophages. Furthermore, PPL effectively enhanced the protein-protein interactions of HDAC3 and p65 with IκBα, which was disrupted by LPS stimulation and were evaluated by Co-IP and molecular modeling. Collectively, our findings indicate that piperlongumine may serve as an anti-proliferative and anti-inflammatory agent and could serve as a potential therapeutic option in treating psoriasis.


Dioxolanes/therapeutic use , Epigenesis, Genetic , Inflammation/drug therapy , Psoriasis/drug therapy , Psoriasis/genetics , Skin/pathology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Chemokines/metabolism , Dioxolanes/pharmacology , Epidermis/pathology , Epigenesis, Genetic/drug effects , HaCaT Cells , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Imiquimod/adverse effects , Inflammation/genetics , Keratin-17/metabolism , Keratinocytes/drug effects , Keratinocytes/pathology , Lipopolysaccharides , Mice , Mice, Inbred BALB C , Models, Biological , Phosphorylation/drug effects , Protein Binding/drug effects , Psoriasis/chemically induced , Psoriasis/pathology , RAW 264.7 Cells , STAT3 Transcription Factor/metabolism
9.
RSC Med Chem ; 11(11): 1295-1302, 2020 Nov 18.
Article En | MEDLINE | ID: mdl-34095841

A library of substituted (1-(benzyl)-1H-1,2,3-triazol-4-yl)(piperazin-1-yl)methanone derivatives were designed, synthesized and screened for their in vitro cytotoxic activity against BT-474, HeLa, MCF-7, NCI-H460 and HaCaT cells by employing 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Among all the synthesized analogues, compound 10ec displayed the highest cytotoxicity with the IC50 value of 0.99 ± 0.01 µM towards BT-474 cancer cell line. The target compound (10ec) was also evaluated for its tubulin polymerization inhibition study. Detailed biological studies such as acridine orange/ethidium bromide (AO/EB), DAPI and annexin V-FITC/propidium iodide staining assay suggested that compound 10ec induced the apoptosis of BT-474 cells. The clonogenic assay revealed that the inhibition of colony formation in BT-474 cells by 10ec in concentration-dependent manner. Moreover, the flow cytometric analysis revealed that 10ec induced apoptosis via cell cycle arrest at the sub-G1 and G2/M phase. In silico studies of sulfonyl piperazine-integrated triazole conjugates unveil that they possess drug-like properties. According to the molecular modelling studies, compound 10ec binds to the colchicine binding site of the tubulin.

10.
J Cell Physiol ; 235(6): 5270-5283, 2020 06.
Article En | MEDLINE | ID: mdl-31846070

Drug discovery is an onerous, extremely expensive, and time-consuming process. Instead, drug repurposing is an attractive strategy for exploiting novel indications for a drug beyond its original use. The untapped potential of drug repurposing compensates the barriers associated with the drug discovery pipeline. Psoriasis is an autoimmune skin disease, where hyperproliferation of keratinocytes and exaggerated immune responses are the important hallmarks of the disease. Extensive in vitro and preclinical research has demonstrated that niclosamide was found to exert potent anticancer and anti-inflammatory properties by targeting STAT3, p65 NF-κB, and NFATc-1 signaling paradigm with minimal host toxicity. From the disease perspective, the static intracellular molecular network in both cancer and psoriasis share overlapping pathological features in terms of hyperproliferation and chronic inflammation, which is mediated by the aforementioned signaling cascade. The plausible mechanistic relevance has prompted us to investigate the implementation of niclosamide for repositioning in psoriasis. Our in vitro and in vivo findings suggest that niclosamide inhibits keratinocytes hyperproliferation by reactive oxygen species-mediated apoptosis through the loss of mitochondrial membrane potential, cell cycle arrest at Sub G1 phase, and DNA fragmentation. Furthermore, niclosamide treatment resulted in abrogation of lipopolysaccharide-induced inflammatory cytokine levels in murine macrophages. Additionally, our results provided a preclinical rationale in imiquimod (IMQ)-induced BALB/c mouse model, where niclosamide diligently mitigated the IMQ-induced epidermal hyperplasia and inflammation by downregulating STAT3, p65 NF-κB, and NFATc-1 transcription factors along with Akt, Ki-67, and ICAM-1 protein expression.


Inflammation/drug therapy , NFATC Transcription Factors/genetics , STAT3 Transcription Factor/genetics , Skin Diseases/drug therapy , Transcription Factor RelA/genetics , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Drug Repositioning , Humans , Inflammation/chemically induced , Inflammation/pathology , Keratinocytes/drug effects , Lipopolysaccharides/toxicity , Mice , Mice, Inbred BALB C , Niclosamide/pharmacology , Psoriasis/drug therapy , Psoriasis/pathology , Signal Transduction/drug effects , Skin Diseases/pathology
11.
Nanomedicine (Lond) ; 14(15): 1991-2010, 2019 08.
Article En | MEDLINE | ID: mdl-31355710

Aim: Selenium nanoparticles (SeNPs) may have a potential role in treating dermal disorders due to its wide therapeutic properties, but there is a need to evaluate its toxicity in keratinocytes. The present study evaluated the molecular mechanism and mode of cell death induced by SeNPs on dermal keratinocytes. Materials & methods: SeNPs were synthesized, characterized and studied in human keratinocytes cells. Oxidative stress and mitochondrial membrane depolarization were evaluated by various techniques. Additionally, autophagy mediated apoptotic cell death was evaluated. Results: SeNPs induced oxidative stress and apoptotic cell death in keratinocytes by increasing autophagy through the formation of acidic lysosomes and autophagosomes. Conclusion: Overall, SeNPs induce the oxidative stress and autophagy mediated apoptotic cell death in human keratinocytes cells.


Autophagic Cell Death/drug effects , Keratinocytes/drug effects , Nanoparticles , Selenium/pharmacology , Apoptosis/drug effects , Cell Line , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Oxidative Stress/drug effects , Selenium/chemistry
12.
Int Immunopharmacol ; 71: 301-312, 2019 Jun.
Article En | MEDLINE | ID: mdl-30933843

The purpose of this study was to investigate the protective effects of ambroxol in psoriasis-like skin inflammation both in vitro and in vivo and delineate the molecular mechanism of ambroxol. Our data demonstrated that ambroxol has an imperative role in inhibiting the lipopolysaccharide (LPS) stimulated nitrite levels, total cellular and mitochondrial reactive oxygen species level which was determined by Griess assay, DCFDA, and MitoSOX Red staining, respectively. We found that ambroxol remarkably reduced imiquimod (IMQ) induced epidermal hyperplasia, psoriasis area and severity index (PASI) scoring, splenomegaly, skin, and ear fold thickness. In addition, the histopathological evaluation revealed that ambroxol topical and subcutaneous treatment eloquently reduced psoriasiform lesions including acanthosis. Moreover, with ambroxol intervention, the levels of antioxidants glutathione (GSH), superoxide dismutase (SOD), and IL-10 were found to be increased along with a reduction in nitrite levels in skin tissues. On the other hand, ambroxol treatment significantly reduced imiquimod-induced levels of inflammatory cytokines such as IL-1ß, IL-6, IL-17, IL-22, IL-23, TGF-ß, and TNF-α. Furthermore, from immunoblotting, we found a decrease in the protein expression of nitrotyrosine, iNOS, NF-κB and MAPKs signaling cascade with a concomitant increase in the expression of Nrf-2 and SOD-1 in RAW 264.7 cells and skin tissues by ambroxol. Similar findings were observed by immunofluorescence in macrophages. Moreover, ambroxol downregulated the ICAM-1 and Ki67 expression observed in skin tissues. Collectively, our results demonstrate that ambroxol may have intriguing therapeutic possibilities in attenuating psoriasis.


Ambroxol/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Macrophages/immunology , Psoriasis/drug therapy , Skin/pathology , Animals , Cytokines/metabolism , Disease Models, Animal , Humans , Hyperplasia , Imiquimod , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred BALB C , Oxidative Stress/drug effects , RAW 264.7 Cells , Signal Transduction
13.
Carbohydr Polym ; 212: 252-259, 2019 May 15.
Article En | MEDLINE | ID: mdl-30832855

Niclosamide, previously used as an anthelmintic drug is currently being repurposed for its anticancer activity. Niclosamide is a brick like biopharmaceutical classification system (BCS) class II drug with poor aqueous solubility and dissolution consequently leading to low bioavailability. By considering the physicochemical properties and geometry of niclosamide, inclusion complex with cyclodextrin was prepared by freeze drying method and characterized using FT-IR, DSC, PXRD, and 1HNMR. In silico molecular modeling study was performed to study the possible interactions between niclosamide and cyclodextrin. The anticancer activity of niclosamide formulation was evaluated through in vitro cell cytotoxicity study using various cancer cell lines. The potential of niclosamide complex for improvement of the bioavailability was evaluated in male BALB/c mice. In vitro cytotoxicity studies indicated significantly higher cytotoxicity at lower concentrations and the pharmacokinetic studies showed significant improvement in Cmax and Tmax of niclosamide from cyclodextrin complex in comparison to pure niclosamide alone.


Antineoplastic Agents/chemical synthesis , Cyclodextrins/chemical synthesis , Drug Compounding/methods , Drug Repositioning/methods , Niclosamide/chemical synthesis , Animals , Anticestodal Agents/chemical synthesis , Anticestodal Agents/metabolism , Antineoplastic Agents/metabolism , Cyclodextrins/metabolism , Drug Evaluation, Preclinical/methods , HCT116 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Niclosamide/metabolism
14.
Cell Death Dis ; 10(2): 81, 2019 01 28.
Article En | MEDLINE | ID: mdl-30692512

Acute respiratory distress syndrome (ARDS) is characterized by an excessive acute inflammatory response in lung parenchyma, which ultimately leads to refractory hypoxemia. One of the earliest abnormalities seen in lung injury is the elevated levels of inflammatory cytokines, among them, the soluble tumor necrosis factor (TNF-α) has a key role, which exerts cytotoxicity in epithelial and endothelial cells thus exacerbates edema. The bacterial lipopolysaccharide (LPS) was used both in vitro (RAW 264.7, THP-1, MLE-12, A549, and BEAS-2B) and in vivo (C57BL/6 mice), as it activates a plethora of overlapping inflammatory signaling pathways involved in ARDS. Nimbolide is a chemical constituent of Azadirachta indica, which contains multiple biological properties, while its role in ARDS is elusive. Herein, we have investigated the protective effects of nimbolide in abrogating the complications associated with ARDS. We showed that nimbolide markedly suppressed the nitrosative-oxidative stress, inflammatory cytokines, and chemokines expression by suppressing iNOS, myeloperoxidase, and nitrotyrosine expression. Moreover, nimbolide mitigated the migration of neutrophils and mast cells whilst normalizing the LPS-induced hypothermia. Also, nimbolide modulated the expression of epigenetic regulators with multiple HDAC inhibitory activity by suppressing the nuclear translocation of NF-κB and HDAC-3. We extended our studies using molecular docking studies, which demonstrated a strong interaction between nimbolide and TNF-α. Additionally, we showed that treatment with nimbolide increased GSH, Nrf-2, SOD-1, and HO-1 protein expression; concomitantly abrogated the LPS-triggered TNF-α, p38 MAPK, mTOR, and GSK-3ß protein expression. Collectively, these results indicate that TNF-α-regulated NF-κB and HDAC-3 crosstalk was ameliorated by nimbolide with promising anti-nitrosative, antioxidant, and anti-inflammatory properties in LPS-induced ARDS.


Azadirachta/chemistry , Limonins/therapeutic use , Molecular Docking Simulation/methods , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Disease Models, Animal , Humans , Limonins/pharmacology , Male , Mice , NF-kappa B/metabolism , Respiratory Distress Syndrome/pathology , Translocation, Genetic
15.
Bioorg Med Chem Lett ; 28(22): 3564-3573, 2018 12 01.
Article En | MEDLINE | ID: mdl-30318440

A facile method for the construction of double bond between 3-ylidene oxindoles and α-azido ketones has been successfully accomplished with a mild base. This method features azido reduction with concomitant double bond formation to provide the new class of bioactive enamino-2-oxindoles. These new compounds were screened for their in vitro cytotoxic potential on selected human cancer cell lines such as colon, lung, breast, and cervical cancer cells. Among them, representative compounds 3a, 3h, 3k, 3p, 3w and 3x showed notable cytotoxicity profile with IC50 values ranging from 1.40 ±â€¯0.10 to 28.7 ±â€¯0.36 µM. Compound 3k displayed most potent cytotoxicity against lung cancer (NCI-H460) cells with an IC50 value of 1.40 ±â€¯0.10 µM. 3k also arrested the G2/M phase of the cell cycle and induced distinctive apoptotic features on lung cancer cells. The apoptosis induction is supported by various cellular assays such as AO/EB, DAPI, and DCFDA staining studies including clonogenic assay. Extent of apoptosis was also analyzed by Annexin binding and JC-1 staining. Moreover, this method is amenable for the generation of a library of new class of stable bioactive enamino-2-oxindoles.


Antineoplastic Agents/chemical synthesis , Apoptosis , Ketones/chemistry , Oxindoles/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Membrane Potential, Mitochondrial/drug effects , Microscopy, Fluorescence , Molecular Conformation , Oxindoles/pharmacology , Reactive Oxygen Species/metabolism
16.
ChemMedChem ; 13(18): 1909-1922, 2018 09 19.
Article En | MEDLINE | ID: mdl-30010248

A new series of (E)-3-[(1-aryl-9H-pyrido[3,4-b]indol-3-yl)methylene]indolin-2-one hybrids were synthesized and evaluated for their in vitro cytotoxic activity against a panel of selected human cancer cell lines, namely, HCT-15, HCT-116, A549, NCI-H460, and MCF-7, including HFL. Among the tested compounds, (E)-1-benzyl-5-bromo-3-{[1-(2,5-dimethoxyphenyl)-9H-pyrido[3,4-b]indol-3-yl]methylene}indolin-2-one (10 s) showed potent cytotoxicity against HCT-15 cancer cells with an IC50 value of 1.43±0.26 µm and a GI50 value of 0.89±0.06 µm. Notably, induction of apoptosis by 10 s on the HCT-15 cell line was characterized by using different staining techniques, such as acridine orange/ethidium bromide (AO/EB) and DAPI. Further, to understand the mechanism of anticancer effects, various assays such as annexin V-FITC/PI, DCFDA, and JC-1were performed. The flow cytometric analysis revealed that compound 10 s arrests the HCT-15 cancer cells at the G0/G1 phase of the cell cycle. Additionally, western blot analysis indicated that treatment of 10 s on HCT-15 cancer cells led to decreased expression of anti-apoptotic Bcl-2 and increased protein expression of both pro-apoptotic Bax and caspase-3, -8, and -9, and cleaved PARP with reference to actin. Next, a clonogenic assay revealed the inhibition of colony formation in HCT-15 cancer cells by 10 s in a dose-dependent manner. Moreover, upon testing on normal human lung cells (HFL), the compounds were observed to be safer with a low toxicity profile. In addition, viscosity and molecular-docking studies showed that compound 10 s has typical intercalation with DNA.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carbolines/pharmacology , DNA/pharmacology , Drug Design , Oxindoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carbolines/chemistry , Cell Proliferation/drug effects , Cells, Cultured , DNA/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Membrane Potential, Mitochondrial/drug effects , Molecular Structure , Oxindoles/chemistry , Structure-Activity Relationship
18.
Bioorg Med Chem ; 26(8): 1996-2008, 2018 05 01.
Article En | MEDLINE | ID: mdl-29525336

A facile one-pot method for the synthesis of new phenanthrene fused-dihydrodibenzo-quinolinone derivatives has been successfully accomplished by employing sulfamic acid as catalyst. These new compounds were evaluated for their in vitro cytotoxic potential against human lung (A549), prostate (PC-3 and DU145), breast (MCF-7) and colon (HT-29 and HCT-116) cancer cell lines. Among all the tested compounds, one of the derivatives 8p showed good anti-proliferative activity against A549 lung cancer cell line with an IC50 of 3.17 ±â€¯0.52 µM. Flow cytometric analyses revealed that compound 8p arrested both Sub G1 and G2/M phases of cell cycle in a dose dependent manner. The compound 8p also displayed significant inhibition of tubulin polymerization and disruption of microtubule network (IC50 of 5.15 ±â€¯0.15 µM). Molecular docking studies revealed that compound 8p efficiently interacted with critical amino acid Cys241 of the α/ß-tubulin by a hydrogen bond (SH…O = 2.4 Å). Further, the effect of 8p on cell viability was also studied by AO/EB, DCFDA and DAPI staining. The apoptotic characteristic features revealed that 8p inhibited cell proliferation effectively through apoptosis by inducing the ROS generation. Analysis of mitochondrial membrane potential through JC-1 staining and annexin V binding assay indicated the extent of apoptosis in A549 cancer cells.


Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Quinolones/chemistry , Sulfonic Acids/chemistry , Tubulin Modulators/chemical synthesis , Tubulin/metabolism , A549 Cells , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Cell Cycle Checkpoints/drug effects , Drug Screening Assays, Antitumor , Humans , Membrane Potential, Mitochondrial/drug effects , Molecular Docking Simulation , Phenanthrenes/chemistry , Protein Structure, Tertiary , Quinolones/metabolism , Quinolones/pharmacology , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Tubulin/chemistry , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacology
19.
Int J Pharm ; 528(1-2): 202-214, 2017 Aug 07.
Article En | MEDLINE | ID: mdl-28596138

Recent reports on the anticancer potential of niclosamide have opened new avenues for anticancer treatment. Niclosamide belongs to the BCS class II, which is indicative of poor solubility and dissolution rate limited absorption. The aim of this study was to improve the dissolution rate of the drug by mesoporous drug delivery system. Porous silica grades (ordered and nonordered) with different pore size, pore volume and surface area were used in the study. The drug was loaded on silica carriers by the solvent evaporation method and characterized by BET surface area analysis, SEM, P-XRD, DSC, and FTIR. A discriminatory dissolution medium was developed for performing the in vitro dissolution of niclosamide. In comparison to the plain drug, all silica based formulations showed improvement in the dissolution rate. Maximum enhancement in the dissolution rate was observed in 1:2 drug:carrier loading ratio when compared to 1:1 ratio. Different properties of mesoporous silica like structural geometry, pore size and microenvironment pH demonstrated a significant impact on drug release from the formulations. Cytotoxicity of the optimized mesoporous formulations of niclosamide was explored in HCT-116, HCT-15, NCI, MDA-MB-231 and A549 cancer cell lines. Nearly 3 fold and 2 fold increase in% cytotoxicity of drug loaded Syloid-244 and Sylysia 350 at 1:2 ratio respectively, were observed when compared to the plain drug.


Drug Carriers/chemistry , Niclosamide/administration & dosage , Silicon Dioxide/chemistry , Cell Line, Tumor , Humans , Porosity , Solubility
...