Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
1.
Phytomedicine ; 119: 154981, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37531902

ABSTRACT

PURPOSE: The declined oxygen tension in the cancer cell leads to the hypoxic adaptive response and favors establishment of tumor micro environment [TEM]. The complex TME consists of interwoven hypoxic HIF-1α and DNA damage repair ATM signaling. The ATM/HIF-1α phosphorylation switch on angiogenesis and abort apoptosis. Targeting this signaling nexus would be a novel therapeutic strategy for the treatment of cancer. BACKGROUND: Steroidal alkaloid solanidine is known for varied pharmacological role but with less molecular evidences. Our earlier findings on solanidine proven its anti-neoplastic activity by inducing apoptosis in lung cancer. In continued research, efforts have been made to establish the underlying molecular signaling in induction of DNA damage in prevailing hypoxic TME. METHODS: The solanidine induced DNA damage was assessed trough alkali COMET assay; signaling nexus and gene expression profile analysis through IB, qRT-PCR, Gelatin Zymography, IHC, IF and ELISA. Pathophysiological modulations assessed through tube formation, migration, invasion assays. Anti-angiogenic studies through CAM, rat aorta, matrigel assays and corneal neovascularization assay. Anti-tumor activity through in-vivo DLA ascites tumor model and LLC model. RESULTS: The results postulates, inhibition of hypoxia driven DDR proteins pATMser1981/pHIF-1αser696 by solanidine induces anti-angiogenesis. Systematic study of both non-tumorigenic and tumorigenic models in-vitro as well as in-vivo experimental system revealed the angio-regression mediated anticancer effect in lung cancer. These effects are due to the impeded expression of angiogenic mediators such as VEGF, MMP2&9 and inflammatory cytokines IL6 and TNFα to induce pathophysiological changes CONCLUSION: The study establishes new role of solanidine by targeting ATM/HIF-1α signaling to induce anti-angiogenesis for the first time. The study highlights the potentiality of plant based phytomedicine solanidine which can targets the multiple hallmarks of cancer by targeting interwoven signaling crosstalk. Such an approach through solanidine necessary to counteract heterogeneous complexity of cancer which could be nearly translated into drug.


Subject(s)
Adenocarcinoma of Lung , Alkaloids , Antineoplastic Agents , Lung Neoplasms , Rats , Animals , Phosphorylation , Antineoplastic Agents/therapeutic use , Hypoxia/drug therapy , Alkaloids/pharmacology , Adenocarcinoma of Lung/drug therapy , Lung Neoplasms/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit , Neovascularization, Pathologic/drug therapy , Cell Line, Tumor , Tumor Microenvironment
2.
Chem Biodivers ; 20(5): e202201152, 2023 May.
Article in English | MEDLINE | ID: mdl-36974341

ABSTRACT

The design, molecular docking, synthesis and structure-activity relationship (SAR) of a series of novel methyl 1-oxo-2-(propan-2-yl)-3-(pyridin-3-yl)-1,2,3,4-tetrahydroisoquinoline-4-carboxylates, were investigated for antiproliferative and cytotoxic studies by screening against cancer cell lines of different origin by MTT, LDH and Trypan Blue Assay. Irrespective of cell lines, among the synthesized nonpeptido-mimetic analogs 5a-e, 5c has executed potent bio-potency with IC50 value of 7.00 to 7.21 µM, which further expressed in-vivo anti-tumor activity against murine T-cell lymphoma cell lines (Daltons Lymphoma-DLA) by regressing tumor growth. The formation of neovessels from the vasculogenesis was diminished reflecting the antitumor activity. The neovessel formation is directly relied on expression of matrix meteloproteases (MMP's) level which was drastically reduced by 5c treatment as evaluated by immonoblot assays. This is further supported by in-silico ADMET studies performed by ACD I-Lab 2.0 were in agreement with Lipinski rule of five. Reporting results were assessed as a positive parameter for further validation of the compound for therapeutic potential of cancer by 5c for preclinical studies in near future.


Subject(s)
Antineoplastic Agents , Tetrahydroisoquinolines , Animals , Mice , Molecular Structure , Molecular Docking Simulation , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Tetrahydroisoquinolines/pharmacology , Metalloproteases/pharmacology , Drug Design , Protein Kinase Inhibitors/pharmacology
3.
Pharmaceuticals (Basel) ; 16(1)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36678622

ABSTRACT

The emerging interest in the field of coordination chemistry and their biological applications has created a novel impact in the field of chemical biology. With this motivation, in this work we have synthesized a novel benzimidazole derived imine ligand, 2-((E)-((1H-benzo[d]-2-yl)methylimino)methyl)-4-fluorophenol (HBMF) and its Co(III) and Cu(II) complexes. The metal complexes (C1-C4) were synthesized in 2:1 (HBMF: metal ion) and 1:1:1 (HBMF: metal ion: 1,10-phen) ratios. Structural elucidations of all the synthesized compounds were performed using FT-IR, UV-Visible, NMR, Mass spectroscopy and elemental analysis techniques. A combination of first principles calculations and molecular dynamics simulations was applied to computationally investigate the structural, reactive, and spectroscopic properties of the newly synthesized HBMF ligand and its complexes with copper and cobalt metal ions. Quantum-mechanical calculations in this study were based on the density functional theory (DFT), while molecular dynamics (MD) simulations were based on the OPLS4 force field. The DFT calculations were used to obtain the reactive and spectroscopic properties of the ligand and its complexes, while molecular dynamics (MD) simulations were used to address the ligand's reactivity with water. Further, the in vitro anti-proliferative activity of the compounds was tested against the A549, Ehrlich-Lettre ascites carcinoma (EAC), SIHA and NIH3T3 cell lines. The biological results depicted that the compound C4, with molecular formula C27H23Cl2CoFN5O3 exhibited profound anti-proliferative activity against the EAC cell line with a significant IC50 value of 10 µm when compared to its parent ligand and other remaining metal complexes under study. Various assays of hematological parameters (alkaline phosphate, creatinine, urea, RBC and WBC) were performed, and significant results were obtained from the experiments. Furthermore, the effect of C4 on neovascularization was evaluated by stimulating the angiogenesis with rVEGF165, which was compared with non-tumor models. The EAC cells were cultured in vivo and administrated with 50 and 75 mg/kg of two doses and tumor parameters were evaluated.

4.
3 Biotech ; 12(11): 306, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36276461

ABSTRACT

Curcumin (diferuloylmethane) is bioactive phenolic compound which exerts diverse antimetastatic effect. Several studies have reported the antimetastatic effect of curcumin by its ability to modulate the epithelial-to-mesenchymal transition (EMT) process in different cancers, but underlying molecular mechanism is poorly understood. EMT is a highly conserved biological process in which epithelial cells acquire mesenchymal-like characteristics by losing their cell-cell junctions and polarity. As a consequence, deviation in cellular mechanism leads to cancer metastasis and thereby death. In this perspective, we explored the antimetastatic potential and mechanism of curcumin on the EMT process by establishing in vitro EMT model in lungs cancer (A549) cells induced by TGF-ß1. Our results showed that curcumin mitigates EMT by regulating the expression of crucial mesenchymal markers such as MMP2, vimentin and N-cadherin. Besides, the transcriptional analysis revealed that the curcumin treatment differentially regulated the expression of 75 genes in NanoString nCounter platform. Further protein-protein interaction network and clusters analysis of differentially expressed genes revealed their involvement in essential biological processes that plays a key role during EMT transition. Altogether, the study provides a comprehensive overview of the antimetastatic potential of curcumin in TGF-ß1-induced EMT in lung cancer cells. Supplementary Information: The online version contains supplementary material available at 10.1007/s13205-022-03360-7.

5.
Preprint in English | bioRxiv | ID: ppbiorxiv-485024

ABSTRACT

Monoclonal antibodies (mAbs) that are specific to SARS-CoV-2 can be useful in diagnosing, preventing, and treating the coronavirus (COVID-19) illness. Strategies for the high-throughput and rapid isolation of these potent neutralizing antibodies are critical toward the development of therapeutically targeting COVID-19 as well as other infectious diseases. In the present study, a single B-cell cloning method was used to screen SARS-CoV-2 receptor-binding domain (RBD) specific, high affinity, and neutralizing mAbs from patients blood samples. An RBD-specific antibody, SAR03, was discovered that showed high binding (ELISA and SPR) and neutralizing activity (competitive ELISA and pseudovirus-based reporter assay) against Sars-CoV-2. Mechanistic studies on human cells revealed that SAR03 competes with the ACE-2 receptor for binding with the RBD domain (S1 subunit) present in the spike protein of Sars-CoV-2. This study highlights the potential of the single B cell cloning method for the rapid and efficient screening of high-affinity and effective neutralizing antibodies for Sars-CoV-2 and other emerging infectious diseases. HighlightsO_LISingle B-cell cloning is a high-throughput and efficient method of generating high affinity neutralizing antibodies C_LIO_LISingle B-cell cloning method was used to screen SARS-CoV-2 receptor-binding domain (RBD) specific, high affinity, and neutralizing monoclonal antibodies from patients blood samples. C_LIO_LIAn RBD-specific antibody, SAR03, was discovered that showed high binding and neutralizing activity against SARS-CoV-2. C_LI

6.
Pharmacol Rep ; 74(2): 353-365, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35001321

ABSTRACT

BACKGROUND: Anomalous activation of intra-cellular signalling cascades confers neoplastic properties on malignant cells. The JAK2/STAT3 proteins play a pivotal role in the pathogenesis of most of the solid malignancies. The over expression of STAT3 in these tumours results in an evasion of apoptosis and thereby pathogenesis. Hence, strategy to target STAT3 to regress tumour development is an emerging new concept. As an approach, anti-neoplastic drug, Azo-hydrozone analogue, BT-1F with potential anti-proliferative effect was evaluated to demonstrate its capacity to counteract STAT3 signal with mechanistic approach. METHODS: Cell based screening for cytotoxicity was performed through MTT, LDH and Trypan blue. The BT-1F induced anti-clonogenic property by clonogenic assay. The apoptotic capacity was examined by crystal violet staining, flow cytometry, Annexin-FITC, DAPI and TUNEL assay. The altered signalling events were studied using immunoblot. The drug-induced anti-tumour effect was evaluated in an in-vivo solid tumour model and molecular interaction was further validated by in-silico studies. RESULTS: The BT-1F exerts chemo-sensitivity specifically against EAC and A549 cells without altering its normal counterpart. The anti-proliferative/anti-clonogenic effect was due to the induction of apoptosis through inhibition of STAT3Tyr705 signal. Eventually downstream signalling proteins p53, Bax, Bad and Bcl-xL were significantly altered. Further in-vivo experimental results validated  in-vitro findings. The computational approaches assures the BT-1F efficiency in binding with STAT3. CONCLUSION: Systemic validation of STAT3 target drug, BT-1F in in-vitro, in-silico and in-vivo models has promising strategy for solid cancer treatment.


Subject(s)
Hydrazones , STAT3 Transcription Factor , Apoptosis , Cell Line, Tumor , Cell Proliferation , Hydrazones/pharmacology , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism
7.
Luminescence ; 37(3): 500-513, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35048496

ABSTRACT

The present communication is strongly focused on the investigation of synthesis, structural and luminescence properties of cerium (Ce3+ )- and europium (Eu3+ )-activated Zn4 Al22 O37 phosphors. Ce3+ - and Eu3+ -doped Zn4 Al22 O37 novel phosphors were prepared using a solution combustion synthesis route. Structural properties were studied using powder X-ray diffraction and high-resolution transverse electron microscopy. The optical properties were studied using ultraviolet-visible light spectroscopy and Fourier transform infrared spectroscopy; luminescence properties were studied using a photoluminescence (PL) technique. The crystal structure of the prepared Zn4 Al22 O37 host and Ce3+ - and Eu3+ -activated Zn4 Al22 O37 phosphors was investigated and was found to have a hexagonal structure. The measured PL emission spectrum of the Ce3+ -doped Zn4 Al22 O37 phosphor showed an intense and broad emission band centred at 421 nm under a 298 nm excitation wavelength. By contrast, the Eu3+ -doped Zn4 Al22 O37 phosphor exhibited two strong and intense emission bands at approximately 594 nm (orange) and 614 nm (red), which were monitored under 395 nm excitation. The Commission Internationale de l'Eclairage (CIE) colour coordinates of the Ce3+ -doped Zn4 Al22 O37 were investigated and found to be x = 0.1567, y = 0.0637 (blue) at 421 nm and for Eu3+ -doped Zn4 Al22 O37 were x = 0.6018, y = 0.3976 (orange) at 594 nm and x = 0.6779, y = 0.3219 (red) at 614 nm emission. The luminescence behaviour of the synthesized phosphors suggested that these phosphors may be used in lighting applications.


Subject(s)
Europium , Luminescence , Europium/chemistry , Ions , X-Ray Diffraction , Zinc
8.
Apoptosis ; 27(1-2): 49-69, 2022 02.
Article in English | MEDLINE | ID: mdl-34837562

ABSTRACT

Reigning of the abnormal gene activation associated with survival signalling in lung cancer leads to the anomalous growth and therapeutic failure. Targeting specific cell survival signalling like JAK2/STAT3 nexus has become a major focus of investigation to establish a target specific treatment. The 2-bromobenzoyl-4-methylphenoxy-acetyl hydra acetyl Coumarin (BP-1C), is new anti-neoplastic agent with apoptosis inducing capacity. The current study was aimed to develop antitumor phramacophore, BP-1C as JAK2 specific inhibitor against lung neoplastic progression. The study validates and identifies the molecular targets of BP-1C induced cell death. Cell based screening against multiple cancer cell lines identified, lung adenocarcinoma as its specific target through promotion of apoptosis. The BP-1C is able to induce, specific hall marks of apoptosis and there by conferring anti-neoplastic activity. Validation of its molecular mechanism, identified, BP-1C specifically targets JAK2Tyr1007/1008 phosphorylation, and inhibits its downstream STAT3Tyr705 signalling pathway to induce cell death. As a consequence, modulation in Akt/Src survival signal and altered expression of interwoven apoptotic genes were evident. The results were reproducible in an in-vivo LLC tumor model and in-ovo xenograft studies. The computational approaches viz, drug finger printing confers, BP-1C as novel class JAK2 inhibitor and molecular simulations studies assures its efficiency in binding with JAK2. Overall, BP-1C is a novel JAK2 inhibitor with experimental evidence and could be effectively developed into a promising drug for lung cancer treatment.


Subject(s)
Apoptosis , Lung Neoplasms , Benzophenones/pharmacology , Cell Line, Tumor , Cell Proliferation , Coumarins/pharmacology , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , STAT3 Transcription Factor/metabolism
9.
Pharmacol Rep ; 73(5): 1344-1360, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34109572

ABSTRACT

BACKGROUND: Imbalance and instability in the structure of the DNA have become major characteristics of cancer. In response to DNA damage, DNA damage response (DDR) protein, ataxia telangiectasia mutated (ATM), plays a pivotal role in the modulation of regulatory regions responsible for inhibition of apoptosis, thereby neoplastic progression. METHODS: A new series of DPA (7a-t) were synthesized, characterized. Anti-proliferative studies to identify the lead compound were carried out by LDH and MTT assay. Apoptosis/DNA damage was measured through FACS, Annexin-v staining, TUNEL and Comet assay. Elucidation of molecular mechanism through immunoblot and further validation of the drug effect through in vivo approaches. RESULTS: Initial in vitro anti-proliferative screening of Compounds DPA (7a-t) against multiple cancer cell lines identified Compound DPA (7n) as a potent cytotoxic molecule with IC50 value of 4.3 µM. Down the line, in vitro and in vivo evaluation of Compound DPA (7n) inferred that it has apoptotic inducing potentiality. Further, evaluation of molecular mechanism inferred that Compound DPA (7n) effectively modulates ATM phosphorylation only, eventually altering downstream signalling pathways. CONCLUSIONS: Compound DPA (7n) emerged as a potent proapoptotic and anti-neoplastic agent by inhibiting ATM kinase activity both in vitro and in vivo. The conferring results ascertain that the drug could be developed as a new ATM kinase inhibitor with anti-cancer capacity.


Subject(s)
Acetamides/chemical synthesis , Acetamides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , DNA Damage , Drug Delivery Systems , Animals , Apoptosis , Cell Line, Tumor , DNA Repair , Humans , Mice , Mice, Inbred BALB C , Molecular Structure , Neoplasms, Experimental/drug therapy , Xenograft Model Antitumor Assays
10.
J Contemp Dent Pract ; 21(2): 190-196, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32381826

ABSTRACT

AIM: The aim of the study was to evaluate the dimensional accuracy of three combinations of polyvinyl siloxane impression material by double-mix single-step impression technique. MATERIALS AND METHODS: Metal master model was made according to the ADA specification no. 19; ISO 4823:2000/AMD 2007. Impressions were made using perforated custom-made metallic trays of 2 mm and 4 mm spacing, the impression materials used were putty, heavy body, regular body and light body. A total of 30 impressions were made by single-step technique and poured in die stone to obtain resultant cast. Ten impressions were made of each combination of polyvinyl siloxane (PVS). Three dimensions (interabutment distance, height and diameter) on resultant cast were measured and compared with metal master model. The results were statistically analyzed and tabulated. RESULTS: Diameter of abutment, the height of abutment and interabutment distance in each group were larger in dimensions as compared with metal master model. The dimensional discrepancies of group I, group II and group III casts when compared with the master model were significantly different from each other. The least difference was found in group I. CONCLUSION: The one-step putty-light body combination (group I) produced the most accurate stone casts compared with one-step heavy body-light body and regular body-light body combinations. CLINICAL SIGNIFICANCE: In everyday dental practice, impression making is imperative. Hence, by doing this study, we tried to find out which material combination is suitable to give us predictable and accurate results.


Subject(s)
Dental Impression Technique , Models, Dental , Polyvinyls , Siloxanes
11.
Biomed Pharmacother ; 112: 108707, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30970513

ABSTRACT

The synthesis and antiproliferative effect of a series of quinoline and thiazole containing coumarin analogs 12a-d and 13a-f respectively, on mice leukemic cells was performed. The chemical structures of newly synthesized compounds were confirmed by IR, 1H NMR, 13C NMR and mass spectral analysis. The result indicates that, 7-methoxy-2-oxo-2H-chromene-3-carboxylic acid [4-(4-methoxy-phenyl)-thiazol-2-yl]-amide (13f) showed potent activity against EAC and DLA cells in MTT (15.3 µM), tryphan blue (15.6 µM) and LDH (14.2 µM) leak assay with 5-fluorouracil as a standard. Further, the anti-neoplastic effect of the compound 13f was verified against Ehrlich ascites tumour by BrdU incorporation, TUNEL, FACS and DNA fragmentation assays. Experimental data showed that compound 13f induces the apoptotic cell death by activating apoptotic factors such as caspase-8 &-3, CAD, Cleaved PARP, γ-H2AX and by degrading genomic DNA of cancer cells and thereby decreasing the ascitic tumour development in mice. Besides, compound 13f was also subjected for docking studies to approve the in vitro and in vivo studies. The data revealed that the compound 13f has very good interaction with caspase 3 protein by binding with amino acid Arg 207 through hydrogen bond.


Subject(s)
Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Carcinoma, Ehrlich Tumor/drug therapy , Coumarins/chemical synthesis , Quinolines/chemistry , Thiazoles/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Carcinoma, Ehrlich Tumor/pathology , Cell Line, Tumor , Computer Simulation , Coumarins/chemistry , Coumarins/therapeutic use , Coumarins/toxicity , Lethal Dose 50 , Mice , Structure-Activity Relationship
12.
Biomed Pharmacother ; 103: 1446-1455, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29864929

ABSTRACT

Ten new 2(4-hydroxy-3-benzoyl) benzamide-5-phenyl-1,3,4-oxadiazole derivatives (10a-j) were synthesized by coupling 3-benzoyl-4-hydroxybenzoic acid (5) with 2-amino-5-phenyl-1,3,4-oxadiazoles (9a-j). The structures of these compounds were confirmed by IR, 1H, 13C NMR, and mass spectra, and also by elemental analyses. The anti-inflammatory activity of the compounds 10a-j were investigated by screening them against human red blood cells (HRBC) in-vitro. The results reveal that among this series, compound 10j with hydroxy substituent, particularly at the ortho position of the phenyl ring attached to the 5th carbon atom of the oxadiazole ring possess significant membrane stabilizing activity in comparison with the control. Further, in-vivo chick chorioallantoic membrane (CAM) and rat corneal anti-angiogenesis assays were performed to assess the effect of compound 10j on endothelial cell migration. This confirmed that compound 10j inhibits the proliferation of endothelial cells. Anti-inflammatory studies detected the amelioration of carrageen induced rat hind paw edema. Further in-vivo and in-silico approaches revealed the inhibition of inflammatory marker enzyme cyclooxygenase-2 (Cox-2) and myleoperoxidase (MPO). The study reports that the compound 10j effectively act against the inflammatory mediated anti-angiogenic disorders which could be translated into a new drug in future.


Subject(s)
Benzophenones/chemical synthesis , Benzophenones/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase 2/metabolism , Edema/drug therapy , Inflammation/drug therapy , Oxadiazoles/chemical synthesis , Oxadiazoles/therapeutic use , Animals , Benzophenones/chemistry , Benzophenones/pharmacology , Chickens , Cyclooxygenase 2 Inhibitors/pharmacology , Edema/complications , Edema/enzymology , Humans , Inflammation/complications , Inflammation/enzymology , Male , Neovascularization, Physiologic/drug effects , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Rats
13.
Life Sci ; 199: 139-150, 2018 Apr 15.
Article in English | MEDLINE | ID: mdl-29524520

ABSTRACT

Aim Deformity in the cellular homeostatic event associated with cell survival and apoptosis are committing factors for carcinogenesis. Interventions of these events by pharmacologically active agent gain predominance in cancer treatment. In current investigation Solanidine, a steroidal alkaloid was evaluated on tumorigenesis by targeting death signal using multiple tumor cells and model systems. MAIN METHODS: Anti-proliferative effect was evaluated using cytotoxic studies. Prolonged cytotoxic effect of Solanidine was examined by colony formation assay. Exhibition of apoptotic hallmark induced by Solanidine was examined using FACS analysis, Annexin-V staining, Acridine orange staining, TUNEL assay. Altered gene expression was evaluated using Immunoblot, Immunofluorescence and Immunohistochemistry technique. In-vitro results were revalidated in EAC solid tumor and CAM xenograft model. KEY FINDINGS: Solanidine exerts its potential effect in a target specific manner. The cytotoxic/anticlonogenic activity was due to induction of typical cellular apoptotic hallmarks and cell cycle blockage at S-G2/M phase. The molecular events underlying this effect is through activation of intrinsic pathway via Bax, Bad and Cytochrome c activation by neutralizing Bcl-2 expression, along with downregulated PI3K/Akt survival signal. As a consequence, downstream pro apoptogenic gene, active Caspase-3 was over expressed by Solanidine to cleave its substrate PARP and promotes nuclear import of DFF-40. Anti-carcinogenic aptitude was further confirmed by murine solid tumors and in-vivo CAM xenograft studies. SIGNIFICANCE: Solanidine emerged as active molecule against tomorigenesis by activating nuclear import of DFF-40 mediated nucleosomal disruption and cell demise. It can be developed as a potential apoptogenic small molecule for cancer therapy.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Alkaloids/pharmacology , Cell Death/drug effects , Deoxyribonucleases/metabolism , Diosgenin/pharmacology , Nucleosomes/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , A549 Cells , Active Transport, Cell Nucleus/physiology , Alkaloids/therapeutic use , Animals , Cell Death/physiology , Diosgenin/therapeutic use , Dose-Response Relationship, Drug , HEK293 Cells , Hep G2 Cells , Humans , MCF-7 Cells , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice , Mice, Inbred BALB C , Xenograft Model Antitumor Assays/methods
14.
Eur J Med Chem ; 143: 1826-1839, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29133037

ABSTRACT

Tumor microenvironment is a complex multistep event which involves several hallmarks that transform the normal cell into cancerous cell. Designing the novel antagonistic molecule to reverse the tumor microenvironment with specific target is essential in modern biological studies. The novel 4-phenyl-2-phenoxyacetamide thiazole analogues 8a-ab were synthesized in multistep process, then screened and assessed for cytotoxic and anti-proliferative effects in vitro against multiple cancer cells of different origin such as MCF-7, A549, EAC and DLA cells which revealed that compound 8f with fluoro and methyl substitute has potential cytotoxic efficacy with an average IC50 value of ˜ 13 µM. The mechanism of cytotoxicity assessed for anti-tumor studies both in ascites and solid tumor models in-vivo inferred the regressed tumor activity. This is due to changes in the cause of tumor microenvironment with crackdown of neovascularization and evoking apoptosis process as assessed by CAM, corneal vascularization and apoptotic hallmarks in 8f treated cells. The molecular gene studies inferred involvement of HIF-1upregulation and stabilization of p53 which are interlinked in signaling as conferred by immunoblot analysis.


Subject(s)
Acetamides/pharmacology , Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Thiazoles/pharmacology , Tumor Hypoxia/drug effects , Acetamides/chemical synthesis , Acetamides/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/pathology , Rats , Rats, Wistar , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry , Tumor Cells, Cultured
15.
Biomed Pharmacother ; 95: 419-428, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28863382

ABSTRACT

Disrupted redox balance is implicated in multiple pathologies including malignant progression and tumor angiogenesis. In this investigation, we report the design and development of novel and effective ROS detoxifying azo-hydrazone molecules targeting malignant pathologies and neoangiogenesis. A series of azo-derivatives conjugated to hydrazones moieties (9a-j) were synthesized using Nano BF3·SiO2. The compounds (9a-j) were screened for in-vitro antioxidant and lipid peroxidation inhibitory activity. Among the series 9a-j, compound 9f potently quenched biologically relevant radicals such as superoxide and hydrogen peroxide which emerged as the lead ROS detoxifying molecules. Compound 9f potently inhibited the proliferative capability of Daltons Lymphoma Ascites (DLA) tumor cells in-vivo in dose dependent manner. Regressed tumor progression was correlated with pronounced endogenous antioxidant enzyme superoxide dismutase and catalase in-vivo. Also, ROS levels were severely suppressed in 9f treated mice as assessed by lapsed lipid peroxidation. Altered enzymic and ROS levels in-vivo by 9f were implicated in suppressed VEGF secretion leading to regressed tumor neovasculature and tumor growth. Considering together, it is evident that the synthetic azo-hydrazone analogue 9f with potent ROS scavenging efficacy inhibits tumor progression and neo-angiogenesis.


Subject(s)
Boranes/chemistry , Carcinogenesis/drug effects , Homeostasis , Hydrazones/chemical synthesis , Hydrazones/therapeutic use , Neovascularization, Pathologic/drug therapy , Reactive Oxygen Species/metabolism , Silicon Dioxide/chemistry , Animals , Antioxidants/metabolism , Ascites/pathology , Disease Progression , Drug Design , Hydrazones/pharmacology , Lipid Peroxidation/drug effects , Mice , Neovascularization, Pathologic/blood , Structure-Activity Relationship , Vascular Endothelial Growth Factor A/metabolism
16.
J Neuroinflammation ; 14(1): 117, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28599652

ABSTRACT

Multiple sclerosis (MS) is a chronic debilitating disease of the central nervous system primarily mediated by T lymphocytes with specificity to neuronal antigens in genetically susceptible individuals. On the other hand, myasthenia gravis (MG) primarily involves destruction of the neuromuscular junction by antibodies specific to the acetylcholine receptor. Both autoimmune diseases are thought to result from loss of self-tolerance, which allows for the development and function of autoreactive lymphocytes. Although the mechanisms underlying compromised self-tolerance in these and other autoimmune diseases have not been fully elucidated, one possibility is numerical, functional, and/or migratory deficits in T regulatory cells (Tregs). Tregs are thought to play a critical role in the maintenance of peripheral immune tolerance. It is believed that Tregs function by suppressing the effector CD4+ T cell subsets that mediate autoimmune responses. Dysregulation of suppressive and migratory markers on Tregs have been linked to the pathogenesis of both MS and MG. For example, genetic abnormalities have been found in Treg suppressive markers CTLA-4 and CD25, while others have shown a decreased expression of FoxP3 and IL-10. Furthermore, elevated levels of pro-inflammatory cytokines such as IL-6, IL-17, and IFN-γ secreted by T effectors have been noted in MS and MG patients. This review provides several strategies of treatment which have been shown to be effective or are proposed as potential therapies to restore the function of various Treg subsets including Tr1, iTr35, nTregs, and iTregs. Strategies focusing on enhancing the Treg function find importance in cytokines TGF-ß, IDO, interleukins 10, 27, and 35, and ligands Jagged-1 and OX40L. Likewise, strategies which affect Treg migration involve chemokines CCL17 and CXCL11. In pre-clinical animal models of experimental autoimmune encephalomyelitis (EAE) and experimental autoimmune myasthenia gravis (EAMG), several strategies have been shown to ameliorate the disease and thus appear promising for treating patients with MS or MG.


Subject(s)
Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Immune Tolerance/drug effects , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunotherapy/methods , Immunotherapy/trends , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Multiple Sclerosis/metabolism , Myasthenia Gravis, Autoimmune, Experimental/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
17.
Clin Exp Immunol ; 189(1): 21-35, 2017 07.
Article in English | MEDLINE | ID: mdl-28268243

ABSTRACT

Neovascularization and jeopardized immunity has been critically emphasized for the establishment of malignant progression. Lectins are the diverse class of carbohydrate interacting proteins, having great potential as immunopotentiating and anti-cancer agents. The present investigation sought to demonstrate the anti-proliferative activity of Dolichos lablab lectin (DLL) encompassing immunomodulatory attributes. DLL specific to glucose and mannose carbohydrate moieties has been purified to homogeneity from the common dietary legume D. lablab. Results elucidated that DLL agglutinated blood cells non-specifically and displayed striking mitogenicity to human and murine lymphocytes in vitro with interleukin (IL)-2 production. The DLL-conditioned medium exerted cytotoxicity towards malignant cells and neoangiogenesis in vitro. Similarly, in-vivo anti-tumour investigation of DLL elucidated the regressed proliferation of ascitic and solid tumour cells, which was paralleled with blockade of tumour neovasculature. DLL-treated mice showed an up-regulated immunoregulatory cytokine IL-2 in contrast to severely declined levels in control mice. Mechanistic validation revealed that DLL has abrogated the microvessel formation by weakening the proangiogenic signals, specifically nuclear factor kappa B (NF-κB), hypoxia inducible factor 1α (HIF-1 α), matrix metalloproteinase (MMP)-2 and 9 and vascular endothelial growth factor (VEGF) in malignant cells leading to tumour regression. In summary, it is evident that the dietary lectin DLL potentially dampens the malignant establishment by mitigating neoangiogenesis and immune shutdown. For the first time, to our knowledge, this study illustrates the critical role of DLL as an immunostimulatory and anti-angiogenic molecule in cancer therapeutics.


Subject(s)
Mitogens/pharmacology , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Plant Lectins/administration & dosage , Plant Lectins/pharmacology , A549 Cells , Agglutination , Animals , Aorta/drug effects , Cell Culture Techniques , Chorioallantoic Membrane/drug effects , Cornea/blood supply , Cornea/drug effects , Disaccharides , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunomodulation , Interleukin-2/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mitogens/immunology , Plant Lectins/immunology , Rats , Rats, Wistar , Seeds/chemistry , Vascular Endothelial Growth Factor A/metabolism
18.
Bioorg Chem ; 71: 55-66, 2017 04.
Article in English | MEDLINE | ID: mdl-28139247

ABSTRACT

A series of novel 4-benzyl-morpholine-2-carboxylic acid N'-[2-(4-benzoyl-phenoxy)-acetyl]-hydrazide derivatives 8a-j has been synthesized from (4-hydroxy-aryl)-aryl methanones through a multi-step reaction sequence and then evaluated for anti-proliferative activity in vitro against various types of neoplastic cells of mouse and human such as DLA, EAC, MCF-7 and A549 cells. From the cytotoxic studies and structural activity relationship of compounds 8a-j, it is clear that methyl group on the B ring of benzophenone is essential for antiproliferative activity and bromo at ortho position (compound 8b) and methyl at para position (compound 8f) on A ring of benzophenone are significant for extensive anti-mitogenic activity. Investigation on clonogenesis and Fluorescence-activated cell sorting suggests that compounds 8b and 8f have the potency to exhibit the prolonged activity with cell cycle arrest on G2/M phase against cancer progression. Further, the compounds 8b and 8f inhibit murine ascites lymphoma through caspase activated DNase mediated apoptosis.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzophenones/chemistry , Benzophenones/pharmacology , Morpholines/chemistry , Morpholines/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Benzophenones/chemical synthesis , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Morpholines/chemical synthesis , Neoplasms/drug therapy , Structure-Activity Relationship
19.
Biochem Biophys Res Commun ; 484(1): 85-92, 2017 02 26.
Article in English | MEDLINE | ID: mdl-28104392

ABSTRACT

Neoplastic cells often reside in distinctive tumor hypoxia armed with a series of adaptive responses including oxidative stress, defective apoptotic machinery and neoangiogenesis, through that further confer cell survival improvement. Plants still acts as reservoir of natural chemicals to provide newer active pharmacophores. Scutellarein is flavones which has wide range of pharmacophoral effects. In our current research, scutellarein employed for targeting oxidative stress mediated tumor angiogenesis and apoptotic nuclear fragmentation. Experimental results revealed that scutellarein has antiproliferative index against multiple cancer cell lines and diminished the oxidative stress and tumor development of murine ascitic lymphoma & inflammatory hepatocellular carcinoma. Eventual consequences lead to reduced neovessel formation by abrogating angiogeneic factors cytokine-VEGF-A, Flt-1, HIF-1α, MMP-2 and MMP-9 and reversing of evading apoptosis by activating caspase-3 activated DNA fragmentation factor (DFF-40) mediated nucleosomal degradation. In summary, our experimental evidences suggest that scutellarein has strong potentiality to attenuate the tumor development by modulating sprouting neovasculature and DFF-40 mediated apoptosis.


Subject(s)
Apigenin/pharmacology , Carcinogenesis/drug effects , Deoxyribonucleases/pharmacology , Neovascularization, Pathologic/chemically induced , Nucleosomes/metabolism , Vascular Endothelial Growth Factor A/physiology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Mice, Inbred BALB C , Rats
20.
Biochem Pharmacol ; 125: 26-40, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27838496

ABSTRACT

Hypoxia is an important module in all solid tumours to promote angiogenesis, invasion and metastasis. Stabilization and subsequent nuclear localization of HIF-1α subunits result in the activation of tumour promoting target genes such as VEGF, MMPs, Flt-1, Ang-1 etc. which plays a pivotal role in adaptation of tumour cells to hypoxia. Increased HIF-α and its nuclear translocation have been correlated with pronounced angiogenesis, aggressive tumour growth and poor patient prognosis leading to current interest in HIF-1α as an anticancer drug target. Benzophenone-1B ([4-(1H-benzimidazol-2-ylmethoxy)-3,5-dimethylphenyl]-(4-methoxyphenyl) methanone, or BP-1B) is a new antineoplastic agent with potential angiopreventive effects. Current investigation reports the cellular biochemical modulation underlying BP-1B cytotoxic/antiangiogenic effects. Experimental evidences postulate that BP-1B exhibits the tumour specific cytotoxic actions against various cancer types with prolonged action. Moreover BP-1B efficiently counteracts endothelial cell capillary formation in in-vitro, in-vivo non-tumour and tumour angiogenic systems. Molecular signaling studies reveal that BP-1B arrests nuclear translocation of HIF-1α devoid of p42/44 pathway under CoCl2 induced hypoxic conditions in various cancer cells thereby leading to abrogated HIF-1α dependent activation of VEGF-A, Flt-1, MMP-2, MMP -9 and Ang-1 angiogenic factors resulting in retarded cell migration and invasions. The in-vitro results were reproducible in the reliable in-vivo solid tumour model. Taken together, we conclude that BP-1B impairs angiogenesis by blocking nuclear localization of HIF-1α which can be translated into a potent HIF-1α inhibitor.


Subject(s)
Benzophenones/pharmacology , Cell Nucleus/metabolism , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/pathology , Animals , Cell Line, Tumor , Chick Embryo , Disease Models, Animal , Disease Progression , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , Mice, Inbred BALB C , Protein Transport , Rats , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...