Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Phytomedicine ; 119: 154981, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37531902

RESUMEN

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.


Asunto(s)
Adenocarcinoma del Pulmón , Alcaloides , Antineoplásicos , Neoplasias Pulmonares , Ratas , Animales , Fosforilación , Antineoplásicos/uso terapéutico , Hipoxia/tratamiento farmacológico , Alcaloides/farmacología , Adenocarcinoma del Pulmón/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neovascularización Patológica/tratamiento farmacológico , Línea Celular Tumoral , Microambiente Tumoral
2.
Chem Biodivers ; 20(5): e202201152, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36974341

RESUMEN

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.


Asunto(s)
Antineoplásicos , Tetrahidroisoquinolinas , Animales , Ratones , Estructura Molecular , Simulación del Acoplamiento Molecular , Línea Celular Tumoral , Proliferación Celular , Ensayos de Selección de Medicamentos Antitumorales , Relación Estructura-Actividad , Antineoplásicos/farmacología , Tetrahidroisoquinolinas/farmacología , Metaloproteasas/farmacología , Diseño de Fármacos , Inhibidores de Proteínas Quinasas/farmacología
3.
3 Biotech ; 12(11): 306, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36276461

RESUMEN

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.

4.
Pharmacol Rep ; 74(2): 353-365, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35001321

RESUMEN

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.


Asunto(s)
Hidrazonas , Factor de Transcripción STAT3 , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Hidrazonas/farmacología , Janus Quinasa 2/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
5.
Apoptosis ; 27(1-2): 49-69, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34837562

RESUMEN

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.


Asunto(s)
Apoptosis , Neoplasias Pulmonares , Benzofenonas/farmacología , Línea Celular Tumoral , Proliferación Celular , Cumarinas/farmacología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Factor de Transcripción STAT3/metabolismo
6.
Pharmacol Rep ; 73(5): 1344-1360, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34109572

RESUMEN

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.


Asunto(s)
Acetamidas/síntesis química , Acetamidas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Daño del ADN , Sistemas de Liberación de Medicamentos , Animales , Apoptosis , Línea Celular Tumoral , Reparación del ADN , Humanos , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biomed Pharmacother ; 112: 108707, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30970513

RESUMEN

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.


Asunto(s)
Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Carcinoma de Ehrlich/tratamiento farmacológico , Cumarinas/síntesis química , Quinolinas/química , Tiazoles/química , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Carcinoma de Ehrlich/patología , Línea Celular Tumoral , Simulación por Computador , Cumarinas/química , Cumarinas/uso terapéutico , Cumarinas/toxicidad , Dosificación Letal Mediana , Ratones , Relación Estructura-Actividad
8.
Biomed Pharmacother ; 103: 1446-1455, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29864929

RESUMEN

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.


Asunto(s)
Benzofenonas/síntesis química , Benzofenonas/uso terapéutico , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Ciclooxigenasa 2/metabolismo , Edema/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Oxadiazoles/síntesis química , Oxadiazoles/uso terapéutico , Animales , Benzofenonas/química , Benzofenonas/farmacología , Pollos , Inhibidores de la Ciclooxigenasa 2/farmacología , Edema/complicaciones , Edema/enzimología , Humanos , Inflamación/complicaciones , Inflamación/enzimología , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Oxadiazoles/química , Oxadiazoles/farmacología , Ratas
9.
Life Sci ; 199: 139-150, 2018 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-29524520

RESUMEN

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.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Alcaloides/farmacología , Muerte Celular/efectos de los fármacos , Desoxirribonucleasas/metabolismo , Diosgenina/farmacología , Nucleosomas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Células A549 , Transporte Activo de Núcleo Celular/fisiología , Alcaloides/uso terapéutico , Animales , Muerte Celular/fisiología , Diosgenina/uso terapéutico , Relación Dosis-Respuesta a Droga , Células HEK293 , Células Hep G2 , Humanos , Células MCF-7 , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
10.
Eur J Med Chem ; 143: 1826-1839, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29133037

RESUMEN

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.


Asunto(s)
Acetamidas/farmacología , Antineoplásicos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Tiazoles/farmacología , Hipoxia Tumoral/efectos de los fármacos , Acetamidas/síntesis química , Acetamidas/química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neovascularización Patológica/patología , Ratas , Ratas Wistar , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/química , Células Tumorales Cultivadas
11.
Biomed Pharmacother ; 95: 419-428, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28863382

RESUMEN

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.


Asunto(s)
Boranos/química , Carcinogénesis/efectos de los fármacos , Homeostasis , Hidrazonas/síntesis química , Hidrazonas/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Dióxido de Silicio/química , Animales , Antioxidantes/metabolismo , Ascitis/patología , Progresión de la Enfermedad , Diseño de Fármacos , Hidrazonas/farmacología , Peroxidación de Lípido/efectos de los fármacos , Ratones , Neovascularización Patológica/sangre , Relación Estructura-Actividad , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Clin Exp Immunol ; 189(1): 21-35, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28268243

RESUMEN

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.


Asunto(s)
Mitógenos/farmacología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Lectinas de Plantas/administración & dosificación , Lectinas de Plantas/farmacología , Células A549 , Aglutinación , Animales , Aorta/efectos de los fármacos , Técnicas de Cultivo de Célula , Membrana Corioalantoides/efectos de los fármacos , Córnea/irrigación sanguínea , Córnea/efectos de los fármacos , Disacáridos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunomodulación , Interleucina-2/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Mitógenos/inmunología , Lectinas de Plantas/inmunología , Ratas , Ratas Wistar , Semillas/química , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Bioorg Chem ; 71: 55-66, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28139247

RESUMEN

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.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Benzofenonas/química , Benzofenonas/farmacología , Morfolinas/química , Morfolinas/farmacología , Animales , Antineoplásicos/síntesis química , Benzofenonas/síntesis química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Morfolinas/síntesis química , Neoplasias/tratamiento farmacológico , Relación Estructura-Actividad
14.
Biochem Biophys Res Commun ; 484(1): 85-92, 2017 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-28104392

RESUMEN

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.


Asunto(s)
Apigenina/farmacología , Carcinogénesis/efectos de los fármacos , Desoxirribonucleasas/farmacología , Neovascularización Patológica/inducido químicamente , Nucleosomas/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratas
15.
Angiogenesis ; 20(1): 55-71, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27743086

RESUMEN

Hypoxia is a feature of all solid tumours, contributing to tumour progression. Activation of HIF-1α plays a critical role in promoting tumour angiogenesis and metastasis. Since its expression is positively correlated with poor prognosis for cancer patients, HIF-1α is one of the most convincing anticancer targets. BP-1T is a novel antiproliferative agent with promising antiangiogenic effects. In the present study, the molecular mechanism underlying cytotoxic/antiangiogenic effects of BP-1T on tumour/non-tumour angiogenesis was evaluated. Evidences show that BP-1T exhibits potent cytotoxicity with prolonged activity and effectively regressed neovessel formation both in reliable non-tumour and tumour angiogenic models. The expression of CoCl2-induced HIF-1α was inhibited by BP-1T in various p53 (WT)-expressing cancer cells, including A549, MCF-7 and DLA, but not in mutant p53-expressing SCC-9 cells. Mechanistically, BP-1T mediates the HIF-1α proteasomal degradation by activating p53/MDM2 pathway and thereby downregulated HIF-1α-dependent angiogenic genes such as VEGF-A, Flt-1, MMP-2 and MMP-9 under hypoxic condition of in vitro and in vivo solid tumour, eventually leading to abolition of migration and invasion. Based on these observations, we conclude that BP-1T acts on HIF-1α degradation through p53/MDM2 proteasome pathway.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Benzofenonas/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Tiazoles/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Benzofenonas/química , Carcinogénesis/metabolismo , Carcinogénesis/patología , Muerte Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Ratones , Microvasos/patología , Modelos Biológicos , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Tiazoles/química
16.
Biochem Pharmacol ; 125: 26-40, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27838496

RESUMEN

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.


Asunto(s)
Benzofenonas/farmacología , Núcleo Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/patología , Animales , Línea Celular Tumoral , Embrión de Pollo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Transporte de Proteínas , Ratas , Factor A de Crecimiento Endotelial Vascular/farmacología
17.
Int Immunopharmacol ; 39: 158-171, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27475665

RESUMEN

Lifestyle and dietary modifications have contributed much to somatic genetic alteration which has concomitantly led to increase in malignant diseases. Henceforth, plant based and dietary interventions to mitigate and impede oncogenic transformation are in great demand. We investigated the latex sap (LSL) of the dietary Lagenaria siceraria vegetable, the first domesticated plant species with the potent lectin activity for its functional role against the tumor progression and its mechanism. LSL has markedly stimulated proliferation of lymphocytes and displayed strong cytotoxic activity against cancer both in-vitro and in-vivo. The tumor regression was paralleled with drastic reduction in tumoral neovasculature as evidenced from angiogenic parameters and abrogated related gene expressions. LSL has also triggered apoptotic signaling cascade in cancer cells through activation of caspase-3 mediated activation of endonuclease and inducing apoptotic cellular events. Collectively our study provides tangible evidences that latex sap from L. siceraria with immunopotentiating ability significantly regresses the tumor progression by targeting angiogenesis and inducing cell death.


Asunto(s)
Aorta/efectos de los fármacos , Cucurbitaceae/inmunología , Látex/uso terapéutico , Linfocitos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Lectinas de Plantas/uso terapéutico , Animales , Aorta/fisiología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfocitos/fisiología , Células MCF-7 , Ratones , Células 3T3 NIH , Trasplante de Neoplasias , Técnicas de Cultivo de Órganos , Ratas , Carga Tumoral/efectos de los fármacos
18.
Eur J Med Chem ; 114: 153-61, 2016 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-26974382

RESUMEN

Mitogenicity is the ability of the natural or synthetic compounds to induce cell division or proliferation. A series of salicylic acid derivatives containing isoxazoline moiety (8a-j) were synthesized and their immunopharmacological activities targeting lymphocyte proliferation and angiogenesis were evaluated. The compounds 8a-j mitogenicity were investigated on immunological cells that include human peripheral blood lymphocytes and murine splenocytes in-vitro. The results implicate that among the series of 8a-j, compound 8e showed a potent proliferative response on both human and murine lymphocytes. The proliferative index of the compound 8e was comparable to the reference mitogen Con A and mitogenecity is due to increased secretion IL-2. In -vivo CAM and rat corneal angiogenesis assays were performed to assess the compound's effect on endothelial cell migration and proliferation which inferred that 8e also induces the proliferation of endothelial cells. The study reports the synthetic immunostimulatory and pro-angiogenic activity of novel mitogen 8e which could be translated into new drug in future.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Factores Inmunológicos/farmacología , Isoxazoles/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Ácido Salicílico/farmacología , Adolescente , Adulto , Inductores de la Angiogénesis/síntesis química , Inductores de la Angiogénesis/química , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células HEK293 , Humanos , Factores Inmunológicos/síntesis química , Factores Inmunológicos/química , Isoxazoles/química , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Masculino , Ratones , Persona de Mediana Edad , Estructura Molecular , Neovascularización Fisiológica/inmunología , Ratas , Ácido Salicílico/química , Bazo/efectos de los fármacos , Bazo/inmunología , Relación Estructura-Actividad , Adulto Joven
19.
Eur J Med Chem ; 115: 342-51, 2016 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-27027818

RESUMEN

A series of diamide-coupled benzophenone, 2-(4-benzoyl-phenoxy)-N-{2-[2-(4-benzoyl-phenoxy)-acetylamino]-phenyl}-acetamide analogues (9a-l) were synthesized by multistep reactions and all compounds were well characterized. Among the series (9a-l), compound 9k with three methyl groups at ortho position in rings A, B, and D and bromo group at the para position in ring E was selected as a lead compound by screening through multiple cancer cell types by in-vitro cytotoxic and antiproliferative assay systems. Also, the cytotoxic nature of the compound 9k resulted the regression of the tumor growth in-vivo, which could be due to decreased vascularisation in the peritoneum lining of the mice which regress the tumor growth. The results were reconfirmed in-vivo chorioallantoic membrane model which indicates a scope of developing 9k into potent anticancer drug in near future.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Benzofenonas/química , Benzofenonas/farmacología , Animales , Antineoplásicos/síntesis química , Benzofenonas/síntesis química , Humanos , Células MCF-7 , Ratones , Relación Estructura-Actividad
20.
Bioorg Chem ; 65: 110-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26918263

RESUMEN

A novel series of benzoic acid N'-[2-(4-benzothiazol-2-yl-piperazin-1-yl)-acetyl]-hydrazides 6a-j were synthesized and characterized by IR, (1)H, (13)C NMR, elemental and mass spectral analyses. The in-vitro cytotoxicity and cell viability assay of the synthesized compounds 6a-j were evaluated against Dalton's lymphoma ascites (DLA) cells. Our results showed that compound 6c with a bromo group on phenyl ring has showed promising antiproliferative efficacy. Further investigation of compound 6c on in-vivo treatment model depicts the increased tumor suppression through inhibition of angiogenesis.


Asunto(s)
Antineoplásicos/farmacología , Hidrazinas/farmacología , Linfoma/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Piperazinas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Hidrazinas/síntesis química , Hidrazinas/química , Linfoma/patología , Masculino , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neovascularización Patológica/patología , Piperazinas/síntesis química , Piperazinas/química , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA