Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Environ Sci Pollut Res Int ; 30(26): 67941-67952, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37140872

ABSTRACT

Desalination is a tried-and-true method for obtaining clean water from the ocean's brackish waters and recycling and reusing water. It requires a fair amount of energy, so it is necessary to create sustainable energy systems to lessen energy use and environmental impact. For thermal desalination procedures, thermal sources can be great heat sources. This paper's research focuses on thermoeconomically optimized multi-effect distillation and geothermal desalination systems. Collecting hot water from subsurface reservoirs is a well-established method of generating electricity through geothermal sources. Low-temperature geothermal sources, which have a temperature of less than 130 °C, can be utilized for thermal desalination systems, for example, multi-effect distillation (MED). Geothermal desalination is affordable, and it is possible to produce power simultaneously. Because it only uses clean, renewable energy and produces no greenhouse gasses or other pollutants, it is also safe for the environment. The viability of any geothermal desalination plant will be influenced by the location of the geothermal resource, feed water supply, cooling water source, water market, and concentrate disposal site. Geothermal energy can directly supply heat for a thermal desalination system or indirectly give electrical power to reverse the osmosis (RO) membrane-based desalination plant.


Subject(s)
Geothermal Energy , Water Purification , Water Purification/methods , Distillation , Water , Cold Temperature
2.
Biomed Pharmacother ; 114: 108855, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31003140

ABSTRACT

The anti-carcinogenic effect of Methylglyoxal (MG) is well established. It generally targets malignant cells by affecting glycolysis and mitochondrial respiration with minimum or no toxicity to normal cells. In an initial study we have reported that MG can synergistically act with 5-Fluorouracil (5-FU) to decreases the number of MCF-7 breast cancer cells (Ghosh S, Pal A, Ray M, 2017). This finding prompted us to study the combination effect of MG and 5-FU extensively in both in vitro and in vivo. Induction of cell apoptosis and cell cycle arrest was systematically studied to reveal the mechanisms of synergy between 5-FU and MG. Our present study revealed that MG can synergistically act with 5-FU and can cause cell death via apoptosis and generated reactive oxygen species (ROS) in MCF-7 cells. Combination of 5-FU and MG resulted in more potent apoptosis induction as revealed by fluorescence microscopy using Hoechst 33342. In comparison to single drug treatment, the co-treatment also increased the number of cells in G0/G1 phase by downregulating the expression of CDK4 and CDK6 as compared to single drug treatment. Levels of Caspase 9 and poly (ADP-ribose) polymerase (PARP) were higher in combination treatment as compared to single drug treatment. These results clearly showed that 5-FU is more effective at lower doses in presence of MG in MCF-7 cells. In case of in vivo studies treatment of EAC (Ehrlich Ascites Carcinoma) bearing mice with MG in combination with 5-FU at various doses, demonstrated the same synergistic effect of MG with 5-FU. The combination study also exhibited tumor regression in BALB/c mouse 4T1 breast tumor model as well. We also clearly demonstrated that MG can decrease the cytotoxic side effects of 5-FU as indicated with acute and chronic toxicity studies and other biochemical analyses of blood and histological studies. Taken together, our results revealed that MG could be a potential candidate for combination therapy to reduce the toxicity burden of 5-FU without any toxic impact on host cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Fluorouracil/pharmacology , Pyruvaldehyde/pharmacology , A549 Cells , Animals , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Xenograft Model Antitumor Assays/methods
3.
Biochem Pharmacol ; 156: 322-339, 2018 10.
Article in English | MEDLINE | ID: mdl-30170097

ABSTRACT

Potent anticancer activity coupled with absence of toxicity at therapeutic dose established the glycolytic metabolite, methylglyoxal, as a promising candidate against malignant neoplasia. In this preclinical study we illustrate the applicability of methylglyoxal in formulating an optimally designed combination regimen with chemotherapeutic drugs against breast cancer. Results demonstrated a synergistic augmentation in doxorubicin and cisplatin mediated cytotoxicity in human breast cancer cell lines MDA MB 231 & MCF 7 with methylglyoxal co-treatment at metronomic concentrations. The cell death due to combination treatment was significantly prevented by N-Acetylcysteine and the synergistic effects were attenuated in presence of inhibitors for apoptosis and necroptosis, in MDA MB 231 and MCF 7 cells, respectively. Additionally, acridine orange staining and immunoblotting with LC3B antibody indicated the suppression of doxorubicin induced autophagy flux with methylglyoxal co-treatment. This report documents for the first time the preferential targeting of breast cancer stem cells by methylglyoxal. Combination treatment with doxorubicin or cisplatin hindered mammosphere forming efficiency and inclusively eliminated both cancer stem as well as non-stem cancer cells. The synergistic effect was validated in Ehrlich mammary carcinoma cell induced murine ascites model and the combination advantage in vivo was achieved without any additional deleterious effect to liver and kidney. Our present study evidences the implications of methylglyoxal inclusion in adjuvant multimodal chemotherapeutics against breast cancer and offers noteworthy insights into the possible outcome.


Subject(s)
Breast Neoplasms/drug therapy , Cisplatin/pharmacology , Doxorubicin/pharmacology , Pyruvaldehyde/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Autophagy , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/administration & dosage , Doxorubicin/administration & dosage , Drug Administration Schedule , Drug Synergism , Female , Humans , Mice , Neoplasms, Experimental , Pyruvaldehyde/administration & dosage , Reactive Oxygen Species , Xenograft Model Antitumor Assays
4.
Amino Acids ; 48(8): 2003-13, 2016 08.
Article in English | MEDLINE | ID: mdl-27138627

ABSTRACT

The anti-cancer effect of methylglyoxal (MG) is now well established in the literature. The main aim of this study was to investigate the effect of creatine as a supplement in combination with MG both in vitro and in vivo. In case of the in vitro studies, two different cell lines, namely MCF-7 (human breast cancer cell line) and C2C12 (mouse myoblast cell line) were chosen. MG in combination with creatine showed enhanced apoptosis as well as higher cytotoxicity in the breast cancer MCF-7 cell line, compared to MG alone. Pre-treatment of well-differentiated C2C12 myotubes with cancerogenic 3-methylcholanthrene (3MC) induced a dedifferentiation of these myotubes towards cancerous cells (that mimic the effect of 3MC observed in solid fibro-sarcoma animal models) and subsequent exposure of these induced cancer cells with MG proved to be cytotoxic. Thus, creatine plus ascorbic acid enhanced the anti-cancer effects of MG. In contrast, when normal C2C12 muscle cells or myotubes (mouse normal myoblast cell line) were treated with MG or MG plus creatine and ascorbic acid, no detrimental effects were seen. This indicated that cytotoxic effects of MG are specifically limited towards cancer cells and are further enhanced when MG is used in combination with creatine and ascorbic acid. For the in vivo studies, tumors were induced by injecting Sarcoma-180 cells (2 × 10(6) cells/mouse) in the left hind leg. After 7 days of tumor inoculation, treatments were started with MG (20 mg/kg body wt/day, via the intravenous route), with or without creatine (150 mg/kg body wt/day, fed orally) and ascorbic acid (50 mg/kg body wt/day, fed orally) and continued for 10 consecutive days. Significant regression of tumor size was observed when Sarcoma-180 tumor-bearing mice were treated with MG and even more so with the aforesaid combination. The creatine-supplemented group demonstrated better overall survival in comparison with tumor-bearing mice without creatine. In conclusion, it may be stated that the anti-cancer effect of MG is enhanced by concomitant creatine supplementation, both in chemically transformed (by 3MC) muscle cells in vitro as well as in sarcoma animal model in vivo. These data strongly suggest that creatine supplementation may gain importance as a safe and effective supplement in therapeutic intervention with the anti-cancer agent MG.


Subject(s)
Creatine/pharmacology , Neoplasms, Experimental/drug therapy , Pyruvaldehyde/pharmacology , Animals , Humans , MCF-7 Cells , Methylcholanthrene/toxicity , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology
5.
Int J Nanomedicine ; 10: 3499-518, 2015.
Article in English | MEDLINE | ID: mdl-25999714

ABSTRACT

PURPOSE: The normal metabolite methylglyoxal (MG) specifically kills cancer cells by inhibiting glycolysis and mitochondrial respiration without much adverse effect upon normal cells. Though the anticancer property of MG is well documented, its gradual enzymatic degradation in vivo has prompted interest in developing a nanoparticulate drug delivery system to protect it and also to enhance its efficacy. MATERIALS AND METHODS: MG-conjugated chitosan nanoparticles (Nano-MG) were prepared by conjugating the carbonyl group of MG with the amino group of chitosan polymer (Schiff's base formation). Nano-MG were characterized in detail using the dynamic light scattering method, zeta potential measurement, Fourier transform infrared spectroscopy, and transmission electron microscopic analysis. Amount of MG anchored to Nano-MG, stability of Nano-MG, and in vitro release of MG from Nano-MG were estimated spectrophotometrically. Ehrlich ascites carcinoma (EAC) cells, human breast cancer cell line HBL-100, and lung epithelial adenocarcinoma cell line A549 were used as test systems to compare Nano-MG with bare MG in vitro. Cytotoxicity to EAC cells was evaluated by the trypan blue dye exclusion test, and cell viability of HBL-100 and A549 cells were studied using 3-(4,5-dimethylthiazol-2-yl) 2,5-diphenyltetrazolium bromide (MTT) assay. Apoptosis of HBL-100 cells was assessed by flow cytometry and confocal microscopy. In vivo studies were performed on both EAC cells inoculated and also in sarcoma-180-induced solid tumor-bearing Swiss albino mice to assess the anticancer activity of Nano-MG in comparison to bare MG with varying doses, times, and administrative routes. RESULTS: Fourier transform infrared spectroscopy revealed the presence of imine groups in Nano-MG due to conjugation of the amino group of chitosan and carbonyl group of MG with diameters of nanoparticles ranging from 50-100 nm. The zeta potential of Nano-MG was +21 mV and they contained approximately 100 µg of MG in 1 mL of solution. In vitro studies with Nano-MG showed higher cytotoxicity and enhanced rate of apoptosis in the HBL-100 cell line in comparison with bare MG, but no detrimental effect on normal mouse myoblast cell line C2C12 at the concerned doses. Studies with EAC cells also showed increased cell death of nearly 1.5 times. Nano-MG had similar cytotoxic effects on A549 cells. In vivo studies further demonstrated the efficacy of Nano-MG over bare MG and found them to be about 400 times more potent in EAC-bearing mice and nearly 80 times more effective in sarcoma-180-bearing mice. Administration of ascorbic acid and creatine during in vivo treatments augmented the anticancer effect of Nano-MG. CONCLUSION: The results clearly indicate that Nano-MG may constitute a promising tool in anticancer therapeutics in the near future.


Subject(s)
Antineoplastic Agents , Chitosan , Nanoparticles , Pyruvaldehyde , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Chitosan/chemistry , Chitosan/toxicity , Humans , Mice , Nanoparticles/chemistry , Nanoparticles/toxicity , Nanotechnology , Pyruvaldehyde/chemistry , Pyruvaldehyde/toxicity
6.
Cell Immunol ; 287(1): 27-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24368179

ABSTRACT

Methylglyoxal (MG), the potent anticancer agent has been conjugated to a nontoxic, biocompatible polymer, chitosan, to protect it from in vivo enzymatic degradation. This polymeric complex, 'Nano-MG' shows remarkable antitumor property and elicits macrophage-mediated immunity in tumor bearing mice on intravenous (0.4 mg/kg body wt/day) treatment more efficiently than MG (20mg/kg body wt/day). These activated macrophages appear more in numbers in the peritoneum and produce more superoxide and nitrite. Moreover, immunomodulatory cytokines and surface receptors of these macrophages like iNOS, IFN-γ, TNF-α, IL-1ß, IL-6, M-CSF, TLR-4 and TLR-9 also exhibit marked up-regulation in Sarcoma-180 tumor bearing mice after Nano-MG treatment compared to untreated tumor bearing counterpart. Hence, Nano-MG acts as an immunostimulant in tumor bearing mice to combat cancer at conspicuously lower dose, probably due to its longer circulation time in blood.


Subject(s)
Chitosan/administration & dosage , Macrophages/drug effects , Nanoparticles/administration & dosage , Pyruvaldehyde/administration & dosage , Sarcoma/therapy , Animals , Cell Line, Tumor , Chitosan/chemistry , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Immunomodulation , Macrophages/immunology , Mice , Nanoparticles/chemistry , Neoplasm Transplantation , Nitrites/metabolism , Pyruvaldehyde/chemistry , Sarcoma/immunology , Superoxides/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
7.
Mol Immunol ; 46(10): 2039-44, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19375802

ABSTRACT

Methylglyoxal profoundly stimulates host's immune response against tumor cell by producing reactive oxygen intermediates (ROI's) and reactive nitrogen intermediates (RNI's) [Bhattacharyya, N., Pal, A., Patra, S., Haldar, A.K., Roy, S., Ray, M., 2008. Activation of macrophages and lymphocytes by methylglyoxal against tumor cells in the host. Int. Immunophar. 8 (11), 1503-1512]. Present study indicated that methylglyoxal stimulates iNOS activation by p38 MAPK-NF-kappa beta dependent pathway and ROS production by ERK and JNK activation in sarcoma-180 tumor bearing mice. Proinflammatory cytokines, for macrophage activation, IL-6 and IL-1 beta were also increased. Production of TLR 4 and TLR 9, which acts through the same signaling pathway, were also upregulated. Hence, concluded that methylglyoxal augmented the IL-6 and IL-1 beta, expression of TLR 4 and TLR 9 and produced MAPKs, important regulators of ROIs and RNIs. Methylglyoxal treatment also increased M-CSF, an upregulator of macrophage production. CD8 and CD4 molecules, associated with T(C) and T(H) cells respectively, were also increased. Overall methylglyoxal treatment is important for enhancement of macrophages and lymphocyte activation or immunomodulation against sarcoma-180 tumor.


Subject(s)
MAP Kinase Signaling System/drug effects , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , NF-kappa B/metabolism , Pyruvaldehyde/pharmacology , Sarcoma 180/immunology , T-Lymphocytes/drug effects , Animals , Biomarkers , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Membrane/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Protein Transport/drug effects , Sarcoma 180/enzymology , Sarcoma 180/pathology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Up-Regulation/drug effects
8.
Int Immunopharmacol ; 8(11): 1503-12, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18617020

ABSTRACT

Methylglyoxal is a normal metabolite and has the potential to affect a wide variety of cellular processes. In particular, it can act selectively against malignant cells. The study described herein was to investigate whether methylglyoxal can enhance the non-specific immunity of the host against tumor cells. Methylglyoxal increased the number of macrophages in the peritoneal cavity of both normal and tumor-bearing mice. It also elevated the phagocytic capacity of macrophages in both these groups of animals. This activation of macrophages was brought about by increased production of Reactive Oxygen Intermediates (ROIs) and Reactive Nitrogen Intermediates (RNIs). The possible mechanism for the production of ROIs and RNIs can be attributed to stimulation of the respiratory burst enzyme NADPH oxidase and iNOS, respectively. IFN-gamma, which is a regulatory molecule of iNOS pathway also showed an elevated level by methylglyoxal. TNF-alpha, which is an important cytokine for oxygen independent killing by macrophage also increased by methylglyoxal in both tumor-bearing and non tumor-bearing animals. Methylglyoxal also played a role in the proliferation and cytotoxicity of splenic lymphocytes. In short, it can be concluded that methylglyoxal profoundly stimulates the immune system against tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Lymphocytes/drug effects , Macrophages, Peritoneal/drug effects , Pyruvaldehyde/pharmacology , Sarcoma 180/immunology , Animals , Cell Proliferation/drug effects , Concanavalin A/pharmacology , Cytotoxicity, Immunologic/drug effects , Female , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Lymphocyte Activation/drug effects , Lymphocytes/immunology , Macrophage Activation/drug effects , Macrophages, Peritoneal/immunology , Mice , NADP/analysis , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , Nitrites/immunology , Nitrites/metabolism , Superoxides/immunology , Superoxides/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...