Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
MedComm (2020) ; 4(2): e247, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37035134

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus, causes coronavirus disease 2019 (COVID-19) which led to neurological damage and increased mortality worldwide in its second and third waves. It is associated with systemic inflammation, myocardial infarction, neurological illness including ischemic strokes (e.g., cardiac and cerebral ischemia), and even death through multi-organ failure. At the early stage, the virus infects the lung epithelial cells and is slowly transmitted to the other organs including the gastrointestinal tract, blood vessels, kidneys, heart, and brain. The neurological effect of the virus is mainly due to hypoxia-driven reactive oxygen species (ROS) and generated cytokine storm. Internalization of SARS-CoV-2 triggers ROS production and modulation of the immunological cascade which ultimately initiates the hypercoagulable state and vascular thrombosis. Suppression of immunological machinery and inhibition of ROS play an important role in neurological disturbances. So, COVID-19 associated damage to the central nervous system, patients need special care to prevent multi-organ failure at later stages of disease progression. Here in this review, we are selectively discussing these issues and possible antioxidant-based prevention therapies for COVID-19-associated neurological damage that leads to multi-organ failure.

2.
Oncogene ; 41(47): 5061-5075, 2022 11.
Article in English | MEDLINE | ID: mdl-36243803

ABSTRACT

Ubiquitin specific peptidase 7 (USP7) is a deubiquitinating enzyme (DUB) that removes ubiquitin tags from specific target protein substrates in order to alter their degradation rate, sub-cellular localization, interaction, and activity. The induction of apoptosis upon USP7 inhibition is well established in cancer containing wild type p53, which operates through the 'USP7-Mdm2-p53' axis. However, in cancers without functional p53, USP7-dependent apoptosis is induced through many other alternative pathways. Here, we have identified another critical p53 independent path active under USP7 to regulate apoptosis. Proteomics analysis identifies XIAP as a potential target of USP7-dependent deubiquitination. GSEA analysis revealed up-regulation of apoptosis signalling upon USP7 inhibition associated with XIAP down-regulation. Modulation of USP7 expression and activity in multiple cancer cell lines showed that USP7 deubiquitinates XIAP to inhibit apoptosis in a caspase-dependent pathway, and the combinatorial inhibition of USP7 and XIAP induces apoptosis in vitro and in vivo. Immunohistochemical staining revealed that grade-wise accumulation of USP7 correlated with an elevated level of XIAP in glioma tissue. This is the first report on the identification and validation of XIAP as a novel substrate of USP7 and together, they involve in the empowerment of the tumorigenic potential of cancer cells by inhibiting apoptosis.


Subject(s)
Glioma , Tumor Suppressor Protein p53 , Humans , Ubiquitin-Specific Peptidase 7/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin Thiolesterase/metabolism , Apoptosis , Glioma/genetics , Cell Line, Tumor , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
3.
Article in English | MEDLINE | ID: mdl-31637020

ABSTRACT

Glioma and cerebral ischemic stroke are two major events that lead to patient death worldwide. Although these conditions have different physiological incidences, ~10% of ischemic stroke patients develop cerebral cancer, especially glioma, in the postischemic stages. Additionally, the high proliferation, venous thrombosis and hypercoagulability of the glioma mass increase the significant risk of thromboembolism, including ischemic stroke. Surprisingly, these events share several common pathways, viz. hypoxia, cerebral inflammation, angiogenesis, etc., but the proper mechanism behind this co-occurrence has yet to be discovered. The hypercoagulability and presence of the D-dimer level in stroke are different in cancer patients than in the noncancerous population. Other factors such as atherosclerosis and coagulopathy involved in the pathogenesis of stroke are partially responsible for cancer, and the reverse is also partially true. Based on clinical and neurosurgical experience, the neuronal structures and functions in the brain and spine are observed to change after a progressive attack of ischemia that leads to hypoxia and atrophy. The major population of cancer cells cannot survive in an adverse ischemic environment that excludes cancer stem cells (CSCs). Cancer cells in stroke patients have already metastasized, but early-stage cancer patients also suffer stroke for multiple reasons. Therefore, stroke is an early manifestation of cancer. Stroke and cancer share many factors that result in an increased risk of stroke in cancer patients, and vice-versa. The intricate mechanisms for stroke with and without cancer are different. This review summarizes the current clinical reports, pathophysiology, probable causes of co-occurrence, prognoses, and treatment possibilities.

4.
Front Biosci (Landmark Ed) ; 24(3): 435-450, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30468665

ABSTRACT

In cerebral tissues, due to continuous and high metabolic demand, energy is produced exclusively by mitochondrial oxidative phosphorylation (OXPHOS). Obstruction of blood flow leads to cerebral ischemia, hypoxia and decreased cellular ATP production. The reactive oxygen species (ROS) generated as by-product of OXPHOS alter many intracellular signaling pathways and result in damaged cellular components. Under such hypoxic conditions, a key factor known as hypoxia inducible factor 1 (HIF1) is stabilized and activated and such activation induces expression of a defined set of target genes which are required for cell survival and angiogenesis. Reperfusion that follows such ischemia alters signaling pathways which are involved in cellular fate. Here, we will review the role of ROS, HIF-1 alpha and other signaling network in mitochondrial dysfunction and cell fate determination in ischemia-reperfusion models in the brain. We will also address both current and future therapeutic strategies for clinical significance that are being developed for treatment of cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Stroke/metabolism , Animals , Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Free Radical Scavengers/therapeutic use , Humans , Mitochondria/metabolism , Platelet Aggregation Inhibitors/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/physiopathology , Stroke/drug therapy , Stroke/physiopathology
5.
Brain Res ; 1704: 164-173, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30326199

ABSTRACT

Cerebral ischemia-reperfusion (CIR) accelerates the progression of neurodegeneration by causing mitochondrial dysfunction to overproduce reactive oxygen species (ROS). Curcumin shows protective effects against CIR-induced oxidative damage. Free curcumin (FC) is effective at high doses due to its poor bioavailability. Also the blood-brain barrier (BBB) limits the passage of substances from circulation into the cerebral region. Thus, formulation of curcumin within polyethylene glycol (PEG)-ylated polylactide-co-glycolide (PLGA) nanoparticles (NC) was applied orally to aged rats to explore its role against CIR injury. Mitochondrial damage was evaluated. The levels of pro-inflammatory cytokines and components of apoptotic pathway were studied. Unlike FC, NC pre-treatment exerted better neuro-protection by ameliorating ROS-mediated oxidative damage and prevented CIR-induced neuronal apoptosis. Therefore, curcumin incorporated PEGylated PLGA nanoparticles may be used as a suitable delivery vehicle to the brain as they can increase curcumin bioavalability and the released curcumin may confer protection to the neurons against CIR-induced oxidative damage.


Subject(s)
Brain/drug effects , Curcumin/therapeutic use , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Reperfusion Injury/drug therapy , Animals , Apoptosis/drug effects , Brain/metabolism , Curcumin/administration & dosage , Female , Lipid Peroxidation/drug effects , Nanocapsules , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism
6.
Nanomedicine ; 13(8): 2439-2450, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28822845

ABSTRACT

Cerebral ischemia-reperfusion is a classic example of reactive oxygen species (ROS) mediated acute damage to brain. Post-ischemic reperfusion induced oxygen free radicals production causes damage to brain cell mitochondria. Antioxidants like quercetin (Qc) have potentials to manage oxidative stress related pathophysiology. However low oral bioavailability and poor cell membrane permeability restrict its therapeutic efficacy. To overcome these hurdles mitochondria specific delivery of Qc nanocapsules was designed to efficiently counteract cerebral ischemia-reperfusion induced cell death and neurodegeneration in young and aged rats. The orally deliverable quercetin loaded polymeric nanocapsules (N1QC) were made mitochondria specific by using triphenylphosphonium cation as one of the matrix components. N1QC demonstrated higher brain uptake and remarkable mitochondrial localization post cerebral ischemia-reperfusion. This unique controlled mitochondrial delivery of quercetin ameliorated histopathological severity by preserving mitochondrial structural and functional integrity through sequestering ROS thus modulating mitochondrial ROS mediated apoptotic cell death in young and aged rats.


Subject(s)
Antioxidants/administration & dosage , Brain Ischemia/drug therapy , Nanocapsules/chemistry , Neuroprotective Agents/administration & dosage , Organophosphorus Compounds/chemistry , Quercetin/administration & dosage , Terphenyl Compounds/chemistry , Animals , Antioxidants/therapeutic use , Brain/drug effects , Brain/metabolism , Brain Ischemia/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Quercetin/therapeutic use , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism
7.
Int J Nanomedicine ; 12: 4059-4071, 2017.
Article in English | MEDLINE | ID: mdl-28603418

ABSTRACT

The importance of fluoride as a natural and industrial toxicant is recognized worldwide. We evaluated the regulating role and biological effect of vesicular (liposomal and nanoencapsulated) melatonin (N-acetyl-5-methoxytryptamine) for drug delivery and controlled release on the depletion of inflammatory mediators, as well as oxidative damage in sodium fluoride (NaF)-treated lungs and liver. Hepatic and bronchial damage was induced in Swiss albino rats with a single acute ingestion of NaF (48 mg/kg body weight, oral gavage). NaF exposure caused the generation of reactive oxygen species (ROS); upregulation of TNF-α and TGF-ß; decreased activities of antioxidant systems (glutathione, glutathione-S-transferase, superoxide dismutase, catalase), succinate dehydrogenase, membrane microviscosity, and membrane potential; increased activity of lipid peroxidation and nicotinamide adenine dinucleotide hydride oxidase; and increased hepatic and nephrite toxicities (P<0.001) compared to those in normal animals. Charge (-ve/+ve)-specific single liposomal (dicetyl phosphate/stearylamine) and nanoencapsulated melatonin (4.46 mg/kg body weight, intravenous) treatments (2 hours after NaF exposure) significantly (P<0.01/0.001) and maximally (P<0.001) inhibited all alterations developed in NaF-mediated oxidative injuries in rat liver (+ve) and lungs (-ve), demonstrating their strong free radical scavenging, antioxidant and antigenotoxic properties, and vesicular efficiencies of targeting. Overall, these results suggest that nanoencapsulated melatonin might be considered as a more powerful remedial therapy in comparison to liposomes, in terms of its efficacy in regulating NaF-intoxicated oxidative injury.


Subject(s)
Drug Delivery Systems/methods , Liposomes/administration & dosage , Melatonin/administration & dosage , Nanocapsules/administration & dosage , Sodium Fluoride/toxicity , Animals , Antioxidants/metabolism , Catalase/metabolism , Glutathione/metabolism , Glutathione Transferase/metabolism , Lipid Peroxidation/drug effects , Liposomes/chemistry , Liposomes/pharmacology , Liver/drug effects , Liver/metabolism , Lung/drug effects , Male , Melatonin/chemistry , Melatonin/pharmacology , Nanocapsules/chemistry , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/metabolism
8.
Exp Gerontol ; 92: 13-22, 2017 06.
Article in English | MEDLINE | ID: mdl-28285147

ABSTRACT

Cerebral ischemia-reperfusion (CIR) injury occurs as a result of oxygen occlusion in the carotid artery through embolus or thrombus formation or cerebrovascular hemorrhage. The oxygen thrust during reperfusion causes the generation of reactive oxidative species (ROS) which exert a potential threat to neuronal survival. ROS may possibly be arrested by antioxidants. After CIR, extracellular matrix remodeling takes place, which is governed by matrix metalloproteinases (MMPs). Augmentation of lipid per oxidation, perturbation of antioxidant enzyme activities and the loss of pyramidal neuronal cells in rat brain were attributed to CIR injury. Melatonin can readily cross the blood-brain barrier (BBB) to exert protective effects as an antioxidant but it is quickly cleared by the circulating blood. Also melatonin is easily degraded by light and hence is found to be ineffective during daytime. Results of the present study showed that unlike free melatonin (FM), the application of nanocapsulated melatonin (NM) exhibited significantly higher potential even at much lower concentrations to rescue neuronal cells and mitochondria during CIR insult and also restored the activities of antioxidative enzymes and MMPs to their normal levels. Hence, nanoencapsulated melatonin may be considered as a suitable drug delivery system for brain to exert protection against CIR injury.


Subject(s)
Aging/metabolism , Antioxidants/pharmacology , Brain Ischemia/drug therapy , Matrix Metalloproteinases/metabolism , Melatonin/pharmacology , Mitochondria/metabolism , Reperfusion Injury/drug therapy , Animals , Antioxidants/chemistry , Blood-Brain Barrier/metabolism , Disease Models, Animal , Female , Melatonin/chemistry , Mitochondria/drug effects , Nanocapsules/chemistry , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
9.
Curr Alzheimer Res ; 13(12): 1363-1373, 2016.
Article in English | MEDLINE | ID: mdl-27357647

ABSTRACT

Recent evidences suggest that cerebral ischemia-reperfusion insult plays significant role in pathogenic diseases like Alzheimer's disease (AD) and other neurodegenerative diseases. Toxic reactive oxygen species (ROS) generated by induced oxidative stress in the episodes of cerebral ischemia-reperfusion (CIR) plays major role in neurodegeneration. As the prime source of ROS generation, neuronal mitochondria, the cellular energy metabolic centre experience severe damage because of CIR-induced oxidative stress. The process of mitochondrial dysfunction is accelerated by CIR that may pave the pathway for neurodegeneration in AD among aged individuals. Prevention of CIR injury may be a shunt in order to minimize the risk of dementia of Alzheimer's type in aged individuals. The use of chemical antioxidants in CIR is not suitable as the blood- brain barrier (BBB) doesn't allow the entry of molecules from blood circulation into the brain. Thus L-ascorbic acid loaded polylactide nanocapsules were prepared and fed orally to assess the role of nanocapsulated ascorbic acid (NAA) against CIR induced oxidative injury in mitochondrial region of rat brains. Mitochondrial injury was assessed by the extent of lipid peroxidation and in situ antioxidant enzyme status. The levels of cytochrome c (cyt c), cyclooxygenase- 2 (COX-2) and iNOS were determined. Results showed that in comparison to free ascorbic acid (AA), NAA exerted better protection to the brain mitochondria by preventing oxidative damage in ROS mediated CIR injury.


Subject(s)
Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Brain Ischemia/drug therapy , Oxidative Stress/drug effects , Reperfusion Injury/drug therapy , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Brain/pathology , Brain/ultrastructure , Brain Ischemia/pathology , Catalase/metabolism , Disease Models, Animal , Female , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Lipid Peroxidation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Nanocapsules/therapeutic use , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
10.
Nanomedicine (Lond) ; 9(15): 2323-37, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24593002

ABSTRACT

AIMS: The aims of our work were to investigate the controlling role and the efficacy of nanocapsulated quercetin drug delivery system on the decrement of inflammatory mediators such as MMP-13 in diethyl nitrosamine (DEN)-induced hepatocarcinogenesis. MATERIALS & METHODS: Hepatocellular carcinoma was developed in the Swiss albino rats by the exposure of DEN. DEN administration caused the generation of reactive oxygen species, upregulation of TNF-α, IL-6, activation of MMP-13, severe oxidative damage, hyperplastic nodules with preneoplastic lesions and the histopathological changes in rat liver. RESULTS & CONCLUSION: Nanocapsulated quercetin treatment restricted all alterations in DEN-mediated development of hepatocarcinogenesis. Therefore, it may be concluded that nanocapsulated quercetin may be accepted as a potent therapeutic formulation in preventing DEN-mediated hepatocarcinogenesis.


Subject(s)
Diethylnitrosamine/toxicity , Down-Regulation/drug effects , Liver Neoplasms, Experimental/prevention & control , Liver/drug effects , Matrix Metalloproteinase 13/metabolism , Nanocapsules , Quercetin/pharmacology , Animals , Liver/enzymology , Liver Neoplasms, Experimental/chemically induced , Male , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Rats
11.
Indian J Med Res ; 137(6): 1163-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23852297

ABSTRACT

BACKGROUND & OBJECTIVES: Malachite green (MG), an environmentally hazardous material, is used as a non permitted food colouring agent, especially in India. Selenium (Se) is an essential nutritional trace element required for animals and humans to guard against oxidative stress induced by xenobiotic compounds of diverse nature. In the present study, the role of the selenium compound diphenylmethyl selenocyanate (DMSE) was assessed on the oxidative stress (OS) induced by a food colouring agent, malachite green (MG) in vivo in mice. METHODS: Swiss albino mice (Mus musculus) were intraperitoneally injected with MG at a standardized dose of 100 µg/ mouse for 30 days. DMSE was given orally at an optimum dose of 3 mg/kg b.w. in pre (15 days) and concomitant treatment schedule throughout the experimental period. The parameters viz. ALT, AST, LPO, GSH, GST, SOD, CAT, GPx, TrxR, CA, MN, MI and DNA damage have been evaluated. RESULTS: The DMSE showed its potential to protect against MG induced hepatotoxicity by controlling the serum alanine aminotransferase and aspartate amino transferase (ALT and AST) levels and also ameliorated oxidative stress by modulating hepatic lipid peroxidation and different detoxifying and antioxidative enzymes such as glutathione-S-transferase (GST), superoxide dismutase (SOD), catalase (CAT), and also the selenoenzymes such as glutathione peroxidase (GPx) and thioredoxin reductase (TrxR) and reduced glutathione level which in turn reduced DNA damage. INTERPRETATION & CONCLUSIONS: The organo-selenium compound DMSE showed significant protection against MG induced heptotoxicity and DNA damage in murine model. Better protection was observed in pretreatment group than in the concomitant group. Further studies need to be done to understand the mechanism of action.


Subject(s)
Antioxidants/chemistry , DNA Damage , Organoselenium Compounds/chemistry , Oxidative Stress , Rosaniline Dyes/adverse effects , Administration, Oral , Animals , Catalase/blood , Chromosome Aberrations , Coloring Agents/adverse effects , Comet Assay , Female , Glutathione/metabolism , Glutathione Peroxidase/blood , Lipid Peroxidation , Liver/drug effects , Liver/enzymology , Mice , Micronucleus Tests , Mitotic Index , Superoxide Dismutase/blood , Thiobarbituric Acid Reactive Substances
12.
Methods Mol Biol ; 1028: 221-30, 2013.
Article in English | MEDLINE | ID: mdl-23740123

ABSTRACT

Oxidative stress, due to the generation of reactive oxygen species, is a major factor in cerebral ischemic damage and changes the activities of antioxidant enzymes and substantially influences the aging process. Free chemical antioxidant is almost ineffective to treat brain ischemia as blood-brain barrier exists in between blood and brain interstitial fluid, limiting component to pass from the circulation into cerebral region. Different compounds have been tested in vivo in different vesiculated forms to prevent cerebral ischemia. Nanoparticle-encapsulated drug treatment resulted in a significant protection of the antioxidant enzymes in both young and old rats. Nanocapsulated drug treatment causes a substantial protection against cerebral ischemia-reperfusion-induced oxidative damage to all parts of brain specifically hippocampal regions of all age groups of rat brain.


Subject(s)
Antioxidants/administration & dosage , Brain Ischemia/metabolism , Drug Carriers/administration & dosage , Aging , Animals , Blood-Brain Barrier , Brain/blood supply , Brain/enzymology , Brain/pathology , Brain Ischemia/drug therapy , Carotid Artery Diseases/drug therapy , Carotid Artery Diseases/metabolism , Disease Models, Animal , Lipid Peroxidation , Male , Mitochondria/metabolism , Nanoparticles/administration & dosage , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Reperfusion Injury
13.
PLoS One ; 8(4): e57735, 2013.
Article in English | MEDLINE | ID: mdl-23620721

ABSTRACT

Cerebral stroke is the leading cause of death and permanent disability among elderly people. In both humans and animals, cerebral ischemia damages the nerve cells in vulnerable regions of the brain, viz., hippocampus, cerebral cortex, cerebellum, and hypothalamus. The present study was conducted to evaluate the therapeutic efficacy of nanoencapsulated quercetin (QC) in combating ischemia-reperfusion-induced neuronal damage in young and aged Swiss Albino rats. Cerebral ischemia was induced by occlusion of the common carotid arteries of both young and aged rats followed by reperfusion. Nanoencapsulated quercetin (2.7 mg/kg b wt) was administered to both groups of animals via oral gavage two hours prior to ischemic insults as well as post-operation till day 3. Cerebral ischemia and 30 min consecutive reperfusion caused a substantial increase in lipid peroxidation, decreased antioxidant enzyme activities and tissue osmolality in different brain regions of both groups of animals. It also decreased mitochondrial membrane microviscosity and increased reactive oxygen species (ROS) generation in different brain regions of young and aged rats. Among the brain regions studied, the hippocampus appeared to be the worst affected region showing increased upregulation of iNOS and caspase-3 activity with decreased neuronal count in the CA1 and CA3 subfields of both young and aged rats. Furthermore, three days of continuous reperfusion after ischemia caused massive damage to neuronal cells. However, it was observed that oral treatment of nanoencapsulated quercetin (2.7 mg/kg b wt) resulted in downregulation of iNOS and caspase-3 activities and improved neuronal count in the hippocampal subfields even 3 days after reperfusion. Moreover, the nanoformulation imparted a significant level of protection in the antioxidant status in different brain regions, thus contributing to a better understanding of the given pathophysiological processes causing ischemic neuronal damage.


Subject(s)
Aging/pathology , Brain Ischemia/drug therapy , Nanocapsules/therapeutic use , Neurons/pathology , Neuroprotective Agents/therapeutic use , Quercetin/therapeutic use , Reperfusion Injury/drug therapy , Animals , Antioxidants/metabolism , Brain Ischemia/complications , Brain Ischemia/enzymology , Brain Ischemia/pathology , Caspase 3/metabolism , Cell Count , Glutathione/metabolism , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Male , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Nanocapsules/ultrastructure , Neurons/drug effects , Neurons/enzymology , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/metabolism , Osmolar Concentration , Quercetin/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reperfusion Injury/complications , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Viscosity
14.
Chem Biol Interact ; 195(3): 206-14, 2012 Feb 05.
Article in English | MEDLINE | ID: mdl-22197969

ABSTRACT

Toxic outcome of chemical therapeutics as well as multidrug resistance are two serious phenomena for their inacceptance in cancer chemotherapy. Antioxidants like curcumin (Cur) have gained immense importance for their excellent anticarcinogenic activities and minimum toxic manifestations in biological system. However, Cur is lipophilic and thus following oral administration hardly appears in blood indicating its potential therapeutic challenge in cancer therapy. Nanocapsulated Cur has been used as a drug delivery vector to focus the effectiveness of these vesicles against hepatocellular carcinoma. The theme of work was to evaluate effectiveness in oral route of polylactide co-glycolide (PLGA) Nanocapsulated curcumin (Nano Cur) against diethylnitrosamine (DEN) induced hepatocellular carcinoma (HCC) in rat. Nano Cur of average diameter 14nm and encapsulation efficiency of 78% were prepared. Fourier Transform Infra Red (FTIR) analysis revealed that there is no chemical interaction between drug and the polymer. Three i.p. injections of the chemical hepatocarcinogen DEN at 15days interval causes hepatotoxicity, the generation of reactive oxygen species (ROS), lipid peroxidation, decrease in plasma membrane microviscosity and depletion of antioxidant enzyme levels in liver. Nano Cur (weekly oral treatment for 16weeks at 20mg/kg b.wt) in DEN induced HCC rats exerted significant protection against HCC and restored redox homeostasis in liver cells. Nanocapsulated Cur caused cancer cell apoptosis as visualized by ApoBrdU analysis. Histopathological analysis confirmed the pathological improvement in the liver. Nano Cur was found to be a potential formulation in oral route in combating the oxidative damage of hepatic cells and eliminating DEN induced hepatocellular cancer cells in rat whereas identical amount of free Cur treatment was found almost ineffective.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Liver Neoplasms, Experimental/prevention & control , Nanocapsules/administration & dosage , Animals , Apoptosis/drug effects , Blotting, Western , Diethylnitrosamine , Glutathione/metabolism , Histocytochemistry , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Microscopy, Atomic Force , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Random Allocation , Rats , Reactive Oxygen Species/metabolism , Spectroscopy, Fourier Transform Infrared , Superoxide Dismutase/analysis
15.
Eur J Cancer Prev ; 21(1): 32-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21968689

ABSTRACT

Hepatocellular carcinoma is the most common primary hepatic malignancy worldwide. N-Nitroso compounds act as strong carcinogens in various animals, including primates. Diethylnitrosamine (DEN) is a well known carcinogenic substance, which induces hepatic carcinoma. The theme of the study was to evaluate the therapeutic efficacy of nanoencapsulated flavonoidal quercetin (3,5,7,3',4'-pentahydroxy flavone, QC) in combating DEN-induced hepatocarcinogenesis in rats. DEN induced a substantial increase in relative liver weights with proliferation and development of hyperplastic nodules. A significant increase in hepatocellular and nephrotoxicity indicated by serum alkaline phosphatase, aspartate transaminase, alanine transaminase, urea, and creatinine was observed in DEN-treated animals. Maximum protection from such toxicity was provided by nanoparticulated QC. Elevated levels of conjugated diene in DEN-treated rats were lowered significantly by nanoparticulated QC. Antioxidant levels in hepatic cells were reduced significantly by the induction of DEN. Nanoparticulated QC was found most potent for complete prevention of DEN-induced reduction in antioxidant levels in the liver. Upregulation of glutathione-S-transferase activity by DEN induction was reduced maximally by nanoencapsulated QC. Nanoencapsulated QC completely protected the mitochondrial membrane of the liver from carcinoma mediated by DEN injection. A significant correlation could be drawn between DEN-induced tissue reactive oxygen species generation and cytochrome C expression in the liver. Nanoencapsulated QC completely prevented the DEN-induced cytochrome C expression in the liver significantly.


Subject(s)
Antioxidants/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Diethylnitrosamine/toxicity , Liver Neoplasms, Experimental/drug therapy , Nanoparticles/chemistry , Nanotechnology , Quercetin/therapeutic use , Alkylating Agents/toxicity , Animals , Blotting, Western , Capsules , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cytochromes c/metabolism , Glutathione/metabolism , Glutathione Transferase/metabolism , Lipid Peroxidation/drug effects , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Mitochondrial Membranes/metabolism , Rats , Rats, Inbred Strains , Reactive Oxygen Species/metabolism
16.
Free Radic Biol Med ; 51(10): 1893-902, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21914470

ABSTRACT

Chronic arsenic exposure causes oxidative stress and mitochondrial dysfunction in the liver and brain. The ideal treatment would be to chelate arsenic and prevent oxidative stress. meso-2,3-Dimercaptosuccinic acid (DMSA) is used to chelate arsenic but its hydrophilicity makes it membrane-impermeative. Conversely, quercetin (QC) is a good antioxidant with limited clinical application because of its hydrophobic nature and limited bioavailability, and it is not possible to solubilize these two compounds in a single nontoxic solvent. Nanocapsules have emerged as a potent drug delivery system and make it feasible to incorporate both hydrophilic and lipophilic compounds. Nanoencapsulated formulations with QC and DMSA either alone or coencapsulated in polylactide-co-glycolide [N(QC+DMSA)] were synthesized to explore their therapeutic application in a rat model of chronic arsenic toxicity. These treatments were compared to administration of quercetin or DMSA alone using conventional delivery methods. Both nanoencapsulated quercetin and nanoencapsulated DMSA were more effective at decreasing oxidative injury in liver or brain compared to conventional delivery methods, but coencapsulation of quercetin and DMSA into nanoparticles had a marked synergistic effect, decreasing liver and brain arsenic levels from 9.5 and 4.8µg/g to 2.2 and 1.5µg/g, respectively. Likewise, administration of coencapsulated quercetin and DMSA virtually normalized changes in mitochondrial function, formation of reactive oxygen species, and liver injury. We conclude that coencapsulation of quercetin and DMSA may provide a more effective therapeutic strategy in the management of arsenic toxicity and also presents a novel way of combining hydrophilic and hydrophobic drugs into a single delivery system.


Subject(s)
Arsenic Poisoning/drug therapy , Arsenic/metabolism , Brain/drug effects , Liver/drug effects , Nanoparticles/therapeutic use , Animals , Antioxidants/administration & dosage , Arsenic Poisoning/pathology , Arsenic Poisoning/physiopathology , Brain/metabolism , Brain/pathology , Chelating Agents/administration & dosage , Disease Models, Animal , Drug Compounding/methods , Drug Delivery Systems , Drug Synergism , Female , Humans , Hydrophobic and Hydrophilic Interactions , Lactic Acid/chemistry , Liver/metabolism , Liver/pathology , Nanoparticles/chemistry , Oxidative Stress/drug effects , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Quercetin/administration & dosage , Rats , Rats, Wistar , Succimer/administration & dosage
17.
Drug Deliv ; 18(6): 451-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21554158

ABSTRACT

CONTEXT: Arsenic is a naturally occurring toxicant that causes acute and chronic adverse health effects, including cancer. OBJECTIVE: The study was performed to evaluate the therapeutic efficacy of liposome entrapped flavonoidal quercetin in combating arsenic toxicity mediated oxidative damage in hepatocytes and brain cells in rat model. MATERIALS AND METHODS: Hepatic and neuronal cell damage in rats was made by daily arsenic (6 mg/kg b wt, 9 mg/kg b wt and 12 mg/kg b wt) treatment via oral route for four consecutive months. Liposomal quercetin (2.71 mg QC/kg b. wt) were injected s.c. on rats treated with 12 mg/kg b. wt. NaAsO(2) twice a week for four months. RESULTS AND DISCUSSION: Inorganic arsenic deposition was found to be most significant in hepatic (9.32 ± 0.100 µg/g tissue) and neuronal (6.21 ± 0.090 µg/g tissue) cells of rats treated with 12 mg/kg b wt of arsenite. Antioxidant levels in hepatic and neuronal cells were reduced significantly by the induction of arsenic. Liposomal quercetin was found most potent for a complete prevention of arsenite-induced reduction in antioxidant levels in the liver and brain of rats. Arsenic induced a substantial increase in hepatic hydroxyproline (HP) and Liposomal quercetin treatment resulted in complete replenishment of the HP level to normal. Liposomal quercetin completely prevented the arsenite-induced upregulation of cytochrome c expression in liver and brain significantly suggesting that the protective effect of Liposomal quercetin could be related to the reduction of arsenic deposition in both the organs. CONCLUSION: Thus, Liposomal quercetin might prove to be of therapeutic potential against arsenite-induced hepatic and neuronal cell damage in rats.


Subject(s)
Brain/drug effects , Liposomes/administration & dosage , Liver/drug effects , Oxidative Stress/drug effects , Quercetin/pharmacology , Animals , Antioxidants/metabolism , Arsenic/toxicity , Brain/metabolism , Brain/pathology , Cytochromes c/metabolism , Female , Liver/metabolism , Liver/pathology , Quercetin/administration & dosage , Rats
18.
Chem Biol Interact ; 186(1): 61-71, 2010 Jun 07.
Article in English | MEDLINE | ID: mdl-20371363

ABSTRACT

Arsenic, the environmental toxicant causes oxidative damage to liver and produces hepatic fibrosis. The theme of our study was to evaluate the therapeutic efficacy of liposomal and nanocapsulated herbal polyphenolic antioxidant quercetin (QC) in combating arsenic induced hepatic oxidative stress, fibrosis associated upregulation of its gene expression and plasma TGF beta (transforming growth factor beta) in rat model. A single dose of arsenic (sodium arsenite-NaAsO(2), 13 mg/kgb.wt) in oral route causes the generation of reactive oxygen species (ROS), arsenic accumulation in liver, hepatotoxicity and decrease in hepatic plasma membrane microviscosity and antioxidant enzyme levels in liver. Arsenic causes fibrosis associated elevation of its gene expression in liver, plasma TGF ss (from normal value 75.2+/-8.67 ng/ml to 196.2+/-12.07 ng/ml) and release of cytochrome c in cytoplasm. Among the two vesicular delivery systems formulated with QC, polylactide nanocapsules showed a promising result compared to liposomal delivery system in controlling arsenic induced alteration of those parameters. A single dose of 0.5 ml of nanocapsulated QC suspension (QC 2.71 mg/kg b.wt) when injected to rats 1h after arsenic administration orally protects liver from arsenic induced deterioration of antioxidant levels as well as oxidative stress associated gene expression of liver. Histopathological examination also confirmed the pathological improvement in liver. Nanocapsulated plant origin flavonoidal compound may be a potent formulation in combating arsenic induced upregulation of gene expression of liver fibrosis through a complete protection against oxidative attack in hepatic cells of rat liver.


Subject(s)
Antioxidants/therapeutic use , Arsenic/adverse effects , Gene Expression/drug effects , Liver Cirrhosis/drug therapy , Liver/pathology , Oxidative Stress/drug effects , Quercetin/therapeutic use , Animals , Antioxidants/administration & dosage , Collagen Type I/metabolism , Cytochromes c/metabolism , Lipid Peroxidation/drug effects , Liposomes/chemistry , Liver/drug effects , Liver Cirrhosis/chemically induced , Male , Nanocapsules/chemistry , Quercetin/administration & dosage , Rats , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/blood
19.
Life Sci ; 84(3-4): 75-80, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19036345

ABSTRACT

AIMS: This study was performed to evaluate the therapeutic efficacy of nanocapsulated flavonoidal quercetin (QC) in combating arsenic-induced reactive oxygen species (ROS)-mediated oxidative damage in hepatocytes and brain cells in a rat model. MAIN METHODS: Hepatic and neuronal cell damage in rats was made by a single injection (sc) of sodium arsenite (NaAsO(2), 13 mg/kg b. wt. in 0.5 ml of physiological saline). A single dose of 500 microl of quercetin suspension (QC) (QC 8.98 micromol/kg) or 500 microl of nanocapsulated QC (NPQC) (QC 8.98 micromol/kg) was given orally to rats at 90 min prior to the arsenite injection. KEY FINDINGS: Inorganic arsenic depositions (182+/-15.6 and 110+/-12.8 ng/g protein) were found in hepatic and neuronal mitochondrial membranes. Antioxidant levels in hepatic and neuronal cells were reduced significantly by arsenic. NPQC prevented the arsenite-induced reduction in antioxidant levels in the liver and brain. Arsenic induced a substantial decrease in liver and brain cell membrane microviscosities, and NPQC treatment resulted in a unique protection against the loss. A significant correlation between mitochondrial arsenic and its conjugated diene level was observed both in liver and brain cells for all experimental rats. SIGNIFICANCE: Arsenic-specific antidotes are used against arsenic-induced toxicity. However, the target site is poorly recognized and therefore achieving an active concentration of drug molecules can be a challenge. Thus, our objective was to formulate NPQC and to investigate its therapeutic potential in an oral route against arsenite-induced hepatic and neuronal cell damage in a rat model.


Subject(s)
Arsenic/toxicity , Arsenites/toxicity , Brain/drug effects , Liver/drug effects , Nanocapsules , Quercetin/administration & dosage , Sodium Compounds/toxicity , Animals , Brain/metabolism , Diet , Female , Glutathione/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Oxidation-Reduction , Rats
20.
Curr Aging Sci ; 1(3): 169-74, 2008 Dec.
Article in English | MEDLINE | ID: mdl-20021389

ABSTRACT

Reactive oxygen species e.g. O(2)(*-), H(2)O(2) and *OH generated by the induction of oxidative stress exert a potential threat on the activity of endogenous antioxidant enzymes and substantially influence the aging process and age-dependant neuropathology. Chemical antioxidant is almost ineffective in protecting neuronal cells from oxidative damage as Blood Brain Barrier exists in between blood and brain interstitial fluid that restricts undegradable influx from the circulation into cerebral region. Quercetin (QC), a flavonoidal antioxidant is known as a potent antioxidant for its polyphenolic configuration. Formulation of QC in polylactide nanocapsule has been done and the efficacy of this vesicular flavonoid has been tested against cerebral ischemia induced oxidative damage in young and old rat brains. Antioxidant potential of QC loaded in nanocapsule (QC 7.2 mmol/kg b.wt., size 50 nm) was investigated by an in vivo model of cerebral ischemia and reperfusion on Sprague Dawley young (2 months, b.wt. 160-180 g) and aged (20 months, b.wt. 415-440 g) rats. Diene level, the index of lipid peroxidation and GSSG/GSH ratio were found to be higher in normal aged, compared to normal young rat brain. Endogenous antioxidants activities were lower in aged rat brain compared to young. Further reduction of these antioxidants were observed in aged rat brain by the induction of cerebral ischemia - reperfusion. Nanocapsule encapsulated QC treatment resulted a significant protection to endogenous antioxidant enzymes against ischemia induced oxidative damage in neuronal cells of young and old rats.


Subject(s)
Aging/metabolism , Antioxidants/administration & dosage , Brain/drug effects , Brain/metabolism , Oxidative Stress/drug effects , Quercetin/administration & dosage , Animals , Blood-Brain Barrier , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Catalase/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Lipid Peroxidation/drug effects , Male , Nanocapsules/chemistry , Polyesters , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...