Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Dev Biol ; 67(4): 115-135, 2023.
Article in English | MEDLINE | ID: mdl-38334179

ABSTRACT

Drug discovery is an extensive process. From identifying lead compounds to approval for clinical application, it goes through a sequence of labor-intensive in vitro, in vivo preclinical screening and clinical trials. Among thousands of drugs screened only a few get approval for clinical trials. Furthermore, these approved drugs are often discontinued due to systemic toxicity and comorbidity at clinically administered dosages. To overcome these limitations, nanoformulations have emerged as the most sought-after strategy to safely and effectively deliver drugs within tumors at therapeutic concentrations. Most importantly, the employment of suitably variable preclinical models is considered highly critical for the therapeutic evaluation of candidate drugs or their formulations. A review of literature from the past 10 years on antiangiogenic nanoformulations shows the employment of limited types of preclinical models mainly the 2-dimensional (2D) monolayer cell culture and murine models as the mainstay for drug uptake, toxicity and efficiency studies. To top it all, murine models are highly expensive, time-consuming and require expertise in handling them. The current review highlights the utilization of the age-old chicken chorioallantoic membrane (CAM), a well-defined angiogenic model in the investigation of antiangiogenic compounds and nanoformulations in an economic framework. For practical applicability, we have evaluated the CAM model to demonstrate the screening of antiangiogenic compounds and that tumor cells can be implanted onto developing CAM for growing xenografts by recruiting host endothelial and other cellular components. In addition, the exploitation of CAM tumor xenograft models for the evaluation of nanoparticle distribution has also been reinforced by demonstrating that intravenously administered iron oxide nanoparticles (IONPs) passively accumulate and exhibit intracellular as well as extracellular compartment accumulation in highly vascular xenografts. Finally, the ethical considerations, benefits, and drawbacks, of using CAM as an experimental model for testing potential therapeutics are also highlighted.


Subject(s)
Chickens , Neoplasms , Humans , Animals , Mice , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/metabolism , Neoplasms/metabolism , Cell Culture Techniques
2.
ACS Omega ; 7(15): 12509-12523, 2022 Apr 19.
Article in English | MEDLINE | ID: mdl-35474820

ABSTRACT

OBJECTIVE: The elevated choline transporters (ChT), choline kinase (ChK), choline uptake, and phosphorylation in certain tumor cells have influenced the development of radiolabeled choline derivatives as diagnostic probes for imaging cell membrane proliferation. We, therefore, aimed to develop a choline-based moiety for imaging choline kinase-overexpressed tumors by single-photon emission tomography (SPECT). A novel choline-based diagnostic probe was synthesized and evaluated preclinically in various ChT- and ChK-overexpressed tumor models for SPECT imaging applications. METHODS: The synthesis of diethylenetriaminepentaacetic acid-bis-choline ethylamine [DTPA-bis(ChoEA)] featured the conjugation of dimethylaminoethanol to a bifunctional chelator DTPA anhydride. [99mTc]Tc-DTPA-bis(ChoEA) was prepared, and its in vivo characteristics were evaluated in BALB/c mice and tumor-xenografted PC3, A549, and HCT116 athymic mouse models. The in vitro parameters, including cell binding and cytotoxicity, were assessed in PC3, A549, and HCT116 cell lines. To evaluate the specificity of the radioprobe, competitive binding studies were performed. Small-animal SPECT/CT diagnostic imaging was performed for in vivo evaluation. The mouse biodistribution data was further investigated to estimate the radiation dose in humans. RESULTS: In silico studies suggested high binding with enhanced specificity. A standard radiolabeling procedure using stannous chloride as a reducing agent showed a labeling yield of 99.5 ± 0.5%. The in silico studies suggested high binding with enhanced specificity. [99mTc]Tc-DTPA-bis(ChoEA) showed high in vitro stability and specificity. The pharmacokinetic studies of [99mTc]Tc-DTPA-bis(ChoEA) in mice showed an increased tumor-to-background ratio after few minutes of intravenous administration. The first-in-human trial was also conducted. The effective dose was estimated to be 0.00467 mSv/MBq (4.67 mSv/GBq), resulting in a radiation dose of up to 1.73 mSv for the 370 MBq injection of [99mTc]Tc-DTPA-bis(ChoEA). CONCLUSIONS: The synthesized radioprobe [99mTc]Tc-DTPA-bis(ChoEA) accumulates specifically in choline kinase-overexpressed tumors with a high signal-to-noise ratio. The preclinical and first-in-man data suggested that [99mTc]Tc-DTPA-bis(ChoEA) could potentially be used as a diagnostic SPECT tracer in the monitoring and staging of cancer.

3.
Biomaterials ; 120: 115-125, 2017 03.
Article in English | MEDLINE | ID: mdl-28056401

ABSTRACT

Magnetic hyperthermia as a treatment modality is acquiring increased recognition for loco-regional therapy of primary and metastatic lung malignancies by pulmonary delivery of magnetic nanoparticles (MNP). The unique characteristic of magnetic nanoparticles to induce localized hyperthermia in the presence of an alternating magnetic field (AMF) allows for preferential killing of cells at the tumor site. In this study we demonstrate the effect of hyperthermia induced by low and high dose of MNP under the influence of an AMF using 3D tumor tissue analogs (TTA) representing the micrometastatic, perfusion independent stage of triple negative breast cancer (TNBC) that infiltrates the lungs. While application of inhalable magnetic nanocomposite microparticles or magnetic nanocomposites (MnMs) to the micrometastatic TNBC model comprised of TTA generated from cancer and stromal cells, showed no measureable adverse effects in the absence of AMF-exposure, magnetic hyperthermia generated under the influence of an AMF in TTA incubated in a high concentration of MNP (1 mg/mL) caused significant increase in cellular death/damage with mechanical disintegration and release of cell debris indicating the potential of these inhalable composites as a promising approach for thermal treatment of diseased lungs. The novelty and significance of this study lies in the development of methods to evaluate in vitro the application of inhalable composites containing MNPs in thermal therapy using a physiologically relevant metastatic TNBC model representative of the microenvironmental characteristics in secondary lung malignancies.


Subject(s)
Hyperthermia, Induced/methods , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Magnetic Field Therapy/methods , Magnetite Nanoparticles/therapeutic use , Triple Negative Breast Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Survival , Lung Neoplasms/pathology , Mice , Neoplasm Micrometastasis/pathology , Neoplasm Micrometastasis/therapy , Treatment Outcome , Triple Negative Breast Neoplasms/therapy
4.
Oncotarget ; 7(27): 41559-41574, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27223428

ABSTRACT

Currently there are no FDA approved targeted therapies for Triple Negative Breast Cancer (TNBC). Ongoing clinical trials for TNBC have focused primarily on targeting the epithelial cancer cells. However, targeted delivery of cytotoxic payloads to the non-transformed tumor associated-endothelium can prove to be an alternate approach that is currently unexplored. The present study is supported by recent findings on elevated expression of stromal galectin-1 in clinical samples of TNBC and our ongoing findings on stromal targeting of radiation induced galectin-1 by the anginex-conjugated arsenic-cisplatin loaded liposomes using a novel murine tumor model. We demonstrate inhibition of tumor growth and metastasis in response to the multimodal nanotherapeutic strategy using a TNBC model with orthotopic tumors originating from 3D tumor tissue analogs (TTA) comprised of tumor cells, endothelial cells and fibroblasts. The 'rigorous' combined treatment regimen of radiation and targeted liposomes is also shown to be well tolerated. More importantly, the results presented provide a means to exploit clinically relevant radiation dose for concurrent receptor mediated enhanced delivery of chemotherapy while limiting overall toxicity. The proposed study is significant as it falls in line with developing combinatorial therapeutic approaches for stroma-directed tumor targeting using tumor models that have an appropriate representation of the TNBC microenvironment.


Subject(s)
Chemoradiotherapy/methods , Galectin 1/antagonists & inhibitors , Galectin 1/metabolism , Molecular Targeted Therapy/methods , Triple Negative Breast Neoplasms/therapy , Adult , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenic Trioxide , Arsenicals/administration & dosage , Cell Line, Tumor , Cells, Cultured , Cisplatin/administration & dosage , Female , Human Umbilical Vein Endothelial Cells , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Nude , Middle Aged , Neoplasm Metastasis , Oxides/administration & dosage , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , Young Adult
5.
Radiat Res ; 185(2): 169-81, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26836231

ABSTRACT

Lung cancer is the most common cause of cancer-related deaths worldwide and non-small cell lung cancer (NSCLC) accounts for ~85% of all lung cancer. While recent research has shown that cancer stem cells (CSC) exhibit radioresistant and chemoresistant properties, current cancer therapy targets the bulk of the tumor burden without accounting for the CSC and the contribution of the tumor microenvironment. CSC interaction with the stroma enhances NSCLC survival, thus limiting the efficacy of treatment. The aim of this study was to elucidate the role of CSC and the microenvironment in conferring radio- or chemoresistance in an in vitro tumor model for NSCLC. The novel in vitro three-dimensional (3D) NSCLC model of color-coded tumor tissue analogs (TTA) that we have developed is comprised of human lung adenocarcinoma cells, fibroblasts, endothelial cells and NSCLC cancer stem cells maintained in low oxygen conditions (5% O2) to recapitulate the physiologic conditions in tumors. Using this model, we demonstrate that a single 5 Gy radiation dose does not inhibit growth of TTA containing CSC and results in elevated expression of cytokines (TGF-α, RANTES, ENA-78) and factors (vimentin, MMP and TIMP), indicative of an invasive and aggressive phenotype. However, combined treatment of single dose or fractionated doses with cisplatin was found to either attenuate or decrease the proliferative effect that radiation exposure alone had on TTA containing CSC maintained in hypoxic conditions. In summary, we utilized a 3D NSCLC model, which had characteristics of the tumor microenvironment and tumor cell heterogeneity, to elucidate the multifactorial nature of radioresistance in tumors.


Subject(s)
Cytokines/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Tumor Microenvironment/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Humans , Lung Neoplasms/pathology , Neoplastic Stem Cells/pathology , Radiation Tolerance , Radiotherapy Dosage
6.
Sci Rep ; 5: 15236, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26468877

ABSTRACT

Low dose metronomic chemotherapy (LDMC) refers to prolonged administration of low dose chemotherapy designed to minimize toxicity and target the tumor endothelium, causing tumor growth inhibition. Topotecan (TPT) when administered at its maximum tolerated dose (MTD) is often associated with systemic hematological toxicities. Liposomal encapsulation of TPT enhances efficacy by shielding it from systemic clearance, allowing greater uptake and extended tissue exposure in tumors. Extended release of TPT from liposomal formulations also has the potential to mimic metronomic therapies with fewer treatments. Here we investigate potential toxicities of equivalent doses of free and actively loaded liposomal TPT (LTPT) and compare them to a fractionated low dose regimen of free TPT in tumor-endothelial spheroids (TES) with/without radiation exposure for a prolonged period of 10 days. Using confocal microscopy, TPT fluorescence was monitored to determine the accumulation of drug within TES. These studies showed TES, being more reflective of the in vivo tumor microenvironment, were more sensitive to LTPT in comparison to free TPT with radiation. More importantly, the response of TES to low-dose metronomic TPT with radiation was comparable to similar treatment with LTPT. This TES study suggests nanoparticle formulations designed for extended release of drug can simulate LDMC in vivo.


Subject(s)
Liposomes/chemistry , Topotecan/chemistry , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gamma Rays , Immunohistochemistry , Mice , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Topotecan/toxicity
7.
Nanomedicine ; 11(8): 2013-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26282381

ABSTRACT

An appropriate representation of the tumor microenvironment in tumor models can have a pronounced impact on directing combinatorial treatment strategies and cancer nanotherapeutics. The present study develops a novel 3D co-culture spheroid model (3D TNBC) incorporating tumor cells, endothelial cells and fibroblasts as color-coded murine tumor tissue analogs (TTA) to better represent the tumor milieu of triple negative breast cancer in vitro. Implantation of TTA orthotopically in nude mice, resulted in enhanced growth and aggressive metastasis to ectopic sites. Subsequently, the utility of the model is demonstrated for preferential targeting of irradiated tumor endothelial cells via radiation-induced stromal enrichment of galectin-1 using anginex conjugated nanoparticles (nanobins) carrying arsenic trioxide and cisplatin. Demonstration of a multimodal nanotherapeutic system and inclusion of the biological response to radiation using an in vitro/in vivo tumor model incorporating characteristics of tumor microenvironment presents an advance in preclinical evaluation of existing and novel cancer nanotherapies. FROM THE CLINICAL EDITOR: Existing in-vivo tumor models are established by implanting tumor cells into nude mice. Here, the authors described their approach 3D spheres containing tumor cells, enodothelial cells and fibroblasts. This would mimic tumor micro-environment more realistically. This interesting 3D model should reflect more accurately tumor response to various drugs and would enable the design of new treatment modalities.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenicals/therapeutic use , Cisplatin/therapeutic use , Coculture Techniques/methods , Drug Delivery Systems , Oxides/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/radiotherapy , Animals , Antineoplastic Agents/administration & dosage , Arsenic Trioxide , Arsenicals/administration & dosage , Breast/drug effects , Breast/pathology , Breast/radiation effects , Cisplatin/administration & dosage , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Galectin 1/analysis , Mice , Mice, Nude , Nanoparticles/chemistry , Oxides/administration & dosage , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects
8.
Cancer Res ; 74(19): 5359-63, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25095784

ABSTRACT

Advanced technologies and biomaterials developed for tissue engineering and regenerative medicine present tractable biomimetic systems with potential applications for cancer research. Recently, the National Cancer Institute convened a Strategic Workshop to explore the use of tissue biomanufacturing for development of dynamic, physiologically relevant in vitro and ex vivo biomimetic systems to study cancer biology and drug efficacy. The workshop provided a forum to identify current progress, research gaps, and necessary steps to advance the field. Opportunities discussed included development of tumor biomimetic systems with an emphasis on reproducibility and validation of new biomimetic tumor models, as described in this report.


Subject(s)
Biomimetics , Neoplasms/therapy , Tissue Engineering , Humans
9.
Transl Cancer Res ; 2(4): 309-319, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-24634853

ABSTRACT

Recent studies delineate a predominant role for the tumor microenvironment in tumor growth and progression. Improved knowledge of cancer biology and investigation of the complex functional interrelation between the cellular and noncellular compartments of the tumor microenvironment have provided an ideal platform for the evolution of novel cancer nanotherapies. In addition, multifunctional "smart" nanoparticles carrying imaging agents and delivering multiple drugs targeted preferentially to the tumor/tumor microenvironment will lead to early diagnosis and better treatment for patients with cancer. The emerging knowledge of the tumor microenvironment has enabled rational designing of nanoparticles for combinatorial treatment strategies that include radiotherapy, antiangiogenesis and chemotherapy. This multimodality approach is thus expected to achieve therapeutic efficacy and enhance the quality of life of cancer patients. This review highlights the unique characteristics of the tumor microenvironment that are exploited by nanotechnology to develop novel drug delivery systems aimed to target the tumor/tumor microenvironment.

10.
Pharmacol Biochem Behav ; 93(1): 31-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19376155

ABSTRACT

Curcumin is a polyphenol extracted from the rhizome of Curcuma longa and well known as a multi-functional drug with antioxidative, anti-cancerous and anti-inflammatory activities. Curcumin's antiaging and neuroprotective potential is widely reported. In the present study, effect of curcumin treatment dose 30 mg kg(-1) day(-1) was investigated against aluminium neurotoxicity in young and old animals. Direct and indirect intakes of aluminium have been reported to be involved in the etiology of several neurodegenerative disorders like Alzheimer's and Parkinson's diseases. Long term Al was administered through drinking water at a dose of 50 mg/kg/day for 6 months in both young (4 months) and old (18 months) male Wistar rats. Result obtained demonstrates that curcumin treatment attenuates the Al-induced alterations at biochemical, behavioral and ultrastructural levels which was well reflected in the electrophysiological recordings. Our results indicate that curcumin's ability to bind redox active metals and cross the blood-brain barrier could be playing crucial role in preventing against Al-induced neurotoxicity.


Subject(s)
Aluminum/antagonists & inhibitors , Aluminum/toxicity , Curcumin/pharmacology , Nervous System/drug effects , Neuroprotective Agents/pharmacology , Aging/pathology , Aging/physiology , Aging/psychology , Aluminum/administration & dosage , Animals , Behavior, Animal/drug effects , Curcumin/administration & dosage , Electroencephalography , Electrophysiological Phenomena , Lipid Peroxidation/drug effects , Male , Maze Learning/drug effects , Microscopy, Electron, Transmission , Nervous System/pathology , Nervous System/physiopathology , Neuroprotective Agents/administration & dosage , Rats , Rats, Wistar , Somatosensory Cortex/drug effects , Somatosensory Cortex/pathology , Somatosensory Cortex/physiopathology
11.
Neurosci Lett ; 453(2): 86-91, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19356599

ABSTRACT

Traumatic brain injury is a major risk of post-traumatic epilepsy in a large number of individuals of different age groups. Lots of research has been done to elucidate the mechanism of post-traumatic epileptogenesis but age-related vulnerability to develop traumatic seizures is still unknown. Therefore, in the present study investigations were carried out to characterize the electrobehavioral seizure manifestation and associated alterations in young and old epileptic groups. FeCl(3) injection model was used to induce post-traumatic seizures as this model closely resembles human post-traumatic epilepsy. Synchronized video-EEG monitoring was performed to diagnose manifestation of seizures in young (4 months) and old (18 months) rats. Biochemical and ultrastructural studies were performed to determine the mechanism behind the altered age-related vulnerability for post-traumatic seizures. Our result shows that old rats were more vulnerable to post-traumatic epilepsy due to faster seizure spread and lower latency for generalization of electro-clinical seizure activity. The observed biochemical and microscopic alterations associated with old age positively correlate with the altered susceptibility to develop seizures in old epileptic groups.


Subject(s)
Aging , Brain/physiopathology , Epilepsy, Post-Traumatic/pathology , Epilepsy, Post-Traumatic/physiopathology , Seizures/physiopathology , Analysis of Variance , Animals , Behavior, Animal , Brain/metabolism , Disease Models, Animal , Electrodes, Implanted , Electroencephalography , Ferric Compounds , Lipid Peroxidation , Male , Membrane Fluidity , Microscopy, Electron , Neurons/pathology , Neurons/ultrastructure , Protein Kinase C/analysis , Rats , Rats, Wistar , Seizures/chemically induced , Sodium-Potassium-Exchanging ATPase/analysis , Video Recording
12.
Biogerontology ; 10(4): 489-502, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19020987

ABSTRACT

This study investigated the effect of curcumin on aluminium-induced alterations in ageing-related parameters: lipid peroxidation, superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione-s-transferase (GST), protein kinase C (PKC), Na(+), K(+)-adenosine triphosphatase (Na(+), K(+)-ATPase) and acetylcholinesterase (AChE) in the cerebral cortex and hippocampus of the brain of 10- and 24-month-old rats. Measurements taken from aluminium-fed rats were compared with those from rats in which curcumin and aluminium were co-administered. In aluminium-treated rats the levels of lipid peroxidation, PKC and AChE were enhanced while the activities of SOD, GPx, GST and Na(+), K(+)-ATPase were significantly decreased in both the brain regions of both age-groups. In animals co-administered with curcumin and aluminium, the levels of lipid peroxidation, activities of PKC and AChE were significantly lowered while the activities of SOD, GPx, GST and Na(+), K(+)-ATPase were significantly enhanced in the two brain regions studied indicating curcumin's protective effects against aluminium toxicity. Though the magnitudes of curcumin-induced alterations varied in young and old animals, the results of the present study also demonstrated that curcumin exerts a protective effect against aluminium-induced elevation of ageing-related changes by modulating the extent of oxidative stress (by upregulating the activities of antioxidant enzymes) and by regulating the activities of Na(+), K(+) ATPase, PKC and AChE. Therefore, it is suggested that curcumin counters aluminium-induced enhancement in ageing-related processes.


Subject(s)
Aging/metabolism , Aluminum Compounds/toxicity , Antioxidants/pharmacology , Cerebral Cortex/drug effects , Chlorides/toxicity , Curcumin/pharmacology , Hippocampus/drug effects , Oxidative Stress/drug effects , Protein Kinase C/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Acetylcholinesterase/metabolism , Age Factors , Aluminum Chloride , Animals , Cerebral Cortex/enzymology , Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , Hippocampus/enzymology , Lipid Peroxidation/drug effects , Male , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
13.
Epilepsy Behav ; 14(2): 300-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19100339

ABSTRACT

The purpose of the study was to investigate whether dietary intake of curcumin can inhibit the onset and progression of seizures and their associated pathophysiology in experimental FeCl(3)-induced epileptogenesis. Curcumin was considered for this study because it can cross the blood-brain barrier and bind redox-active metal ions. It is also well known for its antioxidative, anticancer, and anti-inflammatory properties. In the present study, seizures were induced by intracortical injection of FeCl(3) into young rats. Synchronized video/EEG recordings were obtained to diagnose the progression of seizures. Short-term treatment with a curcumin-supplemented diet (1500 pp mw/w) significantly inhibited the onset of grade III and IV seizures in rats with iron-induced epilepsy. The lower dose of curcumin (500 ppm) was not effective in inhibiting grade III seizures, but retarded the onset and progression of generalized seizures. The seizure-suppressing potential of curcumin is explained by the observed biochemical, behavioral, and ultrastructural results. Our results indicate that curcumin significantly prevents generalization of electroclinical seizure activity as well as the pathogenesis associated with iron-induced epileptogenesis.


Subject(s)
Antioxidants/therapeutic use , Curcumin/therapeutic use , Epilepsy/chemically induced , Epilepsy/prevention & control , Ferric Compounds , Animals , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Electroencephalography/methods , Epilepsy/pathology , Exploratory Behavior/drug effects , Lipid Peroxidation/drug effects , Male , Maze Learning/drug effects , Membrane Fluidity/drug effects , Microscopy, Electron, Transmission/methods , Protein Carbonylation/drug effects , Protein Carbonylation/physiology , Protein Kinase C/metabolism , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Videotape Recording/methods
14.
Neurotoxicology ; 29(6): 1069-79, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18817812

ABSTRACT

Aluminium (Al) is the most abundant metal known for its neurotoxicity in humans. It gains easy access to the central nervous system under normal physiological conditions and accumulates in different brain regions. It has been reported to be involved in the etiology of several neurodegenerative diseases. In this study, we have investigated the effects of long-term intake of aluminium chloride (AlCl(3)) on the electrophysiological, behavioral, biochemical and histochemical functions of hippocampus. Wistar rats were fed with AlCl(3) at a dose of 50mg/(kgday) for 6 months in the drinking water. Effect of long-term intake of Al was studied on the electrical activity of hippocampal CA1 and CA3 regions in brain of young and old rats. Morris water maze and open field tests were performed to investigate the cognitive and anxiety status of aging rats intoxicated with aluminium. Our studies indicate that aluminium intake results in increased multiple unit activity and adversely affect the spatial learning and memory abilities of both young and old rats. Aluminium intake also inflicts oxidative stress-related damage to lipids, membrane associated proteins (Na-K ATPase and PKC) and endogenous antioxidant enzyme activity (SOD, GPx and GST). The compromised antioxidant system might be playing a crucial role in the observed Al-induced alterations. We have observed that the magnitude of AlCl(3)-induced alteration was considerably higher in younger group of rats compared to older group. In conclusion, the results of the present study implicates that aluminium treatment exerts its neurotoxic effects by altering the overall physiology of brain, and the induced changes were strongly correlated with each other.


Subject(s)
Action Potentials/drug effects , Aging/drug effects , Aluminum Compounds/administration & dosage , Brain Chemistry/drug effects , Chlorides/administration & dosage , Cognition/drug effects , Hippocampus/drug effects , Acetates/metabolism , Aluminum Chloride , Analysis of Variance , Animals , Cell Count/methods , Chlorides/metabolism , Electroencephalography , Exploratory Behavior/drug effects , Lipid Peroxidation/drug effects , Male , Maze Learning/drug effects , Memory/drug effects , Protein Kinase C/metabolism , Rats , Rats, Wistar , Spatial Behavior/drug effects
15.
J Ethnopharmacol ; 111(1): 56-62, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17189676

ABSTRACT

Bacopa monniera is a perennial herb, and is used as a nerve tonic in äyurveda, a traditional medicinal system in India. Aluminium-induced neurotoxicity is well known and different salts of aluminium have been reported to accelerate oxidative damage to biomolecules like lipids, proteins and nucleic acids. The objective of the present study was to investigate whether Bacopa monniera could potentially inhibit aluminium toxicity in the cerebral cortex. Male Wister rats (8 months old) were administered with AlCl(3) orally at a dose of 50mg/kg/day in drinking water for 1 month. Experimental rats were given AlCl(3) along with Bacopa monniera extract at a dose of 40 mg/kg/day. One group of rats was treated with l-deprenyl at a dose of 1mg/kg/day along with AlCl(3) treatment. We have observed that Bacopa monniera prevented accumulation of lipid and protein damage significantly, which resulted from aluminium intake. Decline in the activity of endogenous antioxidant enzymes associated with aluminium administration was also inhibited by Bacopa monniera extract. The potential of Bacopa monniera to inhibit Al-induced oxidative stress was observed to be similar to that of l-deprenyl, which was taken as standard. The potential of Bacopa monniera extract to prevent aluminium neurotoxicity was reflected at the microscopic level as well, indicative of its neuroprotective effects. These findings strongly implicate that Bacopa monniera has potential to protect brain from oxidative damage resulting from aluminium toxicity.


Subject(s)
Antioxidants/pharmacology , Bacopa , Cerebral Cortex/drug effects , Lipid Peroxidation/drug effects , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Oxidative Stress/drug effects , Aldehydes/metabolism , Aluminum Chloride , Aluminum Compounds , Animals , Antioxidants/therapeutic use , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Chlorides , Disease Models, Animal , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , India , Male , Medicine, Ayurvedic , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Plant Extracts/pharmacology , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Selegiline/pharmacology , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...