Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Sci Adv ; 8(17): eabm9898, 2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35486721

ABSTRACT

The default mode network (DMN) of the brain is functionally associated with a wide range of behaviors. In this study, we used functional magnetic resonance imaging (fMRI), positron emission tomography (PET), and spectral fiber photometry to investigate the selective neuromodulatory effect of norepinephrine (NE)-releasing noradrenergic neurons in the locus coeruleus (LC) on the mouse DMN. Chemogenetic-induced tonic LC activity decreased cerebral blood volume (CBV) and glucose uptake and increased synchronous low-frequency fMRI activity within the frontal cortices of the DMN. Fiber photometry results corroborated these findings, showing that LC-NE activation induced NE release, enhanced calcium-weighted neuronal spiking, and reduced CBV in the anterior cingulate cortex. These data suggest that LC-NE alters conventional coupling between neuronal activity and CBV in the frontal DMN. We also demonstrated that chemogenetic activation of LC-NE neurons strengthened functional connectivity within the frontal DMN, and this effect was causally mediated by reduced modulatory inputs from retrosplenial and hippocampal regions to the association cortices of the DMN.

2.
Neuroimage ; 238: 118213, 2021 09.
Article in English | MEDLINE | ID: mdl-34116153

ABSTRACT

Superparamagnetic iron-oxide nanoparticles are robust contrast agents for magnetic resonance imaging (MRI) used for sensitive structural and functional mapping of the cerebral blood volume (CBV) when administered intravenously. To date, many CBV-MRI studies are conducted with Feraheme, manufactured for the clinical treatment of iron-deficiency. Unfortunately, Feraheme is currently not available outside the United States due to commercial and regulatory constraints, making CBV-MRI methods either inaccessible or very costly to achieve. To address this barrier, we developed a simple, one-pot recipe to synthesize Carboxymethyl-dextran coated Iron Oxide Nanoparticles, namely, "CION", suitable for preclinical CBV-MRI applications. Here we disseminate a step-by-step instruction of our one-pot synthesis protocol, which allows CION to be produced in laboratories with minimal cost. We also characterized different CION-conjugations by manipulating polymer to metal stoichiometric ratio in terms of their size, surface chemistry, and chemical composition, and shifts in MR relaxivity and pharmacokinetics. We performed several proof-of-concept experiments in vivo, demonstrating the utility of CION for functional and structural MRI applications, including hypercapnic CO2 challenge, visual stimulation, targeted optogenetic stimulation, and microangiography. We also present evidence that CION can serve as a cross-modality research platform by showing concurrent in vivo optical and MRI measurement of CBV using fluorescent-labeled CION. The simplicity and cost-effectiveness of our one-pot synthesis method should allow researchers to reproduce CION and tailor the relaxivity and pharmacokinetics according to their imaging needs. It is our hope that this work makes CBV-MRI more openly available and affordable for a variety of research applications.


Subject(s)
Contrast Media , Dextrans/chemical synthesis , Magnetic Iron Oxide Nanoparticles , Magnetic Resonance Imaging/methods , Humans
3.
Chem Commun (Camb) ; 56(57): 7945-7948, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32531009

ABSTRACT

Two-photon active mitochondriotropic lanthanide nanorods for high resolution fluorescence imaging. The presence of Gd in the nanorods also enabled us to utilize this material as a T1-T2 dual-mode contrast reagent for recording magnetic resonance images of the mouse brain.


Subject(s)
Brain/diagnostic imaging , Lanthanoid Series Elements/chemistry , Magnetic Resonance Imaging , Mitochondria/chemistry , Multimodal Imaging , Nanotubes/chemistry , Animals , Mice , Mice, Inbred C57BL , Photons
4.
Mol Psychiatry ; 24(5): 710-725, 2019 05.
Article in English | MEDLINE | ID: mdl-30214043

ABSTRACT

Noradrenergic signaling plays a well-established role in promoting the stress response. Here we identify a subpopulation of noradrenergic neurons, defined by developmental expression of Hoxb1, that has a unique role in modulating stress-related behavior. Using an intersectional chemogenetic strategy, in combination with behavioral and physiological analyses, we show that activation of Hoxb1-noradrenergic (Hoxb1-NE) neurons decreases anxiety-like behavior and promotes an active coping strategy in response to acute stressors. In addition, we use cerebral blood volume-weighted functional magnetic resonance imaging to show that chemoactivation of Hoxb1-NE neurons results in reduced activity in stress-related brain regions, including the bed nucleus of the stria terminalis, amygdala, and locus coeruleus. Thus, the actions of Hoxb1-NE neurons are distinct from the well-documented functions of the locus coeruleus in promoting the stress response, demonstrating that the noradrenergic system contains multiple functionally distinct subpopulations.


Subject(s)
Adrenergic Neurons/physiology , Homeodomain Proteins/genetics , Stress, Physiological/genetics , Adaptation, Psychological/physiology , Adrenergic Neurons/metabolism , Amygdala/metabolism , Animals , Anxiety/genetics , Anxiety/metabolism , Behavior, Animal/physiology , Brain/metabolism , Female , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism
5.
Neuropsychopharmacology ; 42(3): 615-627, 2017 02.
Article in English | MEDLINE | ID: mdl-27515791

ABSTRACT

Several neuropsychiatric conditions, such as addiction and schizophrenia, may arise in part from dysregulated activity of ventral tegmental area dopaminergic (THVTA) neurons, as well as from more global maladaptation in neurocircuit function. However, whether THVTA activity affects large-scale brain-wide function remains unknown. Here we selectively activated THVTA neurons in transgenic rats and measured resulting changes in whole-brain activity using stimulus-evoked functional magnetic resonance imaging. Applying a standard generalized linear model analysis approach, our results indicate that selective optogenetic stimulation of THVTA neurons enhanced cerebral blood volume signals in striatal target regions in a dopamine receptor-dependent manner. However, brain-wide voxel-based principal component analysis of the same data set revealed that dopaminergic modulation activates several additional anatomically distinct regions throughout the brain, not typically associated with dopamine release events. Furthermore, explicit pairing of THVTA neuronal activation with a forepaw stimulus of a particular frequency expanded the sensory representation of that stimulus, not exclusively within the somatosensory cortices, but brain-wide. These data suggest that modulation of THVTA neurons can impact brain dynamics across many distributed anatomically distinct regions, even those that receive little to no direct THVTA input.


Subject(s)
Benzazepines/pharmacology , Brain/physiology , Cerebrovascular Circulation/physiology , Dopamine/metabolism , Dopaminergic Neurons/physiology , Functional Neuroimaging/methods , Receptors, Dopamine D1/antagonists & inhibitors , Ventral Tegmental Area/physiology , Animals , Benzazepines/administration & dosage , Brain/diagnostic imaging , Brain/drug effects , Cerebrovascular Circulation/drug effects , Magnetic Resonance Imaging/methods , Male , Rats , Rats, Long-Evans , Ventral Tegmental Area/diagnostic imaging , Ventral Tegmental Area/drug effects
7.
Bioconjug Chem ; 25(3): 501-9, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24506698

ABSTRACT

The present study reports the synthesis, characterization, and biological evaluation of a novel macromolecular bipill, synthesized by appending two different anticancer agents, viz., gemcitabine (GEM) and methotrexate (MTX), to the distal ends of a long-circulating poly(ethylene glycol) (PEG) spacer. Covalent conjugation of GEM and MTX via PEG linker not only transformed the solubility profiles of constituent drug molecules, but significantly improved their stability in the presence of plasma. In vitro cytotoxicity studies confirmed that GEM-PEG-MTX exerts higher cytotoxicity (IC50 0.181 µM at 24 h) in human breast adenocarcinoma MCF-7 cell lines, when compared to free drug congeners, i.e., free GEM (IC50 0.294 µM at 24 h) and free MTX (IC50 0.591 µM at 24 h). Tumor growth inhibition studies in chemically induced breast cancer bearing rats established the superiority of GEM-PEG-MTX conjugate over all other pharmaceutical preparations including free drugs, physical mixture of GEM and MTX, and PEGylated GEM/MTX. Toxicity studies in tumor bearing rats as well as healthy mice corroborated that dual drug conjugation is an effective means to synergize the therapeutic indices of potential drug candidates while alleviating drug-associated side effects.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Mammary Neoplasms, Experimental/drug therapy , Methotrexate/pharmacology , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Deoxycytidine/chemical synthesis , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , MCF-7 Cells , Macromolecular Substances/chemical synthesis , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Mammary Neoplasms, Experimental/pathology , Methotrexate/chemical synthesis , Methotrexate/chemistry , Mice , Molecular Structure , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Rats , Rats, Sprague-Dawley , Risk Factors , Structure-Activity Relationship , Gemcitabine
9.
Mol Pharm ; 10(9): 3404-16, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23905512

ABSTRACT

Cancer cell-selective, nuclear targeting is expected to enhance the therapeutic efficacy of a myriad of antineoplastic drugs, particularly those whose pharmacodynamic site of action is the nucleus. In this study, a steroid-macromolecular bioconjugate based on PEG-linked 17ß-Estradiol (E2) was appended to intrinsically cell-penetrable multiwalled carbon nanotubes (MWCNTs) for intranuclear drug delivery and effective breast cancer treatment, both in vitro and in vivo. Taking Doxorubicin (DOX) as a model anticancer agent, we tried to elucidate how E2 appendage influences the cell internalization, intracellular trafficking, and antitumor efficacy of the supramolecularly complexed drug. We observed that the combination of DOX with E2-PEG-MWCNTs not only facilitated nuclear targeting through an estrogen receptor (ER)-mediated pathway but also deciphered to a synergistic anticancer response in vivo. The antitumor efficacy of DOX@E2-PEG-MWCNTs in chemically breast cancer-induced female rats was approximately 18, 17, 5, and 2 times higher compared to the groups exposed to saline, drug-deprived E2-PEG-MWCNTs, free DOX, and DOX@m-PEG-MWCNTs, respectively. While free DOX treatment induced severe cardiotoxicity in animals, animals treated with DOX@m-PEG-MWCNTs and DOX@E2-PEG-MWCNTs were devoid of any perceivable cardiotoxicity, hepatotoxicity, and nephrotoxicity. To the best of our knowledge, this is the first instance in which cancer cell-selective, intranuclear drug delivery, and, subsequently, effective in vivo breast cancer therapy has been achieved using estrogen-appended MWCNTs as the molecular transporter.


Subject(s)
Estradiol/chemistry , Nanotubes, Carbon/chemistry , Ritonavir/chemistry , Calorimetry, Differential Scanning , Drug Delivery Systems/methods , Molecular Structure , Polymers/chemistry , Spectrometry, Fluorescence
10.
Mol Pharm ; 10(7): 2543-57, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23683251

ABSTRACT

The present study reports the design, synthesis, and biological evaluation of a novel, intravenously injectable, theranostic prodrug based on multiwalled carbon nanotubes (MWCNTs) concomitantly decorated with a fluorochrome (Alexa-fluor, AF488/647), radionucleide (Technitium-99m), tumor-targeting module (folic acid, FA), and anticancer agent (methotrexate, MTX). Specifically, MTX was conjugated to MWCNTs via a serum-stable yet intracellularly hydrolyzable ester linkage to ensure minimum drug loss in circulation. Cell uptake studies corroborated the selective internalization of AF-FA-MTX-MWCNTs (1) by folate receptor (FR) positive human lung (A549) and breast (MCF 7) cancer cells through FR mediated endocytosis. Lysosomal trafficking of 1 enabled the conjugate to exert higher anticancer activity as compared to its nontargeted counterpart that was mainly restricted to cytoplasm. Tumor-specific accumulation of 1 in Ehlrich Ascites Tumor (EAT) xenografted mice was almost 19 and 8.6 times higher than free MTX and FA-deprived MWCNTs. Subsequently, the conjugate 1 was shown to arrest tumor growth more effectively in chemically breast tumor induced rats, when compared to either free MTX or nontargeted controls. Interestingly, the anticancer activities of the ester-linked CNT-MTX conjugates (including the one deprived of FA) were significantly higher than their amide-linked counterpart, suggesting that cleavability of linkers between drug and multifunctional nanotubes critically influence their therapeutic performance. The results were also supported by in silico docking and ligand similarity analysis. Toxicity studies in mice confirmed that all CNT-MTX conjugates were devoid of any perceivable hepatotoxicity, cardiotoxicity, and nephrotoxicity. Overall, the delivery property of MWCNTs, high tumor binding avidity of FA, optical detectability of AF fluorochromes, and radio-traceability of (99m)Tc could be successfully integrated and partitioned on a single CNT-platform to augment the therapeutic efficacy of MTX against FR overexpressing cancer cells while allowing a real-time monitoring of treatment response through multimodal imaging.


Subject(s)
Drug Delivery Systems/methods , Folic Acid/chemistry , Methotrexate/chemistry , Nanomedicine/methods , Nanotubes, Carbon/chemistry , Cell Line, Tumor , Endocytosis/physiology , Humans
11.
Bioconjug Chem ; 24(4): 626-39, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23517108

ABSTRACT

The present study explores the possibility of exploiting surface functionality as one of the key regulators for modulating the intracellular trafficking and therapeutic performance of drug loaded carbon nanotubes (CNTs). In line with that approach, a series of biofunctionalized multiwalled carbon nanotubes (f-CNTs 1-6) decorated with various functional molecules including antifouling polymer (PEG), tumor recognition modules (folic acid/hyaluronic acid/estradiol), and fluorophores (rhodamine B isothiocyanate/Alexa Fluor) were synthesized. By loading different anticancer agents (methotrexate (MTX), doxorubicin (DOX), and paclitaxel (PTX)) onto each functionalized CNT preparation, we tried to elucidate how the surface functional molecules associated with each f-CNT influence their therapeutic potential. We observed that antiproliferative or apoptotic activity of drug-loaded CNTs critically depends on their mechanistic pathway of cellular internalization and intracellular trafficking, which in turn had an intimate rapport with their surface chemistry. To our knowledge, for the first time, we have embarked on the possibility of using a surface chemistry dependent "switch" to remote-control the second and third order targeting of chemotherapeutic agents supramolecularly complexed/adsorbed on CNTs, which in turn is expected to benefit the development of futuristic nanobots for cancer theranostics.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Carriers/chemistry , Nanotubes, Carbon/analysis , Nanotubes, Carbon/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Carriers/metabolism , Drug Screening Assays, Antitumor , HeLa Cells , Humans , MCF-7 Cells , Molecular Structure , Structure-Activity Relationship , Surface Properties , Tumor Cells, Cultured
12.
Bioconjug Chem ; 23(11): 2201-13, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23039830

ABSTRACT

The present study reports the optimized synthesis, physicochemical characterization, and biological evaluation of a novel, multiwalled carbon nanotube-hyaluronic acid (MWCNT-HA) conjugate, complexed with an anticancer agent, Doxorubicin (DOX) via π-π stacking interaction. The therapeutic conjugate was concomitantly labeled with a near-infrared fluorescent dye, Alexa-Flour-647 (AF-647), and radiotracer Technetium-99m ((99m)Tc) to track its whereabouts both in vitro and in vivo via optical and scintigraphic imaging techniques. Covalent functionalization of MWCNTs with HA facilitated their internalization into human lung adenocarcinoma, A549 cells via hyaluronan receptors (HR) mediated endocytosis. Internalized nanotubes showed lysosomal trafficking, followed by low pH-triggered DOX release under endolysosomal conditions. Consequently, DOX-loaded HA-MWCNTs exhibited 3.2 times higher cytotoxicity and increased apoptotic activity than free DOX in equivalent concentrations. Organ distribution studies in Ehlrich ascites tumor (EAT) bearing mice model indicated that tumor specific localization of (99m)Tc-MWCNT-HA-DOX is significantly higher than both free drug and nontargeted MWCNTs. Pharmacodynamic studies in chemically breast-cancer-induced rats showed that the tumor-growth inhibitory effect of HA-MWCNT-DOX was 5 times higher than free DOX in equivalent concentration. DOX delivered through HA-MWCNTs was devoid of any detectable cardiotoxity, hepatotoxicity, or nephrotoxicity. All these promising attributes make HA-MWCNTs a "smart" platform for tumor-targeted delivery of anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems , Hyaluronic Acid/chemistry , Nanotubes, Carbon/chemistry , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carbocyanines/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Doxorubicin/chemical synthesis , Doxorubicin/chemistry , Drug Screening Assays, Antitumor , Female , Fluorescent Dyes/chemistry , Humans , Mice , Molecular Structure , Neoplasms/pathology , Organotechnetium Compounds/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tissue Distribution
13.
Chem Res Toxicol ; 25(10): 2127-37, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22994501

ABSTRACT

The present study investigates the effect of functionalization density on the toxicity and cellular uptake of oxidized multiwalled carbon nanotubes (f-MWCNTs) in vitro. The toxicity of f-MWCNTs at varying degrees of carboxylation was assessed in a murine macrophage RAW 264.7 cell line, a model for liver Kupffer cells. In vitro cytotoxicity of oxidized MWCNTs was directly proportional to their functionalization density. The increased cytotoxicity was associated with a concurrent increase in the number of apoptotic cells and production of reactive nitrogen species (RNS). In contrast, reactive oxygen species (ROS) generation was the highest in the case of pristine MWCNTs and decreased with increased functionalization density. Quantitative cellular uptake studies indicated that endogenous ROS production was independent of the concentration of CNTs internalized by a specific cell population and was directly proportional to their surface hydrophobicity. Mechanistic studies suggested that cellular uptake of CNTs was critically charge-dependent and mediated through scavenger receptors, albeit the involvement of nonscavenger receptor mechanisms at low CNT concentrations and their saturation at the experimental concentration cannot be ruled out. A mathematical model was established to correlate between the cellular uptake of CNTs with their length and zeta potential. In an attempt to correlate the results of in vitro toxicity experiments with those of the in vivo toxicity in the mouse model, we found that the toxicity trends in vitro and in vivo are rather opposing. The apparent anomaly was explained on the basis of different experimental conditions and doses associated with cells under in vivo and in vitro culture conditions.


Subject(s)
Macrophages/cytology , Macrophages/drug effects , Nanotubes, Carbon/chemistry , Nanotubes, Carbon/toxicity , Animals , Apoptosis , Cell Line , Macrophages/metabolism , Mice , Models, Biological , Models, Molecular , Oxidation-Reduction , Phagocytosis , Reactive Oxygen Species/metabolism , Surface Properties
14.
Nanomedicine (Lond) ; 7(9): 1311-37, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22583576

ABSTRACT

AIMS: The present study reports a novel approach for enhancing the oral absorption and hypoglycemic activity of insulin via encapsulation in folate-(FA) coupled polyethylene glycol (PEG)ylated polylactide-co-glycolide (PLGA) nanoparticles (NPs; FA-PEG-PLGA NPs). MATERIALS & METHODS: Insulin-loaded FA-PEG-PLGA NPs (size ∼260 nm; insulin loading ∼6.5% [w/w]; encapsulation efficiency: 87.0 ± 1.92%) were prepared by double-emulsion solvent evaporation method. The bioavailability and hypoglycemic activity of orally administered FA-insulin NPs were studied in diabetic rats. RESULTS & CONCLUSION: FA-PEG-PLGA NPs (50 U/kg) exhibited a twofold increase in the oral bioavailability (double hypoglycemia) without any hypoglycemic shock as compared to subcutaneously administered standard insulin solution. Insulin NPs maintained a continual blood glucose level for 24 h, which, however, was transient (<8 h) in the case of subcutaneous insulin and associated with severe hypoglycemic shock. Overall, we have developed a patient-compliant, oral nanoformulation of insulin, once-daily administration of which would be sufficient to control diabetes for at least 24 h.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Folic Acid/chemistry , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Administration, Oral , Animals , Blood Glucose/analysis , Drug Carriers/chemistry , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Insulin/pharmacokinetics , Insulin/therapeutic use , Male , Nanoparticles/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley
15.
Expert Opin Drug Deliv ; 9(4): 383-402, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22432690

ABSTRACT

INTRODUCTION: In situ gel systems refer to a class of novel delivery vehicles, composed of natural, semisynthetic or synthetic polymers, which present the unique property of sol-gel conversion on receipt of biological stimulus. AREAS COVERED: The present review summarizes the latest developments in in situ gel technology, with regard to ophthalmic drug delivery. Starting with the mechanism of ocular absorption, the review expands on the fabrication of various polymeric in situ gel systems, made up of two or more polymers presenting multi-stimuli sensitivity, coupled with other interesting features, such as bio-adhesion, enhanced penetration or sustained release. Various key issues and challenges in this area have been addressed and critically analyzed. EXPERT OPINION: The advent of in situ gel systems has inaugurated a new transom for 'smart' ocular delivery. By virtue of possessing stimuli-responsive phase transition properties, these systems can easily be administered into the eye, similar to normal eye drops. Their unique gelling properties endow them with special features, such as prolonged retention at the site of administration, followed by sustained drug release. Despite the superiority of these systems as compared with conventional ophthalmic formulations, further investigations are necessary to address the toxicity issues, so as to minimize regulatory hurdles during commercialization.


Subject(s)
Biocompatible Materials/administration & dosage , Delayed-Action Preparations/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Ophthalmic Solutions/administration & dosage , Absorption , Adhesiveness , Administration, Ophthalmic , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Biological Availability , Chemistry, Pharmaceutical , Chitosan/chemistry , Drug Carriers/pharmacokinetics , Excipients/administration & dosage , Excipients/pharmacokinetics , Eye/metabolism , Gels , Glucans/chemistry , Humans , Ophthalmic Solutions/chemistry , Ophthalmic Solutions/pharmacokinetics , Poloxamer/chemistry , Polymers/chemistry , Xylans/chemistry
16.
Nanomedicine (Lond) ; 6(10): 1733-54, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22087800

ABSTRACT

UNLABELLED: The present study aims to develop a multifunctional nanoformulation based on technetium-99m labeled, folate conjugated, methotrexate-loaded human serum albumin nanoparticles (HSA NPs) and explore their potential in cancer theragnostics. MATERIALS & METHODS: Methotrexate-loaded HSA NPs were synthesized by a reverse microemulsion technique, followed by chemical crosslinking with glutaraldehyde. These NPs were conjugated with folic acid (FA) through a hydrophilic polyethylene glycol spacer to render them long-circulatory and augment their tumor-specific localization. The therapeutic conjugate was further radiolabeled with a γ-emitter technetium-99m for real-time monitoring of its blood clearance kinetics and biodistribution through the measurement of blood/organ-associated radioactivity and scintigraphic imaging. RESULTS & CONCLUSION: In vitro cell-uptake and cytotoxicity studies corroborated that FA conjugation enabled these NPs to specifically target and kill folate-receptor overexpressing cancer cells via S phase arrest. Blood clearance kinetics and biodistribution studies clearly indicated that circulation time, as well as tumor-specific localization of methotrexate-loaded HSA nanocarriers, could be significantly augmented upon polyethylene glycolylation and conjugation of FA. Finally, we demonstrated that these targeted HSA NPs inhibited tumor growth more effectively, as compared with the nontargeted controls.


Subject(s)
Drug Carriers/chemical synthesis , Folic Acid/analysis , Methotrexate/pharmacokinetics , Nanoparticles/chemistry , Serum Albumin/analysis , Antimetabolites, Antineoplastic/pharmacokinetics , Apoptosis , Cell Line, Tumor , Cell Survival , Drug Stability , Folic Acid/biosynthesis , HeLa Cells , Humans , Magnetic Resonance Spectroscopy/methods , Methotrexate/analysis , Serum Albumin/biosynthesis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Spectroscopy, Fourier Transform Infrared/methods , Surface Properties , Technetium Tc 99m Aggregated Albumin/chemical synthesis , Technetium Tc 99m Aggregated Albumin/chemistry , Technetium Tc 99m Aggregated Albumin/pharmacokinetics , Tissue Distribution
17.
Chem Res Toxicol ; 24(11): 2028-39, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-21978239

ABSTRACT

Carboxylated carbon nanotubes stand as the most promising nanovectors for biomedical and pharmaceutical applications due to their ease of covalent conjugation with eclectic functional molecules including therapeutic drugs, proteins, and oligonucleotides. In the present study, we attempt to investigate how the toxicity of acid-oxidized multiwalled carbon nanotubes (MWCNTs) can be tweaked by altering their degree of functionalization and correlate the toxicity trend with their biodistribution profile. In line with that rationale, mice were exposed to 10 mg/kg of pristine (p) and acid-oxidized (f) MWCNTs with varying degrees of carboxylation through a single dose of intravenous injection. Thereafter, extensive toxicity studies were carried out to comprehend the short-term (7 day) and long-term (28 day) impact of p- and various f-MWCNT preparations on the physiology of healthy mice. Pristine MWCNTs with a high aspect ratio, surface hydrophobicity, and metallic impurities were found to induce significant hepatotoxicity and oxidative damage in mice, albeit the damage was recovered after 28 days of treatment. Conversely, acid-oxidized carboxylated CNTs with shorter lengths, hydrophilic surfaces, and high aqueous dispersibility proved to be less toxic and more biocompatible than their pristine counterparts. A thorough scrutiny of various biochemical parameters, inflammation indexes, and histopathological examination of liver indicated that toxicity of MWCNTs systematically decreased with the increased functionalization density. The degree of shortening and functionalization achieved by refluxing p-MWCNTs with strong mineral acids for 4 h were sufficient to render the CNTs completely hydrophilic and biocompatible, while inducing minimal hepatic accumulation and inflammation. Quantitative biodistribution studies in mice, intravenously injected with Tc-99m labeled MWCNTs, clearly designated that clearance of CNTs from reticuloendothelial system (RES) organs such as liver, spleen, and lungs was critically functionalization density dependent. Well-individualized MWCNTs with shorter lengths (<500 nm) and higher degrees of oxidation (surface carboxyl density >3 µmol/mg) were not retained in any of the RES organs and rapidly cleared out from the systematic circulation through renal excretion route without inducing any obvious nephrotoxicity. As both p- and f-MWCNT-treated groups were devoid of any obvious nephrotoxicity, CNTs with larger dimensions and lower degrees of functionalization, which fail to clear out from the body via renal excretion route, were thought to be excreted via biliary pathway in faeces.


Subject(s)
Biocompatible Materials/toxicity , Fullerenes/toxicity , Nanotechnology/methods , Nanotubes, Carbon/toxicity , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Fullerenes/chemistry , Fullerenes/pharmacokinetics , Hydrophobic and Hydrophilic Interactions , Injections, Intravenous , Kidney/chemistry , Liver/chemistry , Lung/chemistry , Mice , Nanotubes, Carbon/chemistry , Oxidation-Reduction , Spleen/chemistry , Technetium/analysis , Tissue Distribution
18.
Expert Opin Drug Deliv ; 8(11): 1407-24, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21831007

ABSTRACT

INTRODUCTION: The therapeutic efficacy of perorally administered drugs is often obscured by their poor oral bioavailability (BA) and low metabolic stability in the gastrointestinal tract (GIT). Solid lipid nanoparticles (SLNs) have emerged as potential BA enhancer vehicles for various Class II, III and IV drug molecules. AREA COVERED: This review examines the recent advancements in SLN technology, with regards to oral drug delivery. The discussion critically examines the effect of various key constituents on SLN absorption and their applications in oral drug delivery. The relationship between the complexity of absorption (and various factors involved during absorption, including particle size), stability and the self-emulsifying ability of the lipids used has been explored. EXPERT OPINION: The protective effect of SLNs, coupled with their sustained/controlled release properties, prevents drugs/macromolecules from premature degradation and improves their stability in the GIT. An extensive literature survey reveals that direct peroral administration of SLNs improves the BA of drugs by 2- to 25-fold. Overall, the ease of large-scale production, avoidance of organic solvents and improvement of oral BA make SLNs a potential BA enhancer vehicle for various Class II, III and IV drugs.


Subject(s)
Drug Carriers/pharmacokinetics , Lipids/pharmacokinetics , Nanoparticles/chemistry , Pharmacokinetics , Administration, Oral , Animals , Biological Availability , Cell Membrane Permeability , Chemistry, Pharmaceutical , Drug Carriers/administration & dosage , Drug Stability , Drug-Related Side Effects and Adverse Reactions/prevention & control , Emulsifying Agents/chemistry , Humans , Intestinal Absorption , Lipids/administration & dosage , Lipids/chemistry , Nanoparticles/administration & dosage , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Solubility
19.
Nanomedicine (Lond) ; 6(4): 599-603, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21718173

ABSTRACT

For the seventh time, nanomedicine experts from around the globe congregated in SAS Nagar, Punjab, for the Fourth Winter School on Nanotechnology in Advanced Drug Delivery, organized by the National Institute of Pharmaceutical Education and Research (NIPER), Mohali, India. The program covered almost all the scintillating areas of nanomedicine, including novel nanosystems for oral, ocular and transdermal drug delivery, nanostructured surfaces for medical applications, 'smart' nanobullets for site-specific drug and gene delivery, designer nanoparticles for therapeutic delivery, tissue engineering and nanobiocomposites, cancer nanotherapy, and novel analytical and diagnostic tools. Special emphasis was given to the commercialization of nanomedical products, including issues related to intellectual property and risk management.


Subject(s)
Nanomedicine/methods , Nanostructures/chemistry , Nanotechnology/methods
20.
Biomaterials ; 32(28): 6860-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21704368

ABSTRACT

The present investigation consists in the development and characterization of CoQ10 loaded PLGA nanoparticles (CoQ10-NPs, size < 100 nm) by a scalable emulsion-diffusion-evaporation method. Thermal and crystallinity analysis collectively corroborated that CoQ10 was entrapped into the NPs in amorphous form. The lyophilized CoQ10-NPs were found to be stable for a period of 6 months (at room temperature). In vitro cell culture studies indicated that CoQ10-NPs significantly quenched ROS with nearly 10 fold higher efficacy than free CoQ10. Further, positively charged CoQ10-NPs were localized in two major sources of ROS generation: mitochondria and lysosomes. CoQ10-NPs showed improved oral bioavailability (4.28 times) as compared to free CoQ10. Finally remarkably higher hepatoprotective and anti-inflammatory activity of CoQ10-NPs as compared to free CoQ10 was observed due to mitigation of deleterious effects associated with the generation of free radicals. As elucidated by live noninvasive animal imaging, the higher anti-inflammatory activity of CoQ10-NPs can be attributed to significant accumulation of these NPs in the inflamed tissues.


Subject(s)
Inflammation/drug therapy , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polymers , Reactive Oxygen Species/metabolism , Ubiquinone/analogs & derivatives , Animals , Biological Availability , Cell Line , Female , Freeze Drying , Humans , Materials Testing , Mice , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , Particle Size , Polymers/chemistry , Polymers/pharmacokinetics , Random Allocation , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism , Ubiquinone/chemistry , Ubiquinone/metabolism , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...