Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Microencapsul ; 39(2): 95-109, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35147068

ABSTRACT

AIM: Baclofen and Lamotrigine via PLGA nanoparticles were developed for nose-to-brain delivery for the treatment of Neuropathic pain. METHODS: Nanoparticles were prepared using the modified nano-precipitation method. The prepared NPs were characterised and further in vitro and in vivo studies were performed. RESULTS: The Bcf-Ltg-PLGA-NPs were ∼177.7 nm with >75%(w/w) drugs encapsulated. In vitro dissolution studies suggested zero-order release profiles following the Korsmeyer-Peppas model. In vitro cytotoxicity and staining studies on mammalian cells showed dose dependant cytotoxicity where nanoparticles were significantly less toxic (>95% cell-viability). ELISA studies on RAW-macrophages showed Bcf-Ltg-PLGA-NPs as a potential pro-inflammatory-cytokines inhibitor. In vivo gamma-scintigraphy studies on rats showed intra-nasal administration of 99mTc-Bcf-Ltg-PLGA-NPs showed Cmax 3.6%/g at Tmax = 1.5h with DTE% as 191.23% and DTP% = 38.61% in brain. Pharmacodynamics evaluations on C57BL/6J mice showed a significant reduction in licks/bites during inflammation-induced phase II pain. CONCLUSION: The findings concluded that the combination of these drugs into a single nanoparticle-based formulation has potential for pain management.


Baclofen and Lamotrigine loaded PLGA nanoparticles were prepared with a size of 177.7nm, PDI 0.057 and Zeta Potential −15.8 mVIn vitro cell lines based studies showed dose dependant cytotoxicity and Bcf-Ltg-PLGA-NPs were found to be pro-inflammatory cytokines inhibitorsIn vivo Pharmacokinetic studies showed Cmax 3.6%/g at Tmax = 1.5 h with Drug Targeting Efficiency 191.23% and Drug Target Organ Transport 38.61% in the brain for prepared nanoparticlesIn vivo pharmacodynamics studies showed a significant reduction in licks/bites during inflammation-induced phase II pain.


Subject(s)
Nanoparticles , Neuralgia , Animals , Baclofen/therapeutic use , Drug Carriers/therapeutic use , Lamotrigine/therapeutic use , Mammals , Mice , Mice, Inbred C57BL , Neuralgia/drug therapy , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Rats
2.
Toxicol Appl Pharmacol ; 409: 115294, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33069748

ABSTRACT

PP2A, a trimeric Serine/Threonine Protein Phosphatase 2A highly expressed in brain, is a master regulator of cellular functions. Reduction in PP2A activity has been linked to progression of microglial mediated neuroinflammatory diseases. Inflammatory conditions are characterized by increased population of CD86+ve M1 cells and a therapeutic strategy to polarize microglial cells towards CD206+ve M2 cells is the need of hour. In this paper we analyzed A: whether the level of PP2A is altered in CD86+ve cells, B: whether FTY720, a known modulator of PP2A, is able to restore the level of PP2A in inflamed CD86+ve cells. Results revealed that PP2A activity was significantly diminished in inflamed cells but the surprising observation was the cell viability of only 35.99% upon FTY720 treatment in inflamed cells lacking basal PP2A activity. A sharp increase at mRNA level of CD95 and ASK-1 indicated that apoptosis occurred in these cells through CD95/ASK-1/JNK pathway. Importantly, flow cytometric analysis revealed apoptosis of not only CD86+ve cells but also CD206+ve cells. Previous studies have reported that FTY720 polarizes microglial cells towards M2 states; however apoptosis of M2 cells was not studied. As western blot analysis revealed that FTY720 failed to completely restore PP2A, another PP2A modulator, Memantine, was used for co-treatment. Upon co-treatment, the level of PP2A was completely restored and also viability of microglial cells was significantly improved with a significant reduction in apoptosis of M2 cells. These findings suggest that co-treatment strategy may prove beneficial to balance M1/M2 microglial population, thereby improving neuronal functions.


Subject(s)
Cell Survival/physiology , Inflammation/metabolism , Microglia/metabolism , Neuroprotection/physiology , Protein Phosphatase 2/metabolism , Animals , Antigens, CD/metabolism , Apoptosis/physiology , Cells, Cultured , Mice , Neurons/metabolism , Signal Transduction/physiology
3.
Mol Neurobiol ; 60(7): 4049-4063, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37017907

ABSTRACT

Protein phosphatase 2A (PP2A), the activity of which is dictated by the composition of its regulatory subunit, is strongly related to the progression of neurodegenerative disease. The potential role of PP2A on the phenotypic transition of microglial cells under obese conditions is poorly explored. An understanding of the role of PP2A and identification of regulatory subunits contributing to microglial phenotypic transitions in obese condition may serve as a therapeutic target for obesity-associated neurodegeneration. C57BL/6 mice were exposed to obese-associated vascular dementia conditions by performing unilateral common carotid artery occlusion on obese mice of microglial polarization and PP2A activity using flow cytometry, real-time PCR, western blotting, and immunoprecipitation enzymatic assay, followed identifications of PP2A regulatory subunits using LCMS and RT-PCR. Chronic HFD feeding significantly increased the populations of infiltrated macrophages, showing a high percentage of CD86+ in VaD mice, and the expression of pro-inflammatory cytokines, and we observed that PP2A modulates metabolic reprogramming of microglia by regulating OXPHOS/ECAR activity. Using Co-IP and LCMS, we identified the six specific regulatory subunits, namely PPP2R2A, PPP2R2D, PPP2R5B, PPP2R5C, PPP2R5D, and PPP2R5E, that are associated with microglial-activation during obesity-associated-VaD. Interestingly, pharmacological up-regulation of PP2A more significantly suppressed the expression of TNF-alpha than other pro-inflammatory-cytokines and increased the expression of Arginase-1, suggesting that PP2A modulates microglial-phenotypic transitions through TNF-α/Arg-1 axis. Our present findings demonstrate microglial polarization in HFD associated with VaD, and point towards a therapeutic target by providing specific PP2A regulatory-subunits implicated in microglial activation during obesity-related-vascular-dementia.


Subject(s)
Dementia, Vascular , Neurodegenerative Diseases , Mice , Animals , Protein Phosphatase 2/metabolism , Microglia/metabolism , Tumor Necrosis Factor-alpha/metabolism , Dementia, Vascular/metabolism , Neurodegenerative Diseases/metabolism , Mice, Inbred C57BL , Cytokines/metabolism
4.
Int J Biol Macromol ; 191: 548-559, 2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34536476

ABSTRACT

The usefulness of sirolimus (SIR) in the treatment of diseases that involve retinal degeneration like age-related macular degeneration (AMD) has been well documented. However, the problem still remains probably owing to the peculiar environment of the eye and/or unfavourable physiochemical profile of SIR. In the present work, we aimed to fabricate sirolimus loaded PLGA nanoparticles (SIR-PLGA-NP) and chitosan decorated PLGA nanoparticles (SIR-CH-PLGA-NP) to be administered via non-invasive subconjunctival route. Both the nanoparticles were characterized in terms of size, zeta potential, DSC, FTIR and XRD analysis. Quality by Design (QbD) approach was employed during the preparation of nanoparticles and the presence of chitosan coating was confirmed through thermogravimetric analysis and contact angle studies. Cationic polymer modification showed sustained in-vitro SIR release and enhanced ex-vivo scleral permeation and penetration. Further, SIR-CH-PLGA-NP revealed enhanced cellular uptake and thus, reduced lipopolysaccharide (LPS)-induced free-radicals generation by RAW 264.7 cells. The prepared nanoparticles were devoid of residual solvent and were found to be safe in HET-CAM analysis, RBCs damage analysis and histopathology studies. Moreover, high anti-angiogenic potential was observed in SIR-CH-PLGA-NP compared with SIR-PLGA-NP in chorioallantoic membrane (CAM) test. Overall, the current work opens up an avenue for further investigation of CH-PLGA-NP as SIR nanocarrier in the treatment of AMD.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Chitosan/analogs & derivatives , Macular Degeneration/drug therapy , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Sirolimus/administration & dosage , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Chick Embryo , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Macular Degeneration/metabolism , Male , Mice , RAW 264.7 Cells , Rats , Rats, Wistar , Sclera/drug effects , Sclera/metabolism , Sirolimus/pharmacology , Sirolimus/therapeutic use
5.
Drug Deliv Transl Res ; 9(5): 879-890, 2019 10.
Article in English | MEDLINE | ID: mdl-30887226

ABSTRACT

Direct nose-to-brain delivery of drugs and faster onset of action have made intra-nasal route a much sought-after alternative to conventional routes of drug delivery to the brain. Lamotrigine is used for the treatment and management of neuropathic pain, and in the present work, lamotrigine (LTG)-PLGA nanoparticles were developed for intra-nasal delivery. The LTG-PLGA nanoparticles were prepared using modified nanoprecipitation method via high-speed homogenization and ultra-sonication techniques. Entrapment efficiency (EE%) of developed LTG-PLGA-NPs was found to be 84.87 ± 1.2% with drug loading of 10.21 ± 0.89%. The particle size of developed nanoparticles was found to be 184.6 nm with PDI value of 0.082 and zeta potential of - 18.8 mV. Dissolution profiles were studied in PBS (pH 7.4), simulated nasal fluid, and simulated cerebrospinal fluid where almost complete release was observed within 5 h in CSF. In vitro, cytotoxicity was analyzed using MTT assay where dose-dependent cytotoxicity was observed for developed LTG-PLGA-NPs. In vitro cytokine analysis showed positive effects of LTG-PLGA-NPs as pro-inflammatory cytokine suppressors. Further, in vivo studies were performed for radiolabeled formulation and drug (99mTc-LTG-PLGA-NPs and 99mTc-LTG-aqueous) using Sprague Dawley rats where with the help of gamma scintigraphy studies, various routes of administration viz. oral, intra-nasal, and intra-venous were compared. Various pharmacokinetic parameters were evaluated using biodistribution studies to estimate the drug levels in blood and brain. For 99mTc-LTG-PLGA-NPs via intra-nasal route, drug targeting efficiency (DTE%) was found to be 129.81% and drug target organ transport (DTP%) to be 22.81% in brain with Cmax of 3.82%/g within Tmax 1.5 h. Thus, the developed PLGA nanoparticles for intra-nasal delivery provide a possible alternative for existing available drug formulation for neuropathic pain management.


Subject(s)
Anticonvulsants/administration & dosage , Brain/metabolism , Drug Carriers/administration & dosage , Lamotrigine/administration & dosage , Nanoparticles/administration & dosage , Nasal Mucosa/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Administration, Intranasal , Animals , Anticonvulsants/pharmacokinetics , Cell Line, Tumor , Cytokines/metabolism , Drug Carriers/pharmacokinetics , Lamotrigine/pharmacokinetics , Mice , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , RAW 264.7 Cells , Rats, Sprague-Dawley
6.
PLoS One ; 14(10): e0223070, 2019.
Article in English | MEDLINE | ID: mdl-31622373

ABSTRACT

The present study evaluates the effect of flaxseed oil (FXO) supplementation on adipose tissue macrophages (ATM's), E and D series resolvin (Rv) levels and adipose tissue inflammation. Male C57BL/6J mice were divided into five groups (n = 5): lean group (given standard chow diet), HFD group given high fat diet (approx. 18 weeks) till they developed insulin resistance and 4, 8 or 16 mg/kg group (HFD group later orally supplemented with 4, 8 or 16 mg/kg body weight flaxseed oil) for 4 weeks.The present study showed that FXO supplementation led to enhanced DHA, EPA, RvE1-E2, RvD2, RvD5- D6, IL-4, IL-10 and arginase 1 levels in ATMs together with altered immune cell infiltration and reduced NF-κB expression. The FXO supplementation suppresses immune cell infiltration into adipose tissue and alters adipose tissue macrophage phenotype towards the anti-inflammatory state via enhancement of E and D series resolvins, arginase 1 expression and anti-inflammatory cytokines level (IL-4 and IL-10.) leading to amelioration of insulin resistance in flaxseed oil supplemented HFD mice.


Subject(s)
Adipose Tissue/metabolism , Inflammation/diet therapy , Linseed Oil/pharmacology , Obesity/diet therapy , Adipose Tissue/drug effects , Animals , Arginase/metabolism , Chemokine CCL2/metabolism , Cytokines/metabolism , Diet, High-Fat/adverse effects , Dietary Supplements , Disease Models, Animal , Fatty Acids/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/genetics , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Obesity/metabolism , Obesity/pathology
7.
Mol Neurobiol ; 55(2): 1750-1761, 2018 02.
Article in English | MEDLINE | ID: mdl-28224476

ABSTRACT

Protein phosphatase 2A (PP2A), a ubiquitously expressed serine/threonine phosphatase, is a vitally important phosphatase for the cellular system. Structurally, it is constituted of three different subunits, namely catalytic subunit (PP2Ac), structural scaffold subunit (PP2A-A), and regulatory subunit (PP2A-B). All subunits have various isoforms, and catalytic and scaffold subunits are ubiquitously expressed, whereas regulatory subunits are more specific to tissue and cell type. It is the numerous possibilities of PP2A holoenzyme assembly with varying isoform components that make it possess a dual nature of activator or the inhibitory character in different signaling pathways, namely neural developmental pathways, Akt/protein kinase B pathway, NF-kB pathway, MAPK pathway, apoptosis pathway, and cell cycle progression to name a few. Importantly, the expression of PP2A in the brain is highest among the serine phosphatases and is known to actively participate in the neural development process. However, the exact mechanism of action of PP2A is still debated and enunciating the holoenzyme components, especially the regulatory subunit of PP2A involved in regulating neural developmental process is still poorly understood. In this review, we try to throw some light on the involvement of various PP2A holoenzyme forms in the process of neurogenesis and progression of neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases/metabolism , Protein Phosphatase 2/metabolism , Signal Transduction/physiology , Animals , Apoptosis/physiology , Humans , Neurogenesis/physiology , Proteolysis
SELECTION OF CITATIONS
SEARCH DETAIL