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










Publication year range
1.
Mol Pharm ; 20(6): 2899-2910, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37116080

ABSTRACT

Oxidative stress, reactive oxygen species generation, and overexpression of VEGF are signatory events in diabetic retinopathy. The downregulation of VEGF and anti-inflammatory action pave the way for diabetic retinopathy (DR) therapy. In that, lower absorption kinetics of melatonin limits its immense therapeutic potential. Hence, we have demonstrated a reverse microemulsion method to synthesize melatonin-loaded polydopamine nanoparticles to replenish both at a single platform with an improved melatonin delivery profile. The study has evaluated in vitro and in vivo protection efficiency of biocompatible melatonin-loaded polydopamine nanoparticles (MPDANPs). The protection mechanism was explained by downregulation of VEGF, CASPASE3, and PKCδ against high-glucose/streptozotocin (STZ)-induced insults, in vitro and in vivo. The anti-inflammatory and antiangiogenic effect and potential of MPDANPs to enhance melatonin in vivo stability with prolonged circulation time have proved MPDANPs as a potential therapeutic candidate in DR management. The DR therapeutic potential of MPDANPs has been arbitrated by improving the bioavailability of melatonin and inhibition of VEGF-PKCδ crosstalk in vivo.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Melatonin , Humans , Diabetic Retinopathy/drug therapy , Melatonin/pharmacology , Melatonin/therapeutic use , Retina , Vascular Endothelial Growth Factor A
2.
ACS Appl Mater Interfaces ; 15(10): 12708-12718, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36857164

ABSTRACT

Transferrin is an iron transporting protein consisting of bilobal protein shells (apotransferrin) with dual domains in each lobe, holding an interdomain iron binding cleft. This cleft is useful in synthesizing an iron oxide core inside the transferrin shell. In vitro reconstitution chemistry provides a nano-dimensional synthesis of the mineral core inside the protein shell. The present study demonstrates the synthesis of magnetotransferrin with reconstitution of apotransferrin to form iron oxide nanoparticles within the transferrin. Transmission electron microscopy investigations along with analysis of electronic diffraction patterns and magnetometry studies indicate entrapment of superparamagnetic iron (III) oxide nanoparticles. In vivo/ex vivo imaging of the brain and immunogold staining of brain sections further validate the brain targeting potential of "magnetotransferrin". The in vivo therapeutic potential of magneto transferrin has been demonstrated by induction of TRPV1 magnetic stimuli protein, having an important regulatory role in Parkinsonism management. In an exploration of neuroprotective mechanisms, deacetylation of H3K27 of synuclein has been revealed through the TRPV1-mediated HDAC3 activation in the treatment of Parkinsonism. Thus, this magnetic protein could be a potent candidate for brain targeting, bio-imaging, and therapy of neurological infirmities.


Subject(s)
Iron , Transferrin , Transferrin/chemistry , Iron/metabolism , Brain/metabolism , Magnetics
3.
Biomater Sci ; 10(18): 5301-5317, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-35917200

ABSTRACT

Glioblastoma (GBM) is a complex brain cancer with frequent relapses and high mortality and still awaits effective treatment. Mitochondria dysfunction is a pathogenic condition in GBM and could be a prime therapeutic target for ceasing GBM progression. Strategies to overcome brain solid tumor barriers and selectively target mitochondria within specific cell types may improve GBM treatment. Here, we present hypericin-conjugated gold nanoparticles (PEG-AuNPs@Hyp) where hypericin is a mitochondrion-targeting agent exhibiting multimodal therapy by critically impacting the IDH2 gene (Isocitrate dehydrogenase) and its interaction with polycomb methyltransferase EZH1/2 for GBM therapy. It significantly localizes in mitochondria by enhanced cellular uptake in the human GBM cell lines/three-dimensional (3D) culture model under red-light exposure. It triggers oxidative stress and changes the mitochondrial potential, with increased Bax/Bcl2 ratio enhancing GBM cell death. The suppressed expression of mutated IDH2 and polycomb group of proteins upon PEG-AuNPs@Hyp/light exposure regulates mitochondria-targeting-mediated GBM metabolism with epigenetic repression of complex machinery function. Polyubiquitination and proteasomal degradation of EZH1 indicate the implication of these polycomb proteins in GBM progression. Chromatin immunoprecipitation reveals the IDH2 and EZH1/EZH2 direct interaction, confirming the role played by IDH2 in modulating the expression of EZH1 and EZH2. In vivo studies further displayed better tumor ablation in a GBM tumor-bearing nude mouse model. The present multimodal nanoformulation compromised the functional dependency of polycomb on mitochondrial IDH2 and established the mechanism of GBM inhibition.


Subject(s)
Brain Neoplasms , Glioblastoma , Isocitrate Dehydrogenase/metabolism , Metal Nanoparticles , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Gold/metabolism , Humans , Mice , Mice, Nude , Mitochondria/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
4.
ACS Biomater Sci Eng ; 8(9): 3810-3818, 2022 Sep 12.
Article in English | MEDLINE | ID: mdl-36005299

ABSTRACT

The imbalance in the bone remodeling process with more bone resorption by osteoclasts compared to bone formation by osteoblasts results in a metabolic bone disorder known as osteoporosis. This condition reduces the bone mineral density and increases the risk of fractures due to low bone mass and disrupted bone microarchitecture. Osteoclastogenesis increases when the receptor activator NFκB ligand (RANKL) on the osteoblast surface binds to the receptor activator NFκB (RANK) on the osteoclast surface and the function of the decoy receptor of RANKL, osteoprotegrin, is compromised due to external stimuli such as heparin and lipopolysaccharides. The RANK/RANKL axis promotes the nuclear factor kappa B (NFκB) expression, which in turn increases the histone methyltransferase activity of EzH2 and EzH1 for the epigenetic regulation of osteoclastogenesis-related genes. Genistein counteracts NFκB-induced osteoclastogenesis and downstream signaling through the direct regulation of histone methyltransferase, EzH2 and EzH1, transcription. However, genistein possesses limitations like low bioavailability, low water solubility, high estrogen activity, and thyroid side effects, which obstruct its therapeutic usage. Here, the nanoemulsified formulation of genistein with vitamin D was utilized to circumvent the limitations of genistein so that it can be utilized for therapeutic purposes in osteoporosis management. The nanoemulsification of genistein and vitamin D was performed through the spontaneous emulsification using Tween 80 and medium chain triglyceride oil as an organic phase. The physiologically stable and biocompatible combination of the genistein and vitamin D nanoemulsion (GVNE) exhibited the controlled release pattern of genistein with Korsmeyer-Peppas and Higuchi models under different pH conditions (7.4, 6.5, and 1.2). The GVNE potentially enhanced the therapeutic efficacy under in vitro osteoporosis models and helped restore disease parameters like alkaline phosphatase activity, tartrate-resistant acid phosphatase activity, and the formation of multinuclear giant cells. Molecularly, the GVNE overturned the LPS-induced osteoclastogenesis by downregulation of NFκB expression along with its binding on EzH2 and EzH1 promoters. GVNE effects on the osteoporosis model established it as an efficient antiosteoporotic therapy. This nanonutraceutical-based formulation provides an epigenetic regulation of osteoporosis management and opens new avenues for alternate epigenetic therapies for osteoporosis.


Subject(s)
Genistein , Osteoporosis , Epigenesis, Genetic , Genistein/therapeutic use , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B/therapeutic use , Osteoporosis/drug therapy , Osteoporosis/genetics , Osteoporosis/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RANK Ligand/therapeutic use , Vitamin D/therapeutic use
5.
ACS Appl Mater Interfaces ; 12(23): 25633-25644, 2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32453568

ABSTRACT

Bmi1 is associated with advanced prognosis of acute myeloid leukemia (AML), and polyethylenimine (PEI)-stabilized Bmi1 siRNA-entrapped human serum albumin (HSA) nanocarriers (PEI@HSANCs) were used to protect siRNA from degradation and also to control epigenetic regulation-based AML therapy. The nanoform increased the transfection efficiency of Bmi1 siRNA through caveolae-mediated endocytosis and enhanced Bax translocation into the mitochondria. It enhanced the caspase 3-mediated apoptosis through the Bax activation and Bcl-2 inhibition. The molecular analysis reveals the downregulation of polycomb proteins, Bmi1 and EzH2, along with inhibition of H3K27me3 and H2AK119ub1. The signaling cascade revealed downregulation of Bmi1 through ubiquitin-mediated degradation and is reversed by a proteasome inhibitor. Further mechanistic studies established a crucial role of transcription factor, C-Myb and Bmi1, as its direct targets for maintenance and progression of AML. Chromatin immunoprecipitation (ChIP) assay confirmed Bmi1 as a direct target of C-Myb as it binds to promoter sequence of Bmi1 between -235 to +43 and -111 to +43. The in vivo studies performed in the AML xenograft model evidence a decrease in the population of leukemic stem cells marker (CD45+) and an increase in the myeloid differentiating marker expression (CD11b+) in the bone marrow after the Bmi1 siRNA nanoconjugated therapy. Activation of apoptotic pathways and withdrawal of epigenetic repression through a ubiquitin proteasomal pathway potentiating a novel antileukemic therapy were established.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Carriers/chemistry , Leukemia, Myeloid, Acute/drug therapy , Nanocomposites/therapeutic use , Polycomb Repressive Complex 1/metabolism , RNA, Small Interfering/therapeutic use , Animals , Apoptosis/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Epigenesis, Genetic/drug effects , Female , Humans , Mice, Inbred BALB C , Nanocomposites/chemistry , Polycomb Repressive Complex 1/genetics , Polyethyleneimine/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Serum Albumin, Human/chemistry , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
6.
Biomater Sci ; 8(5): 1345-1363, 2020 Mar 07.
Article in English | MEDLINE | ID: mdl-31912833

ABSTRACT

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra and localized deposition of cytoplasmic fibrillary inclusions as Lewy bodies in the brain. The aberrant phosphorylation of α-synuclein at serine 129 is the key process on its early onset, which alters the cellular conformation to oligomers and insoluble aggregates, underpinning cellular oxidative stress and mitochondrial dysfunction, leading to devastating PD synucleinopathy. The multiple neuroprotective roles of dopamine and melatonin are often demonstrated separately; however, this approach suffers from low and short bioavailability and is associated with side-effects upon overdosing. Herein, highly pleiotropic melatonin-enriched polydopamine nanostructures were fabricated, which showed efficient brain tissue retention, sustainable and prolonged melatonin release, and prevented neuroblastoma cell death elicited by Parkinson's disease-associated and mitochondrial damaging stimuli. The synergistic neuroprotection re-established the mitochondrial membrane potential, reduced the generation of cellular reactive oxygen species (ROS), inhibited the activation of both the caspase-dependent and independent apoptotic pathways, and exhibited an anti-inflammatory effect. At the molecular level, it suppressed α-synuclein phosphorylation at Ser 129 and reduced the cellular deposition of high molecular weight oligomers. The therapeutic assessment on ex vivo organotypic brain slice culture, and in vivo experimental PD model confirmed the superior brain targeting, collective neuroprotection on dopaminergic neurons with reduced alpha-synuclein phosphorylation and deposition in the hippocampal and substantia nigra region of the brain. Thus, nature-inspired melatonin-enriched polydopamine nanostructures conferring collective neuroprotective effects attributes activation of anti-oxidative, anti-inflammatory, and anti-apoptotic pathways may be superior for application in a nanomedicine-based PD therapy.


Subject(s)
Indoles/pharmacology , Melatonin/pharmacology , Nanostructures/chemistry , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Polymers/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Indoles/chemistry , Melatonin/chemistry , Membrane Potential, Mitochondrial/drug effects , Neuroprotective Agents/chemistry , Parkinson Disease/pathology , Polymers/chemistry , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
7.
ACS Appl Mater Interfaces ; 12(5): 5658-5670, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31986005

ABSTRACT

Alzheimer's disease (AD) is one of the common causes of dementia and mild cognitive impairments, which is progressively expanding among the elderly population worldwide. A short Amyloid-ß (Aß) peptide generated after amyloidogenic processing of amyloid precursor protein exist as intermolecular ß-sheet rich oligomeric, protofibriler, and fibrillar structures and believe to be toxic species which instigate neuronal pathobiology in the brain and deposits as senile plaque. Enormous efforts are being made to develop an effective anti-AD therapy that can target Aß processing, aggregation, and propagation and provide a synergistic neuroprotective effect. However, a nanodrug prepared from natural origin can confer a multimodal synergistic chemo/photothermal inhibition of Aß pathobiology is not yet demonstrated. In the present work, we report a dopamine-melatonin nanocomposite (DM-NC), which possesses a synergistic near-infrared (NIR) responsive photothermal and pharmacological modality. The noncovalent interaction-mediated self-assembly of melatonin and dopamine oxidative intermediates leads to the evolution of DM-NCs that can withstand variable pH and peroxide environment. NIR-activated melatonin release and photothermal effect collectively inhibit Aß nucleation, self-seeding, and propagation and can also disrupt the preformed Aß fibers examined using in vitro Aß aggregation and Aß-misfolding cyclic amplification assays. The DM-NCs display a higher biocompatibility to neuroblastoma cells, suppress the AD-associated generation of intracellular reactive oxygen species, and are devoid of any negative impact on the axonal growth process. In okadaic acid-induced neuroblastoma and ex vivo midbrain slice culture-based AD model, DM-NCs exposure suppresses the intracellular Aß production, aggregation, and accumulation. Therefore, this nature-derived nanocomposite demonstrates a multimodal NIR-responsive synergistic photothermal and pharmacological modality for effective AD therapy.


Subject(s)
Amyloid beta-Peptides/chemistry , Dopamine/chemistry , Melatonin/chemistry , Nanocomposites/radiation effects , Neurons/drug effects , Alzheimer Disease/metabolism , Amyloid/drug effects , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain Chemistry , Cell Line, Tumor , Dopamine/pharmacology , Female , Humans , Infrared Rays , Melatonin/pharmacology , Mice , Mice, Inbred BALB C , Nanocomposites/chemistry , Neuroblastoma , Neurons/metabolism , Reactive Oxygen Species/metabolism
8.
Mol Pharm ; 17(2): 604-621, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31904978

ABSTRACT

Acute myeloid leukemia (AML) is a malignant disorder of hematopoietic progenitor cells with a poor prognosis of 26% of patients surviving 5 years after diagnosis. Poor bioavailability and solubility are significant factors limiting the efficacy of chemopreventive agents. In AML, the epigenetic regulator polycomb group of protein member EZH2 is highly expressed and is essential for the survival of leukemic cells. An EZH2-specific inhibitor, EPZ011989, encapsulated in human serum albumin nanoparticles (HSANPs) was synthesized for the first time via the desolvation method. The noncovalent interactions between EPZ011989 and HSANPs in nanocomposites facilitating the efficient loading and sustainable release of the drug showed enhanced cellular uptake and nuclear localization of EPZ011989-loaded HSANPs in human AML cell lines. The reduction of cell viability, colony formation inhibition, cell cycle arrest at the G2/M phase, and cell proliferation assay promoting apoptosis through the loss of mitochondrial homeostasis exerting antileukemic activity were evident. The real-time polymerase chain reaction (PCR) and western blot-based studies showed that the present nanoformulation reduces the level of PcG proteins, including EZH2, BMI-1, etc. This downregulation is associated with reduced H3K27me3 and H2AK119ub modifications conferring chromatin compaction. The immunoprecipitation study showed the physical interaction of EZH2 and c-Myb can be linked to the regulation of leukemogenesis. Further investigation revealed the mechanism of EZH2 and c-Myb downregulation via ubiquitination and proteasomal degradation pathway, confirmed by using proteasome inhibitor, suggesting the key role of proteasomal degradation machinery. Moreover, c-Myb interacted with the EZH2 promoter, which is evident by the chromatin immunoprecipitation assay and siRNA silencing. Furthermore, the formulation of EPZ011989 in HSANPs improved its biodistribution in vivo and showed excellent aqueous dispersibility and biocompatibility. In vivo studies further showed that EPZ011989-loaded HSANPs reduce the expression of CD11b+ and CD45+ markers in immunophenotyping from peripheral blood and bone marrow in engrafted nude mice. Targeted depletion of EZH2 alleviated the disease progression in nude mice and prolonged their survival. The findings provide valuable experimental evidence for the targeted epigenetic therapy of AML. The present results demonstrate an epigenetic regulation-based superior antileukemic therapy.


Subject(s)
Drug Delivery Systems/methods , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Nanoparticles/administration & dosage , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-myb/genetics , Animals , Cell Survival/drug effects , Drug Compounding/methods , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Serum Albumin, Human/chemistry , Tissue Distribution , Transfection , U937 Cells , Xenograft Model Antitumor Assays
9.
ACS Biomater Sci Eng ; 6(5): 3139-3153, 2020 05 11.
Article in English | MEDLINE | ID: mdl-33463265

ABSTRACT

Epigenetically regulated therapeutic intervention of cancer is an emerging era of research in the development of a promising therapy. Epigenetic changes are intrinsically reversible and providing the driving force to drug resistance in colorectal cancer (CRC). The regulation of polycomb group (PcG) proteins, BMI1 and EZH2, and the associated CRC progression hold promises for a novel treatment regime. The present study enlightens targeted photodynamic therapy (PDT) with potential photosensitizer hypericin nanocomposite in the development of epigenetic-based CRC therapy. We have synthesized hypericin-loaded transferrin nanoformulations (HTfNPs) overcoming the compromised hydrophobicity and poor bioavailability of the placebo drug. Targeted PDT with hypericin nanocomposite-induced BMI1 degradation assisted CRC retardation. In the present study, transferrin nanoparticles were reported to control the premature release of hypericin and improve its availability with better targeting at the disease site. Targeted intracellular internalization to colon cancer cells having a differential expression of transferrin receptors, in vivo biodistribution, stability, and pharmacokinetics provide promising applications in the nanodelivery system. Indeed, in vitro anticancer efficiency, cell cycle arrest at the G0/G1 phase, and elevated reactive oxygen species (ROS) generation confirm the anticancer effect of nanoformulation. In the exploration of mechanism, nanotherapeutic intervention by activation of PP2A, Caspase3 and inhibition of BMI1, EZH2, 3Pk, NFκB was evident. An exciting outcome of this study uncovered the camouflaged role of PP2A in the regulation of BMI1. PP2A mediates the ubiquitination/degradation of BMI1, which is revealed by changes in the physical interaction of PP2A and BMI1. Our study confirms the anticancer effect of HTfNP-assisted PDT by inducing PP2A-mediated BMI1 ubiquitination/degradation demonstrating an epigenetic-driven nanotherapeutic approach in CRC treatment.


Subject(s)
Colorectal Neoplasms , Nanoparticles , Photochemotherapy , Anthracenes , Colorectal Neoplasms/drug therapy , Humans , Perylene/analogs & derivatives , Tissue Distribution , Transferrin
10.
ACS Chem Neurosci ; 10(8): 3375-3385, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31244053

ABSTRACT

Optogenetics have evolved as a promising tool to control the processes at a cellular level via photons. Specially, it confers a specific control over cellular function through real-time cytomodulation even in freely moving animals. Neuronal stimulation is prerequisite for deep tissue light penetration or insertion of optrode for light illumination to the neurons that have been proven to be compromised due to poor light penetration and invasiveness of the procedure, respectively. In this review, the application of nanotechnology is being elaborated by the use of metal nanoparticles (AuNPs), upconversion nanocrystals (UCNPs), and quantum dots (CdSe) for targeting particular organs or tissues, and their potential to emit a specific light on excitation to overcome the limitations associated with earlier methods has been elucidated. The optothermal and magnetothermal properties, photoluminescence, and higher photostability of nanomaterials are explored in context of therapeutic applicability of optogenetics. The nanostructure characteristics and specific ion channel targeting have shown promising therapeutic potential against neurodegenerative disorders (Alzheimer's, Parkinson's, Huntington's), epilepsy, and blindness. This review compiles mechanical and optical characteristics of nanomaterials that endow superior optogenetic therapeutic potentials to cure immedicable infirmities.


Subject(s)
Nanoparticles/therapeutic use , Nanotechnology/methods , Nanotechnology/trends , Optogenetics/methods , Optogenetics/trends , Animals , Humans , Neurology/methods , Neurology/trends
11.
ACS Appl Mater Interfaces ; 10(35): 29325-29337, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30096228

ABSTRACT

Pseudomonas aeruginosa, a Gram-negative rod-shaped bacterium is a notorious pathogen causing chronic infections. Its ability to form antibiotic-resistant biofilm has raised the need for the development of alternative treatment approaches. An ideal alternate can be silver nanoparticles known for their strong yet tunable bactericidal activity. However, their use in commercial in vivo medicine could not see the light of the day because of the unwanted toxicity of silver in the host cells at higher concentrations. Thus, strategies which can modulate the bacterial cell-silver nanoparticle interactions thereby reducing the amount of nanoparticles required to kill a typical number of bacterial cells are utmost welcomed. The current work showcases one such strategy by functionalizing the silver nanoparticles with l-fucose to increase their interactions with the LecB lectins present on P. aeruginosa PAO1. The advantage of this approach lies in the higher bactericidal and antibiofilm activity of fucose-functionalized silver nanoparticles (FNPs) as compared to the citrate-capped silver nanoparticles (CNPs) of similar size and concentrations. The superior bactericidal potential of FNPs as demonstrated by fluorescence-assisted cell sorting, confocal laser scanning microscopy, and transmission electron microscopy analyses may be attributed to the higher reactive oxygen species generation and oxidative membrane damage. Additionally, FNPs prevented the formation of biofilms by downregulating the expression of various virulence genes at lower concentrations as compared to CNPs. The practical applicability of the approach was demonstrated by preventing bacterial colonization on artificial silicone rubber surfaces. These results can be extrapolated in the treatment of catheter-associated urinary tract infections caused by P. aeruginosa. In conclusion, the present work strongly advocates the use of antivirulence targets and their corresponding binding residues for the augmentation of the bactericidal effect of silver nanoparticles.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Metal Nanoparticles , Pseudomonas aeruginosa/drug effects , Silver/pharmacology , Urinary Catheters/microbiology , Anti-Bacterial Agents/chemistry , Fucose/chemistry , Metal Nanoparticles/chemistry
12.
Nanotechnology ; 28(36): 365102, 2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28820142

ABSTRACT

Melatonin (MEL) has promising medicinal value as an anticancer agent in a variety of malignancies, but there are difficulties in achieving a therapeutic dose due to its short half-life, low bioavailability, poor solubility and extensive first-pass metabolism. In this study chitosan/tripolyphosphate (TPP) nanoparticles were prepared by an ionic gelation method to overcome the therapeutic challenges of melatonin and to improve its anticancer efficacy. Characterization of the melatonin-loaded chitosan (MEL-CS) nanoformulation was performed using transmission and scanning electron microscopies, dynamic light scattering, Fourier transform infrared spectroscopy, Raman spectroscopy and x-ray diffraction. In vitro release, cellular uptake and efficacy studies were tested for their enhanced anticancer potential in human U87MG glioblastoma cells. Confocal studies revealed higher cellular uptake of MEL-CS nanoparticles and enhanced anticancer efficacy in human malignant glioblastoma cancer cells than in healthy non-malignant human HEK293T cells in mono- and co-culture models. Our study has shown for the first time that MEL-CS nanocomposites are therapeutically more effective as compared to free MEL at inducing functional anticancer efficacy in the human brain tumour U87MG cell line.

13.
J Oncol ; 2012: 782020, 2012.
Article in English | MEDLINE | ID: mdl-21876694

ABSTRACT

Neuroblastoma, a progressive solid tumor in childhood, continues to be a clinical challenge. It is highly vascular, heterogeneous, and extracranial tumor that originates from neural crest. Angiogenesis, genetic abnormalities, and oncogene amplification are mainly responsible for malignant phenotype of this tumor. Survivability of malignant neuroblastoma patients remains poor despite the use of traditional therapeutic strategies. Angiogenesis is a very common and necessary pre-requisite for tumor progression and metastasis. Angiogenesis is also a major factor in making malignant neuroblastoma. Thus, prevention of angiogenesis can be a highly significant strategy in the treatment of malignant neuroblastoma. Here, we summarize our current understanding of angiogenesis in malignant neuroblstoma and describe the use of experimental anti-angiogenic agents either alone or in combination therapy. This review will clearly indicate the importance of angiogenesis in the pathogenesis of malignant neuroblastoma, its prevention as a promising therapy in preclinical models of malignant neuroblastoma, and prospective clinical trials.

14.
Neurochem Res ; 36(12): 2292-305, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21786169

ABSTRACT

Glioblastoma shows poor response to current therapies and warrants new therapeutic strategies. We examined the efficacy of combination of valproic acid (VPA) and taxol (TX) or nanotaxol (NTX) in human glioblastoma LN18 and T98G cell lines. Cell differentiation was manifested in changes in morphological features and biochemical markers. Cell growth was controlled with down regulation of vascular endothelial growth factor (VEGF), epidermal growth factor receptor (EGFR), nuclear factor-kappa B (NF-κB), phospho-Akt (p-Akt), and multi-drug resistance (MDR) marker, indicating suppression of angiogenic, survival, and multi-drug resistance pathways. Cell cycle analysis showed that combination therapy (VPA and TX or NTX) increased the apoptotic sub G1 population and apoptosis was further confirmed by Annexin V-FITC/PI binding assay and scanning electron microscopy. Combination therapy caused activation of caspase-8 and cleavage of Bid to tBid and increased Bax:Bcl-2 ratio and mitochondrial release of cytochrome c and apoptosis-inducing factor (AIF). Upregulation of calpain and caspases (caspase-9 and caspase-3) and substrate degradation were also detected in course of apoptosis. The combination of VPA and NTX most effectively controlled the growth of LN18 and T98G cells. Therefore, this combination of drugs can be used as an effective treatment for controlling growth of human glioblastoma cells.


Subject(s)
Cell Differentiation/drug effects , Glioblastoma/drug therapy , Paclitaxel/therapeutic use , Valproic Acid/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Albumin-Bound Paclitaxel , Albumins/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Inducing Factor/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Down-Regulation , Drug Therapy, Combination , Humans , Paclitaxel/administration & dosage , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
15.
Invest New Drugs ; 28(6): 812-24, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19777160

ABSTRACT

Neuroblastoma is an extracranial, solid, and heterogeneous malignancy in children. The conventional therapeutic modalities are mostly ineffective and thus new therapeutic strategies for malignant neuroblastoma are urgently warranted. We examined the synergistic efficacy of combination of sorafenib (SF) and genistein (GST) in human malignant neuroblastoma SK-N-DZ (N-Myc amplified) and SH-SY5Y (N-Myc non-amplified) cell lines. MTT assay showed dose-dependent decrease in cell viability and the combination therapy more prominently inhibited the cell proliferation in both cell lines than either treatment alone. Apoptosis was confirmed morphologically by Wright staining. Flow cytometric analysis of cell cycle phase distribution and Annexin V-FITC/PI staining showed increase in subG1 DNA content and early apoptosis, respectively, after treatment with the combination of drugs. Apoptosis was further confirmed by scanning electron microscopy. Combination therapy showed activation of caspase-8, cleavage of Bid to tBid, increase in p53 and p21 expression, down regulation of anti-apoptotic Mcl-1, and increase in Bax:Bcl-2 ratio to trigger apoptosis. Down regulation of MDR, hTERT, N-Myc, VEGF, FGF-2, NF-κB, p-Akt, and c-IAP2 indicated suppression of angiogenic and survival pathways. Mitochondrial release of cytochrome c and Smac into cytosol indicated involvement of mitochondia in apoptosis. Increases in proteolytic activities of calpain and caspase-3 were also confirmed. Our results suggested that combination of SF and GST inhibited angiogenic and survival factors and increased apoptosis via receptor and mitochondria mediated pathways in both neuroblastoma SK-N-DZ and SH-SY5Y cell lines. Thus, this combination of drugs could be a potential therapeutic strategy against human malignant neuroblastoma cells having N-Myc amplification or non-amplification.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Apoptosis/drug effects , Benzenesulfonates/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Genistein/pharmacology , Neuroblastoma/pathology , Pyridines/pharmacology , Tumor Suppressor Protein p53/metabolism , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Drug Synergism , Flow Cytometry , Gene Amplification/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/ultrastructure , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/drug effects , Sorafenib , Telomerase/metabolism , Treatment Outcome , Up-Regulation/drug effects , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...