Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Front Pharmacol ; 14: 1265440, 2023.
Article in English | MEDLINE | ID: mdl-37745070

ABSTRACT

Cancer has remained one of the leading causes of death worldwide, with a lack of effective treatment. The intrinsic shortcomings of conventional therapeutics regarding tumor specificity and non-specific toxicity prompt us to look for alternative therapeutics to mitigate these limitations. In this regard, we developed multifunctional bimetallic (FeCo) bi-MIL-88B-FC MOFs modified with folic acid-conjugated chitosan (FC) as drug delivery systems (DDS) for targeted delivery of 5-Fluorouracil (5-FU). The bi-MIL-88B nanocarriers were characterized through various techniques, including powder X-ray diffraction, scanning electron microscopy, energy-dispersive X-ray, thermogravimetric analysis, and Fourier transform infrared spectroscopy. Interestingly, 5-FU@bi-MIL-88B-FC showed slower release of 5-FU due to a gated effect phenomenon endowed by FC surface coating compared to un-modified 5-FU@bi-MIL-88B. The pH-responsive drug release was observed, with 58% of the loaded 5-FU released in cancer cells mimicking pH (5.2) compared to only 24.9% released under physiological pH (5.4). The in vitro cytotoxicity and cellular internalization experiments revealed the superiority of 5-FU@bi-MIL-88B-FC as a highly potent targeted DDS against folate receptor (FR) positive SW480 cancer cells. Moreover, due to the presence of Fe and Co in the structure, bi-MIL-88B exhibited peroxidase-like activity for chemodynamic therapy. Based on the results, 5-FU@bi-MIL-88B-FC could serve as promising candidate for smart DDS by sustained drug release and selective targeting.

2.
Genes (Basel) ; 13(3)2022 03 21.
Article in English | MEDLINE | ID: mdl-35328105

ABSTRACT

Genomic epidemiology of SARS-CoV-2 is imperative to explore the transmission, evolution, and also pathogenicity of viruses. The emergence of SARS-CoV-2 variants of concern posed a severe threat to the global public health efforts. To assess the potential consequence of these emerging variants on public health, continuous molecular epidemiology is of vital importance. The current study has been designed to investigate the major SARS-CoV-2 variants and emerging mutations in virus structural and non-structural proteins (NSP) during the fourth wave in September 2021 from the Punjab province of Pakistan. Twenty SARS-CoV-2 positive samples have been collected from major cities were subjected to next-generation sequencing. Among the 20 whole genomes (GenBank Accession SRR16294858-SRR16294877), 2 samples failed to be completely sequenced. These genome sequences harbored 207 non-synonymous mutations, among which 19 were unique to GISAID. The genome sequences were detected: Delta 21I, 21J variants (B.1.617.2). Mutation's spike_F157del, spike_P681R, spike_T478K, spike_T19R, spike_L452R, spike_D614G, spike_G142D, spike_E156G, and spike_R158del have been detected in all samples where K1086Q, E554K, and C1250W were unique in spike protein. These genomic sequences also harbored 129 non-synonymous mutations in NSP. The most common were NSP3_P1469S (N = 17), NSP3_A488S (N = 17), NSP3_P1228L (N = 17), NSP4_V167L (N = 17), NSP4_T492I (N = 17), NSP6_T77A (N = 17), NSP14_A394V (N = 17), NSP12_G671S (N = 18), and NSP13_P77L (N = 18). The mutation, F313Y in NSP12, detected in the current study, was found in a single isolate from Belgium. Numerous other unique mutations have been detected in the virus papain-like protease (NSP3), main protease (NSP5), and RNA-dependent RNA polymerase (NSP12). The most common non-synonymous mutations in the spike protein were subjected to stability analysis, exhibiting a stabilizing effect on structures. The presence of Delta variants may affect therapeutic efforts and vaccine efficacy. Continuous genomic epidemiology of SARS-CoV-2 in Pakistan may be useful for better management of SARS-CoV-2 infections.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/epidemiology , COVID-19/genetics , Genome, Viral , Humans , Mutation , Pakistan/epidemiology , Pandemics , SARS-CoV-2/genetics
3.
Bioconjug Chem ; 31(5): 1474-1485, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32286806

ABSTRACT

Exploring a combined phototherapeutic strategy to overcome the limitations of a single mode therapy and inducing high anticancer efficiency is highly promising for precision cancer nanomedicine. However, a single-wavelength laser activates dual photothermal/photodynamic therapy (PTT/PDT) treatment is still a formidable challenge. Herein, we strategically design and fabricate a multifunctional theranostic nanosystem based on chlorin e6-functionalized polydopamine (PDA) coated prussian blue/manganese dioxide nanoparticles (PB-MnO2@PDA-Ce6 NPs). Interestingly, the obtained PB-MnO2@PDA NPs not only offer an effective delivery system for Ce6 but also provide strong optical absorption in the near-infrared range, endowing high antitumor efficacy of PTT. More importantly, the as-prepared PB-MnO2@PDA-Ce6 nanoagents exhibit an effective oxygen generation, superior reactive oxygen species (ROS), and outstanding photothermal conversion ability to greatly improve PTT and PDT treatments. As a result, both in vitro and in vivo treatments guided by MR imaging on liver cancer cells reveal the complete cell/tumor eradication under a single wavelength of 660 nm laser irradiation, implying the simultaneous synergistic PDT/PTT effects triggered by PB-MnO2@PDA-Ce6 nanoplatform, which are much higher than individual treatment. Taken together, our phototherapeutic nanoagents exhibit an excellent therapeutic performance, which may act as a nanoplatform to find safe and clinically translatable routes to accelerate cancer therapeutics.


Subject(s)
Ferrocyanides/chemistry , Indoles/chemistry , Infrared Rays , Manganese Compounds/chemistry , Nanoparticles/chemistry , Oxides/chemistry , Oxygen/metabolism , Photochemotherapy/methods , Polymers/chemistry , Cell Line, Tumor , Drug Design , Humans , Indoles/pharmacology , Lasers , Polymers/pharmacology
4.
ACS Nano ; 13(8): 8890-8902, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31291092

ABSTRACT

Intratumoral glucose depletion-induced cancer starvation represents an important strategy for anticancer therapy, but it is often limited by systemic toxicity, nonspecificity, and adaptive development of parallel energy supplies. Herein, we introduce a concept of cascaded catalytic nanomedicine by combining targeted tumor starvation and deoxygenation-activated chemotherapy for an efficient cancer treatment with reduced systemic toxicity. Briefly, nanoclustered cascaded enzymes were synthesized by covalently cross-linking glucose oxidase (GOx) and catalase (CAT) via a pH-responsive polymer. The release of the enzymes can be first triggered by the mildly acidic tumor microenvironment and then be self-accelerated by the subsequent generation of gluconic acid. Once released, GOx can rapidly deplete glucose and molecular oxygen in tumor cells while the toxic side product, i.e., H2O2, can be readily decomposed by CAT for site-specific and low-toxicity tumor starvation. Furthermore, the enzymatic cascades also created a local hypoxia with the oxygen consumption and reductase-activated prodrugs for an additional chemotherapy. The current report represents a promising combinatorial approach using cascaded catalytic nanomedicine to reach concurrent selectivity and efficiency of cancer therapeutics.


Subject(s)
Glucose Oxidase/chemistry , Neoplasms/drug therapy , Prodrugs/pharmacology , Tumor Microenvironment/drug effects , Catalysis , Glucose/chemistry , Glucose Oxidase/pharmacology , Humans , Hydrogen Peroxide/chemistry , Nanomedicine , Nanoparticles/chemistry , Neoplasms/pathology , Oxidation-Reduction , Oxidative Stress/drug effects , Prodrugs/chemistry
5.
Front Pharmacol ; 10: 447, 2019.
Article in English | MEDLINE | ID: mdl-31156425

ABSTRACT

All-trans retinoic acid (ATRA) is an effective agent that induces differentiation, inhibits cell proliferation, and acts as an anticancer agent. ATRA was successfully conjugated with Pluronic F127 via esterification to enhance its anticancer effects. Pluronic-ATRA showed high cytotoxicity and inhibitory concentrations (IC50) 50% lower than those of ATRA in various breast cancer cell lines (4T1:31.16-8.57 µg/mL; EMT6: 50.48-7.08 µg/mL; MDA-MB-231:37.58-8.99 µg/mL; BT474:25.27-9.09 µg/mL). In combination with chemotherapy, Pluronic-ATRA synergistically enhanced the cytotoxic effects of cisplatin (CDDP). Pluronic-ATRA combined with CDDP effectively suppressed breast tumor growth in vivo. The results of this study demonstrate the potential of Pluronic-ATRA as an anticancer agent that can be used in combination therapy against solid tumors.

6.
Biomater Sci ; 7(7): 2740-2748, 2019 Jun 25.
Article in English | MEDLINE | ID: mdl-30994642

ABSTRACT

The tumor hypoxic microenvironment (THME) has a profound impact on tumor progression, and modulation of the THME has become an essential strategy to promote photodynamic therapy (PDT). Here, an oxygen self-supplied nanodelivery system that is based on nanometal-organic frameworks (nMOFs) with embedded AuNPs (Au@ZIF-8) on the nMOF surface as a catalase (CAT)-like nanozyme and encapsulating Ce6 inside as a photosensitizer was found to mitigate tumor hypoxia and reinforce PDT. As soon as Au@ZIF-8 reaches the tumor site, the AuNP nanozyme can catalyze excessive H2O2 to produce O2 to alleviate tumor hypoxia, promoting the production of 1O2 with strong toxicity toward tumor cells under irradiation. Our study demonstrates that nMOFs embellished with a nanozyme have great potential for overcoming the THME for cancer therapeutics, which provides a facile strategy for accurate bioimaging and cancer therapy in vivo.


Subject(s)
Catalase/metabolism , Drug Carriers/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Organometallic Compounds/chemistry , Photochemotherapy/methods , Animals , Biomimetic Materials/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Chlorophyllides , Hydrogen Peroxide/metabolism , Mice , Oxygen/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology , Tumor Burden/drug effects , Tumor Burden/radiation effects , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects
7.
Biomater Sci ; 7(5): 1933-1939, 2019 Apr 23.
Article in English | MEDLINE | ID: mdl-30775753

ABSTRACT

Polymeric nanoparticles as drug delivery systems have the potential to improve the therapeutic efficacy and reduce the toxicity of chemotherapeutic drugs by enhancing the drug selectivity in vivo. The efficacy is directly dependent on the polymeric nanoparticles' in vivo fate. Therefore, it is very important to develop a method to stably label the polymeric nanoparticles for detecting the in vivo fate. Here, we report a method to stably label self-assembled nanoparticles by the incorporation of rhodamine B-conjugated poly(ε-caprolactone) (PCL-RhoB). Only 1% of PCL-RhoB was released from the RhoB-labeled polymeric nanoparticles (RhoB-PNPs) in phosphate buffer within 12 hours, which suggested that the signal of PCL-RhoB can be used to represent the behaviors of polymeric nanoparticles both in vitro and in vivo. PCL-RhoB could be effectively extracted and quantitatively detected by ultra-high-performance liquid chromatography (UPLC) in various media, such as PBS, a cell culture medium containing 10% FBS (pH = 7.4 and pH = 6.8), mouse serum, simulated intestinal fluid and cell or tissue lysis. The intracellular contents of PCL-RhoB in MDA-MB-231 cells detected by UPLC were linearly correlated to the concentration of the RhoB-PNPs. In addition, the contents of PCL-RhoB in plasma and the spleen were proportional to the injected dose of RhoB-PNPs in vivo. As an application example, the pharmacokinetics and biodistribution of the nanoparticles over time in vivo were analyzed following intravenous injection to confirm the feasibility of this method.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Polyesters/chemistry , Rhodamines/chemistry , Animals , Biological Transport , Cell Line, Tumor , Drug Carriers/pharmacokinetics , Fluorescent Dyes/chemistry , Humans , Mice , Polyesters/pharmacokinetics , Tissue Distribution
8.
Nanotechnology ; 30(6): 065102, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30543196

ABSTRACT

Transition metal dichalogenides (TMDCs) with unique layered structures hold promising potential as transducers for photothermal therapy. However, the low photothermal conversion efficiency and poor stability in some cases limit their practical applications. Herein, we demonstrate the fabrication of ultrathin homogeneous hybridized TMDC nanosheets and their use for highly efficient photothermal tumor ablation. In particular, the nanosheets were composed of metallic WSe2 intercalated with polyvinylpyrrolidone (PVP), which was facilely prepared through a solvothermal process from the mixture of selenourea crystals, WCl6 powder along with PVP polymeric nanogel. Our characterizations revealed that the obtained nanosheets exhibited excellent photothermal conversion efficiency, therapeutic demonstration with improved biocompatibility and physiological stability attributing to the combined merits of metallic phase of WSe2 and hydrophilic PVP insertion. Both the histological analysis of vital organs and in vitro/in vivo tests confirmed the nanosheets as actively effective and biologically safe in this phototherapeutic technique. Findings from this non-invasive experiment clearly emphasize the explorable therapeutic efficacy of the layered-based hybrid agents in future cancer treatment planning procedures.


Subject(s)
Photosensitizing Agents/therapeutic use , Phototherapy/methods , Povidone/chemistry , Selenium/chemistry , Tungsten/chemistry , Animals , Cell Line, Tumor , Female , Infrared Rays/therapeutic use , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Neoplasms, Experimental/therapy , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Temperature , Xenograft Model Antitumor Assays
9.
Biomacromolecules ; 19(4): 1130-1141, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29514006

ABSTRACT

Multidrug resistance (MDR) is the major cause for chemotherapy failure, which constitutes a formidable challenge in the field of cancer therapy. The synergistic chemo-photothermal treatment has been reported to be a potential strategy to overcome MDR. In this work, rationally designed enzyme-degradable, hyperbranched polyphosphoester nanomedicines were developed for reversing MDR via the codelivery of doxorubicin and IR-780 (hPPEDOX&IR) as combined chemo-photothermal therapy. The amphiphilic hyperbranched polyphosphoesters with phosphate bond as the branching point were synthesized via a simple but robust one-step polycondensation reaction. The self-assembled hPPEDOX&IR exhibited good serum stability, sustained release, preferable tumor accumulation, and enhanced drug influx of doxorubicin in resistant MCF-7/ADR cells. Moreover, the degradation of hPPEDOX&IR was accelerated in the presence of alkaline phosphatase, which was overexpressed in various cancers, resulting in the fast release of encapsulated doxorubicin. The enzyme-degradable polymer generated synergistic chemo-photothermal cytotoxicity against MCF-7/ADR cells and, thus, the efficient ablation of DOX-resistant tumor without regrowth. This delivery system may open a new avenue for codelivery of chemo- and photothermal therapeutics for MDR tumor therapy.


Subject(s)
Drug Delivery Systems , Drug Resistance, Neoplasm/drug effects , Nanomedicine , Neoplasms/drug therapy , Combined Modality Therapy , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Drug Therapy/methods , Humans , Indoles/chemistry , Indoles/pharmacology , MCF-7 Cells , Phototherapy
10.
Int J Nanomedicine ; 11: 2451-61, 2016.
Article in English | MEDLINE | ID: mdl-27330288

ABSTRACT

Lipid and protein oxidation are well-known manifestations of free radical activity and oxidative stress. The current study investigated extermination of Leishmania tropica promastigotes induced by lipid and protein oxidation with reactive oxygen species produced by PEGylated metal-based nanoparticles. The synthesized photodynamic therapy-based doped and nondoped zinc oxide nanoparticles were activated in daylight that produced reactive oxygen species in the immediate environment. Lipid and protein oxidation did not occur in dark. The major lipid peroxidation derivatives comprised of conjugated dienes, lipid hydroperoxides, and malondialdehyde whereas water, ethane, methanol, and ethanol were found as the end products. Proteins were oxidized to carbonyls, hydroperoxides, and thiol degrading products. Interestingly, lipid hydroperoxides were produced by more than twofold of the protein hydroperoxides, indicating higher degradation of lipids compared to proteins. The in vitro evidence represented a significant contribution of the involvement of both lipid and protein oxidation in the annihilated antipromastigote effect of nanoparticles.


Subject(s)
Leishmania tropica/drug effects , Light , Lipid Peroxidation/drug effects , Metal Nanoparticles/chemistry , Proteins/metabolism , Reactive Oxygen Species/pharmacology , Zinc Oxide/pharmacology , Gas Chromatography-Mass Spectrometry , Leishmania tropica/metabolism , Lipid Peroxides/metabolism , Malondialdehyde/metabolism , Metal Nanoparticles/ultrastructure , Oxidation-Reduction/drug effects , Polyenes/metabolism , Protein Carbonylation/drug effects , Spectroscopy, Fourier Transform Infrared , Time Factors , X-Ray Diffraction
11.
Int J Nanomedicine ; 10: 6891-903, 2015.
Article in English | MEDLINE | ID: mdl-26604755

ABSTRACT

Human beings suffer from several infectious agents such as viruses, bacteria, and protozoans. Recently, there has been a great interest in developing biocompatible nanostructures to deal with infectious agents. This study investigated benign ZnCuO nanostructures that were visible-light-responsive due to the resident copper in the lattice. The nanostructures were synthesized through a size-controlled hot-injection process, which was adaptable to the surface ligation processes. The nanostructures were then characterized through transmission electron microscopy, X-ray diffraction, diffused reflectance spectroscopy, Rutherford backscattering, and photoluminescence analysis to measure crystallite nature, size, luminescence, composition, and band-gap analyses. Antiprotozoal efficiency of the current nanoparticles revealed the photodynamic killing of Leishmania protozoan, thus acting as efficient metal-based photosensitizers. The crystalline nanoparticles showed good biocompatibility when tested for macrophage toxicity and in hemolysis assays. The study opens a wide avenue for using toxic material in resident nontoxic forms as an effective antiprotozoal treatment.


Subject(s)
Copper/chemistry , Leishmania/drug effects , Macrophages/parasitology , Metal Nanoparticles/chemistry , Photosensitizing Agents/chemistry , Protozoan Infections/drug therapy , Zinc/chemistry , Apoptosis , Crystallization , Dose-Response Relationship, Drug , Drug Design , Hemolysis , Humans , Inhibitory Concentration 50 , Light , Luminescence , Macrophages/cytology , Microscopy, Electron, Transmission , Necrosis , Particle Size , Permeability , Photochemistry , Reactive Oxygen Species/metabolism , Temperature , X-Ray Diffraction
12.
Free Radic Biol Med ; 77: 230-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25266330

ABSTRACT

We describe daylight responsive silver (Ag) doped semiconductor nanoparticles of zinc oxide (DSNs) for photodynamic therapy (PDT) against Leishmania. The developed materials were characterized by X-ray diffraction analysis (XRD), Rutherford backscattering (RBS), diffused reflectance spectroscopy (DRS), and band-gap analysis. The Ag doped semiconductor nanoparticles of zinc oxide were PEGylated to enhance their biocompatibility. The DSNs demonstrated effective daylight response in the PDT of Leishmania protozoans, through the generation of reactive oxygen species (ROS) with a quantum yield of 0.13 by nondoped zinc oxide nanoparticles (NDSN) whereas 0.28 by DSNs. None of the nanoparticles have shown any antileishmanial activity in dark, confirming that only ROS produced in the daylight were involved in the killing of leishmanial cells. Furthermore, the synthesized nanoparticles were found biocompatible. Using reactive oxygen species scavengers, cell death was attributable mainly to 77-83% singlet oxygen and 18-27% hydroxyl radical. The nanoparticles caused permeability of the cell membrane, leading to the death of parasites. Further, the uptake of nanoparticles by Leishmania cells was confirmed by inductively coupled plasma atomic emission spectroscopy (ICP-AES). We believe that these DSNs are widely applicable for the PDT of leishmaniasis, cancers, and other infections due to daylight response.


Subject(s)
Antiprotozoal Agents/pharmacology , Leishmaniasis/drug therapy , Nanoparticles/chemistry , Photosensitizing Agents/pharmacology , Silver/chemistry , Zinc Oxide/chemistry , Animals , Artemia , Cell Membrane Permeability , Cell Survival , Cells, Cultured , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Leishmania tropica/drug effects , Leishmania tropica/metabolism , Macrophages/drug effects , Particle Size , Photochemotherapy , Polyethylene Glycols/chemistry , Reactive Oxygen Species/metabolism , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL