Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Nanobiotechnology ; 22(1): 347, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898529

ABSTRACT

BACKGROUND: Silica nanoparticles (SNPs) have immense potential in biomedical research, particularly in drug delivery and imaging applications, owing to their stability and minimal interactions with biological entities such as tissues or cells. RESULTS: With synthesized and characterized cyanine-dye-doped fluorescent SNPs (CSNPs) using cyanine 3.5, 5.5, and 7 (Cy3.5, Cy5.5, and Cy7). Through systematic analysis, we discerned variations in the surface charge and fluorescence properties of the nanoparticles contingent on the encapsulated dye-(3-aminopropyl)triethoxysilane conjugate, while their size and shape remained constant. The fluorescence emission spectra exhibited a redshift correlated with increasing dye concentration, which was attributed to cascade energy transfer and self-quenching effects. Additionally, the fluorescence signal intensity showed a linear relationship with the particle concentration, particularly at lower dye equivalents, indicating a robust performance suitable for imaging applications. In vitro assessments revealed negligible cytotoxicity and efficient cellular uptake of the nanoparticles, enabling long-term tracking and imaging. Validation through in vivo imaging in mice underscored the versatility and efficacy of CSNPs, showing single-switching imaging capabilities and linear signal enhancement within subcutaneous tissue environment. CONCLUSIONS: This study provides valuable insights for designing fluorescence imaging and optimizing nanoparticle-based applications in biomedical research, with potential implications for targeted drug delivery and in vivo imaging of tissue structures and organs.


Subject(s)
Carbocyanines , Fluorescent Dyes , Nanoparticles , Optical Imaging , Silicon Dioxide , Silicon Dioxide/chemistry , Nanoparticles/chemistry , Carbocyanines/chemistry , Animals , Mice , Optical Imaging/methods , Fluorescent Dyes/chemistry , Humans , Silanes/chemistry , Particle Size , Propylamines , Benzothiazoles
2.
Biosens Bioelectron ; 175: 112868, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33281048

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a newly emerged human infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In a global pandemic, development of a cheap, rapid, accurate, and easy-to-use diagnostic test is necessary if we are to mount an immediate response to this emerging threat. Here, we report the development of a specific lateral flow immunoassay (LFIA)-based biosensor for COVID-19. We used phage display technology to generate four SARS-CoV-2 nucleocapsid protein (NP)-specific single-chain variable fragment-crystallizable fragment (scFv-Fc) fusion antibodies. The scFv-Fc antibodies bind specifically and with high affinity to the SARS-CoV-2 NP antigen, but not to NPs of other coronaviruses. Using these scFv-Fc antibodies, we screened three diagnostic antibody pairs for use on a cellulose nanobead (CNB)-based LFIA platform. The detection limits of the best scFv-Fc antibody pair, 12H1 as the capture probe and 12H8 as the CNB-conjugated detection probe, were 2 ng antigen protein and 2.5 × 104 pfu cultured virus. This LFIA platform detected only SARS-CoV-2 NP, not NPs from MERS-CoV, SARS-CoV, or influenza H1N1. Thus, we have successfully developed a SARS-CoV-2 NP-specific rapid diagnostic test, which is expected to be a simple and rapid diagnostic test for COVID-19.


Subject(s)
Antigens, Viral/isolation & purification , Biosensing Techniques , COVID-19/diagnosis , SARS-CoV-2/isolation & purification , Antibodies, Viral/blood , Antigens, Viral/immunology , COVID-19/immunology , COVID-19/virology , Humans , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Single-Chain Antibodies/immunology
3.
Biosens Bioelectron ; 171: 112715, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33099241

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), a newly emerging human infectious disease. Because no specific antiviral drugs or vaccines are available to treat COVID-19, early diagnostics, isolation, and prevention are crucial for containing the outbreak. Molecular diagnostics using reverse transcription polymerase chain reaction (RT-PCR) are the current gold standard for detection. However, viral RNAs are much less stable during transport and storage than proteins such as antigens and antibodies. Consequently, false-negative RT-PCR results can occur due to inadequate collection of clinical specimens or poor handling of a specimen during testing. Although antigen immunoassays are stable diagnostics for detection of past infection, infection progress, and transmission dynamics, no matched antibody pair for immunoassay of SARS-CoV-2 antigens has yet been reported. In this study, we designed and developed a novel rapid detection method for SARS-CoV-2 spike 1 (S1) protein using the SARS-CoV-2 receptor ACE2, which can form matched pairs with commercially available antibodies. ACE2 and S1-mAb were paired with each other for capture and detection in a lateral flow immunoassay (LFIA) that did not cross-react with SARS-CoV Spike 1 or MERS-CoV Spike 1 protein. The SARS-CoV-2 S1 (<5 ng of recombinant proteins/reaction) was detected by the ACE2-based LFIA. The limit of detection of our ACE2-LFIA was 1.86 × 105 copies/mL in the clinical specimen of COVID-19 Patients without no cross-reactivity for nasal swabs from healthy subjects. This is the first study to detect SARS-CoV-2 S1 antigen using an LFIA with matched pair consisting of ACE2 and antibody. Our findings will be helpful to detect the S1 antigen of SARS-CoV-2 from COVID-19 patients.


Subject(s)
Betacoronavirus/isolation & purification , Biosensing Techniques/instrumentation , Clinical Laboratory Techniques , Coronavirus Infections/diagnosis , Peptidyl-Dipeptidase A/chemistry , Pneumonia, Viral/diagnosis , Spike Glycoprotein, Coronavirus/analysis , Angiotensin-Converting Enzyme 2 , Antibodies, Monoclonal/chemistry , Biosensing Techniques/economics , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques/economics , Clinical Laboratory Techniques/instrumentation , Coronavirus Infections/economics , Equipment Design , Humans , Immunoassay/economics , Immunoassay/instrumentation , Immunoconjugates/chemistry , Pandemics , SARS-CoV-2 , Sensitivity and Specificity , Time Factors
5.
ACS Nano ; 14(4): 5135-5142, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32293168

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a newly emerging human infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, previously called 2019-nCoV). Based on the rapid increase in the rate of human infection, the World Health Organization (WHO) has classified the COVID-19 outbreak as a pandemic. Because no specific drugs or vaccines for COVID-19 are yet available, early diagnosis and management are crucial for containing the outbreak. Here, we report a field-effect transistor (FET)-based biosensing device for detecting SARS-CoV-2 in clinical samples. The sensor was produced by coating graphene sheets of the FET with a specific antibody against SARS-CoV-2 spike protein. The performance of the sensor was determined using antigen protein, cultured virus, and nasopharyngeal swab specimens from COVID-19 patients. Our FET device could detect the SARS-CoV-2 spike protein at concentrations of 1 fg/mL in phosphate-buffered saline and 100 fg/mL clinical transport medium. In addition, the FET sensor successfully detected SARS-CoV-2 in culture medium (limit of detection [LOD]: 1.6 × 101 pfu/mL) and clinical samples (LOD: 2.42 × 102 copies/mL). Thus, we have successfully fabricated a promising FET biosensor for SARS-CoV-2; our device is a highly sensitive immunological diagnostic method for COVID-19 that requires no sample pretreatment or labeling.


Subject(s)
Betacoronavirus/isolation & purification , Biosensing Techniques , Coronavirus Infections/diagnosis , Pneumonia, Viral/diagnosis , Transistors, Electronic , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques , Graphite , Humans , Nanotechnology/instrumentation , Nasal Cavity , Pandemics , SARS-CoV-2 , Specimen Handling
6.
Biomolecules ; 10(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-32131481

ABSTRACT

Gintonin, a novel ginseng-derived lysophosphatidic acid receptor ligand, improves brain functions and protects neurons from oxidative stress. However, little is known about the effects of gintonin against Pb-induced brain maldevelopment. We investigated the protective effects of gintonin on the developing cerebellum after prenatal and postnatal Pb exposure. Pregnant female rats were randomly divided into three groups: control, Pb (0.3% Pb acetate in drinking water), and Pb plus gintonin (100 mg/kg, p.o.). Blood Pb was increased in dams and pups; gintonin treatment significantly decreased blood Pb. On postnatal day 21, the number of degenerating Purkinje cells was remarkably increased while the number of calbindin-, GAD67-, NMDAR1-, LPAR1-immunoreactive intact Purkinje cells, and GABA transporter 1-immunoreactive pinceau structures were significantly reduced in Pb-exposed offspring. Following Pb exposure, gintonin ameliorated cerebellar degenerative effects, restored increased pro-apoptotic Bax, and decreased anti-apoptotic Bcl2. Gintonin treatment attenuated Pb-induced accumulation of oxidative stress (Nrf2 and Mn-SOD) and inflammation (IL-1ß and TNFα,), restoring the decreased cerebellar BDNF and Sirt1. Gintonin ameliorated Pb-induced impairment of myelin basic protein-immunoreactive myelinated fibers of Purkinje cells. Gintonin attenuated Pb-induced locomotor dysfunctions. The present study revealed the ameliorating effects of gintonin against Pb, suggesting the potential use of gintonin as a preventive agent in Pb poisoning during pregnancy and lactation.


Subject(s)
Lactation/metabolism , Lead Poisoning , Maternal Exposure/adverse effects , Panax/chemistry , Plant Extracts/pharmacology , Purkinje Cells/metabolism , Animals , Female , Lead Poisoning/drug therapy , Lead Poisoning/embryology , Lead Poisoning/pathology , Plant Extracts/chemistry , Pregnancy , Purkinje Cells/pathology , Rats
7.
Sci Rep ; 9(1): 8512, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186474

ABSTRACT

Brain-derived neurotrophic factor (BDNF) plays crucial roles in memory impairments including Alzheimer's disease (AD). Previous studies have reported that tetrasialoganglioside GQ1b is involved in long-term potentiation and cognitive functions as well as BDNF expression. However, in vitro and in vivo functions of GQ1b against AD has not investigated yet. Consequently, treatment of oligomeric Aß followed by GQ1b significantly restores Aß1-42-induced cell death through BDNF up-regulation in primary cortical neurons. Bilateral infusion of GQ1b into the hippocampus ameliorates cognitive deficits in the triple-transgenic AD mouse model (3xTg-AD). GQ1b-infused 3xTg-AD mice had substantially increased BDNF levels compared with artificial cerebrospinal fluid (aCSF)-treated 3xTg-AD mice. Interestingly, we also found that GQ1b administration into hippocampus of 3xTg-AD mice reduces Aß plaque deposition and tau phosphorylation, which correlate with APP protein reduction and phospho-GSK3ß level increase, respectively. These findings demonstrate that the tetrasialoganglioside GQ1b may contribute to a potential strategy of AD treatment.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/metabolism , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Gangliosides/therapeutic use , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Disease Models, Animal , Gangliosides/administration & dosage , Gangliosides/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Rats , Up-Regulation , tau Proteins/metabolism
8.
Am J Alzheimers Dis Other Demen ; 34(2): 104-111, 2019 03.
Article in English | MEDLINE | ID: mdl-30497273

ABSTRACT

BACKGROUND: The aim of this study was to identify white matter structural networks of amnestic mild cognitive impairment (aMCI) dichotomized by ß amyloid (Aß) status and compare them using network-based statistics (NBS). METHODS: Patients underwent whole-brain diffusion-weighted magnetic resonance imaging, detailed neuropsychological test and [18F]-Florbetaben amyloid positron emission tomography. We performed the NBS analysis to compare the whole-brain white matter structural networks extracted from diffusion tensor images. RESULTS: One hundred sixteen participants (Aß- cognitively normal [CN], n = 35; Aß- aMCI, n = 42; Aß+ aMCI, n = 39) were included. There was no subnetwork showing significant difference between Aß+ aMCI and Aß- aMCI. However, by comparing each aMCI group with control group, we found that supplementary motor areas were common hub regions. Intriguingly, Aß+ aMCI showed reduced connectivity mainly in the medial frontal regions, while Aß- aMCI showed somewhat uniform disruption when compared to CN. CONCLUSION: Structural network analysis using network-based approach in aMCI may shed light on further understanding of white matter disruption in the prodromal stage of Alzheimer's disease.


Subject(s)
Amnesia/metabolism , Amyloid/metabolism , Brain Mapping , Cognitive Dysfunction/metabolism , White Matter , Aged , Alzheimer Disease/metabolism , Aniline Compounds , Brain/metabolism , Cerebral Cortex/metabolism , Diffusion Tensor Imaging , Female , Humans , Male , Neuropsychological Tests/statistics & numerical data , Positron-Emission Tomography , Stilbenes
9.
Mol Pharm ; 16(1): 165-172, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30521347

ABSTRACT

A number of cancer-targeting peptide-drug conjugates (PDCs) have been explored as alternatives to antibody-drug conjugates (ADCs) for targeted cancer therapy. However, the much shorter circulation half-life of PDCs compared with ADCs in vivo has limited their therapeutic value and thus their translation into the clinic, highlighting the need to develop new approaches for extending the half-life of PDCs. Here, we report a new strategy for targeted cancer therapy of a PDC based on a molecular hybrid between an antihapten antibody and a hapten-labeled PDC. An anticotinine antibody (Abcot) was used as a model antihapten antibody. The anticancer drug SN38 was linked to a cotinine-labeled aptide specific to extra domain B of fibronectin (cot-APTEDB), yielding the model PDC, cot-APTEDB-SN38. The cotinine-labeled PDC showed specific binding to and cytotoxicity toward an EDB-overexpressing human glioblastoma cell line (U87MG) and also formed a hybrid complex (HC) with Abcot in situ, designated HC[cot-APTEDB-SN38/Abcot]. In glioblastoma-bearing mice, in situ HC[cot-APTEDB-SN38/Abcot] significantly extended the circulation half-life of cot-APTEDB-SN38 in blood, and it enhanced accumulation and penetration within the tumor and, ultimately, inhibition of tumor growth. These findings suggest that the present platform holds promise as a new, targeted delivery strategy for PDCs in anticancer therapy.


Subject(s)
Antibodies/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Peptides/chemistry , Animals , Cell Line, Tumor , Drug Delivery Systems , Female , Glioma/drug therapy , Humans , Immunoconjugates/chemistry , In Situ Nick-End Labeling , Irinotecan/chemistry , Irinotecan/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude
10.
Cancer Res ; 78(24): 6890-6902, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30352813

ABSTRACT

: Although cancer stem cells (CSC) are thought to be responsible for tumor recurrence and resistance to chemotherapy, CSC-related research and drug development have been hampered by the limited supply of diverse, patient-derived CSC. Here, we present a functional polymer thin film (PTF) platform that promotes conversion of cancer cells to highly tumorigenic three-dimensional (3D) spheroids without the use of biochemical or genetic manipulations. Culturing various human cancer cells on the specific PTF, poly(2,4,6,8-tetravinyl-2,4,6,8-tetramethyl cyclotetrasiloxane) (pV4D4), gave rise to numerous multicellular tumor spheroids within 24 hours with high efficiency and reproducibility. Cancer cells in the resulting spheroids showed a significant increase in the expression of CSC-associated genes and acquired increased drug resistance compared with two-dimensional monolayer-cultured controls. These spheroids also exhibited enhanced xenograft tumor-forming ability and metastatic capacity in nude mice. By enabling the generation of tumorigenic spheroids from diverse cancer cells, the surface platform described here harbors the potential to contribute to CSC-related basic research and drug development. SIGNIFICANCE: A new cell culture technology enables highly tumorigenic 3D spheroids to be easily generated from various cancer cell sources in the common laboratory.


Subject(s)
Neoplastic Stem Cells/cytology , Polymers/chemistry , Spheroids, Cellular/cytology , Animals , Carcinogenesis/metabolism , Cell Culture Techniques , Cell Line, Tumor , Female , Genome , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Materials Testing , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Recurrence, Local/pathology , Reproducibility of Results
11.
ACS Nano ; 12(7): 6904-6916, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29949348

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in psoriatic skin inflammation and acts as a key player in the pathogenesis and progression of this autoimmune disease. Although numerous inhibitors that intervene in STAT3-associated pathways have been tested, an effective, highly specific inhibitor of STAT3 has yet to be identified. Here, we evaluated the in vitro and in vivo biological activity and therapeutic efficacy of a high-affinity peptide specific for STAT3 (APTstat3) after topical treatment via intradermal and transcutaneous delivery. Using a preclinical model of psoriasis, we show that intradermal injection of APTstat3 tagged with a 9-arginine cell-penetrating peptide (APTstat3-9R) reduced disease progression and modulated psoriasis-related cytokine signaling through inhibition of STAT3 phosphorylation. Furthermore, by complexing APTstat3-9R with specific lipid formulations led to formation of discoidal lipid nanoparticles (DLNPs), we were able to achieve efficient skin penetration of the STAT3-inhibiting peptide after transcutaneous administration, thereby effectively inhibiting psoriatic skin inflammation. Collectively, these findings suggest that DLNP-assisted transcutaneous delivery of a STAT3-inhibiting peptide could be a promising strategy for treating psoriatic skin inflammation without causing adverse systemic events. Moreover, the DLNP system could be used for transdermal delivery of other therapeutic peptides.


Subject(s)
Drug Delivery Systems , Inflammation/drug therapy , Nanoparticles/chemistry , Peptides/pharmacology , Psoriasis/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Administration, Cutaneous , Animals , Cells, Cultured , Female , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Peptides/administration & dosage , Psoriasis/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Skin/drug effects , Skin/metabolism
12.
Chemistry ; 24(21): 5623-5629, 2018 Apr 11.
Article in English | MEDLINE | ID: mdl-29405457

ABSTRACT

Cysteine plays a crucial role in cellular functions and in human pathologies. However, the development of cysteine probes with extremely accurate detection is still a key challenge for the field. Herein, we have fully characterized and developed a novel selective fluorescent probe: red emission, aqueous detection and large Stokes' shift for cysteine (Reals-C). Key in the probe synthesis is a Michael addition onto an acroylate group and subsequent intramolecular cyclization. The probe exhibits analyte detection via an intricate role set up by the leaving groups so to discriminate and form the red-emissive analyte sensing platform (λex =471 nm, λem =637 nm) through a chemical cascade pathway. Furthermore, the sensing ability of the probe was demonstrated by both in vitro and in vivo assays. This probe enables for successfully endogenous cysteine sensing in HaCaT human keratinocytes through comparison with a commercial thiol-sensitive probe; Reals-C shows excellent in vivo cysteine detection in a drug-induced animal liver injury model.


Subject(s)
Cysteine/analysis , Fluorescent Dyes/chemistry , Animals , Chemical and Drug Induced Liver Injury , Cyclization , Cysteine/chemistry , Cysteine/metabolism , Disease Models, Animal , Fluorescent Dyes/chemical synthesis , Humans , Keratinocytes/drug effects , Spectrometry, Fluorescence/methods , Sulfhydryl Compounds/chemistry
13.
Anal Chem ; 90(4): 2648-2654, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29359562

ABSTRACT

The development of novel fluorescent probes for monitoring the concentration of various biomolecules in living systems has great potential for eventual early diagnosis and disease intervention. Selective detection of competitive species in biological systems is a great challenge for the design and development of fluorescent probes. To improve on the design of fluorescent coumarin-based biothiol sensing technologies, we have developed herein an enhanced dual emission doubly activated system (DACP-1 and the closely related DACP-2) for the selective detection of glutathione (GSH) through the use of one optical channel and the detection of cysteine (Cys) by another channel. A phenylselenium group present at the 4-position completely quenches the fluorescence of the probe via photoinduced electron transfer to give a nonfluorescent species. Probes are selective for glutathione (GSH) in the red region and for cysteine/homocysteine (Cys/Hcy) in the green region. When they were treated with GSH, DACP-1 and DACP-2 showed strong fluorescence enhancement in comparison to that for closely related species such as amino acids, including Cys/Hcy. Fluorescence quantum yields (ΦF) increased for the red channel (<0.001 to 0.52 (DACP-1) and 0.48 (DACP-2)) and green channel (Cys) (<0.001 to 0.030 (DACP-1) and 0.026 (DACP-2)), respectively. Competing fluorescent enhancements upon addition of closely related species were negligible. Fast responses, improved water solubility, and good cell membrane permeability were all properly established with the use of DACP-1 and DACP-2. Live human lung cancer cells and fibroblasts imaged by confocal microscopy, as well as live mice tumor model imaging, confirmed selective detection.


Subject(s)
Cysteine/analysis , Fibroblasts/chemistry , Fluorescent Dyes/chemistry , Glutathione/analysis , Lung Neoplasms/chemistry , Optical Imaging , Animals , Cell Survival/drug effects , Density Functional Theory , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/pharmacology , Humans , Injections, Intravenous , Lung Neoplasms/diagnostic imaging , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Molecular Structure , Neoplasms, Experimental/chemistry , Neoplasms, Experimental/diagnostic imaging , Tumor Cells, Cultured
14.
Mol Pharm ; 14(11): 3772-3779, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28969419

ABSTRACT

Tumor necrosis factor-α has shown potent antitumor effects in preclinical and clinical studies. However, severe side effects at less than therapeutic doses have limited its systemic delivery, prompting the need for a new strategy for targeted delivery of the protein to tumors. Here, we report a fusion protein of mouse tumor necrosis factor (TNF)-α (mTNFα) and a cancer-targeting, high-affinity aptide and investigate its therapeutic efficacy in tumor-bearing mice. A fusion protein consisting of mTNFα, a linker, and an aptide specific to extra domain B (EDB) of fibronectin (APTEDB), designated mTNFα-APTEDB, was successfully produced by expression in Escherichia coli. mTNFα-APTEDB retained specificity and affinity for its target, EDB. In mice bearing EDB-overexpressing fibrosarcomas, mTNFα-APTEDB showed greater efficacy in inhibiting tumor growth than mTNFα alone or mTNFα linked to a nonrelevant aptide, without causing an appreciable loss in body weight. Moreover, in vivo antitumor efficacy was further significantly increased by combination treatment with the chemotherapeutic drug, melphalan, suggesting a synergistic effect attributable to enhanced drug uptake into the tumor as a result of TNFα-mediated enhanced vascular permeability. These results suggest that a fusion protein of mTNFα with a cancer-targeting peptide could be a new anticancer therapeutic option for ensuring potent antitumor efficacy after systemic delivery.


Subject(s)
Fibronectins/metabolism , Peptides/metabolism , Recombinant Fusion Proteins/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Animals , Fibronectins/chemistry , Fibrosarcoma/drug therapy , Melphalan/chemistry , Melphalan/metabolism , Mice , Peptides/chemistry , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor-alpha/chemistry
15.
J Control Release ; 256: 56-67, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28428066

ABSTRACT

Although it has been shown that the size of nanoparticle-based vaccines is a key determining factor for the induction of immune responses, few studies have provided detailed analyses of thresholds or critical sizes of nanoparticle vaccines. Here we report effects of the size of gold nanoparticle (GNP)-based vaccines on their efficiency of delivery to lymph nodes (LNs) and induction of CD8+ T-cell responses. We further propose a threshold size of GNPs for use as an effective vaccine. To examine the effects of GNP size, we synthesized GNPs with diameters of 7, 14 and 28nm, and then conjugated them with recombinant ovalbumin (OVA) as a model antigen. The resulting OVA-GNPs had hydrodynamic diameter (HD) of ~10, 22, and 33nm for 7, 14 and 28nm GNPs, respectively and exhibited a size-dependent increase in cellular uptake by dendritic cells (DCs) and subsequent T-cell cross-priming and activation. Upon injection into a mouse footpad, both 22- and 33-nm OVA-GNPs showed much higher delivery efficiency to draining LNs than did 10-nm OVA-GNPs. An ex vivo restimulation assay using OVA as an antigen revealed that frequencies of OVA-specific CD8+ T cells were higher in mice immunized with 22- and 33-nm OVA-GNPs than in those immunized with 10-nm OVA-GNPs; moreover, these cells were shown to be poly-functional. In a tumor-prevention study, 22-nm OVA-GNPs showed greater antitumor efficacy, and higher infiltration of CD8+ T-cells and greater tumor cell apoptosis and cell death than 10-nm OVA-GNPs. Taken together, our results suggest that the size threshold for induction of potent cellular responses and T-cell poly-functionality by GNPs lies between 10nm and 22nm, and highlight the importance of nanoparticle size as a critical parameter in designing and developing nanoparticle-based vaccines.


Subject(s)
Antigens/administration & dosage , Gold/administration & dosage , Metal Nanoparticles/administration & dosage , Ovalbumin/administration & dosage , Vaccines/administration & dosage , Animals , Antigens/chemistry , Antigens/genetics , Cell Line , Cell Survival/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Escherichia coli/genetics , Female , Gold/chemistry , Lymph Nodes/metabolism , Metal Nanoparticles/chemistry , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/pathology , Ovalbumin/chemistry , Ovalbumin/genetics , Particle Size , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/drug effects , Vaccines/chemistry
16.
Biomaterials ; 123: 118-126, 2017 04.
Article in English | MEDLINE | ID: mdl-28167390

ABSTRACT

Although PEGylated liposomes (PEG-LS) have been intensively studied as drug-delivery vehicles, the rigidity and the hydrophilic PEG corona of liposomal membranes often limits cellular uptake, resulting in insufficient drug delivery to target cells. Thus, it is necessary to develop a new type of lipid-based self-assembled nanoparticles capable of enhanced cellular uptake, tissue penetration, and drug release than conventional PEGylated liposomes. Herein, we describe a simple modification of bicellar formulation in which the addition of a PEGylated phospholipid produced a dramatic physicochemical change in morphology, i.e., the disc-shaped bicelle became a uniformly distributed ultra-small (∼12 nm) spherical micelle. The transformed lipid-based nanoparticles, which we termed hyper-cell-permeable micelles (HCPMi), demonstrated not only prolonged stability in serum but also superior cellular and tumoral uptake compared to a conventional PEGylated liposomal system (PEG-LS). In addition, HCPMi showed rapid cellular uptake and subsequent cargo release into the cytoplasm of cancer cells. Cells treated with HCPMi loaded with docetaxel (DTX) had an IC50 value of 0.16 µM, compared with 0.78 µM with PEG-LS loaded with DTX, a nearly five-fold decrease in cell viability, indicating excellent efficiency in HCPMi uptake and release. In vivo tumor imaging analysis indicated that HCPMi penetrated deep into the tumor core and achieved greater uptake than PEG-LS. Results of HCPMi (DTX) treatment of allograft and xenograft mice in vivo showed high tumoral uptake and appreciable tumor retardation, with ∼70% tumor weight reduction in the SCC-7 allograft model. Taken together, these findings indicate that HCPMi could be developed further as a highly competent lipid-based drug-delivery system.


Subject(s)
Liposomes/chemistry , Nanocapsules/chemistry , Neoplasms, Experimental/chemistry , Neoplasms, Experimental/drug therapy , Taxoids/administration & dosage , Taxoids/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Docetaxel , Emulsions , Liposomes/ultrastructure , Mice , Mice, Inbred BALB C , Mice, Nude , Micelles , Nanocapsules/ultrastructure , Neoplasms, Experimental/pathology , Treatment Outcome
17.
Theranostics ; 6(13): 2367-2379, 2016.
Article in English | MEDLINE | ID: mdl-27877241

ABSTRACT

Combination of photodynamic therapy (PDT) with photothermal therapy (PTT) has achieved significantly improved therapeutic efficacy compared to a single phototherapy modality. However, most nanomaterials used for combined PDT/PTT are made of non-biodegradable materials (e.g., gold nanorods, carbon nanotubes, and graphenes) and may remain intact in the body for long time, raising concerns over their potential long-term toxicity. Here we report a new combined PDT/PTT nanomedicine, designated SP3NPs, that exhibit photo-decomposable, photodynamic and photothermal properties. SP3NPs were prepared by self-assembly of PEGylated cypate, comprising FDA-approved PEG and an ICG derivative. We confirmed the ability of SP3NPs to generate both singlet oxygen for a photodynamic effect and heat for photothermal therapy in response to NIR laser irradiation in vitro. Also, the unique ability of SP3NPs to undergo irreversible decomposition upon NIR laser irradiation was demonstrated. Further our experimental results demonstrated that SP3NPs strongly accumulated in tumor tissue owing to their highly PEGylated surface and relatively small size (~60 nm), offering subsequent imaging-guided combined PDT/PTT treatment that resulted in tumor eradication and prolonged survival of mice. Taken together, our SP3NPs described here may represent a novel and facile approach for next-generation theranostics with great promise for translation into clinical practice in the future.


Subject(s)
Hyperthermia, Induced/methods , Melanoma/diagnosis , Melanoma/therapy , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Optical Imaging/methods , Phototherapy/methods , Animals , Cell Line, Tumor , Heterografts , Humans , Infrared Rays , Lasers , Mice , Treatment Outcome
18.
ACS Nano ; 10(11): 9909-9918, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27792310

ABSTRACT

In vitro culture systems for primary neurons have served as useful tools for neuroscience research. However, conventional in vitro culture methods are still plagued by challenging problems with respect to applications to neurodegenerative disease models or neuron-based biosensors and neural chips, which commonly require long-term culture of neural cells. These impediments highlight the necessity of developing a platform capable of sustaining neural activity over months. Here, we designed a series of polymeric bilayers composed of poly(glycidyl methacrylate) (pGMA) and poly(2-(dimethylamino)ethyl methacrylate) (pDMAEMA), designated pGMA:pDMAEMA, using initiated chemical vapor deposition (iCVD). Harnessing the surface-growing characteristics of iCVD polymer films, we were able to precisely engraft acetylcholine-like functionalities (tertiary amine and quaternary ammonium) onto cell culture plates. Notably, pGD3, a pGMA:pDMAEMA preparation with the highest surface composition of quaternary ammonium, fostered the most rapid outgrowth of neural cells. Clear contrasts in neural growth and survival between pGD3 and poly-l-lysine (PLL)-coated surfaces became apparent after 30 days in vitro (DIV). Moreover, brain-derived neurotrophic factor level continuously accumulated in pGD3-cultured neurons, reaching a 3-fold increase at 50 DIV. Electrophysiological measurements at 30 DIV revealed that the pGD3 surface not only promoted healthy maturation of hippocampal neurons but also enhanced the function of hippocampal ionotropic glutamate receptors in response to synaptic glutamate release. Neurons cultured long-term on pGD3 also maintained their characteristic depolarization-induced Ca2+ influx functions. Furthermore, primary hippocampal neurons cultured on pGD3 showed long-term survival in a stable state up to 90 days-far longer than neurons on conventional PLL-coated surfaces. Taken together, our findings indicate that a polymer thin film with optimal acetylcholine-like functionality enables a long-term culture and survival of primary neurons.


Subject(s)
Acetylcholine/pharmacology , Cell Culture Techniques , Hippocampus/cytology , Neurons , Polymers , Cells, Cultured , Humans
19.
Nanotechnology ; 27(48): 48LT01, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27804918

ABSTRACT

Self-assembled nanoparticles (NPs) have been intensively utilized as cancer drug delivery carriers because hydrophobic anticancer drugs may be efficiently loaded into the particle cores. In this study, we synthesized and evaluated the therapeutic index of self-assembled NPs chemically conjugated to a fibronectin extra domain B-specific peptide (APTEDB) and an anticancer agent SN38. The APTEDB-SN38 formed self-assembled structures with a diameter of 58 ± 3 nm in an aqueous solution and displayed excellent drug loading, solubility, and stability properties. A pharmacokinetic study revealed that the blood circulation half-life of SN38 following injection of the APTEDB-SN38 NPs was markedly higher than that of the small molecule CPT-11. The APTEDB-SN38 NPs delivered SN38 to tumor sites by both passive and active targeting. Finally, the APTEDB-SN38 NPs exhibited potent antitumor activities and low toxicities against EDB-expressing tumors (LLC, U87MG) in mice. This system merits further preclinical and clinical investigations for SN38 delivery.


Subject(s)
Nanoparticles , Animals , Antineoplastic Agents , Cell Line, Tumor , Drug Carriers , Mice , Mice, Inbred BALB C , Neoplasms
20.
Chem Asian J ; 11(24): 3598-3605, 2016 Dec 19.
Article in English | MEDLINE | ID: mdl-27863045

ABSTRACT

Small-molecule organoselenium-based fluorescent probes possess great capacity in understanding biological processes through the detection of various analytes such as reactive oxygen/nitrogen species (ROS/RNS), biothiols (cysteine, homocysteine and glutathione), lipid droplets, etc. Herein, we present how substituents on the BODIPY system play a significant part in the detection of biologically important analytes for in vitro conditions and live cell imaging studies. The fluorescence of the probe was quenched by 2-chloro and 6-phenyl selenium groups; the probe shows high selectivity with NaOCl among other ROS/RNS, and gives a turn-on response. The maximum fluorescence intensity is attained within ≈1-2 min with a low detection limit (19.6 nm), and shows a ≈110-fold fluorescence enhancement compared to signals generated for other ROS/RNS. Surprisingly, in live cell experiments, the probe specifically located and accumulated in lipid droplets, and showed a fluorescence turn-on response. We believe this turn-on response occurred because of aggregation-induced emission (AIE), which surprisingly occurred only by introducing one lipophilic mesityl group at the meso position of the BODIPY.


Subject(s)
Boron Compounds/chemistry , Fluorescent Dyes/chemistry , Organoselenium Compounds/chemistry , Cell Line , Cell Survival/drug effects , Humans , Light , Microscopy, Confocal , Organoselenium Compounds/chemical synthesis , Organoselenium Compounds/therapeutic use , Quantum Theory , Reactive Nitrogen Species/chemistry , Reactive Oxygen Species/chemistry , Selenium/chemistry , Spectrometry, Fluorescence , Stem Cells/cytology , Stem Cells/drug effects , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...