Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
J Nanobiotechnology ; 22(1): 293, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802812

ABSTRACT

BACKGROUND: The exogenous delivery of miRNA to mimic and restore miRNA-34a activity in various cancer models holds significant promise in cancer treatment. Nevertheless, its effectiveness is often impeded by challenges, including a short half-life, propensity for off-target accumulation, susceptibility to inactivation by blood-based enzymes, concerns regarding patient safety, and the substantial cost associated with scaling up. As a means of overcoming these barriers, we propose the development of miRNA-loaded Tat-A86 nanoparticles by virtue of Tat-A86's ability to shield the loaded agent from external environmental factors, reducing degradation and inactivation, while enhancing circulation time and targeted accumulation. RESULTS: Genetically engineered Tat-A86, featuring 16 copies of the interleukin-4 receptor (IL-4R)-binding peptide (AP1), Tat for tumor penetration, and an elastin-like polypeptide (ELP) for presenting target ligands and ensuring stability, served as the basis for this delivery system. Comparative groups, including Tat-E60 and A86, were employed to discern differences in binding and penetration. The designed ELP-based nanoparticle Tat-A86 effectively condensed miRNA, forming stable nanocomplexes under physiological conditions. The miRNA/Tat-A86 formulation bound specifically to tumor cells and facilitated stable miRNA delivery into them, effectively inhibiting tumor growth. The efficacy of miRNA/Tat-A86 was further evaluated using three-dimensional spheroids of lewis lung carcinoma (LLC) as in vitro model and LLC tumor-bearing mice as an in vivo model. It was found that miRNA/Tat-A86 facilitates effective cell killing by markedly improving miRNA penetration, leading to a substantial reduction in the size of LLC spheroids. Compared to other controls, Tat-A86 demonstrated superior efficacy in suppressing the growth of 3D cellular aggregates. Moreover, at equivalent doses, miRNA-34a delivered by Tat-A86 inhibited the growth of LLC cells in allograft mice. CONCLUSIONS: Overall, these studies demonstrate that Tat-A86 nanoparticles can deliver miRNA systemically, overcoming the basic hurdles impeding miRNA delivery by facilitating both miRNA uptake and stability, ultimately leading to improved therapeutic effects.


Subject(s)
Elastin , MicroRNAs , Nanoparticles , Peptides , Animals , MicroRNAs/genetics , Elastin/chemistry , Mice , Peptides/chemistry , Humans , Nanoparticles/chemistry , Cell Line, Tumor , Neoplasms/therapy , Neoplasms/drug therapy , Drug Carriers/chemistry , Female , Elastin-Like Polypeptides
2.
Int J Nanomedicine ; 16: 5039-5052, 2021.
Article in English | MEDLINE | ID: mdl-34335025

ABSTRACT

BACKGROUND: Thermal-responsive self-assembled elastin-like polypeptide (ELP)-based nanoparticles are an emerging platform for controlled delivery of therapeutic peptides, proteins and small molecular drugs. The antitumor effect of bioengineered chimeric polypeptide AP1-ELP-KLAK containing an interleukin-4 receptor (IL-4R) targeting peptide and pro-apoptotic peptide (KLAKLAK) was evaluated in glioblastoma (GBM) in vitro and in vivo. METHODS AND RESULTS: Herein, the therapeutic effect of AP1-ELP-KLAK was tested in advanced, and less curable glioblastoma cells with higher expression of IL-4R. Glioblastoma cell lines stably expressing different reporter systems i.e., caspase-3 sensor (surrogate marker for cellular apoptosis) or effluc/enhanced firefly luciferase (cellular viability) were established to measure cell death non-invasively. Bioluminescence imaging (BLI) of D54/effluc and U97MG/effluc treated with AP1-ELP-KLAK exhibited higher cell death up to 2~3-fold than the control. Treatment with AP1-ELP-KLAK resulted in time-dependent increase of caspase-3 sensor BLI activity in D54/C cells and D54/C tumor-bearing mice. Intravenous injection of AP1-ELP-KLAK dramatically reduced tumor growth by inducing cellular apoptosis in D54/effluc tumor-bearing mice. Further, the immuno-histological examination of the excised tumor tissue confirmed the presence of apoptotic cells as well as caspase-3 activation. CONCLUSION: Collectively, AP1-ELP-KLAK effectively induced cellular apoptosis of glioblastoma cells and non-invasive imaging provides a window for real-time monitoring of anti-tumor effect with the provision of improving therapeutic efficacy in a glioblastoma mice model.


Subject(s)
Glioblastoma , Animals , Cell Line, Tumor , Glioblastoma/drug therapy , Humans , Mice , Peptides , Receptors, Interleukin-4
3.
Nanotheranostics ; 4(2): 57-70, 2020.
Article in English | MEDLINE | ID: mdl-32190533

ABSTRACT

In order to improve clinical outcomes for novel drug delivery systems, distinct optimization of size, shape, multifunctionality, and site-specificity are of utmost importance. In this study, we designed various multivalent elastin-like polypeptide (ELP)-based tumor-targeting polymers in which multiple copies of IL-4 receptor (IL-4R)-targeting ligand (AP1 peptide) were periodically incorporated into the ELP polymer backbone to enhance the affinity and avidity towards tumor cells expressing high levels of IL-4R. Several ELPs with different molecular sizes and structures ranging from unimer to micelle-forming polymers were evaluated for their tumor accumulation as well as in vivo bio-distribution patterns. Different percentages of cell binding and uptake were detected corresponding to polymer size, number of targeting peptides, or unimer versus micelle structure. As compared to low molecular weight polypeptides, high molecular weight AP1-ELP showed superior binding activity with faster entry and efficient processing in the IL-4R-dependent endocytic pathway. In addition, in vivo studies revealed that the high molecular weight micelle-forming AP1-ELPs (A86 and A100) displayed better tumor penetration and extensive retention in tumor tissue along with reduced non-specific accumulation in vital organs, when compared to low molecular weight non-micelle forming AP1-ELPs. It is suggested that the superior binding activities shown by A86 and A100 may depend on the multiple presentation of ligands upon transition to a micelle-like structure rather than a larger molecular weight. Thus, this study has significance in elucidating the different patterns underlying unimer and micelle-forming ELP-mediated tumor targeting as well as the in vivo biodistribution.


Subject(s)
Antineoplastic Agents , Drug Carriers , Elastin , Neoplasms/metabolism , Peptides , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Carriers/pharmacokinetics , Elastin/chemistry , Elastin/metabolism , Elastin/pharmacokinetics , Female , Humans , Mice, Inbred BALB C , Micelles , Molecular Weight , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacokinetics , Protein Conformation , Receptors, Interleukin-4/chemistry , Receptors, Interleukin-4/metabolism , Tissue Distribution , Xenograft Model Antitumor Assays
4.
ACS Biomater Sci Eng ; 6(9): 5024-5031, 2020 09 14.
Article in English | MEDLINE | ID: mdl-33455295

ABSTRACT

Acquired drug resistance is a common occurrence and the main cause of melanoma treatment failure. Melanoma cells frequently developed resistance against cisplatin during chemotherapy, and thus, targeting delivery systems have been devised to decrease drug resistance, increase therapeutic efficacy, and reduce side effects. We genetically engineered a macromolecular carrier using the recursive directional ligation method that specifically targets cisplatin-resistant (Cis-R) melanoma. This carrier is composed of an elastin-like polypeptide (ELP) and multiple copies of Cis-R melanoma-targeting ligands (M-peptide). The designed M16E108 contains 16 targeting ligands incorporated within an ELP and has an ideal thermal phase transition at 39 °C. When treated to melanoma cells, M16E108 specifically accumulated in Cis-R B16F10 melanoma cells and accumulated to a lesser extent in parental B16F10 cells. Consistently, M16E108 exhibited efficient homing and longer retention in tumor tissues in Cis-R melanoma-bearing mice than in parental B16F10 melanoma-bearing mice. Thus, M16E108 was found to display considerable potential as a novel agent that specifically targets cisplatin-resistant melanoma.


Subject(s)
Elastin , Melanoma , Animals , Cisplatin/pharmacology , Elastin/genetics , Ligands , Melanoma/drug therapy , Mice , Peptides
5.
Sci Rep ; 8(1): 16070, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30375433

ABSTRACT

Exosomes are small extracellular membrane vesicles released from endosomes of various cells and could be found in most body fluids. The main functions of exosomes have been recognized as important mediators of intercellular communication and as potential biomarkers of various disease states. This study investigated whether exogenous exosomes from mice with acetaminophen (APAP)-induced liver injury can damage the recipient hepatic cells or promote hepatotoxicity in mice. We observed that exogenous exosomes derived from APAP-exposed mice were internalized into the primary mouse hepatocytes or HepG2 hepatoma cells and significantly decreased the viability of these recipient cells. They also elevated mRNA transcripts and proteins associated with the cell death signaling pathways in primary hepatocytes or HepG2 cells via exosomes-to-cell communications. In addition, confocal microscopy of ex vivo liver section showed that exogenously added exosomes were accumulated in recipient hepatocytes. Furthermore, plasma reactive oxygen species and hepatic TNF-α/IL-1ß production were elevated in APAP-exosomes recipient mice compared to control-exosomes recipient mice. The levels of apoptosis-related proteins such as phospho-JNK/JNK, Bax, and cleaved caspase-3 were increased in mouse liver received APAP-exosomes. These results demonstrate that exogenous exosomes from APAP-exposed mice with acute liver injury are functional and stimulate cell death or toxicity of the recipient hepatocytes and mice.


Subject(s)
Acetaminophen/adverse effects , Chemical and Drug Induced Liver Injury/genetics , Exosomes/genetics , Hepatocytes/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Exosomes/drug effects , Extracellular Vesicles/drug effects , Hep G2 Cells , Hepatocytes/pathology , Humans , Interleukin-1beta/genetics , Liver/drug effects , Liver/pathology , Mice , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Transplant Recipients , Tumor Necrosis Factor-alpha/genetics
6.
Transl Oncol ; 11(4): 1044-1052, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29982102

ABSTRACT

Accurate and timely visualization of apoptotic status in response to radiation is necessary for deciding whether to continue radiation or change to another mode of treatment. This is especially critical in patients with colorectal cancer, which requires a delicate combination of surgery, radiation, and chemotherapy in order to achieve optimal outcome. In this study, we investigated the potential of phosphatidylserine-recognizing peptide 1 (PSP1) as an apoptosis-targeting probe, which identifies phosphatidylserine on cell surfaces. We first screened colon cancer cell lines for their sensitivity to radiation and selected two cell lines: HCT116 and HT29. Cell binding assay using fluorescence-activated cell sorting and optical imaging showed that HCT116 cells had better binding to PSP1 than HT29 cells. Thus, mouse xenograft model using HCT116 cells was generated and was topically irradiated with either single or fractionated dose of radiation followed by systemic administration of PSP1 for subsequent molecular optical imaging. We confirmed that the PSP1 probe was selectively bound to apoptosis-induced tumor in a radiation dose-dependent manner. We also observed that fractionated radiation regimen, which is recently being used in clinical situation, was more effective in inducing tumor apoptosis than corresponding single-dose radiation treatment. We then evaluated the correlation between tumor targeting of PSP1 and suppression effect of tumor development and found that tumor volume and fluorescence intensity were correlated before (correlation coefficient r2 = 0.534) and after (r2 = 0.848) radiation therapy. Our study shows that PSP1 peptide is an efficient index probe for deciding "go or no-go" for radiation therapy in colorectal cancer.

7.
Sci Rep ; 8(1): 3892, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29497090

ABSTRACT

Expression of various molecules on the surface of cancer cells compared to normal cells creates a platform for the generation of various drug vehicles for targeted therapy. Multiple interactions between ligands and their receptors mediated by targeting peptide-modified polymer could enable simultaneous delivery of a drug selectively to target tumor cells, thus limiting side effects resulting from non-specific drug delivery. In this study, we synthesized a novel tumor targeting system by using two key elements: (1) Bld-1 peptide (SNRDARRC), a recently reported bladder tumor targeting peptide identified by using a phage-displayed peptide library, and (2) ELP, a thermally responsive polypeptide. B5V60 containing five Bld-1 peptides and non-targeted ELP77 with a thermal phase-transition over 37 °C were analyzed to determine their bioactivities. Further studies confirmed the superior binding ability of B5V60 to bladder tumor cells and the cellular accumulation of B5V60 in cancer cells was dependent on the expression level of sialyl-Tn antigen (STn), a tumor-associated carbohydrate antigen. Additionally, B5V60 displayed excellent localization in bladder tumor xenograft mice after intravenous injection and was strictly confined to sialyl-Tn antigen-overexpressing tumor tissue. Thus, our newly designed B5V60 showed high potential as a novel carrier for STn-specific targeted cancer therapy or other therapeutic applications.


Subject(s)
Drug Delivery Systems/methods , Molecular Targeted Therapy/methods , Peptides/pharmacology , Amino Acid Sequence/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Elastin/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Peptides/chemistry , Peptides/genetics , Protein Binding/physiology , Urinary Bladder Neoplasms/metabolism
8.
Nanomedicine ; 14(3): 633-642, 2018 04.
Article in English | MEDLINE | ID: mdl-29309907

ABSTRACT

The use of thrombolytic therapies is limited by an increased risk of systemic hemorrhage due to lysis of hemostatic clots. We sought to develop a plasmin-based thrombolytic nanocage that efficiently dissolves the clot without causing systemic fibrinolysis or disrupting hemostatic clots. Here, we generated a double chambered short-length ferritin (sFt) construct that has an N-terminal region fused to multivalent clot targeting peptides (CLT: CNAGESSKNC) and a C-terminal end fused to a microplasmin (µPn); CLT recognizes fibrin-fibronectin complexes in clots, µPn efficiently dissolves clots, and the assembly of double chambered sFt (CLT-sFt-µPn) into nanocage structure protects the activated-µPn from its circulating inhibitors. Importantly, activated CLT-sFt-µPn thrombolytic nanocage showed a prolonged circulatory life over activated-µPn and efficiently lysed the preexisting clots in both arterial and venous thromboses models. Thus, CLT-sFt-µPn thrombolytic nanocage platform represents the prototype of a targeted clot-busting agent with high efficacy and safety over existing thrombolytic therapies.


Subject(s)
Coronary Thrombosis/prevention & control , Ferritins/chemistry , Fibrinolysin/chemistry , Fibrinolytic Agents/administration & dosage , Nanoparticles/administration & dosage , Peptide Fragments/chemistry , Thrombolytic Therapy/methods , Venous Thrombosis/prevention & control , Animals , Coronary Thrombosis/pathology , Disease Models, Animal , Fibrinolytic Agents/chemistry , Male , Mice , Mice, Inbred ICR , Nanoparticles/chemistry , Rats , Rats, Sprague-Dawley , Venous Thrombosis/pathology
9.
Biomol Ther (Seoul) ; 25(4): 367-373, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28208010

ABSTRACT

This study was performed to evaluate whether microRNAs (miRNAs) in circulating exosomes may serve as biomarkers of drug-induced liver, kidney, or muscle-injury. Quantitative PCR analyses were performed to measure the amounts of liver-specific miRNAs (miR-122, miR-192, and miR-155), kidney-specific miR-146a, or muscle-specific miR-206 in plasma and exosomes from mice treated with liver, kidney or muscle toxicants. The levels of liver-specific miRNAs in circulating plasma and exosomes were elevated in acetaminophen-induced liver injury and returned to basal levels by treatment with antioxidant N-acetyl-cysteine. Circulating miR-146a and miR-206 were increased in cisplatin-induced nephrotoxicity and bupivacaine-induced myotoxicity, respectively. Taken together, these results indicate that circulating plasma and exosomal miRNAs can be used as potential biomarkers specific for drug-induced liver, kidney or muscle injury.

10.
J Appl Toxicol ; 37(5): 554-562, 2017 05.
Article in English | MEDLINE | ID: mdl-27682001

ABSTRACT

Perfluorooctanoic acid (PFOA) has wide applications, including as a raw material for converted paper and packaging products. With the widespread use of PFOA, concerns regarding its potential environmental and health impacts have increased. In spite of the known hepatotoxicity and genotoxicity of PFOA, correlation with PFOA and allergic inflammation is not well known. In this study, the effect of PFOA on the degranulation of mast cells and mast cell-mediated allergic inflammation in the presence of FcεRI cross-linking was evaluated. In immunoglobulin (Ig) E-stimulated mast cells, PFOA increased the release of histamine and ß-hexosaminidase by the up-regulation of intracellular calcium levels. PFOA enhanced gene expression of several pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, and IL-8 by the activation of nuclear factor (NF)-κB in IgE-stimulated mast cells. Also, PFOA exacerbated allergic symptoms via hypothermia, and an increase of serum histamine, TNF-α, IgE and IgG1 in the ovalbumin-induced systemic anaphylaxis. The present data indicate that PFOA aggravated FcɛRI-mediated mast cell degranulation and allergic symptoms. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Caprylates/toxicity , Cell Degranulation/drug effects , Fluorocarbons/toxicity , Hypersensitivity/pathology , Inflammation/pathology , Mast Cells/drug effects , Anaphylaxis/chemically induced , Anaphylaxis/pathology , Animals , Calcium/metabolism , Cell Survival/drug effects , Cytokines/biosynthesis , Histamine Release/drug effects , Immunoglobulin E/immunology , Immunoglobulin Fc Fragments/metabolism , Male , Mast Cells/pathology , Mice , Mice, Inbred ICR , beta-N-Acetylhexosaminidases/metabolism
11.
Theranostics ; 6(12): 2235-2249, 2016.
Article in English | MEDLINE | ID: mdl-27924160

ABSTRACT

Elastin-like polypeptide (ELP)-based drug delivery has been utilized for various applications including cancer therapies for many years. Genetic incorporation of internalization ligands and cell-targeting peptides along with ELP polymer enhanced tumor accumulation and retention time as well as stability and activities of the drug conjugates. Herein, we described a unique delivery system comprised of genetically engineered ELP incorporated with multiple copies of IL-4 receptor targeting peptide (AP1) periodically and proapoptotic peptide (KLAKLAK)2 referred to as AP1-ELP-KLAK. It triggered thermal-responsive self-assembly into a nanoparticle-like structure at physiological body temperature and stabilized its helical conformation, which is critical for its membrane-disrupting activities. Increased IL-4 receptor specific cellular internalization was associated with the enhanced cytotoxic effect of (KLAKLAK)2 peptide. Additionally, multivalent presentation of targeting ligands by AP1-ELP-KLAK significantly enhanced intratumoral localization and prolonged the retention time compared to ELP-KLAK, non-targeted control. Systemic administration of AP1-ELP-KLAK significantly inhibited tumor growth by provoking cell apoptosis in various tumor xenograft models without any specific organ toxicity. Thus, our newly designed AP1-ELP-KLAK polymer nanoparticle is a promising candidate for effective cancer therapy and due to the simple preparative procedures of ELPs, this platform can be used as a good carrier for tumor-specific delivery of other therapeutics.


Subject(s)
Antimicrobial Cationic Peptides/administration & dosage , Antineoplastic Agents/administration & dosage , Drug Carriers/administration & dosage , Elastin/administration & dosage , Nanoparticles/administration & dosage , Neoplasms/therapy , Peptides/administration & dosage , Animals , Antimicrobial Cationic Peptides/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Elastin/metabolism , Female , Heterografts , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/metabolism , Protein Multimerization , Treatment Outcome
12.
Brain Res ; 1646: 514-521, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27369450

ABSTRACT

With the increasing worldwide incidence of Alzheimer's disease (AD), there is a critical need for the discovery of more effective diagnostic methods. However, development of diagnostic tools in AD has been hindered by obstacles such as the absence of exact biomarkers. Apoptosis caused by amyloid-ß (Aß) plays an important role in AD pathology; therefore, provides an attractive biological target for the diagnosis of AD. The present study aimed to evaluate the potential of small peptide, named ApoPep-1 (Apoptosis-targeting peptide-1) as a new apoptosis imaging agent in AD. The fluorescein-conjugated ApoPep-1, but not the control peptide, targeted apoptotic cells in the brain of amyloid precursor protein (APP)/presenilin 1 (PS1) mice. We also observed fluorescence signals during in vivo imaging of apoptotic cells using ApoPep-1, and fluorescence levels increased in an age-dependent manner in APP/PS1 mice. Ex vivo imaging of isolated brains in APP/PS1 mice further confirmed the targeting of ApoPep-1 to apoptotic cells. The fluorescein-labeled ApoPep-1 co-localized with brain cells such as neurons, astrocytes, and microglia, all of which undergo apoptosis in the APP/PS1 mice brain. These findings demonstrate that ApoPep-1 can target apoptotic brain cells, and be used for experimental investigations relevant to apoptosis in AD.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/administration & dosage , Apoptosis , Brain/physiopathology , Immunohistochemistry/methods , Oligopeptides/administration & dosage , Peptide Fragments/administration & dosage , Alzheimer Disease/metabolism , Animals , Astrocytes/metabolism , Astrocytes/physiology , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Fluorescein/administration & dosage , Fluorescent Dyes/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/physiology , Neurons/metabolism , Neurons/physiology , Oligopeptides/metabolism
13.
Int Arch Allergy Immunol ; 170(1): 35-45, 2016.
Article in English | MEDLINE | ID: mdl-27355819

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is the most common allergic inflammatory skin disease. The activation of innate immunity by house dust mite (Dermatophagoides farinae extract, DFE) allergen plays an important role in the pathogenesis of AD. We previously showed the inhibitory effect of an extract of Amomum xanthioides on allergic diseases, and isolated 1,2,4,5-tetramethoxybenzene (TMB) as a major active component. In this study, we investigated whether TMB relieves DFE-induced allergic inflammation symptoms. METHODS: We established a DFE-induced allergic inflammation model in BALB/c mice by repeated skin exposure to DFE. To define the underlying mechanisms of action, we used a tumor necrosis factor-α and interferon-x03B3;-activated human keratinocytes (HaCaT cell line) and mouse keratinocytes (3PC cell line) cell line model. RESULTS: Oral administration of TMB suppressed allergic inflammation symptoms, such as histopathological analysis and ear thickness, in addition to serum IgE, DFE-specific IgE and IgG2a levels. TMB decreased the serum histamine levels and tissue infiltration of inflammatory cells, including mast cells and eosinophils. TMB also inhibited CD4+IFN-x03B3;+, CD4+IL-4+, and CD4+IL-17A+ lymphocyte expansion in the draining lymph nodes and expression of the Th2 cytokines in the ear tissue. TMB significantly inhibited the expression of cytokines and chemokines by the downregulation of the mitogen-activated protein kinases and nuclear factor of activated cytoplasmic T cells in HaCaT cells. CONCLUSIONS: TMB improved DFE-induced allergic inflammation by suppressing the production of proinflammatory cytokines and chemokines. Our results suggest that TMB might be a potential therapeutic agent for AD.


Subject(s)
Allergens/immunology , Anisoles/pharmacology , Hypersensitivity/immunology , Hypersensitivity/pathology , Pyroglyphidae/immunology , Animals , Cell Line , Cytokines/blood , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Disease Models, Animal , Female , Histamine Release , Humans , Hypersensitivity/drug therapy , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunophenotyping , Keratinocytes/metabolism , Mice , Mice, Inbred BALB C , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
14.
Oncol Rep ; 36(1): 131-6, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27220374

ABSTRACT

Gas (SF6)-filled microbubbles (MBs) were prepared by emulsion and solvent-evaporation method. The prepared MBs were further conjugated with doxorubicin (Dox)-loaded nano-sized liposome and peptide ligands to interleukin-4 receptor (IL4R) for targeting brain tumor cells. The final MB-liposome (Dox)-IL4R targeting peptide ligand [MB-Lipo (Dox)-IL4RTP] had a spherical structure with the mean size of 1,500 nm. The MB-Lipo (Dox)­IL4RTP exhibited cellular uptake in U87MG brain tumor cells (a brain tumor cell line expressing strongly IL4R) with frequency ultrasound energy suggesting that MB-Lipo (Dox)­IL4RTP provided effective targeting ability for brain tumor cells. In addition, WST-1 assay results showed that MB-Lipo (Dox)­IL4RTP inhibited the proliferation of U87MG cells IL4R­dependently. This was confirmed by western blotting of γH2AX, phospho (Ser15)-p53, p53 and p21 which are signal transduction proteins involved in DNA damage response and cell cycle arrest. Taken together, these results indicate that MB-Lipo (Dox)-IL4RTP represents a promising ultrasonic contrast agent for tumor-targeting ultrasonic imaging.


Subject(s)
Brain Neoplasms/drug therapy , Doxorubicin/analogs & derivatives , Drug Carriers/therapeutic use , Interleukin-4 Receptor alpha Subunit/metabolism , Microbubbles/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair/genetics , Doxorubicin/therapeutic use , Histones/metabolism , Humans , Liposomes/therapeutic use , Peptides/therapeutic use , Polyethylene Glycols/therapeutic use , Tumor Suppressor Protein p53/metabolism , Ultrasonography
15.
Biomacromolecules ; 17(3): 1150-9, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26899206

ABSTRACT

Ferritin cage nanoparticles are promising platforms for targeted delivery of imaging and therapeutic agents because their cage structure can accommodate small molecules and their surfaces can be decorated with multiple functionalities. However, selective targeting is still a challenge for translating ferritin-based nanomedicines into the clinic, especially for heterogeneous diseases such as cancer. Targeting peptides can be genetically fused onto the surface of a ferritin cage, forming peptide bunches on nanocages (PBNCs) that offer synergistic increases in binding avidity. Here, we utilized two sites of the ferritin monomer, the N-terminus and the loop between the fourth and fifth helices, which are exposed on the surface of the assembled 24-subunit ferritin cage, to ligate one or two types of peptides to achieve "super affinity" and bispecificity, respectively. PBNCs formed by ligation of the IL-4 receptor-targeting peptide, AP1, to both sites (48AP1-PBNCs) tethered IL-4R, expressing tumor cells with greater affinity than did PBNCs with AP1 ligated to a single site (24AP1-PBNCs). Moreover, bispecific PBNCs containing 24 RGD peptides and 24 AP1 peptides (24RGD/24AP1-PBNCs) were capable of independently targeting cells expressing the corresponding receptors. Bispecific and superaffinity PBNCs could be useful for efficient targeting of ferritin-based therapeutic/diagnostic agents in a clinical setting.


Subject(s)
Ferritins/chemistry , Metal Nanoparticles/chemistry , Oligopeptides/chemistry , Cell Line, Tumor , Humans , Ligands , Oligopeptides/metabolism , Protein Binding , Receptors, Interleukin-4/metabolism
16.
J Control Release ; 231: 68-76, 2016 06 10.
Article in English | MEDLINE | ID: mdl-26780174

ABSTRACT

This work demonstrates the development of magnetically guided drug delivery systems and its potential on efficient anticancer therapy. The magnetically guided drug delivery system was successfully developed by utilizing superparamagnetic iron oxide nanoparticle, ß-cyclodextrin, and polymerized paclitaxel. Multivalent host-guest interactions between ß-cyclodextrin-conjugated superparamagnetic iron oxide nanoparticle and polymerized paclitaxel allowed to load the paclitaxel and the nanoparticle into the nano-assembly. Clusterized superparamagnetic iron oxide nanoparticles in the nano-assembly permitted the rapid and efficient targeted drug delivery. Compared to the control groups, the developed nano-assembly showed the enhanced anticancer effects in vivo as well as in vitro. Consequently, the strategy of the use of superparamagnetic nanoparticles and multivalent host-guest interactions has a promising potential for developing the efficient drug delivery systems.


Subject(s)
Antineoplastic Agents/chemistry , Magnetite Nanoparticles/chemistry , Paclitaxel/chemistry , beta-Cyclodextrins/chemistry , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Magnetics , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Paclitaxel/pharmacology , Particle Size , Polymers/chemistry , Surface Properties , beta-Carotene 15,15'-Monooxygenase/immunology
17.
Biomacromolecules ; 17(1): 12-9, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26646195

ABSTRACT

Ferritin cage nanoparticles are promising platforms for targeted delivery of imaging and therapeutic agents. One of the main advantages of cage nanoparticles is the ability to display multiple functionalities through genetic modification so as to achieve desired therapeutic or diagnostic functions. Ferritin complexes formed from short ferritin (sFt) lacking the fifth helix can accommodate dual peptide and protein functionalities on N- and C-terminal sites in sFt monomers. The resulting double-chambered NanoCage (DCNC) offers the potential of dual activities; these activities are augmented by the avidity of the ligands, which do not impede each other's function. Here we demonstrated proof-of-concept of DCNCs, loading the tumor-targeting proapoptotic peptide CGKRK(KLAKLAK)2 onto the N-terminal chamber and green fluorescent protein (GFP) onto the C-terminal chamber. The resulting KLAK-sFt-GFP DCNCs were internalized into the human breast adenocarcinoma cell line MDA-MB-231 and induced apoptosis. These findings suggest that DCNCs containing various combinations of peptides and proteins could be applied as therapeutics in different diseases.


Subject(s)
Ferritins/chemistry , Green Fluorescent Proteins/chemistry , Peptides/chemistry , Animals , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Humans , Ligands , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanotechnology/methods
18.
Arthritis Res Ther ; 17: 309, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26530111

ABSTRACT

INTRODUCTION: Current methods for early diagnosis of osteoarthritis (OA) are limited. We assessed whether in vivo detection of chondrocyte death by ApoPep-1 (CQRPPR), a peptide that binds to histone H1 of apoptotic and necrotic cells, could be used to detect the initiation of OA. METHODS: Apoptosis-induced ATDC5 cells were labeled with Annexin V and ApoPep-1. Surgical destabilization of the medial meniscus (DMM) was performed on both knees of 12-week-old male mice and severity of OA was determined by histological analysis according to the Osteoarthritis Research Society International (OARSI) guidelines. At 1, 2, 4, and 8 weeks post-surgery, mice were intravenously injected with fluorescence-labeled ApoPep-1 or control peptide and in vivo imaging was performed within 30 minutes of injection by near-infrared fluorescence (NIRF). Binding of ApoPep-1 to OA joints was demonstrated by ex vivo imaging and immunofluorescent staining using TUNEL and histone H1 and type II collagen antibodies. RESULTS: Strong signals of ApoPep-1 were observed on the apoptotic ATDC5 cells. Knees corresponded to grade II, III, and V OA at 2, 4, and 8 weeks after DMM, respectively. Between 2 and 8 weeks after surgery, the in vivo NIRF signal at OA-ApoPep1-injected joints was consistently stronger than sham-operated or OA-control peptide-injected joints. ApoPep-1, TUNEL, and histone H1 signals were stronger in grade II OA cartilage than sham-operated cartilage when detected by immunofluorescent staining. Type II collagen expression was similar between grade II OA and sham group. CONCLUSION: ApoPep-1 can be used to detect OA in vivo by binding to apoptotic chondrocytes. This is a novel, sensitive, and rapid method which can detect apoptotic cells in OA rodent models soon after its onset.


Subject(s)
Chondrocytes/pathology , Diagnostic Imaging/methods , Early Diagnosis , Oligopeptides/metabolism , Osteoarthritis/diagnosis , Animals , Apoptosis , Disease Models, Animal , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL
19.
Anal Sci ; 31(7): 699-704, 2015.
Article in English | MEDLINE | ID: mdl-26165294

ABSTRACT

In this study, a simple, highly sensitive electrochemical biosensor for myoglobin was developed using a myoglobin-specific binding peptide as a sensing probe. A peptide (Myo-3R7, CPSTLGASC, 838 Da) identified by phage display and that specifically binds to myoglobin was covalently immobilized on a gold electrode functionalized via a dithiobis(succinimidyl propionate) (DSP) self-assembled monolayer (SAM). The peptide immobilization was confirmed with fluorescence microarray scanning and cyclic voltammetry (CV). The electrochemical performance of the biosensor with respect to myoglobin was characterized by CV and differential pulse voltammetry (DPV) using Fe(CN)6(3-)/Fe(CN)6(4-) as a redox probe. We successfully detected myoglobin in a broad working range of 17.8 to 1780 ng mL(-1) with a correlation coefficient (R(2)) of 0.998. The estimated limit of detection (LOD) was fairly low, 9.8 ng mL(-1) in 30 min. The electrochemical biosensor based on a myoglobin-specific binding peptide offers sensitivity, selectivity, and rapidity, making it an attractive tool for the early detection of cardiac infarction.


Subject(s)
Biosensing Techniques/methods , Myocardial Infarction/diagnosis , Myoglobin/metabolism , Oligopeptides/metabolism , Acute Disease , Amino Acid Sequence , Early Diagnosis , Electrochemistry , Ferricyanides/chemistry , Gold/chemistry , Humans , Limit of Detection , Models, Molecular , Myoglobin/chemistry , Oligopeptides/chemistry , Protein Conformation , Substrate Specificity , Succinimides/chemistry , Time Factors
20.
J Control Release ; 209: 327-36, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25979323

ABSTRACT

A growing body of evidence suggests that pathological lesions express tissue-specific molecular targets or biomarkers within the tissue. Interleukin-4 receptor (IL-4R) is overexpressed in many types of cancer cells, including lung cancer. Here we investigated the properties of IL-4R-binding peptide-1 (IL4RPep-1), a CRKRLDRNC peptide, and its ability to target the delivery of liposomes to lung tumor. IL4RPep-1 preferentially bound to H226 lung tumor cells which express higher levers of IL-4R compared to H460 lung tumor cells which express less IL-4R. Mutational analysis revealed that C1, R2, and R4 residues of IL4RPep-1 were the key binding determinants. IL4RPep-1-labeled liposomes containing doxorubicin were more efficiently internalized in H226 cells and effectively delivered doxorubicin into the cells compared to unlabeled liposomes. In vivo fluorescence imaging of nude mice subcutaneously xenotransplanted with H226 tumor cells indicated that IL4RPep-1-labeled liposomes accumulate more efficiently in the tumor and inhibit tumor growth more effectively compared to unlabeled liposomes. Interestingly, expression of IL-4R was high in vascular endothelial cells of tumor, while little was detected in vascular endothelial cells of control organs including the liver. IL-4R expression in cultured human vascular endothelial cells was also up-regulated when activated by a pro-inflammatory cytokine tumor necrosis factor-α. Moreover, the up-regulation of IL-4R expression was observed in primary human lung cancer tissues. These results indicate that IL-4R-targeting nanocarriers may be a useful strategy to enhance drug delivery through the recognition of IL-4R in both tumor cells and tumor endothelial cells.


Subject(s)
Lung Neoplasms/metabolism , Oligopeptides/administration & dosage , Receptors, Interleukin-4/metabolism , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Liposomes , Lung Neoplasms/drug therapy , Mice, Nude , Oligopeptides/pharmacology , Oligopeptides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...