Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Virology ; 595: 110098, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38705084

ABSTRACT

Acinetobacter baumannii is one of the most important pathogens of healthcare-associated infections. The rising prevalence of multidrug-resistant A. baumannii (MRAB) strains and biofilm formation impact the outcome of conventional treatment. Phage-related therapy is a promising strategy to tame troublesome multidrug-resistant bacteria. Here, we isolated and evaluated a highly efficient lytic phage called MRABP9 from hospital sewage. The phage was a novel species within the genus Friunavirus and exhibited lytic activity against 2 other identified MRAB strains. Genomic analysis revealed it was a safe virulent phage and a pectate lyase domain was identified within its tail spike protein. MRABP9 showed potent bactericidal and anti-biofilm activity against MRAB, significantly delaying the time point of bacterial regrowth in vitro. Phage administration could rescue the mice from acute lethal MRAB infection. Considering its features, MRABP9 has the potential as an efficient candidate for prophylactic and therapeutic use against acute infections caused by MRAB strains.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Bacteriophages , Drug Resistance, Multiple, Bacterial , Phage Therapy , Acinetobacter baumannii/virology , Acinetobacter baumannii/drug effects , Animals , Acinetobacter Infections/microbiology , Acinetobacter Infections/therapy , Mice , Bacteriophages/genetics , Bacteriophages/physiology , Phage Therapy/methods , Genome, Viral , Biofilms/drug effects , Biofilms/growth & development , Humans , Female , Sewage/virology
2.
J Control Release ; 368: 595-606, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38185333

ABSTRACT

Ferroptosis, a unique iron-dependent mode of cell death characterized by lipid peroxide accumulation, holds significant potential for the treatment of glioblastoma (GBM). However, the effectiveness of ferroptosis is hindered by the limited intracellular ferrous ions (Fe2+) and hydrogen peroxide (H2O2). In this study, a novel near-infrared (NIR)-light-responsive nanoplatform (ApoE-UMSNs-GOx/SRF) based on upconversion nanoparticles (UCNPs) was developed. A layer of mesoporous silica and a lipid bilayer were coated on UCNPs sequentially and loaded with glucose oxidase (GOx) and sorafenib, respectively. Further attachment of the ApoE peptide endowed the nanoplatform with BBB penetration and GBM targeting capabilities. Our results revealed that ApoE-UMSNs-GOx/SRF could efficiently accumulated in the orthotopic GBM and induce amplified ferroptosis when combining with NIR irradiation. The UCNPs mediated the photoreduction of Fe3+ to Fe2+ by converting NIR to UV light, and excess H2O2 was produced by the reaction of glucose with the loaded GOx. These processes greatly promoted the production of ROS, which together with inhibition of system Xc- by the loaded sorafenib, leading to enhanced accumulation of lipid peroxides and significantly improved the antiglioma effect both in vitro and in vivo. Our strategy has the potential to enhance the effectiveness of ferroptosis as a therapeutic approach for GBM.


Subject(s)
Ferroptosis , Glioblastoma , Nanoparticles , Neoplasms , Photochemotherapy , Humans , Glioblastoma/drug therapy , Photochemotherapy/methods , Sorafenib , Hydrogen Peroxide , Apolipoproteins E/therapeutic use , Regeneration , Cell Line, Tumor , Nanoparticles/chemistry , Neoplasms/drug therapy
3.
Int J Mol Sci ; 24(17)2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37686068

ABSTRACT

Microglia are believed to be the key immune effectors of the central immune microenvironment, and their dysregulation is associated with neuroinflammation and mood disorders. Nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain-containing five (NLRC5) is a new member of the Nod-like receptor family. Recently, NLRC5 has been reported to be expressed by microglia. Nonetheless, the exact roles of NLRC5 in microglial activation and its function in depression have not been investigated yet. Herein, we found that reducing NLRC5 decreased lipopolysaccharide (LPS)-induced secretion of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) in primary cultured microglia and microglial cell lines but not in bone marrow-derived macrophages (BMDMs). In more detail, reducing NLRC5 diminished the secretion of LPS-induced cytokines by attenuating IKKα/ß phosphorylation and inhibiting NF-κB signaling. Moreover, the expression of Nlrc5 in the hippocampus of LPS- or chronic unpredictable mild stress (CUMS)-induced depressive mice was increased. In line with the in vitro findings, Nlrc5 deficiency inhibited microglial activation in the mouse hippocampus and improved LPS- or CUMS-induced depressive-like behaviors. In summary, we demonstrated the critical role of NLRC5 in LPS-induced microglial activation and LPS- or CUMS-induced depressive mouse models.


Subject(s)
Lipopolysaccharides , NF-kappa B , Animals , Mice , Lipopolysaccharides/toxicity , Microglia , Signal Transduction , Cytokines , Intracellular Signaling Peptides and Proteins/genetics
4.
Acta Histochem ; 124(7): 151939, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35952483

ABSTRACT

Nucleotide oligomerization domain-like receptors (NLRs), belonging to a large family of pattern recognition receptors, participate in the host's first line of defense against invading pathogens. Caspase recruitment domain containing 5 (NLRC5), the largest member in the NLR family, is demonstrated to be involved in the innate immune response and inflammatory diseases far and wide. Recent studies report that NLRC5 is associated with some central nervous system (CNS) diseases. Besides, NLRC5 is a mastery regulator for the expression of MHC class I both in the immune system and the CNS, while MHC class I is expressed and exerts its function in the brain. Therefore, it is necessary to investigate the expression pattern of NLRC5 in the developing and adult CNS. In our study, postnatal brain sections of C57BL/6 J mice are analyzed for the expression of NLRC5 protein by immunofluorescence. In the postnatal stages of developing telencephalon, NLRC5 exhibits a spatial and temporal expression pattern. NLRC5 is time-specifically expressed in subfields of hippocampus and different layers of prefrontal cortex. Moreover, it is shown that NLRC5 is highly cell type specific. It can be expressed in large quantities by neurons and microglia, but rarely expressed by astrocytes. Taken together, our research is important for further understanding the biological characteristics of NLRC5 and its function in the CNS.


Subject(s)
Immunity, Innate , Intracellular Signaling Peptides and Proteins , Animals , Brain , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Nucleotides
5.
J Nanobiotechnology ; 20(1): 214, 2022 May 06.
Article in English | MEDLINE | ID: mdl-35524277

ABSTRACT

Immunotherapy has gradually emerged as the most promising anticancer therapy. In addition to conventional anti-PD-1/PD-L1 therapy, anti-CTLA-4 therapy, CAR-T therapy, etc., immunotherapy can also be induced by stimulating the maturation of immune cells or inhibiting negative immune cells, regulating the tumor immune microenvironment and cancer vaccines. Lipid nanovesicle drug delivery system includes liposomes, cell membrane vesicles, bacterial outer membrane vesicles, extracellular vesicles and hybrid vesicles. Lipid nanovesicles can be used as functional vesicles for cancer immunotherapy, and can also be used as drug carriers to deliver immunotherapy drugs to the tumor site for cancer immunotherapy. Here, we review recent advances in five kinds of lipid nanovesicles in cancer immunotherapy and assess the clinical application prospects of various lipid nanovesicles, hoping to provide valuable information for clinical translation in the future.


Subject(s)
Immunotherapy , Neoplasms , Drug Delivery Systems , Humans , Lipids , Neoplasms/drug therapy , Tumor Microenvironment
6.
ACS Appl Mater Interfaces ; 13(20): 23396-23409, 2021 May 26.
Article in English | MEDLINE | ID: mdl-33982563

ABSTRACT

Paclitaxel (PTX) is a first-line chemotherapeutic drug for breast cancer, but PTX resistance often occurs in metastatic breast cancer. In addition, due to the poor targeting of chemotherapeutic drugs and the presence of the blood-brain barrier (BBB), it is hard to effectively treat brain metastatic breast cancer using paclitaxel. Thus, it is urgent to develop an effective drug delivery system for the treatment of brain metastatic breast cancer. The current study found that TWF1 gene, an epithelial-mesenchymal transition-associated gene, was overexpressed in brain metastatic breast cancer (231-BR) cells and was associated with the PTX resistance of 231-BR cells. Knockdown of TWF1 by small interference RNA (siRNA) in 231-BR cells could effectively increase the sensitivity of brain metastatic breast cancer cells to paclitaxel. Then, a liposome-based drug delivery system was developed for PTX delivery across BBB, enhancing PTX sensitivity and brain metastases targeting via BRBP1 peptide modification. The results showed that BRBP1-modified liposomes could effectively cross the BBB, specifically accumulate in brain metastases, and effectively interfere TWF1 gene expression in vitro and in vivo, and thus they enhanced proliferation inhibition, cell cycle arrest, and apoptosis induction, thereby inhibiting the formation and growth of brain metastases. In summary, our results indicated that BRBP1-modified and PTX- and TWF1 siRNA-loaded liposomes have the potential for the treatment of brain metastatic breast cancer, which lays the foundation for the development of a new targeted drug delivery system.


Subject(s)
Brain Neoplasms , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Liposomes , Paclitaxel , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Cell Line, Tumor , Female , Humans , Liposomes/chemistry , Liposomes/pharmacokinetics , Mice , Mice, Inbred BALB C , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nanoparticles/chemistry , Nanoparticles/metabolism , Oligopeptides/chemistry , Paclitaxel/chemistry , Paclitaxel/pharmacology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology
7.
Front Cell Neurosci ; 14: 573208, 2020.
Article in English | MEDLINE | ID: mdl-33192317

ABSTRACT

In the immune system, Major Histocompatibility Complex class I (MHC-I) molecules are located on the surface of most nucleated cells in vertebrates where they mediate immune responses. Accumulating evidence indicates that MHC-I molecules are also expressed in the central nervous system (CNS) where they play important roles that are significantly different from their immune functions. Classical MHC-I molecules are temporally and spatially expressed in the developing and adult CNS, where they participate in the synaptic formation, remodeling and plasticity. Therefore, clarifying the regulation of MHC-I expression is necessary to develop an accurate understanding of its function in the CNS. Here, we show that microRNA 34a (miR34a), a brain enriched noncoding RNA, is temporally expressed in developing hippocampal neurons, and its expression is significantly increased after MHC-I protein abundance is decreased in the hippocampus. Computational algorithms identify putative miR34a target sites in the 3'UTR of MHC-I mRNA, and here we demonstrate direct targeting of miR34a to MHC-I mRNA using a dual-luciferase reporter assay system. MiR34a targeting can decrease constitutive MHC-I expression in both Neuro-2a neuroblastoma cells and primary hippocampal neurons. Finally, miR34a mediated reduction of MHC-I results in increased dendritic growth and branching in cultured hippocampal neurons. Taken together, our findings identify miR34a as a novel regulator of MHC-I for shaping neural morphology in developing hippocampal neurons.

8.
Mater Sci Eng C Mater Biol Appl ; 116: 111188, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32806329

ABSTRACT

Tumor metastasis to brain is the main clinical manifestation of patients with advanced breast cancer, leading to poor survival prognosis. In order to detect the early incidence of brain metastasis, it is urgent to develop hypersensitive contrast agents for multimode imaging. In this study, PEG-phospholipids coated, a phage play derived peptide, BRBP1 peptide modified ultra-small iron oxide nanoparticles were prepared for targeted NIRF and MR imaging of breast cancer brain metastasis. The nanoparticles showed 10 nm core-shell, high relaxivity values and photon emission efficiency in vitro. The nanoparticles offered a T2 contrast imaging effect and near-infrared fluorescent signal enhancement. Compared with control peptide modified nanoparticles, the MR/NIRF imaging signal of BRBP1-modified nanoparticles in tumor tissue was significantly enhanced, which should be induced by the targeting ability of BRBP1 peptide. These results indicated that BRBP1-SPIO@mPEG (DiR) nanoparticles could be applied as an effective targeted delivery system for diagnosis of breast cancer brain metastasis.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Magnetite Nanoparticles , Nanoparticles , Brain Neoplasms/diagnostic imaging , Breast Neoplasms/diagnostic imaging , Cell Line, Tumor , Contrast Media , Humans , Magnetic Iron Oxide Nanoparticles , Magnetic Resonance Imaging
9.
J Neurochem ; 152(2): 182-194, 2020 01.
Article in English | MEDLINE | ID: mdl-31549732

ABSTRACT

Major histocompatibility Complex class I (MHC I) molecules are ubiquitously expressed, being found in most nucleated cells, where they are central mediators of both the adaptive and innate immune responses. Recent studies have shown that MHC I are also expressed in the developing brain where they participate in synapse elimination and plasticity. Up-regulation of MHC I within the developing brain has been reported, however, the mechanism(s) regulating this developmental up-regulation of neuronal MHC I remains unknown. Here, we show NLR family CARD domain containing 5 (NLRC5), a newly identified member of the NLR family, is widely expressed in hippocampal neurons, and the expression pattern of NLRC5 coincides with increased MHC I mRNA in the developing hippocampus. Using a luciferase assay in Neuro-2a cells we demonstrate that NLRC5 can induce the activation of MHC I and this induction requires the W/S-X-Y motif. Further studies show that transcription factors regulatory factor X (RFX) and CREB1, which bind to X1 and X2 box, are crucial for NLRC5-mediated induction. Moreover immunoprecipitation experiments reveal that NLRC5 interacts with RFX subunits RFX5 and RFXANK. Knockout of Nlrc5 dramatically impairs basal expression of MHC I in mouse hippocampus. Taken together, our findings identify NLRC5 as a key regulator of MHC I up-regulation in the developing hippocampus and suggest an important role for NLRC5 in neurons. Cover Image for this issue: doi: 10.1111/jnc.14729.


Subject(s)
Hippocampus/growth & development , Hippocampus/metabolism , Histocompatibility Antigens Class I/biosynthesis , Intracellular Signaling Peptides and Proteins/deficiency , Animals , Animals, Newborn , Base Sequence , Cell Line, Tumor , Histocompatibility Antigens Class I/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism
10.
Neurochem Res ; 44(2): 312-322, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30406910

ABSTRACT

Neuronal MHC class I proteins have been previously reported to regulate synaptic plasticity. Several reports indicate MHC class I proteins are expressed early during development of the nervous system, suggesting they may also play a role in neuronal development. Using cultured cortical neurons, we show MHC class I proteins aggregate at specific sites in neuronal cell bodies, which overlap with the actin cytoskeleton. Knockout of MHC class I in cultured neurons increases total dendritic length and the number of branch points. These effects are abolished by reintroducing MHC class I expression. Similarly, blocking of MHC class I proteins or PirB by an MHCI antibody or a soluble PirB ectodomain respectively, mimics the knock out phenotype of increased dendritic branching. This effect is correlated with decreased phosphorylation of both LIMK and cofilin, suggesting it may be mediated by an induction of cofilin activity. Finally, layer II and III cortical neurons in the sensorimotor region of an MHC class I deficiency mouse model show increased dendritic growth and branching. Altogether, our results suggest MHC class I plays a role in inhibiting or limiting the degree of dendrite arborization during the development of cortical neurons.


Subject(s)
Dendrites/metabolism , Histocompatibility Antigens Class I/metabolism , Neurons/pathology , Receptors, Immunologic/metabolism , Animals , Cells, Cultured , Dendritic Spines/metabolism , Histocompatibility Antigens Class I/genetics , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/physiology , Neurons/metabolism
11.
J Cancer Res Ther ; 14(Supplement): S473-S479, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29970709

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) is the fifth most common tumor worldwide. The discovery of new therapies against HCC is highly dependable on finding molecules which play essential roles in cancer development. The objective of this study was to evaluate the activity of gamma secretase (γ-secretase), and the antitumor effects of a γ-secretase inhibitor (GSI) in HCC. METHODS: The expression of presenilin 1 (PS1), a core component of γ-secretase, was examined by Western blot. Activity of γ-secretase was measured by a luciferase-based reporter system, and cancer cells were transfected either with PS1 dominant negative mutant (PS1D385A) or treated with GSI. RESULTS: Expression of PS1 was increased in HCC tissue and several HCC cell lines, which were accompanied by elevated γ-secretase activity. Cell colony formation and cell proliferation were decreased upon treatment with GSI but not with PS1D385A transfection. CONCLUSION: GSIs may be appealing candidates for the development of new therapies against HCC.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Enzyme Inhibitors/pharmacology , Liver Neoplasms/metabolism , Adult , Amyloid Precursor Protein Secretases/genetics , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Female , Gene Expression , Genes, Reporter , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Male , Middle Aged , Molecular Targeted Therapy , Mutation , Neoplasm Grading , Presenilin-1/genetics , Presenilin-1/metabolism , Tumor Burden
12.
Theranostics ; 7(11): 2837-2848, 2017.
Article in English | MEDLINE | ID: mdl-28824719

ABSTRACT

Ischemic stroke is one of the leading causes of morbidity and mortality worldwide. The expression of major histocompatibility complex class I (MHC-I) molecules in the central nervous system, which are silenced under normal physiological conditions, have been reported to be induced by injury stimulation. The purpose of this study was to determine whether MHC-I molecules could serve as molecular targets for the acute phase of ischemic stroke and to assess whether a high-affinity peptide specific for MHC-I molecules could be applied in the near-infrared imaging of cerebral ischemic mice. Quantitative real-time PCR and Western blotting were used to detect the expression of MHC-I molecules in two mouse models of cerebral ischemic stroke and an in vitro model of ischemia. The NetMHC 4.0 server was used to screen a high-affinity peptide specific for mouse MHC-I molecules. The Rosetta program was used to identify the specificity and affinity of the screened peptide (histocompatibility-2 binding peptide, H2BP). The results demonstrated that MHC-I molecules could serve as molecular targets for the acute phase of ischemic stroke. Cy5.5-H2BP molecular probes could be applied in the near-infrared imaging of cerebral ischemic mice. Research on the expression of MHC-I molecules in the acute phase after ischemia and MHC-I-targeted imaging may not only be helpful for understanding the mechanism of ischemic and hypoxic brain injury and repair but also has potential application value in the imaging of ischemic stroke.


Subject(s)
Central Nervous System/diagnostic imaging , Central Nervous System/pathology , Histocompatibility Antigens Class I/analysis , Ischemia/pathology , Molecular Diagnostic Techniques/methods , Stroke/diagnostic imaging , Stroke/pathology , Animals , Blotting, Western , Disease Models, Animal , Gene Expression Profiling , Mice , Models, Biological , Molecular Probe Techniques , Real-Time Polymerase Chain Reaction
13.
Oncotarget ; 7(12): 14199-206, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26883101

ABSTRACT

Alzheimer's disease (AD) is one of the most common neurodegenerative diseases worldwide. The main features of AD are the pathological changes of density and distribution of intracellular neurofibrillary tangles (NFT) and extracellular amyloid plaques. The processing of amyloid beta precursor protein (APP) to ß-amyloid peptide (Aß) is one of the critical events in the pathogenesis of AD. In this study, we evaluated the role of the interaction of neural cell adhesion molecule (NCAM) and APP in neurite outgrowth using two different experimental systems: PC12E2 cells and hippocampal neurons that were isolated from wild type, APP knock-in and APP knock-out mice. PC12E2 cells or hippocampal neurons were co-cultured with NCAM-negative or NCAM-positive fibroblasts L929 cells. We found that APP promoted neurite outgrowth of PC12E2 cells and hippocampal neurons in either the presence or absence of NCAM. Secreted APP can rescue the neurite outgrowth in hippocampal neurons from APP knock-out mice. The interaction of APP and NCAM had synergic effect in promoting neurite outgrowth in both PC12E2 cells and hippocampal neurons. Our results suggested that the interaction of APP with NCAM played an important role in AD development and therefore could be a potential therapeutic target for AD treatment.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Embryo, Mammalian/cytology , Fibroblasts/cytology , Hippocampus/cytology , Neural Cell Adhesion Molecules/metabolism , Neurites/metabolism , Neuronal Outgrowth/physiology , Animals , Cells, Cultured , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Hippocampus/metabolism , Mice , Mice, Knockout , Rats
14.
Sci Rep ; 5: 17871, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26639364

ABSTRACT

A new species of spiroplasma, Spiroplasma eriocheiris (S. eriocheiris), was identified as a lethal pathogen of tremor disease (TD) in Chinese mitten crab recently. In order to acquire appropriate biological and diagnostic tools for characterizing this newly discovered pathogen, 5 monoclonal antibodies (mAbs) and a polyclonal antibody (pAb) against S. eriocheiris were produced. Among the mAbs, 6F5, 7C8 and 12H5 lead to the deformation of S. eriocheiris. A peptide sequence, YMRDMQSGLPRY was identified as a mimic motif of MreB that is the cell shape determining protein of S. eriocheiris interacting with 3 mAbs. Furthermore, a double antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA) for detection of S. eriocheiris was established using the mAb and pAb we prepared. It detected as low as 0.1 µg/mL of S. eriocheiris. No cross-reaction was observed with three other common bacteria (Pseudomonas aeruginosa, Escherichia coli, and Bacillus subtilis) and the hemolymph samples of healthy Eriocheir sinensis. Collectively, our results indicated that the mAbs and pAb we prepared could be used in the analysis of S. eriocheiris membrane proteins mimotope and development of a diagnostic kit for S. eriocheiris infections.


Subject(s)
Antibodies, Bacterial/biosynthesis , Antibodies, Monoclonal/biosynthesis , Spiroplasma/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Peptide Library , Rabbits , Recombination, Genetic/genetics , Reproducibility of Results , Sensitivity and Specificity
15.
Int J Nanomedicine ; 10: 7345-58, 2015.
Article in English | MEDLINE | ID: mdl-26677324

ABSTRACT

PURPOSE: To explore the thermoresistance and expression of heat-shock protein 90 (HSP90) in magnetic hyperthermia-treated human liver cancer stem-like cells (LCSCs) and the effects of a heat-shock protein HSP90 inhibitor 17-allylamino-17-demethoxgeldanamycin (17-AAG) on hepatocellular carcinoma-burdened nude mice. METHODS: CD90(+) LCSCs were isolated by magnetic-activated cell sorting from BEL-7404. Spheroid formation, proliferation, differentiation, drug resistance, and tumor formation assays were performed to identify stem cell characteristics. CD90-targeted thermosensitive magnetoliposomes (TMs)-encapsulated 17-AAG (CD90@17-AAG/TMs) was prepared by reverse-phase evaporation and its characteristics were studied. Heat tolerance in CD90(+) LCSCs and the effect of CD90@17-AAG/TMs-mediated heat sensitivity were examined in vitro and in vivo. RESULTS: CD90(+) LCSCs showed significant stem cell-like properties. The 17-AAG/TMs were successfully prepared and were spherical in shape with an average size of 128.9±7.7 nm. When exposed to magnetic hyperthermia, HSP90 was up-regulated in CD90(+) LCSCs. CD90@17-AAG/TMs inhibited the activity of HSP90 and increased the sensitivity of CD90(+) LCSCs to magnetic hyperthermia. CONCLUSION: The inhibition of HSP90 could sensitize CD90(+) LCSCs to magnetic hyperthermia and enhance its anti-tumor effects in vitro and in vivo.


Subject(s)
Benzoquinones/pharmacology , Carcinoma, Hepatocellular/pathology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hyperthermia, Induced , Lactams, Macrocyclic/pharmacology , Liver Neoplasms/pathology , Magnetic Phenomena , Neoplastic Stem Cells/drug effects , Animals , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Female , Humans , Liver Neoplasms/therapy , Mice , Mice, Nude
16.
PLoS One ; 10(8): e0135223, 2015.
Article in English | MEDLINE | ID: mdl-26263390

ABSTRACT

MHC class I (MHC-I) molecules are important components of the immune system. Recently MHC-I have been reported to also play important roles in brain development and synaptic plasticity. In this study, we examine the molecular mechanism(s) underlying activity-dependent MHC-I expression using hippocampal neurons. Here we report that neuronal expression level of MHC-I is dynamically regulated during hippocampal development after birth in vivo. Kainic acid (KA) treatment significantly increases the expression of MHC-I in cultured hippocampal neurons in vitro, suggesting that MHC-I expression is regulated by neuronal activity. In addition, KA stimulation decreased the expression of pre- and post-synaptic proteins. This down-regulation is prevented by addition of an MHC-I antibody to KA treated neurons. Further studies demonstrate that calcium-dependent protein kinase C (PKC) is important in relaying KA simulation activation signals to up-regulated MHC-I expression. This signaling cascade relies on activation of the MAPK pathway, which leads to increased phosphorylation of CREB and NF-κB p65 while also enhancing the expression of IRF-1. Together, these results suggest that expression of MHC-I in hippocampal neurons is driven by Ca2+ regulated activation of the MAPK signaling transduction cascade.


Subject(s)
Calcium Signaling , Gene Expression Regulation , Genes, MHC Class I , Neurons/metabolism , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Interferon Regulatory Factor-1/metabolism , Kainic Acid/pharmacology , MAP Kinase Signaling System/drug effects , Mice , NF-kappa B/metabolism , Protein Kinase C/metabolism , Pyramidal Cells/metabolism , RNA, Messenger , Synapses/metabolism
17.
Exp Brain Res ; 233(9): 2733-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26169100

ABSTRACT

Recent animal studies have found neuronal expression of major histocompatibility complex (MHC) class I in the central nervous system (CNS). However, the developmental expression profiles of MHC class I in human CNS remain unclear. Here, we systemically evaluate the expression and subcellular localization of MHC class I molecules during human CNS development using immunohistochemistry and immunofluorescence. Between the age of 20-33 gestational weeks (GW), MHC class I expression was relatively absent in the cerebral cortex with the exception of a few neurons; however, expression increased rapidly in the cochlear nuclei and in the cerebellar cortical Purkinje cells while increasing slowly in the substantia nigra. Expression was also detected in some nuclei and nerve fibers of the brain stem including the ambiguus nucleus, the locus coeruleus and the solitary tract as early as 20 GW and persisted through 33 GW. These early-stage neural cells with MHC class I protein expression later developed neuronal morphology. 30-33 GW is an important period of MHC class I expression in neurons, and during this period, MHC class I molecules were found to be enriched not only in neuronal cell bodies and neurites but also in nerve fibers and in the surrounding stroma. No expression was detected in the adult brain with exception of the cerebrovascular endothelium. MHC class I molecules displayed greater postsynaptic colocalization in cerebellar Purkinje cells, in the lateral geniculate nucleus and in the cochlear nuclei. These results demonstrate diverse spatiotemporal expression patterns for MHC class I molecules in the prenatal human CNS and strongly support the notion that MHC class I molecules play important roles in both CNS development and plasticity.


Subject(s)
Central Nervous System , Gene Expression Regulation, Developmental/physiology , Histocompatibility Antigens Class I/metabolism , Adult , Age Factors , Central Nervous System/embryology , Central Nervous System/growth & development , Central Nervous System/metabolism , Disks Large Homolog 4 Protein , Gestational Age , Humans , Infant, Newborn , Infant, Premature/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Synaptophysin/metabolism
18.
Neurochem Res ; 40(7): 1487-96, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26040564

ABSTRACT

Recent studies clearly demonstrate major histocompatibility complex (MHC) class I expression in the brain plays an important functional role in neural development and plasticity. A previous study from our laboratory demonstrated the temporal and spatial expression patterns of classical MHC class I molecules in the brain of C57 mice. Studies regarding non-classical MHC class I molecules remain limited. Here we examine the expression of non-classical MHC class I molecules in mouse central nervous system (CNS) during embryonic and postnatal developmental stages using in situ hybridization and immunofluorescence. We find non-classical MHC class I molecules, M3/T22/Q1, are expressed in the cerebral cortex, neuroepithelium of the lateral ventricle, neuroepithelium of aquaeductus and developing cerebellum during embryonic developmental stages. During the postnatal period from P0 to adult, non-classical MHC class I mRNAs are detected in olfactory bulb, hippocampus, cerebellum and some nerve nuclei. Overall, the expression patterns of non-classical MHC class I molecules are similar to those of classical MHC class I molecules in the developing mouse brain. In addition, non-classical MHC class I molecules are present in the H2-K(b) and H2-D(b) double knock-out mice where their expression levels are greatly increased within the same locations as compared to wild type mice. The elucidation and discovery of the expression profile of MHC class I molecules during development is important for supporting an enhanced understanding of their physiological and potential pathological roles within the CNS.


Subject(s)
Brain/immunology , Histocompatibility Antigens Class I/metabolism , Animals , Brain/embryology , Brain/growth & development , In Situ Hybridization , Mice , Mice, Inbred C57BL
19.
Sci Rep ; 5: 8029, 2015 Jan 26.
Article in English | MEDLINE | ID: mdl-25619721

ABSTRACT

Novel molecularly targeted agents that block the development and metastasis of human brain metastatic breast cancer hold great promise for their translational value. In this study, we constructed a novel targeting composite peptide BRBP1-TAT-KLA comprising of three elements: a brain metastatic breast carcinoma cell (231-BR)-binding peptide BRBP1, a cell penetrating peptide TAT, and a proapoptotic peptide KLA. This composite peptide efficiently internalized in 231-BR cells and consequently induced mitochondrial damage and cellular apoptosis. Exposure of 231-BR cells to BRBP1-TAT-KLA significantly decreased cell viability and increased apoptosis compared with the cells treated with the control peptides. In vivo relevance of these findings was further corroborated in the 231-BR tumor-bearing mice that demonstrated significantly delayed tumor development and metastasis following administration of BRBP1-TAT-KLA compared with those treated with TAT-KLA alone. Interestingly, BRBP1-TAT-KLA inhibited the formation of both large and micro-metastases, while TAT-KLA alone failed to significantly reduce micro-metastases in the breast cancer brain metastasis mice. BRBP1-TAT-KLA selectively homed to the tumors in vivo where it induced cellular apoptosis without significant toxicity on non-tumor tissues. Our findings therefore demonstrated the enhanced antitumor effects of the BRBP1 compound peptide BRBP1-TAT-KLA, providing insights toward development of a potential therapeutic strategy for brain metastatic breast cancer.


Subject(s)
Brain Neoplasms/genetics , Breast Neoplasms/genetics , Gene Products, tat/genetics , Oligopeptides/genetics , Peptide Fragments/administration & dosage , Peptides/genetics , Animals , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Female , Gene Products, tat/administration & dosage , Humans , Intercellular Signaling Peptides and Proteins , Mice , Peptide Fragments/genetics , Peptides/administration & dosage , Xenograft Model Antitumor Assays
20.
Int J Nanomedicine ; 9: 4231-43, 2014.
Article in English | MEDLINE | ID: mdl-25228802

ABSTRACT

BACKGROUND: This study aimed to generate targeted folic acid-conjugated, doxorubicin-loaded, magnetic iron oxide bovine serum albumin nanospheres (FA-DOX-BSA MNPs) that lower the side effects and improve the therapeutic effect of antitumor drugs when combined with hyperthermia and targeting therapy. A new nanodrug using magnetic nanospheres for heating and addition of the folate receptor with cancer cell specificity was prepared. The characteristics of these nanospheres and their antitumor effects in nasopharyngeal carcinoma were explored. METHODS: FA-DOX-BSA MNPs comprising encapsulated magnetic iron oxide nanoparticles were prepared using a desolvation cross-linking method. Activated folic acid (N-hydroxysuccinimide ester of folic acid) was conjugated to the surface of albumin nanospheres via amino groups. RESULTS: Folic acid was successfully expressed on the surface of the nanospheres. Electron microscopy revealed that the FA-DOX-BSA MNPs were nearly spherical and uniform in size, with an average diameter of 180 nm. The nanomaterial could deliver doxorubicin at clinically relevant doses with an entrapment efficiency of 80%. An increasing temperature test revealed that incorporation of magnetic iron oxide into nanospheres could achieve a satisfactory heat treatment temperature at a significantly lower dose when placed in a high-frequency alternating magnetic field. FA-DOX-BSA MNPs showed greater inhibition of tumors than in the absence of folic acid in vitro and in vivo. Compared with chemotherapy alone, hyperthermia combined with chemotherapy was more effective against tumor cells. CONCLUSION: Folic acid-conjugated bovine serum albumin nanospheres composed of mixed doxorubicin and magnetic iron oxide cores can enable controlled and targeted delivery of anticancer drugs and may offer a promising alternative to targeted doxorubicin therapy for nasopharyngeal carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Folic Acid/chemistry , Magnetite Nanoparticles/chemistry , Serum Albumin, Bovine/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cattle , Doxorubicin/chemistry , Humans , KB Cells , Male , Mice , Mice, Nude , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...