Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 22(1): 138, 2024 02 19.
Article in English | MEDLINE | ID: mdl-38374138

ABSTRACT

BACKGROUND: Applications of nonthermal plasma have expanded beyond the biomedical field to include antibacterial, anti-inflammatory, wound healing, and tissue regeneration. Plasma enhances epithelial cell repair; however, the potential damage to deep tissues and vascular structures remains under investigation. RESULT: This study assessed whether liquid plasma (LP) increased nitric oxide (NO) production in human umbilical vein endothelial cells by modulating endothelial NO synthase (eNOS) phosphorylation and potential signaling pathways. First, we developed a liquid plasma product and confirmed the angiogenic effect of LP using the Matrigel plug assay. We found that the NO content increased in plasma-treated water. NO in plasma-treated water promoted cell migration and angiogenesis in scratch and tube formation assays via vascular endothelial growth factor mRNA expression. In addition to endothelial cell proliferation and migration, LP influenced extracellular matrix metabolism and matrix metalloproteinase activity. These effects were abolished by treatment with NG-L-monomethyl arginine, a specific inhibitor of NO synthase. Furthermore, we investigated the signaling pathways mediating the phosphorylation and activation of eNOS in LP-treated cells and the role of LKB1-adenosine monophosphate-activated protein kinase in signaling. Downregulation of adenosine monophosphate-activated protein kinase by siRNA partially inhibited LP-induced eNOS phosphorylation, angiogenesis, and migration. CONCLUSION: The present study suggests that LP treatment may be a novel strategy for promoting angiogenesis in vascular damage. Video Abstract.


Subject(s)
Extracellular Matrix , Nitric Oxide Synthase Type III , Plasma , Vascular System Injuries , Humans , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Angiogenesis , Extracellular Matrix/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase/pharmacology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Protein Kinases/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Vascular System Injuries/metabolism , Vascular System Injuries/therapy , Plasma/metabolism
2.
Cell Death Dis ; 14(2): 119, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36781835

ABSTRACT

The skin functions as the outermost protective barrier to the internal organs and major vessels; thus, delayed regeneration from acute injury could induce serious clinical complications. For rapid recovery of skin wounds, promoting re-epithelialization of the epidermis at the initial stage of injury is essential, wherein epithelial keratinocytes act as leading cells via migration. This study applied plasma technology, which has been known to enable wound healing in the medical field. Through in vitro and in vivo experiments, the study elucidated the effect and molecular mechanism of the liquid plasma (LP) manufactured by our microwave plasma system, which was found to improve the applicability of existing gas-type plasma on skin cell migration for re-epithelialization. LP treatment promoted the cytoskeletal transformation of keratinocytes and migration owing to changes in the expression of integrin-dependent focal adhesion molecules and matrix metalloproteinases (MMPs). This study also identified the role of increased levels of intracellular reactive oxygen species (ROS) as a driving force for cell migration activation, which was regulated by changes in NADPH oxidases and mitochondrial membrane potential. In an in vivo experiment using a murine dorsal full-thickness acute skin wound model, LP treatment helped improve the re-epithelialization rate, reaffirming the activation of the underlying intracellular ROS-dependent integrin-dependent signaling molecules. These findings indicate that LP could be a valuable wound management material that can improve the regeneration potential of the skin via the activation of migration-related molecular signaling within the epithelial cell itself with plasma-driven oxidative eustress.


Subject(s)
Keratinocytes , Skin , Animals , Mice , Reactive Oxygen Species/metabolism , Skin/metabolism , Keratinocytes/metabolism , Wound Healing/physiology , Cell Movement , Integrins/metabolism , Oxidation-Reduction
3.
Adv Sci (Weinh) ; 9(23): e2201212, 2022 08.
Article in English | MEDLINE | ID: mdl-35694866

ABSTRACT

Recent multi-omics analyses paved the way for a comprehensive understanding of pathological processes. However, only few studies have explored Alzheimer's disease (AD) despite the possibility of biological subtypes within these patients. For this study, unsupervised classification of four datasets (genetics, miRNA transcriptomics, proteomics, and blood-based biomarkers) using Multi-Omics Factor Analysis+ (MOFA+), along with systems-biological approaches following various downstream analyses are performed. New subgroups within 170 patients with cerebral amyloid pathology (Aß+) are revealed and the features of them are identified based on the top-rated targets constructing multi-omics factors of both whole (M-TPAD) and immune-focused models (M-IPAD). The authors explored the characteristics of subtypes and possible key-drivers for AD pathogenesis. Further in-depth studies showed that these subtypes are associated with longitudinal brain changes and autophagy pathways are main contributors. The significance of autophagy or clustering tendency is validated in peripheral blood mononuclear cells (PBMCs; n = 120 including 30 Aß- and 90 Aß+), induced pluripotent stem cell-derived human brain organoids/microglia (n = 12 including 5 Aß-, 5 Aß+, and CRISPR-Cas9 apolipoprotein isogenic lines), and human brain transcriptome (n = 78). Collectively, this study provides a strategy for precision medicine therapy and drug development for AD using integrative multi-omics analysis and network modelling.


Subject(s)
Alzheimer Disease , Amyloidosis , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins/metabolism , Amyloidosis/metabolism , Autophagy/genetics , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Microglia/metabolism , Microglia/pathology
4.
Exp Mol Med ; 53(6): 1046-1054, 2021 06.
Article in English | MEDLINE | ID: mdl-34108650

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia, and many studies have focused on finding effective blood biomarkers for the accurate diagnosis of this disease. Predicting cerebral amyloid deposition is considered the key for AD diagnosis because a cerebral amyloid deposition is the hallmark of AD pathogenesis. Previously, blood biomarkers were discovered to predict cerebral amyloid deposition, and further efforts have been made to increase their sensitivity and specificity. In this study, we analyzed blood-test factors (BTFs) that can be commonly measured in medical health check-ups from 149 participants with cognitively normal, 87 patients with mild cognitive impairment, and 64 patients with clinically diagnosed AD dementia with brain amyloid imaging data available. We demonstrated that four factors among regular health check-up blood tests, cortisol, triglyceride/high-density lipoprotein cholesterol ratio, alanine aminotransferase, and free triiodothyronine, showed either a significant difference by or correlation with cerebral amyloid deposition. Furthermore, we made a prediction model for Pittsburgh compound B-positron emission tomography positivity, using BTFs and the previously discovered blood biomarkers, the QPLEXTM Alz plus assay kit biomarker panel, and the area under the curve was significantly increased up to 0.845% with 69.4% sensitivity and 90.6% specificity. These results show that BTFs could be used as co-biomarkers and that a highly advanced prediction model for amyloid plaque deposition could be achieved by the combinational use of diverse biomarkers.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides , Brain/metabolism , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/pathology , Hematologic Tests , Humans , Positron-Emission Tomography
5.
Prog Neurobiol ; 203: 102075, 2021 08.
Article in English | MEDLINE | ID: mdl-34004220

ABSTRACT

Amyloid-ß (Aß) and tau are major pathological hallmarks of Alzheimer's disease (AD). Several studies have revealed that Aß accelerates pathological tau transition and spreading during the disease progression, and that reducing tau can mitigate pathological features of AD. However, molecular links between Aß and tau pathologies remain elusive. Here, we suggest a novel role for the plexin-A4 as an Aß receptor that induces aggregated tau pathology. Plexin-A4, previously known as proteins involved in regulating axon guidance and synaptic plasticity, can bound to Aß with co-receptor, neuropilin-2. Genetic downregulation of plexin-A4 in neurons was sufficient to prevent Aß-induced activation of CDK5 and reduce tau hyperphosphorylation and aggregation, even in the presence of Aß. In an AD mouse model that manifests both Aß and tau pathologies, genetic downregulation of plexin-A4 in the hippocampus reduced tau pathology and ameliorated spatial memory impairment. Collectively, these results indicate that the plexin-A4 is capable of mediating Aß-induced tau pathology in AD pathogenesis.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Animals , Cell Adhesion Molecules , Disease Models, Animal , Mice , Mice, Transgenic , Nerve Tissue Proteins , tau Proteins
6.
Nat Commun ; 12(1): 280, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436582

ABSTRACT

Developing effective drugs for Alzheimer's disease (AD), the most common cause of dementia, has been difficult because of complicated pathogenesis. Here, we report an efficient, network-based drug-screening platform developed by integrating mathematical modeling and the pathological features of AD with human iPSC-derived cerebral organoids (iCOs), including CRISPR-Cas9-edited isogenic lines. We use 1300 organoids from 11 participants to build a high-content screening (HCS) system and test blood-brain barrier-permeable FDA-approved drugs. Our study provides a strategy for precision medicine through the convergence of mathematical modeling and a miniature pathological brain model using iCOs.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Brain/pathology , Drug Evaluation, Preclinical , Gene Regulatory Networks , Organoids/pathology , Alzheimer Disease/genetics , Cinnamates/pharmacology , Cinnamates/therapeutic use , Gene Regulatory Networks/drug effects , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Reproducibility of Results , Risk Factors
7.
Cancer Res Treat ; 52(4): 1019-1030, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32599978

ABSTRACT

PURPOSE: Radiation-induced oral mucositis limits delivery of high-dose radiation to targeted cancers. Therefore, it is necessary to develop a treatment strategy to alleviate radiation-induced oral mucositis during radiation therapy. We previously reported that inhibiting reactive oxygen species (ROS) generation suppresses autophagy. Irradiation induces autophagy, suggesting that antioxidant treatment may be used to inhibit radiation-induced oral mucositis. MATERIALS AND METHODS: We determined whether treatment with N-acetyl cysteine (NAC) could attenuate radiation-induced buccal mucosa damage in vitro and in vivo. The protective effects of NAC against oral mucositis were confirmed by transmission electron microscopy and immunocytochemistry. mRNA and protein levels of DNA damage and autophagy-related genes were measured by quantitative real-time polymerase chain reaction and western blot analysis, respectively. RESULTS: Rats manifesting radiation-induced oral mucositis showed decreased oral intake, loss of body weight, and low survival rate. NAC intake slightly increased oral intake, body weight, and the survival rate without statistical significance. However, histopathologic characteristics were markedly restored in NAC-treated irradiated rats. LC3B staining of rat buccal mucosa revealed that NAC treatment significantly decreased the number of radiation-induced autophagic cells. Further, NAC inhibited radiation-induced ROS generation and autophagy signaling. In vitro, NAC treatment significantly reduced the expression of NRF2, LC3B, p62, and Beclin-1 in keratinocytes compared with that after radiation treatment. CONCLUSION: NAC treatment significantly inhibited radiation-induced autophagy in keratinocytes and rat buccal mucosa and may be a potentially safe and effective option for the prevention of radiation-induced buccal mucosa damage.


Subject(s)
Acetylcysteine/administration & dosage , Free Radical Scavengers/administration & dosage , Radiation Injuries, Experimental/prevention & control , Stomatitis/prevention & control , Administration, Inhalation , Animals , Autophagy/drug effects , Autophagy/radiation effects , Cell Line , Female , Humans , Keratinocytes , Mouth Mucosa/drug effects , Mouth Mucosa/pathology , Mouth Mucosa/radiation effects , Nebulizers and Vaporizers , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/pathology , Rats , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Stomatitis/etiology , Stomatitis/pathology
8.
Cells ; 9(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-32131492

ABSTRACT

Non-thermal plasma (NTP) has been studied as a novel therapeutic tool for cancer that does not damage healthy cells. In this study, we show that NTP-treated solutions (NTS) can induce death in various leukemia cells through mechanistic target of rapamycin (mTOR) ubiquitination. Previously, we manufactured and demonstrated the efficacy of NTS in solid cancers. NTS did not exhibit any deleterious side effects, such as acute death or weight loss in nude mice. In the present study, NTS induced cell death in myeloid leukemia cells, including acute myeloid leukemia (AML) and chronic myeloid leukemia (CML). We found that mTOR was downregulated in NTS-treated cells via the ubiquitin-proteasome system (UPS). We also identified 'really interesting new gene' finger protein 126 (RNF126) as a novel binding protein for mTOR through protein arrays and determined the role of E3 ligase in NTS-induced mTOR ubiquitination. NTS-derived reactive oxygen species (ROS) affected RNF126 expression and lysosomal dysfunction. These findings suggest that NTS has potential antileukemic effects through RNF126-mediated mTOR ubiquitination with no deleterious side effects. Thus, NTS may represent a new therapeutic method for chemotherapy-resistant leukemia.


Subject(s)
Leukemia/metabolism , Leukemia/therapy , Plasma Gases/pharmacology , TOR Serine-Threonine Kinases/metabolism , Aged , Animals , Cell Line, Tumor , Down-Regulation/drug effects , Humans , K562 Cells , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
Gut ; 69(2): 283-294, 2020 02.
Article in English | MEDLINE | ID: mdl-31471351

ABSTRACT

OBJECTIVE: Cerebral amyloidosis and severe tauopathy in the brain are key pathological features of Alzheimer's disease (AD). Despite a strong influence of the intestinal microbiota on AD, the causal relationship between the gut microbiota and AD pathophysiology is still elusive. DESIGN: Using a recently developed AD-like pathology with amyloid and neurofibrillary tangles (ADLPAPT) transgenic mouse model of AD, which shows amyloid plaques, neurofibrillary tangles and reactive gliosis in their brains along with memory deficits, we examined the impact of the gut microbiota on AD pathogenesis. RESULTS: Composition of the gut microbiota in ADLPAPT mice differed from that of healthy wild-type (WT) mice. Besides, ADLPAPT mice showed a loss of epithelial barrier integrity and chronic intestinal and systemic inflammation. Both frequent transfer and transplantation of the faecal microbiota from WT mice into ADLPAPT mice ameliorated the formation of amyloid ß plaques and neurofibrillary tangles, glial reactivity and cognitive impairment. Additionally, the faecal microbiota transfer reversed abnormalities in the colonic expression of genes related to intestinal macrophage activity and the circulating blood inflammatory monocytes in the ADLPAPT recipient mice. CONCLUSION: These results indicate that microbiota-mediated intestinal and systemic immune aberrations contribute to the pathogenesis of AD in ADLPAPT mice, providing new insights into the relationship between the gut (colonic gene expression, gut permeability), blood (blood immune cell population) and brain (pathology) axis and AD (memory deficits). Thus, restoring gut microbial homeostasis may have beneficial effects on AD treatment.


Subject(s)
Alzheimer Disease/microbiology , Alzheimer Disease/therapy , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , Alzheimer Disease/metabolism , Animals , Behavior, Animal , Chronic Disease , Disease Models, Animal , Humans , Inflammation/microbiology , Intestines/microbiology , Memory, Short-Term , Mice, Transgenic , Permeability , Plaque, Amyloid/microbiology , Plaque, Amyloid/pathology , Spatial Learning , tau Proteins/analysis
10.
Cell Mol Immunol ; 17(8): 856-864, 2020 08.
Article in English | MEDLINE | ID: mdl-31217526

ABSTRACT

It has been reported that neutrophil extracellular traps (NETs) impair wound healing in diabetes and that inhibiting NET generation (NETosis) improves wound healing in diabetic mice. Gonadotropin-releasing hormone (GnRH) agonists are associated with a greater risk of diabetes. However, the role of GnRH in diabetic wound healing is unclear. We determined whether GnRH-promoted NETosis and induced more severe and delayed diabetic wound healing. A mouse model of diabetes was established using five injections with streptozotocin. Mice with blood glucose levels >250 mg/dL were then used in the experiments. GnRH agonist treatment induced delayed wound healing and increased NETosis at the skin wounds of diabetic mice. In contrast, GnRH antagonist treatment inhibited GnRH agonist-induced delayed wound healing. The expression of NETosis markers PAD4 and citrullinated histone H3 were increased in the GnRH-treated diabetic skin wounds in diabetic mice and patients. In vitro experiments also showed that neutrophils expressed a GnRH receptor and that GnRH agonist treatment increased NETosis markers and promoted phorbol myristate acetate (PMA)-induced NETosis in mouse and human neutrophils. Furthermore, GnRH antagonist treatment suppressed the expression of NETosis markers and PMA-induced NETosis, which were increased by GnRH treatment. These results indicated that GnRH-promoted NETosis and that increased NETosis induced delayed wound healing in diabetic skin wounds. Thus, inhibition of GnRH might be a novel treatment of diabetic foot ulcers.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Extracellular Traps/metabolism , Gonadotropin-Releasing Hormone/adverse effects , Wound Healing , Animals , Citrullination/drug effects , Disease Models, Animal , Extracellular Traps/drug effects , Gonadotropin-Releasing Hormone/agonists , HL-60 Cells , Histones/metabolism , Humans , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/ultrastructure , Protein-Arginine Deiminase Type 4/metabolism , Receptors, LHRH/metabolism , Wound Healing/drug effects
11.
Aging Cell ; 19(1): e13081, 2020 01.
Article in English | MEDLINE | ID: mdl-31763743

ABSTRACT

Alzheimer's disease (AD) is an age-related neurodegenerative disease. The most common pathological hallmarks are amyloid plaques and neurofibrillary tangles in the brain. In the brains of patients with AD, pathological tau is abnormally accumulated causing neuronal loss, synaptic dysfunction, and cognitive decline. We found a histone deacetylase 6 (HDAC6) inhibitor, CKD-504, changed the tau interactome dramatically to degrade pathological tau not only in AD animal model (ADLPAPT ) brains containing both amyloid plaques and neurofibrillary tangles but also in AD patient-derived brain organoids. Acetylated tau recruited chaperone proteins such as Hsp40, Hsp70, and Hsp110, and this complex bound to novel tau E3 ligases including UBE2O and RNF14. This complex degraded pathological tau through proteasomal pathway. We also identified the responsible acetylation sites on tau. These dramatic tau-interactome changes may result in tau degradation, leading to the recovery of synaptic pathology and cognitive decline in the ADLPAPT mice.


Subject(s)
Alzheimer Disease/genetics , Neurodegenerative Diseases/genetics , Protein Processing, Post-Translational/genetics , tau Proteins/metabolism , Acetylation , Animals , Disease Models, Animal , Humans , Mice
12.
Sci Rep ; 9(1): 13510, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31534179

ABSTRACT

Non-thermal plasma (NTP) has many functional activities such as, sterilization, wound healing and anti-cancer activity. Despite of its wide spread biomedical application, the effect of NTP on immune cells and allergic response has not been well studied. In this study, we determined whether NTP suppresses mast cell activation, which is important for allergic response, and ameliorates an atopic dermatitis (AD)-like skin inflammatory disease in mice. Exposure to NTP-treated medium during mast cell activation inhibited the expression and production of IL-6, TNF-α and suppressed NF-κB activation. We also investigated whether NTP treatment ameliorates house dust mite (HDM)-induced AD-like skin inflammation in mice. NTP treatment inhibited increases in epidermal thickness and recruitment of mast cells and eosinophils, which are important cell types in AD pathogenesis. In addition, Th2 cell differentiation was induced by application of HDM and the differentiation was also inhibited in the draining lymph node of NTP-treated mice. Finally, the expression of AD-related cytokines and chemokines was also decreased in NTP-treated mice. Taken together, these results suggest that NTP might be useful in the treatment of allergic skin diseases, such as AD.


Subject(s)
Mast Cells/drug effects , Mast Cells/metabolism , Plasma Gases/pharmacology , Animals , Chemokines/metabolism , Cytokines/metabolism , Dermatitis, Atopic/therapy , Disease Models, Animal , Eosinophils/metabolism , Hypersensitivity/pathology , Male , Mice , Mice, Inbred C57BL , Plasma Gases/metabolism , Skin/pathology , Th2 Cells/immunology
13.
Behav Brain Res ; 372: 112069, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31271817

ABSTRACT

Although epidemiological studies have identified an association between hearing loss and cognitive impairment, there is a lack of biological evidence detailing the mechanisms underlying this association. The present study investigated the effects of hearing loss on cognitive impairment using an at-risk model. In this animal model, amyloid-ß (Aß) was administered to the brain to such an extent that it did not cause cognitive impairments but made the brain vulnerable to risk factors. This study included four experimental groups based on hearing level and Aß administration. Behavioral tests were conducted to evaluate cognitive function, and synaptic protein levels were measured in the hippocampus and prefrontal cortex. The group with hearing loss and Aß administration showed significantly greater deficits on cognitive tests associated with the hippocampus than the other three groups (only Aß administration, only hearing loss, and without hearing loss or Aß administration). The hearing loss and Aß administration group also had significantly lower levels of synaptic proteins in the hippocampus than the other groups. The present results suggest that hearing loss may act as a risk factor for cognitive impairment in Alzheimer's disease. Additionally, the present findings indicate hearing loss may cause hippocampal synapses to be more vulnerable to Aß-induced damage.


Subject(s)
Cognitive Dysfunction/etiology , Hearing Loss/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cognition Disorders/psychology , Cognitive Dysfunction/metabolism , Hearing Loss/complications , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Male , Peptide Fragments/pharmacology , Rats , Rats, Wistar , Risk Factors , Synapses/metabolism , Temporal Lobe/metabolism
14.
Aging Cell ; 18(1): e12872, 2019 02.
Article in English | MEDLINE | ID: mdl-30515991

ABSTRACT

Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive memory loss resulting from cumulative neuronal cell death. O-linked ß-N-acetyl glucosamine (O-GlcNAc) modification of the proteins reflecting glucose metabolism is altered in the brains of patients with AD. However, the link between altered O-GlcNAc modification and neuronal cell death in AD is poorly understood. Here, we examined the regulation of O-GlcNAcylation of c-Fos and the effects of O-GlcNAcylated c-Fos on neuronal cell death during AD pathogenesis. We found that amyloid beta (Aß)-induced O-GlcNAcylation on serine-56 and 57 of c-Fos was resulted from decreased interaction between c-Fos and O-GlcNAcase and promoted neuronal cell death. O-GlcNAcylated c-Fos increased its stability and potentiated the transcriptional activity through higher interaction with c-Jun, resulting in induction of Bim expression leading to neuronal cell death. Taken together, Aß-induced O-GlcNAcylation of c-Fos plays an important role in neuronal cell death during the pathogenesis of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-fos/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Cell Death/drug effects , Cell Line , Gene Expression Regulation/drug effects , Glycosylation/drug effects , Humans , Mice, Transgenic , Neurons/drug effects , Protein Stability/drug effects , Proto-Oncogene Proteins c-fos/genetics , Rats, Sprague-Dawley , Transcription, Genetic/drug effects , beta-N-Acetylhexosaminidases/metabolism
15.
Sci Rep ; 8(1): 13754, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30213992

ABSTRACT

Adequate and rapid mucosal regeneration is one of the most important factors in the healing process of nasal mucosa after surgery or trauma. In particular, delayed mucosal regeneration after surgery is an important cause of surgical failure. However, no effective treatment is available yet. Non-thermal plasma (NTP) has several medical effects, but the existing probe type is limited to local direct treatment. Therefore, we investigated the various effects using liquid type plasma to overcome this limitation. In addition, the therapeutic effects of non-thermal plasma treated solution (NTS) on nasal mucosa have yet to be determined. Experiments were carried out using BEAS-2B, a human bronchial epithelial cell line similar to nasal mucosa epithelium. NTS had no cytotoxicity to the BEAS-2B cells and enhanced cell proliferation. NTS also promoted migration of BEAS-2B cells. NTS increased cell proliferation and migration via epidermal growth factor receptor (EGFR) activities and epithelial-to-mesenchymal transition (EMT) signaling. Furthermore, NTS enhanced wound healing of nasal mucosa in an animal model. Accordingly, NTS promotes nasal mucosa wound healing by increasing cell proliferation and migration. These findings suggest the therapeutic potential of NTS in nasal mucosa wound healing.


Subject(s)
Cell Proliferation/radiation effects , Nasal Mucosa/physiopathology , Plasma Gases , Regeneration , Animals , Bronchi/pathology , Bronchi/radiation effects , Cell Movement/radiation effects , Disease Models, Animal , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Epithelial-Mesenchymal Transition/radiation effects , Epithelium/pathology , Epithelium/radiation effects , Genes, erbB-1/genetics , Humans , Nasal Mucosa/radiation effects , Nasal Mucosa/surgery , Rats , Signal Transduction/radiation effects , Wound Healing/radiation effects
16.
Sci Rep ; 8(1): 12520, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30131570

ABSTRACT

Although TRAIL can directly induce cell death in some cancer cells, it appears that TRAIL resistance exists in many cancers. This study focuses on anti-cancer drugs for TRAIL-resistant head and neck cancer (HNC) to provide further progress toward effective cancer therapy. Results indicate in TRAIL-resistant HNC cells, that combined TRAIL and VPA treatment greatly reduced cell viability and therefore induced cell death, relative to treatment with TRAIL or VPA alone. A caspase-dependent signaling pathway was demonstrated, and combined treatment with TRAIL and VPA also significantly decreased the expression of HDAC4. When we pretreated cells with z-VAD followed by combined treatment with TRAIL and VPA, cell death was blocked with no reduction in expression of HDAC4. To confirm that cell death involved HDAC4 in HNC cells, we knocked down expression of HDAC4 with siRNA, followed by treatment with TRAIL and VPA. Results showed that loss of HDAC4 sensitized the TRAIL-resistant HNC cells to apoptotic cell death. Finally, we showed elevated expression of HDAC4 in HNC tissues compared to normal tissues obtained from the same patients. In conclusion, we suggest that combined VPA and TRAIL treatment may be a promising therapy for HNC via HDAC4 degradation.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Head and Neck Neoplasms/drug therapy , Histone Deacetylases/chemistry , Repressor Proteins/chemistry , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Valproic Acid/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Histone Deacetylases/genetics , Humans , Proteolysis , Repressor Proteins/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
17.
Oncol Rep ; 40(3): 1734-1742, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30015943

ABSTRACT

Lung cancer is the first leading cause of cancer­related death in the United States. Non­small cell lung cancer (NSCLC) is the most common type of lung cancer and is associated with a poor patient prognosis. Identification of promising molecular targets is required for the effective prevention and therapy of NSCLC. Epithelial­specific ETS­1 (ESE­1) belongs to the superfamily of ETS transcription factors. The effect of ESE­1 on tumorigenesis is controversial in several types of cancer while its role in lung cancer remains unknown. The present study was designed to investigate whether ESE­1 expression affects tumorigenic activity using human NSCLC cells and a mouse xenograft model. ESE­1 expression suppressed anchorage­independent growth in soft agar assay and led to an increase in G1 arrest and apoptosis in human NSCLC cells. ESE­1 expression suppressed the invasion and migration of human NSCLC cells. Western blot analysis, RT­PCR and promoter assay indicated that ESE­1 expression was transcriptionally downregulated by treatment of transforming growth factor (TGF)­ß, an EMT (epithelial­mesenchymal transition) stimulator. The xenograft study indicated that ESE­1 expression inhibited tumor formation and development. Our data demonstrated that ESE­1 plays a key role as a tumor suppressor in human NSCLC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement , Cell Proliferation , DNA-Binding Proteins/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , Carcinogenesis , Carcinoma, Non-Small-Cell Lung/metabolism , Epithelial-Mesenchymal Transition , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Sci Rep ; 8(1): 2277, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396442

ABSTRACT

The accumulation and differentiation of adipocytes contribute to the development of obesity and metabolic diseases. It is well-known that interactions of transcription factors such as peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer binding protein α (C/EBPα), and endoplasmic reticulum (ER) stress are required for adipogenesis. Recently, use of nonthermal atmospheric plasma (NTP) is expanding from the biomedical field into various other fields. In this study, we investigated whether nonthermal plasma-treated solution (NTS) has an inhibitory effect on adipogenesis and elucidated its mechanisms. Our results demonstrated that NTS significantly inhibited pre-adipocyte differentiation into adipocytes based on Oil Red O staining and triglyceride accumulation. Moreover, NTS treatment suppressed the mRNA and protein expression levels of key adipogenic transcription factors, and adipocyte-specific genes. NTS also down-regulated endoplasmic reticulum stress-related proteins. Consistent with in vitro studies, an animal study using a mouse model of diet-induced obesity showed that NTS treatment reduced body weight and fat, ER stress/UPR, triglyceride, and adipogenic marker level without altering food intake. These findings indicate that NTS inhibits adipogenic differentiation, and provide a mechanistic explanation of the inhibitory effect of NTS on adipogenesis. Taken together, our results suggest that NTS might be useful to treat obesity and obesity-related diseases.


Subject(s)
Adipocytes/drug effects , Cell Differentiation/drug effects , Endoplasmic Reticulum/drug effects , Lipogenesis/drug effects , Plasma Gases , Solutions/chemistry , Stress, Physiological/drug effects , 3T3-L1 Cells , Adipocytes/physiology , Animals , Body Weight , Disease Models, Animal , Mice , Obesity/prevention & control , Solutions/administration & dosage
19.
Autophagy ; 14(3): 385-403, 2018.
Article in English | MEDLINE | ID: mdl-29260979

ABSTRACT

HSPA5/GRP78/BiP plays an important role in cell survival or tumor progression. For these reasons, HSPA5 is an emerging therapeutic target in cancer development. Here we report that HSPA5 contributes to head and neck cancer (HNC) survival via maintenance of lysosomal activity; however, a nonthermal plasma (NTP, considered as a next-generation cancer therapy)-treated solution (NTS) inhibits HNC progression through HSPA5-dependent alteration of lysosomal activity. HSPA5 prevents NTS-induced lysosome inhibition through lysosomal-related proteins or regulation of gene expression. However, NTS-induced MUL1/MULAN/GIDE/MAPL (mitochondrial ubiquitin ligase activator of NFKB 1) leads to downregulation of HSPA5 via K48-linked ubiquitination at the lysine 446 (K446) residue. MUL1 knockdown hinders NTS-induced lysosome inhibition or cytotoxicity through the reduction of HSPA5 ubiquitination in HNC cells. While MUL1 was suppressed, HSPA5 was overexpressed in tissues of HNC patients. NTS strongly inhibited HNC progression via alterations of expression of MUL1 and HSPA5, in vivo in a xenograft model. However, NTS did not induce inhibition of tumor progression or HSPA5 reduction in MUL1 knockout (KO) HNC cells which were generated by CRISPR/Cas9 system. The data provide compelling evidence to support the idea that the regulation of the MUL1-HSPA5 axis can be a novel strategy for the treatment of HNC.


Subject(s)
Autophagy/physiology , Head and Neck Neoplasms/metabolism , Heat-Shock Proteins/metabolism , Lysosomes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line, Tumor , Cell Survival/physiology , Endoplasmic Reticulum Chaperone BiP , Humans , Mice , Ubiquitination/physiology , Xenograft Model Antitumor Assays
20.
Mol Neurodegener ; 12(1): 23, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28241840

ABSTRACT

BACKGROUND: Reduction or inhibition of histone deacetylase 6 (HDAC6) has been shown to rescue memory in mouse models of Alzheimer's disease (AD) and is recently being considered a possible therapeutic strategy. However, the restoring mechanism of HDAC6 inhibition has not been fully understood. METHODS AND RESULTS: Here, we found that an anti-oxidant protein Peroxdiredoxin1 (Prx1), a substrate of HDAC6, malfunctions in Aß treated cells, the brains of 5xFAD AD model mice and AD patients. Malfunctioning Prx1, caused by reduced Prx1 acetylation levels, was recovered by HDAC6 inhibition. Increasing acetylation levels of Prx1 by HDAC6 inhibition recovered elevated reactive oxygen species (ROS) levels, elevated Ca2+ levels and impaired mitochondrial axonal transport, sequentially, even in the presence of Aß. Prx1 mutant studies on the K197 site for an acetylation mimic or silencing mutation support the results showing that HDAC6 inhibitor restores Aß-induced disruption of ROS, Ca2+ and axonal transport. CONCLUSIONS: Taken together, increasing acetylation of Prx1 by HDAC6 inhibition has several beneficial effects in AD pathology. Here, we present the novel mechanism by which elevated acetylation of Prx1 rescues mitochondrial axonal transport impaired by Aß. Therefore, our results suggest that modulation of Prx1 acetylation by HDAC6 inhibition has great therapeutic potential for AD and has further therapeutic possibilities for other neurodegenerative diseases as well.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Axonal Transport , Histone Deacetylases/metabolism , Peroxiredoxins/metabolism , Acetylation/drug effects , Alzheimer Disease/pathology , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...