Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
IBRO Neurosci Rep ; 16: 336-344, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38390232

ABSTRACT

Alzheimer's disease (AD) is the most common age-related progressive neurodegenerative disorder. The accumulation of amyloid beta-peptide is a neuropathological marker of AD. While melatonin is recognized to have protective effects on aging and neurodegenerative disorders, the therapeutic effect of melatonin on calcineurin in AD is poorly understood. In this study, we examined the effect and underlying molecular mechanisms of melatonin treatment on amyloid beta-mediated neurotoxicity in neuroblastoma cells. Melatonin treatment decreased calcineurin and autophagy in neuroblastoma cells. Electron microscopy images showed that melatonin inhibited amyloid beta-induced autophagic vacuoles. The increase in the amyloid beta-induced apoptosis rate was observed more in PrPC-expressing ZW cells than in PrPC-silencing Zpl cells. Taken together, the results suggest that by mitigating the effect of calcineurin and autophagy flux activation, melatonin could also rescue amyloid beta-induced neurotoxic effects. These findings may be relevant to therapy for neurodegenerative diseases, including AD.

2.
BMC Neurosci ; 24(1): 59, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37932682

ABSTRACT

BACKGROUND: Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus that has no specific treatment except for supportive medical care. JEV is a neurotropic virus that affects the nervous system and triggers inflammation in the brain. METHODS: Melatonin is used as a sleep-inducing agent in neurophysiology and may serve as a protective agent against neurological and neurodegenerative diseases. Herein, we investigated the effects of melatonin and the critical roles of the serine/threonine protein phosphatase calcineurin during JEV infection in SK-N-SH neuroblastoma cells. RESULTS: Melatonin treatment decreased JEV replication and JEV-mediated neurotoxicity. Calcineurin activity was increased by JEV infection and inhibited by melatonin treatment. Through calcineurin regulation, melatonin decreased the JEV-mediated neuroinflammatory response and attenuated JEV-induced autophagy. CONCLUSIONS: Calcineurin inactivation has a protective effect in JEV-infected neuronal cells, and melatonin is a novel resource for the development of anti-JEV agents.


Subject(s)
Encephalitis Virus, Japanese , Encephalitis, Japanese , Melatonin , Animals , Humans , Encephalitis Virus, Japanese/physiology , Calcineurin/pharmacology , Melatonin/pharmacology , Autophagy
3.
Virus Res ; 338: 199249, 2023 12.
Article in English | MEDLINE | ID: mdl-37858731

ABSTRACT

Flaviviruses are a major cause of viral diseases worldwide, for which effective treatments have yet to be discovered. The prion protein (PrPc) is abundantly expressed in brain cells and has been shown to play a variety of roles, including neuroprotection, cell homeostasis, and regulation of cellular signaling. However, it is still unclear whether PrPc can protect against flaviviruses. In this study, we investigated the role of PrPc in regulating autophagy flux and its potential antiviral activity during Japanese encephalitis virus (JEV) infection. Our in vivo experiment showed that JEV was more lethal to the PrPc knocked out mice which was further supported by histological analysis, western blot and rtPCR results from infected mice brain samples. Role of PrPc against viral propagation in vitro was verified through cell survival study, protein expression and RNA replication analysis, and adenoviral vector assay by overexpressing PrPc. Further analysis indicated that after virus entry, PrPc inhibited autophagic flux that prevented JEV replication inside the host cell. Our results from in vivo and in vitro investigations demonstrate that prion protein effectively inhibited JEV propagation by regulating autophagy flux which is used by JEV to release its genetic material and replication after entering the host cell, suggesting that prion protein may be a promising therapeutic target for flavivirus infection.


Subject(s)
Encephalitis Virus, Japanese , Encephalitis, Japanese , Animals , Mice , Prion Proteins/genetics , Prion Proteins/pharmacology , Cell Line , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Virus Replication
4.
ACS Chem Neurosci ; 12(17): 3277-3283, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34424663

ABSTRACT

Prion diseases are mortal neurodegenerative pathologies that are caused by the accumulation of abnormal prion protein (PrPSc) in the brain. Recent advances reveal that calcineurin may play a critical role in regulating nuclear factor kappa B (NF-κB) in the calcium-calmodulin pathway. However, the exact mechanism by calcineurin remains unclear. In the present study, we observed that the prion peptide induces calcineurin and autophagy activation. Also, NF-κB and proinflammatory cytokines like interleukin (IL)-6 and tumor necrosis factor (TNF)-α are upregulated upon exposure to prion peptide in human neuroblastoma. The results show that the prion peptide induces calcineurin activation, leading to the activation of NF-κB transcription factor via autophagy signaling. Expression of TNF-α and IL-6 was increased by calcineurin activation and blocked by calcineurin inhibitor and autophagy inhibitor treatments. Collectively, these findings indicate that calcineurin activation mediated by prion protein induces NF-κB-driven neuroinflammation via autophagy pathway, suggesting that calcineurin and autophagy may be possible therapeutic targets for neuroinflammation in neurodegeneration diseases including prion disease.


Subject(s)
NF-kappa B , Prions , Autophagy , Calcineurin , Calcium , Humans , Peptides
5.
Oxid Med Cell Longev ; 2021: 5572129, 2021.
Article in English | MEDLINE | ID: mdl-34394828

ABSTRACT

Prion diseases are caused by PrPsc accumulation in the brain, which triggers dysfunctional mitochondrial injury and reactive oxygen species (ROS) generation in neurons. Recent studies on prion diseases suggest that endoplasmic reticulum (ER) stress induced by misfolding proteins such as misfolded prion protein results in activation of calcineurin. Calcineurin is a calcium-related protein phosphatase of type 2B that exists in copious quantities in the brain and acts as a critical nodal component in the control of cellular functions. To investigate the relationship between calcineurin and intracellular ROS, we assessed the alteration of CaN and ROS induced by prion peptide (PrP) 106-126. Human prion peptide increased mitochondrial ROS by activating calcineurin, and the inhibition of calcineurin activity protected mitochondrial function and neuronal apoptosis in neuronal cells. These results suggest that calcineurin plays a pivotal role in neuronal apoptosis by mediating mitochondrial injury and ROS in prion diseases.


Subject(s)
Calcineurin/metabolism , Mitochondria/drug effects , Peptides/pharmacology , Prion Proteins/chemistry , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cytosol/drug effects , Cytosol/metabolism , Humans , Mitochondria/metabolism , Peptides/chemical synthesis , Tacrolimus/pharmacology , Up-Regulation/drug effects
6.
Mol Med Rep ; 23(6)2021 06.
Article in English | MEDLINE | ID: mdl-33846779

ABSTRACT

Prion diseases, which involve the alteration of cellular prion protein into a misfolded isoform, disrupt the central nervous systems of humans and animals alike. Prior research has suggested that peroxisome proliferator­activator receptor (PPAR)γ and autophagy provide some protection against neurodegeneration. PPARs are critical to lipid metabolism regulation and autophagy is one of the main cellular mechanisms by which cell function and homeostasis is maintained. The present study examined the effect of troglitazone, a PPARγ agonist, on autophagy flux in a prion peptide (PrP) (106­126)­mediated neurodegeneration model. Western blot analysis confirmed that treatment with troglitazone increased LC3­II and p62 protein expression, whereas an excessive increase in autophagosomes was verified by transmission electron microscopy. Troglitazone weakened PrP (106­126)­mediated neurotoxicity via PPARγ activation and autophagy flux inhibition. A PPARγ antagonist blocked PPARγ activation as well as the neuroprotective effects induced by troglitazone treatment, indicating that PPARγ deactivation impaired troglitazone­mediated protective effects. In conclusion, the present study demonstrated that troglitazone protected primary neuronal cells against PrP (106­126)­induced neuronal cell death by inhibiting autophagic flux and activating PPARγ signals. These results suggested that troglitazone may be a useful therapeutic agent for the treatment of neurodegenerative disorders and prion diseases.


Subject(s)
Autophagy/drug effects , Hypoglycemic Agents/pharmacology , Neurons/metabolism , PPAR gamma/metabolism , Peptide Fragments/adverse effects , Prions/adverse effects , Troglitazone/pharmacology , Animals , Autophagy-Related Protein 5/genetics , Cell Line , Humans , Mice , Mice, Inbred ICR , Neurons/drug effects , Neuroprotective Agents/pharmacology , PPAR gamma/agonists , Prion Proteins
7.
Cell Commun Signal ; 18(1): 109, 2020 07 11.
Article in English | MEDLINE | ID: mdl-32650778

ABSTRACT

BACKGROUND: The distinctive molecular structure of the prion protein, PrPsc, is established only in mammals with infectious prion diseases. Prion protein characterizes either the transmissible pathogen itself or a primary constituent of the disease. Our report suggested that prion protein-mediated neuronal cell death is triggered by the autophagy flux. However, the alteration of intracellular calcium levels, AMPK activity in prion models has not been described. This study is focused on the effect of the changes in intracellular calcium levels on AMPK/autophagy flux pathway and PrP (106-126)-induced neurotoxicity. METHODS: Western blot and Immunocytochemistry was used to detect AMPK and autophagy-related protein expression. Flow cytometry and a TdT-mediated biotin-16-dUTP nick-end labeling (TUNEL) assay were used to detect the percentage of apoptotic cells. Calcium measurement was employed using fluo-4 by confocal microscope. RESULTS: We examined the effect of calcium homeostasis alterations induced by human prion peptide on the autophagy flux in neuronal cells. Treatment with human prion peptide increased the intracellular calcium concentration and induced cell death in primary neurons as well as in a neuronal cell line. Using pharmacological inhibitors, we showed that the L-type calcium channel is involved in the cellular entry of calcium ions. Inhibition of calcium uptake prevented autophagic cell death and reduction in AMP-activated protein kinase (AMPK) activity induced by human prion peptide. CONCLUSION: Our data demonstrated that prion peptide-mediated calcium inflow plays a pivotal role in prion peptide-induced autophagic cell death, and reduction in AMPK activity in neurons. Altogether, our results suggest that calcium influx might play a critical role in neurodegenerative diseases, including prion diseases. Video Abstract.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy , Calcium/metabolism , Neurons/metabolism , Neurons/pathology , Peptides/pharmacology , Prions/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Calcium Channels, L-Type/metabolism , Down-Regulation/drug effects , Intracellular Space/metabolism , Mice, Inbred ICR , Neurons/drug effects , Phosphorylation/drug effects
8.
Int J Biochem Cell Biol ; 119: 105680, 2020 02.
Article in English | MEDLINE | ID: mdl-31866508

ABSTRACT

It is usually accepted that prion proteins induce apoptosis in nerve cells. However, the mechanisms of PrPSc-neurotoxicity are not completely clear. Calcineurin is a Ca2+/calmodulin-dependent phosphatase. It activates autophagy, and may represent a link between deregulation of Ca2+ homeostasis and neuronal cell death. In this study, the effect of calcineurin activation mediated by human prion protein induced neuronal cell death via AMPK dephosphorylation and autophagy, was investigated. Synthetic peptides of PrP (PrP 106-126) increased calcineurin activity, without changing the levels of this protein phosphatase. Furthermore, these peptides reduced the levels of AMPK phosphorylation at threonine residue 172 and in autophagy activation. Calcineurin inhibitor, FK506, prevented this effect. The data showed that PrP-treated neurons had lower levels of AMPK than control neurons. This decrease in AMPK levels was matched via activation of autophagy. FK506 prevented the changes in AMPK and autophagy levels induced by PrP peptides. Taken together, the data demonstrated that prion peptides triggered an apoptotic cascade via calcineurin activation, which mediated AMPK dephosphorylation and autophagy activation. Therefore, these data suggest that therapeutic strategies targeting calcineurin inhibition might facilitate the management of neurodegenerative disorders including prion disease.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Calcineurin/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Neurons/drug effects , Peptide Fragments/pharmacology , Prions/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Calcineurin Inhibitors/pharmacology , Cell Line, Tumor , Humans , Neuroblastoma/enzymology , Neuroblastoma/metabolism , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Phosphorylation/drug effects , Tacrolimus/pharmacology
9.
Mol Neurobiol ; 56(6): 4192-4202, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30288697

ABSTRACT

Accumulation of prion protein (PrPc) into a protease-resistant form (PrPsc) in the brains of humans and animals affects the central nervous system. PrPsc occurs only in mammals with transmissible prion diseases. Prion protein refers to either the infectious pathogen itself or the main component of the pathogen. Recent studies suggest that autophagy is one of the major functions that keep cells alive and which has a protective effect against neurodegeneration. In this study, we investigated whether the anti-hypertensive drug, captopril, could attenuate prion peptide PrP (106-126)-induced calcium alteration-mediated neurotoxicity. Treatment with captopril increased both LC3-II (microtubule-associated protein 1A/1B-light chain 3-II) and p62 protein levels, indicating autophagy flux inhibition. Electron microscopy confirmed the occurrence of autophagic flux inhibition in neuronal cells treated with captopril. Captopril attenuated PrP (106-126)-induced neuronal cell death via AMPK activation and autophagy inhibition. Compound C suppressed AMPK activation as well as the neuroprotective effects of captopril. Thus, these data showed that an anti-hypertensive drug has a protective effect against prion-mediated neuronal cell death via autophagy inhibition and AMPK activation, and also suggest that anti-hypertensive drugs may be effective therapeutic agents against neurodegenerative disorders, including prion diseases.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Captopril/pharmacology , Neurons/enzymology , Neurons/pathology , Peptides/metabolism , Prions/metabolism , Animals , Calcium/metabolism , Enzyme Activation/drug effects , Mice , Neurons/drug effects , Neurons/ultrastructure , Neurotoxins/toxicity
10.
Int J Biochem Cell Biol ; 102: 20-30, 2018 09.
Article in English | MEDLINE | ID: mdl-29929000

ABSTRACT

Telmisartan broadly used for the treatment of hypertension that is also known for its anticancer properties. TRAIL has the potential to kill tumor cells with minimal toxicity in normal cells by binding to death receptors, DR4 and DR5. Unfortunately, these TRAIL-death receptors have failed as most human cancers are resistant to TRAIL-mediated apoptosis. In this study, we evaluated telmisartan as a novel TRAIL-DR5-targeting agent with the aim of rendering TRAIL-based cancer therapies more active. Herein, we demonstrated that telmisartan could sensitize TRAIL and enhance NSCLC tumor cell death. The molecular mechanism includes the blocking of AMPK phosphorylation causes inhibition of autophagy flux by telmisartan resulting in ROS generation leading to death receptor (DR5) upregulation and subsequent activation of the caspase cascade by TRAIL treatment. Furthermore, using chloroquine and siATG5 significantly enhances ROS production and application of the ROS scavenger N-acetyl-cysteine (NAC) rescues the cells undergoing apoptosis by abrogating the expression of DR5 and finally the caspase cascade. Additionally, NAC treatment also maintains autophagy flux and makes the cells unresponsive to TRAIL. In summary, telmisartan in combination with TRAIL exhibits enhanced cytotoxic capacity toward lung cancer cells, thereby providing the potential for effective and novel therapeutic approaches to treat lung cancer.


Subject(s)
Autophagy/drug effects , Lung Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Telmisartan/pharmacology , Up-Regulation/drug effects , A549 Cells , AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Targeted Therapy , Phosphorylation/drug effects , Telmisartan/therapeutic use
11.
Oncotarget ; 8(59): 100021-100033, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29245957

ABSTRACT

The combination of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) with subsidiary agents is a promising anticancer strategy to conquer TRAIL resistance in malignant cells. Glipizide is a second-generation oral hypoglycemic medicine for the cure of type II diabetes because of its capability to selectively stimulate insulin secretion from ß-cells. In this study, we revealed that glipizide could trigger TRAIL-mediated apoptotic cell death in human lung adenocarcinoma cells. Pretreatment with glipizide downregulation of p-Akt and p-mTOR in different concentrations. In addition, LC3-II and p-Akt was suppressed in the presence of LY294002, a well-known inhibitor of P13K. Treatment with glipizide commenced in a slight increase in conversion rate of LC3-I to LC3-II and significantly decreased p62 expression levels in a dose-dependent manner. This indicates that glipizide encouraged autophagy flux activation in human lung cancer cells. Inhibition of autophagy flux applying a specific inhibitor and genetically modified ATG5 siRNA enclosed glipizide-mediated enhancing effect of TRAIL. These data demonstrate that inhibition of Akt/mTOR by glipizide sensitizes TRAIL-induced tumor cell death through activating autophagy flux and also suggest that glipizide may be a combination therapeutic target with TRAIL protein in TRAIL-resistant cancer cells.

12.
Oncotarget ; 8(16): 26819-26831, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28460464

ABSTRACT

Members of the tumor necrosis factor (TNF) transmembrane cytokine superfamily, such as TNFα and Fas ligand (FasL), play crucial roles in inflammation and immunity. TRAIL is a member of this superfamily with the ability to selectively trigger cancer cell death but does not motive cytotoxicity to most normal cells. Troglitazone are used in the cure of type II diabetes to reduce blood glucose levels and improve the sensitivity of an amount of tissues to insulin. In this study, we revealed that troglitazone could trigger TRAIL-mediated apoptotic cell death in human lung adenocarcinoma cells. Pretreatment of troglitazone induced activation of PPARγ in a dose-dependent manner. In addition conversion of LC3-I to LC3-II and PPARγ was suppressed in the presence of GW9662, a well-characterized PPARγ antagonist. Treatment with troglitazone resulted in a slight increase in conversion rate of LC3-I to LC3-II and significantly decreased p62 expression levels in a dose-dependent manner. This indicates that troglitazone induced autophagy flux activation in human lung cancer cells. Inhibition of autophagy flux applying a specific inhibitor and genetically modified ATG5 siRNA enclosed troglitazone-mediated enhancing effect of TRAIL. These data demonstrated that activation of PPARγ mediated by troglitazone enhances human lung cancer cells to TRAIL-induced apoptosis via autophagy flux and also suggest that troglitazone may be a combination therapeutic target with TRAIL protein in TRAIL-resistant cancer cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Chromans/pharmacology , Lung Neoplasms/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thiazolidinediones/pharmacology , Cell Line, Tumor , Humans , Troglitazone
13.
J Invest Dermatol ; 137(6): 1257-1266, 2017 06.
Article in English | MEDLINE | ID: mdl-28257794

ABSTRACT

Skin inflammation is a response of the immune system to infection and injury. In this study, we report that hinokitiol, a tropolone-related natural compound that exhibits antioxidant, anti-inflammatory, and anticancer properties in various cell types, can modulate the inflammatory responses of primary human keratinocytes challenged with lipopolysaccharide (LPS). Hinokitiol treatment inhibited LPS-mediated up-regulation of proinflammatory factors including tumor necrosis factor alpha, IL-6, and prostaglandin E2 (PGE2). NF-κB activation and cell migration induced by LPS were blocked in keratinocytes treated with hinokitiol. Sirt1, a class Ⅲ histone deacetylase, was up-regulated by hinokitiol treatment, and the inhibition of Sirt1 activity using a pharmacological inhibitor or genetic silencing blocked hinokitiol-mediated anti-inflammatory effects. Further, hyperactivation of Sirt1 deacetylase using an adenoviral vector also attenuated LPS-induced inflammatory responses. We thus show that hinokitiol can attenuate LPS-mediated proinflammatory signals via Sirt1 histone deacetylase activation in primary human keratinocytes and suggest that hinokitiol may be a potential therapeutic agent in skin inflammatory diseases like psoriasis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/prevention & control , Lipopolysaccharides/pharmacology , Monoterpenes/pharmacology , Sirtuin 1/metabolism , Tropolone/analogs & derivatives , Analysis of Variance , Cells, Cultured/cytology , Cells, Cultured/drug effects , Drug Interactions , Humans , Immunohistochemistry , Inflammation/pathology , Keratinocytes/cytology , Keratinocytes/drug effects , Real-Time Polymerase Chain Reaction/methods , Sirtuin 1/drug effects , Tropolone/pharmacology
14.
Oncotarget ; 7(52): 85697-85708, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27911875

ABSTRACT

Mitochondrial quality control is a process by which mitochondria undergo successive rounds of fusion and fission with dynamic exchange of components to segregate functional and damaged elements. Removal of mitochondrion that contains damaged components is accomplished via autophagy. In this study, we investigated whether ginsenoside Rg3, an active ingredient of the herbal medicine ginseng that is used as a tonic and restorative agent, could attenuate prion peptide, PrP (106-126)-induced neurotoxicity and mitochondrial damage. To this end, western blot and GFP-LC3B puncta assay were performed to monitor autophagy flux in neuronal cells; LC3B-II protein level was found to increase after Rg3 treatment. In addition, electron microscopy analysis showed that Rg3 enhanced autophagic vacuoles in neuronal cells. By using autophagy inhibitors wortmannin and 3-methyladenine (3MA) or autophagy protein 5 (Atg5) small interfering RNA (siRNA), we demonstrated that Rg3 could protect neurons against PrP (106-126)-induced cytotoxicity via autophagy flux. We found that Rg3 could also attenuate PrP (106-126)-induced mitochondrial damage via autophagy flux. Taken together, our results suggest that Rg3 is a possible therapeutic agent in neurodegenerative disorders, including prion diseases.


Subject(s)
Autophagy/drug effects , Ginsenosides/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Prions/toxicity , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis/drug effects , Autophagy/physiology , Cell Line, Tumor , Humans , Mitochondria/physiology , Proto-Oncogene Proteins c-akt/physiology
15.
Oncotarget ; 7(40): 65660-65668, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27582540

ABSTRACT

Epigallocatechin gallate (EGCG) is a major polyphenol in green tea. Recent studies have reported that EGCG can inhibit TRAIL-induced apoptosis and activate autophagic flux in cancer cells. However, the mechanism behind these processes is unclear. The present study found that EGCG prevents tumor cell death by antagonizing the TRAIL pathway and activating autophagy flux. Our results indicate that EGCG dose-dependently inhibits TRAIL-induced apoptosis and decreases the binding of death receptor 4 and 5 (DR4 and 5) to TRAIL. In addition, EGCG activates autophagy flux, which is involved in the inhibition of TRAIL cell death. We confirmed that the protective effect of EGCG can be reversed using genetic and pharmacological tools through re-sensitization to TRAIL. The inhibition of autophagy flux affects not only the re-sensitization of tumor cells to TRAIL, but also the restoration of death receptor proteins. This study demonstrates that EGCG inhibits TRAIL-induced apoptosis through the manipulation of autophagic flux and subsequent decrease in number of death receptors. On the basis of these results, we suggest further consideration of the use of autophagy activators such as EGCG in combination anti-tumor therapy with TRAIL.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Catechin/analogs & derivatives , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Receptors, Death Domain/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/metabolism , Anticarcinogenic Agents/pharmacology , Catechin/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Tumor Cells, Cultured
16.
Oncotarget ; 7(21): 29944-57, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27074563

ABSTRACT

Prion diseases are fatal neurodegenerative disorders that are derived from structural changes of the native PrPc. Recent studies indicated that hinokitiol induced autophagy known to major function that keeps cells alive under stressful conditions. We investigated whether hinokitiol induces autophagy and attenuates PrP (106-126)-induced neurotoxicity. We observed increase of LC3-II protein level, GFP-LC3 puncta by hinokitiol in neuronal cells. Addition to, electron microscopy showed that hinokitiol enhanced autophagic vacuoles in neuronal cells. We demonstrated that hinokitiol protects against PrP (106-126)-induced neurotoxicity via autophagy by using autophagy inhibitor, wortmannin and 3MA, and ATG5 small interfering RNA (siRNA). We checked hinokitiol activated the hypoxia-inducible factor-1α (HIF-1α) and identified that hinokitiol-induced HIF-1α regulated autophagy. Taken together, this study is the first report demonstrating that hinokitiol protected against prion protein-induced neurotoxicity via autophagy regulated by HIF-1α. We suggest that hinokitiol is a possible therapeutic strategy in neuronal disorders including prion disease.


Subject(s)
Autophagy/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Monoterpenes/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/physiology , Prion Diseases/drug therapy , Prions/physiology , Tropolone/analogs & derivatives , Adenine/analogs & derivatives , Adenine/metabolism , Androstadienes/pharmacology , Animals , Apoptosis/drug effects , Autophagy/genetics , Autophagy-Related Protein 5/genetics , Cells, Cultured , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Microscopy, Electron, Transmission , Neurons/ultrastructure , Primary Cell Culture , Protein Kinase Inhibitors/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction/drug effects , Tropolone/pharmacology , Wortmannin
17.
Oncotarget ; 7(21): 29989-30002, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27102156

ABSTRACT

An unusual molecular structure of the prion protein, PrPsc is found only in mammals with transmissible prion diseases. Prion protein stands for either the infectious pathogen itself or a main component of it. Recent studies suggest that autophagy is one of the major functions that keep cells alive and has a protective effect against the neurodegeneration. In this study, we investigated that the effect of human prion protein on autophagy-lysosomal system of primary neuronal cells. The treatment of human prion protein induced primary neuron cell death and decreased both LC3-II and p62 protein amount indicating autophagy flux activation. Electron microscope pictures confirmed the autophagic flux activation in neuron cells treated with prion protein. Inhibition of autophagy flux using pharmacological and genetic tools prevented neuron cell death induced by human prion protein. Autophagy flux induced by prion protein is more activated in prpc expressing cells than in prpc silencing cells. These data demonstrated that prion protein-induced autophagy flux is involved in neuron cell death in prion disease and suggest that autophagy flux might play a critical role in neurodegenerative diseases including prion disease.


Subject(s)
Apoptosis/physiology , Autophagy , Neurons/physiology , Peptide Fragments/physiology , Prion Diseases/metabolism , Prions/physiology , Animals , Cells, Cultured , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Neurons/ultrastructure , Primary Cell Culture , Sequestosome-1 Protein/metabolism
18.
Oncotarget ; 7(17): 23468-81, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-26992204

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily. TRAIL is regarded as one of the most promising anticancer agents, because it can destruct cancer cells without showing any toxicity to normal cells. Metformin is an anti-diabetic drug with anticancer activity by inhibiting tumor cell proliferation. In this study, we demonstrated that metformin could induce TRAIL-mediated apoptotic cell death in TRAIL-resistant human lung adenocarcinoma A549 cells. Pretreatment of metformindownregulation of c-FLIP and markedly enhanced TRAIL-induced tumor cell death by dose-dependent manner. Treatment with metformin resulted in slight increase in the accumulation of microtubule-associated protein light chain LC3-II and significantly decreased the p62 protein levels by dose-dependent manner indicated that metformin induced autophagy flux activation in the lung cancer cells. Inhibition of autophagy flux using a specific inhibitor and genetically modified ATG5 siRNA blocked the metformin-mediated enhancing effect of TRAIL. These data demonstrated that downregulation of c-FLIP by metformin enhanced TRAIL-induced tumor cell death via activating autophagy flux in TRAIL-resistant lung cancer cells and also suggest that metformin may be a successful combination therapeutic strategy with TRAIL in TRAIL-resistant cancer cells including lung adenocarcinoma cells.


Subject(s)
Adenocarcinoma/pathology , Apoptosis/drug effects , Autophagy/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Lung Neoplasms/pathology , Metformin/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Humans , Hypoglycemic Agents/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Tumor Cells, Cultured
19.
Oncotarget ; 7(11): 12075-88, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26918354

ABSTRACT

Melatonin (N-acetyl-5-methoxytryptamine), which is primarily synthesized in and secreted from the pineal gland, plays a pivotal role in cell proliferation as well as in the regulation of cell metastasis and cell survival in a diverse range of cells. The aim of this study is to investigate protection effect of melatonin on H2O2-induced cell damage and the mechanisms of melatonin in human keratinocytes. Hydrogen peroxide dose-dependently induced cell damages in human keratinocytes and co-treatment of melatonin protected the keratinocytes against H2O2-induced cell damage. Melatonin treatment activated the autophagy flux signals, which were identified by the decreased levels of p62 protein. Inhibition of autophagy flux via an autophagy inhibitor and ATG5 siRNA technique blocked the protective effects of melatonin against H2O2-induced cell death in human keratinocytes. And we found the inhibition of sirt1 using sirtinol and sirt1 siRNA reversed the protective effects of melatonin and induces the autophagy process in H2O2-treated cells. This is the first report demonstrating that autophagy flux activated by melatonin protects human keratinocytes through sirt1 pathway against hydrogen peroxide-induced damages. And this study also suggest that melatonin could potentially be utilized as a therapeutic agent in skin disease.


Subject(s)
Hydrogen Peroxide/pharmacology , Keratinocytes/drug effects , Melatonin/pharmacology , Sirtuin 1/metabolism , Autophagy/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Membrane Proteins/metabolism , Signal Transduction/drug effects , Skin/cytology , Skin/drug effects , Skin/metabolism , Transfection
20.
Oncotarget ; 7(4): 4356-68, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26517672

ABSTRACT

Niacin, also known as vitamin B3 or nicotinamide is a water-soluble vitamin that is present in black beans and rice among other foods. Niacin is well known as an inhibitor of metastasis in human breast carcinoma cells but the effect of niacin treatment on TRAIL-mediated apoptosis is unknown. Here, we show that niacin plays an important role in the regulation of autophagic flux and protects tumor cells against TRAIL-mediated apoptosis. Our results indicated that niacin activated autophagic flux in human colon cancer cells and the autophagic flux activation protected tumor cells from TRAIL-induced dysfunction of mitochondrial membrane potential and tumor cell death. We also demonstrated that ATG5 siRNA and autophagy inhibitor blocked the niacin-mediated inhibition of TRAIL-induced apoptosis. Taken together, our study is the first report demonstrating that niacin inhibits TRAIL-induced apoptosis through activation of autophagic flux in human colon cancer cells. And our results also suggest that autophagy inhibitors including genetic and pharmacological tools may be a successful therapeutics during anticancer therapy using TRAIL.


Subject(s)
Apoptosis/drug effects , Autophagy , Colonic Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Niacin/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Vasodilator Agents/pharmacology , Blotting, Western , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , RNA, Small Interfering/genetics , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...