Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
FEBS Lett ; 598(11): 1366-1374, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38553939

ABSTRACT

The interleukin (IL)-1 family of cytokines plays a pivotal role in immune responses. Among the members of IL-1 family, IL-1ß is synthesized as an inactive precursor (pro-IL-1ß) and becomes active upon cleavage, which is typically facilitated by inflammasomes through caspase-1. In our research, we explored the potential role of caspase-3 in the cleavage of pro-IL-1ß and found that caspase-3 cleaves pro-IL-1ß, specifically at Asp26. Moreover, we found that in the absence of caspase-3 cleavage, the release of active IL-1ß via the inflammasome is increased. Our study introduces pro-IL-1ß as a new substrate for caspase-3 and suggests that caspase-3-mediated cleavage has the potential to suppress IL-1ß-mediated inflammatory responses.


Subject(s)
Caspase 3 , Inflammasomes , Inflammation , Interleukin-1beta , Interleukin-1beta/metabolism , Inflammation/metabolism , Inflammation/immunology , Inflammasomes/metabolism , Humans , Caspase 3/metabolism , Animals , Protein Precursors/metabolism , Mice , Caspase 1/metabolism , HEK293 Cells , Interleukin-1
2.
Antioxidants (Basel) ; 13(1)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38275656

ABSTRACT

Peiminine is the main natural alkaloid compound extracted from the Chinese herb Fritillaria. Although peiminine is known for its antioxidant and anti-inflammatory effects in conditions such as mastitis and arthritis, its impact on inflammation induced by Cutibacterisum acnes (C. acnes) has not been explored. The aim of this study was to investigate the effect of peiminine on C. acnes-induced inflammatory responses in the skin and to identify the underlying mechanism involved. We discovered that peiminine inhibits the C. acnes-induced expression of inflammatory mediators such as pro-interleukin-1ß (pro-IL-1ß), cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in mouse bone marrow-derived macrophages (BMDMs). Peiminine suppressed the activation of nuclear factor-kappa B (NF-κB) without affecting the activation of mitogen-activated protein kinase (MAPK) pathways such as JNK, ERK, and p38 MAPK. In addition, we found that peiminine suppressed inflammatory cytokine expression and ameliorated histological symptoms in C. acnes-induced mouse skin. Our study is the first to provide evidence that peiminine has an inhibitory effect on acne, and it points toward the potential of incorporating peiminine into cosmetic and pharmaceutical formulations for acne treatment.

3.
Int J Mol Sci ; 24(5)2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36901873

ABSTRACT

Cutibacterium acnes (C. acnes), a Gram-positive anaerobic bacterium, proliferates in hair follicles and pores and causes inflammation in the skin of young people. The rapid growth of C. acnes triggers macrophages to secrete proinflammatory cytokines. Pyrrolidine dithiocarbamate (PDTC) is a thiol compound that exerts antioxidant and anti-inflammatory effects. Although the anti-inflammatory function of PDTC in several inflammatory disorders has been reported, the effect of PDTC on C. acnes-induced skin inflammation remains unexplored. In the present study, we examined the effect of PDTC on C. acnes-induced inflammatory responses and determined the mechanism by using in vitro and in vivo experimental models. We found that PDTC significantly inhibited the expression of C. acnes-induced proinflammatory mediators, such as interleukin-1ß (IL-1ß), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and NOD-like receptor (NLR) pyrin domain-containing 3 (NLRP3), in mouse-bone-marrow-derived macrophage (BMDM) cells. PDTC suppressed C. acnes-induced activation of nuclear factor-kappa B (NF-κB), which is the major transcription factor for proinflammatory cytokine expression. In addition, we found that PDTC inhibited caspase-1 activation and IL-1ß secretion through suppressing NLRP3 and activated the melanoma 2 (AIM2) inflammasome but not the NLR CARD-containing 4 (NLRC4) inflammasome. Moreover, we found that PDTC improved C. acnes-induced inflammation by attenuating C. acnes-induced IL-1ß secretion in a mouse acne model. Therefore, our results suggest that PDTC has potential therapeutic value for the amelioration of C. acnes-induced skin inflammation.


Subject(s)
Dermatitis , Inflammasomes , Mice , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Base Composition , Phylogeny , RNA, Ribosomal, 16S , Sequence Analysis, DNA , Cytokines/metabolism , Interleukin-6/genetics , Inflammation/pathology , NLR Proteins , Anti-Inflammatory Agents
4.
Immunology ; 168(1): 110-119, 2023 01.
Article in English | MEDLINE | ID: mdl-36054548

ABSTRACT

We recently reported that lactoferrin (LF) induces Foxp3+ Treg differentiation through binding to TGFß receptor III (TßRIII), and this activity was further enhanced by TGFß1. Generally, a low T-cell receptor (TCR) signal strength is favourable for Foxp3+ Treg differentiation. In the present study, we explored the effect of lactoferrin chimera (LFch, containing lactoferricin [aa 17-30] and lactoferrampin [aa 265-284]), along with TGFß1 on Foxp3+ Treg differentiation. LFch alone did not induce Foxp3 expression, yet LFch dramatically enhanced TGFß1-induced Foxp3 expression. LFch had little effect on the phosphorylation of Smad3, a canonical transcriptional factor of TGFß1. Instead, LFch attenuated the phosphorylation of S6 (a target of mTOR), IκB and PI3K. These activities of LFch were completely abrogated by pretreatment of LFch with soluble TGFß1 receptor III (sTßRIII). Consistent with this, the activity of LFch on TGFß1-induced Foxp3 expression was also abrogated by treatment with sTßRIII. Finally, the TGFß1/LFch-induced T cell population substantially suppressed the proliferation of responder CD4+ T cells. These results indicate that LFch robustly enhances TGFß1-induced Foxp3+ Treg differentiation by diminishing TCR/CD28 signal intensity.


Subject(s)
CD28 Antigens , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/metabolism , Lactoferrin/pharmacology , Lactoferrin/metabolism , Receptors, Antigen, T-Cell/metabolism , Cell Differentiation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
5.
Antioxidants (Basel) ; 11(9)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36139814

ABSTRACT

Atopic dermatitis (AD) is a chronic inflammatory skin disease accompanied by severe itching and dry skin. Currently, the incidence of AD due to excessive activation of immune cells by various environmental factors is increasing worldwide, and research on inflammatory response inhibitors with fewer side effects is continuously needed. Cynanoside F (CF) is one of the pregnane-type compounds in the root of Cynanchum atratum, an oriental medicinal herb that has been shown to have antioxidant, antitumor, and anti-inflammatory effects. Although CF has been isolated as a component in Cynanchum atratum, the scientific role of CF has not yet been explored. In this study, we evaluated the effect of CF on AD and revealed the mechanism using in vitro and in vivo experimental models. CF significantly reduced lipopolysaccharide (LPS)-induced protein expression levels of interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and cyclooxygenase-2 (COX-2), which are important proinflammatory mediators in the RAW264.7 macrophage cell line. CF did not inhibit the nuclear factor-kappa B (NF-κB) signaling activated by LPS but significantly reduced the phosphorylation of mitogen-activated protein kinases (MAPKs), such as p38 MAPK, JNK, and ERK. CF consistently inhibited the activity of the activator protein-1 (AP-1) transcription factor, a downstream molecule of MAPK signaling. In addition, in an experiment using an oxazolone-induced AD mouse model, the CF-treated group showed a marked decrease in epidermal thickness, the number of infiltrated mast cells, and the amount of histamine. The mRNA levels of IL-1ß, interleukin-4 (IL-4), and thymic stromal lymphopoietin (TSLP) were consistently lowered in the group treated with CF. Moreover, the phosphorylation of c-Jun and c-Fos protein levels, which are the AP-1 components, were lowered in the skin tissues of CF-treated mice. These results provide the first evidence that CF has an inhibitory effect on AD and suggest the possibility of CF being developed as a potential therapeutic agent for AD.

6.
Antioxidants (Basel) ; 11(2)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35204088

ABSTRACT

Atopic dermatitis (AD) is a common inflammatory skin disorder, and numerous pharmacological approaches are employed to reduce symptoms. Natural products of plant-derived materials have been accepted as complementary therapy for the treatment of a wide range of inflammatory diseases. Cynanchi atrati (CA) is an oriental medicinal herb used in the treatment of acute urinary infection, febrile diseases, and laryngopharyngitis. However, the role of CA root extract in skin inflammation such as AD has not been explored yet. In this study, we examined the possible effect of CA root extract on skin inflammation and evaluated the underlying signaling mechanism using in vitro and in vivo modeling systems. Raw264.7 macrophages were used for in vitro experiments, and an oxazolone-induced AD mouse model was used to evaluate in vivo effects. CA extract significantly inhibited the expression levels of lipopolysaccharide (LPS)-induced pro-inflammatory cytokines such as interleukin-6 (IL-6) and interleukin-1ß (IL-1ß) in RAW264.7 macrophages. The CA root extract mediated suppression of pro-inflammatory cytokine expression and was associated with the decreased nuclear factor kappa B (NF-κB) gene transcriptional activation. Moreover, CA root extract attenuated the in vivo expression of IL-6 and tumor necrosis factor-α (TNF-α) and ear swelling in the AD mouse models. We also observed that the inhibitory effect of CA root extract on skin inflammation was accompanied by the upregulation of calcineurin 1 (RCAN1) expression, which functions in the inflammatory pathways by suppressing NF-κB signaling. We consistently observed that the immunosuppressive effect of CA root extract in AD was significantly perturbed in the RCAN1 knockout mice. In addition, we isolated a phenolic acid compound, sinapic acid (SA), from the CA root extract and found that SA consistently exerted an immunosuppressive effect in RAW264.7 macrophages by inducing RCAN1 expression. Our results provide the first evidence that CA root extract and its phenolic acid constituent, SA, modulate NF-κB signaling pathways by inducing RCAN1 expression in the skin inflammation process. Thus, we suggest that CA root extract has a therapeutic value for the treatment of AD by targeting endogenous immune regulators.

7.
J Cell Physiol ; 237(2): 1406-1417, 2022 02.
Article in English | MEDLINE | ID: mdl-34647615

ABSTRACT

Regulator of calcineurin 1 (RCAN1) is located close to the Down syndrome critical region (DSCR) on human chromosome 21 and is related to the Down syndrome (DS) phenotype. To identify a novel binding partner of RCAN1, we performed yeast two-hybrid screening and identified mitogen-activated protein (MAP) kinase/extracellular signal-regulated kinase (ERK) kinase 1 (MEK1) as a partner. MEK1 was able to bind and phosphorylate RCAN1 in vitro and in vivo. MEK1-dependent RCAN1 phosphorylation caused an increase in RCAN1 expression by increasing the protein half-life. Nerve growth factor (NGF)-dependent activation of the MEK1 pathway consistently induced RCAN1 expression. Moreover, we found that RCAN1 overexpression inhibited NGF-induced neurite outgrowth and expression of neuronal marker genes, such as growth cone-associated protein 43 (GAP43) and synapsin I, via inhibition of MEK1-ERK1/2 pathways. Our findings provide evidence that MEK1-dependent RCAN1 phosphorylation acts as an important molecular mechanism in the control of neuronal differentiation.


Subject(s)
Calcineurin , Nerve Growth Factor , Calcineurin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Nerve Growth Factor/metabolism , Phosphorylation , Transcription Factors/metabolism
8.
Cells ; 10(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34571975

ABSTRACT

A natural phenolic acid compound, sinapic acid (SA), is a cinnamic acid derivative that contains 3,5-dimethoxyl and 4-hydroxyl substitutions in the phenyl ring of cinnamic acid. SA is present in various orally edible natural herbs and cereals and is reported to have antioxidant, antitumor, anti-inflammatory, antibacterial, and neuroprotective activities. Although the anti-inflammatory function of SA has been reported, the effect of SA on the NOD-like receptor pyrin domain-containing 3 (NLRP3) inflammasome has not been explored. In the present study, to elucidate the anti-inflammatory mechanism of SA, we examined whether SA modulates the NLRP3 inflammasome. We found that SA blocked caspase-1 activation and IL-1ß secretion by inhibiting NLRP3 inflammasome activation in bone marrow-derived macrophages (BMDMs). Apoptosis-associated speck-like protein containing CARD (ASC) pyroptosome formation was consistently blocked by SA treatment. SA specifically inhibited NLRP3 activation but not the NLRC4 or AIM2 inflammasomes. In addition, SA had no significant effect on the priming phase of the NLRP3 inflammasome, such as pro-IL-1ß and NLRP3 inflammasome expression levels. Moreover, we found that SA attenuated IL-1ß secretion in LPS-induced systemic inflammation in mice and reduced lethality from endotoxic shock. Our findings suggest that the natural compound SA has potential therapeutic value for the suppression of NLRP3 inflammasome-associated inflammatory diseases.


Subject(s)
Coumaric Acids/pharmacology , Inflammasomes/drug effects , Inflammation/drug therapy , Inflammation/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Caspase 1/metabolism , Cells, Cultured , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells
9.
Int J Mol Sci ; 21(20)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081375

ABSTRACT

Y-27632 is known as a selective Rho-associated coiled coil-forming kinase (ROCK) inhibitor. Y-27632 has been shown to induce neurite outgrowth in several neuronal cells. However, the precise molecular mechanisms linking neurite outgrowth to Y-27632 are not completely understood. In this study, we examined the ability of Y-27632 to induce neurite outgrowth in PC12 cells and evaluated the signaling cascade. The effect of Y-27632 on the neurite outgrowth was inhibited by reactive oxygen species (ROS) scavengers such as N-acetyl cysteine (NAC) and trolox. Furthermore, Y-27632-induced neurite outgrowth was not triggered by NADPH oxidase 1 (NOX1) knockdown or diphenyleneiodonium (DPI), a NOX inhibitor. Suppression of the Rho-family GTPase Rac1, which is under the negative control of ROCK, with expression of the dominant negative Rac1 mutant (Rac1N17) prevented Y-27632-induced neurite outgrowth. Moreover, the Rac1 inhibitor NSC23766 prevented Y-27632-induced AKT and p21-activated kinase 1 (PAK1) activation. AKT inhibition with MK2206 suppressed Y-27632-induced PAK1 phosphorylation and neurite outgrowth. In conclusion, our results suggest that Rac1/NOX1-dependent ROS generation and subsequent activation of the AKT/PAK1 cascade contribute to Y-27632-induced neurite outgrowth in PC12 cells.


Subject(s)
Amides/pharmacology , Neuronal Outgrowth/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction , Acetylcysteine/pharmacology , Animals , Chromans/pharmacology , Free Radical Scavengers/pharmacology , NADPH Oxidase 1/metabolism , Onium Compounds/pharmacology , PC12 Cells , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
10.
Int J Mol Sci ; 20(14)2019 Jul 17.
Article in English | MEDLINE | ID: mdl-31319552

ABSTRACT

Acne is an inflammatory skin disorder in puberty with symptoms including papules, folliculitis, and nodules. Propionibacterium acnes (P. acnes) is the main anaerobic bacteria that cause acne. It is known to proliferate within sebum-blocked skin hair follicles. P. acnes activates monocytic cell immune responses to induce the expression of proinflammatory cytokines. Although the anti-inflammatory function of the Laurus nobilis (L. nobilis) extract (LNE) on several immunological disorders have been reported, the effect of LNE in P. acnes-mediated skin inflammation has not yet been explored. In the present study, we examined the ability of the LNE to modulate the P. acnes-induced inflammatory signaling pathway, and evaluated its mechanism. LNE significantly suppressed the expression of P. acnes-mediated proinflammatory cytokines, such as IL-1ß, IL-6, and NLRP3. We also found that LNE inhibited the inflammatory transcription factor NF-κB in response to P. acnes. In addition, eucalyptol, which is the main constituent of LNE, consistently inhibited P. acnes-induced inflammatory signaling pathways. Moreover, LNE significantly ameliorated P. acnes-induced inflammation in a mouse model of acne. We suggest for the first time that LNE hold therapeutic value for the improvement of P. acnes-induced skin inflammation.


Subject(s)
Acne Vulgaris/drug therapy , Anti-Inflammatory Agents/pharmacology , Eucalyptol/pharmacology , Gram-Positive Bacterial Infections/drug therapy , Laurus/chemistry , Plant Extracts/pharmacology , Propionibacterium acnes/growth & development , Acne Vulgaris/metabolism , Acne Vulgaris/microbiology , Acne Vulgaris/pathology , Animals , Anti-Inflammatory Agents/chemistry , Cell Line , Eucalyptol/chemistry , Gram-Positive Bacterial Infections/metabolism , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Mice , Plant Extracts/chemistry
11.
Mol Cell Endocrinol ; 494: 110488, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31207272

ABSTRACT

Hydrocortisone exerts adverse effects on various organs, including the heart. This study investigated the still unclear effects of hydrocortisone on electrophysiological and biochemical aspects of cardiac excitation-contraction coupling. In guinea pigs' hearts, hydrocortisone administration reduced the QT interval of ECG and the action potential duration (APD). In guinea pig ventricular myocytes, hydrocortisone reduced contraction and Ca2+ transient amplitudes. These reductions and the effects on APD were prevented by pretreatment with the protein kinase C (PKC) inhibitor staurosporine. In an overexpression system of Xenopus oocytes, hydrocortisone increased hERG K+ currents and reduced Kv1.5 K+ currents; these effects were negated by pretreatment with staurosporine. Western blot analysis revealed dose- and time-dependent changes in PKCα/ßII, PKCε, and PKCγ phosphorylation by hydrocortisone in guinea pig ventricular myocytes. Therefore, hydrocortisone can acutely affect cardiac excitation-contraction coupling, including ion channel activity, APD, ECG, Ca2+ transients, and contraction, possibly via biochemical changes in PKC.


Subject(s)
Action Potentials/drug effects , Calcium/metabolism , Electrocardiography , Heart/physiology , Hydrocortisone/pharmacology , Intracellular Space/metabolism , Myocardial Contraction/drug effects , Protein Kinase C/metabolism , Animals , Diastole/drug effects , Ether-A-Go-Go Potassium Channels/metabolism , Guinea Pigs , Heart/diagnostic imaging , Heart/drug effects , Heart Ventricles/cytology , Ion Channel Gating/drug effects , Kv1.5 Potassium Channel/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oocytes/drug effects , Oocytes/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Staurosporine/pharmacology , Time Factors , Xenopus laevis
12.
J Cell Physiol ; 234(5): 6854-6864, 2019 05.
Article in English | MEDLINE | ID: mdl-30387132

ABSTRACT

Laurus nobilis Linn. (Lauraceae), commonly known as Bay, has been used as a traditional medicine in the Mediterranean and Europe to treat diverse immunological disorders. Although the effects of L. nobilis on immunosuppression have been reported, the detailed underlying mechanism remains unclear. In this study, to elucidate the anti-inflammatory mechanism of L. nobilis, we examined the effect of L. nobilis leaf extract on inflammasome activation in mouse bone marrow-derived macrophages. L. nobilis leaf extract inhibited NOD-like receptor pyrin domain-containing 3 (NLRP3) inflammasome activation, which was associated with caspase-1 activation, interleukin-1ß secretion, and apoptosis-associated speck-like protein containing a CARD (ASC) pyroptosome complex formation. We also observed that 1,8-cineole, the major component of L. nobilis extract, consistently suppressed NLRP3 inflammasome activation. Furthermore, L. nobilis leaf extract attenuated the in vivo expression of proinflammatory cytokines in an acute lung injury mouse model. Our results provide the first evidence that L. nobilis leaf extract modulates inflammatory signaling by suppressing inflammasome activation.


Subject(s)
Inflammasomes/drug effects , Inflammation/drug therapy , Lauraceae/chemistry , Laurus/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plant Extracts/pharmacology , Plant Leaves/chemistry , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Caspase 1/metabolism , Cell Line , Cytokines/metabolism , HEK293 Cells , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
13.
Biochem Pharmacol ; 143: 107-117, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28712932

ABSTRACT

Pyrrolidine dithiocarbamate (PDTC) is a thiol compound that elicits anti-inflammatory effects by inhibiting NF-κB signaling. In this study, we report that regulator of calcineurin activity 1 (RCAN1) expression is induced by PDTC treatment and that increased RCAN1 expression is dependent on the generation of reactive oxygen species (ROS) and activation of p38 MAPK and JNK signaling. We also report that the ability of PDTC to induce RCAN1 is mediated by activator protein-1 (AP-1)-dependent gene transcription, and identified a functional AP-1 binding site in the RCAN1 promoter by producing mutations and conducting chromatin immunoprecipitation (ChIP) analyses. Moreover, we show that the PDTC-mediated inhibitory effect on NF-κB signaling is significantly perturbed by knocking out RCAN1. Our data provide the first evidence that PDTC prevents in vivo expression of pro-inflammatory cytokines by inducing RCAN1 expression.


Subject(s)
Antioxidants/pharmacology , Cytokines/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Muscle Proteins/biosynthesis , NF-kappa B/metabolism , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Calcium-Binding Proteins , Chromatin Immunoprecipitation , Cytokines/blood , Cytokines/immunology , DNA-Binding Proteins , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Muscle Proteins/genetics , NF-kappa B/immunology , Signal Transduction , Transcription, Genetic
14.
J Biol Chem ; 291(19): 10277-92, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26912655

ABSTRACT

Sirtuin 3 (SIRT3) is an NAD(+)-dependent protein deacetylase. Recent studies have shown that SIRT3 expression is decreased in nonalcoholic fatty liver disease (NAFLD). Moreover, SIRT3 is a key regulator of succinate dehydrogenase (SDH), which catalyzes the oxidation of succinate to fumarate. Increased succinate concentrations and the specific G protein-coupled receptor 91 (GPR91) are involved in the activation of hepatic stellate cells (HSCs). In this study, we aimed to establish whether SIRT3 regulated the SDH activity, succinate, and GPR91 expression in HSCs and an animal model of NAFLD. Our goal was also to determine whether succinate released from hepatocytes regulated HSC activation. Inhibiting SIRT3 using SIRT3 siRNA exacerbated HSC activation via the SDH-succinate-GPR91 pathway, and SIRT3 overexpression or honokiol treatment attenuated HSC activation in vitro In isolated liver and HSCs from methionine- and choline-deficient (MCD) diet-induced NAFLD, the expression of SIRT3 and SDH activity was decreased, and the succinate concentrations and GPR91 expression were increased. Moreover, we found that GPR91 knockdown or resveratrol treatment improved the steatosis in MCD diet-fed mice. This investigation revealed a novel mechanism of the SIRT3-SDH-GPR91 cascade in MCD diet-induced HSC activation in NAFLD. These findings highlight the biological significance of novel strategies aimed at targeting SIRT3 and GPR91 in HSCs with the goal of improving NAFLD treatment.


Subject(s)
Actins/metabolism , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Sirtuin 3/physiology , Succinate Dehydrogenase/metabolism , Actins/genetics , Animals , Blotting, Western , Cells, Cultured , Choline/metabolism , Choline Deficiency , Diet , Fluorescent Antibody Technique , Hepatic Stellate Cells/cytology , Immunoenzyme Techniques , Male , Methionine/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Succinate Dehydrogenase/genetics
15.
J Biol Chem ; 290(34): 21019-21031, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26157140

ABSTRACT

Pituitary adenylate cyclase-activating peptide (PACAP) is a neurotrophic peptide involved in a wide range of nervous functions, including development, differentiation, and survival, and various aspects of learning and memory. Here we report that PACAP induces the expression of regulator of calcineurin 1 (RCAN1, also known as DSCR1), which is abnormally expressed in the brains of Down syndrome patients. Increased RCAN1 expression is accompanied by activation of the PKA-cAMP response element-binding protein pathways. EMSA and ChIP analyses demonstrate the presence of a functional cAMP response element in the RCAN1 promoter. Moreover, we show that PACAP-dependent neuronal differentiation is significantly disturbed by improper RCAN1 expression. Our data provide the first evidence of RCAN1, a Down syndrome-related gene, as a novel target for control of the neurotrophic function of PACAP.


Subject(s)
Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Base Sequence , Calcium-Binding Proteins , Cell Differentiation , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Genes, Reporter , Humans , Intracellular Signaling Peptides and Proteins/genetics , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Muscle Proteins/genetics , Neurons/cytology , PC12 Cells , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Primary Cell Culture , Protein Binding , Rats , Response Elements , Signal Transduction
16.
Biochem Biophys Res Commun ; 459(4): 604-9, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25753203

ABSTRACT

Calcineurin is a calcium/calmodulin-dependent phosphatase that has been implicated in T cell activation through the induction of nuclear factors of activated T cells (NFAT). We have previously suggested that endogenous regulator of calcineurin (RCAN1, also known as DSCR1) is targeted by protein kinase A (PKA) for the control of calcineurin activity. In the present study, we characterized the PKA-mediated phosphorylation site in RCAN1 by mass spectrometric analysis and revealed that PKA directly phosphorylated RCAN1 at the Ser 93. PKA-induced phosphorylation and the increase in the half-life of the RCAN1 protein were prevented by the substitution of Ser 93 with Ala (S93A). Furthermore, the PKA-mediated phosphorylation of RCAN1 at Ser 93 potentiated the inhibition of calcineurin-dependent pro-inflammatory cytokine gene expression by RCAN1. Our results suggest the presence of a novel phosphorylation site in RCAN1 and that its phosphorylation influences calcineurin-dependent inflammatory target gene expression.


Subject(s)
Calcineurin/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Amino Acid Sequence , DNA-Binding Proteins , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Molecular Sequence Data , Muscle Proteins/chemistry , Phosphorylation
17.
BMB Rep ; 47(7): 369-75, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24856828

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic bioactive peptide that was first isolated from an ovine hypothalamus in 1989. PACAP belongs to the secretin/glucagon/vasoactive intestinal polypeptide (VIP) superfamily. PACAP is widely distributed in the central and peripheral nervous systems and acts as a neurotransmitter, neuromodulator, and neurotrophic factor via three major receptors (PAC1, VPAC1, and VPAC2). Recent studies have shown a neuroprotective role of PACAP using in vitro and in vivo models. In this review, we briefly summarize the current findings on the neurotrophic and neuroprotective effects of PACAP in different brain injury models, such as cerebral ischemia, Parkinson's disease (PD), and Alzheimer's disease (AD). This review will provide information for the future development of therapeutic strategies in treatment of these neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases/pathology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Humans , Neurodegenerative Diseases/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Reactive Oxygen Species/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Signal Transduction
18.
Phytother Res ; 27(4): 493-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22627996

ABSTRACT

Xanthorrhizol, a natural sesquiterpenoid compound isolated from Curcuma xanthorrhiza Roxb, has been known to inhibit the growth of human colon, breast, liver and cervical cancer cells. In this study, xanthorrhizol decreased cell viability, induced apoptosis and decreased the level of full-length PARP in SCC-15 oral squamous cell carcinoma (OSCC) cells. A decrease in cell viability and PARP degradation was not prevented by treatment with the caspase inhibitor Z-VAD-fmk in xanthorrhizol-treated cells. Xanthorrhizol treatment elevated intracellular Ca(2+) and ROS levels in SCC-15 cells. Treatment with a Ca(2+) chelator, EGTA/AM, did not affect xanthorrhizol- induced cytotoxicity, but cell viability was partly recovered by treatment with endogenous antioxidant, GSH, or hydroxy radical trapper, MCI-186. Furthermore, the viability of xanthorrhizol-treated SCC-15 cells was significantly restored by treatment with SB203580 and/or SP600125 but not significantly by PD98059 treatment. Xanthorrhizol-induced activation of p38 MAPK and JNK was blocked by MCI-186. Finally, xanthorrhizol suppressed the number of tumors in buccal pouches and increased the survival rate in hamsters treated with 7,12-dimethylbenz[a]anthracene. In conclusion, xanthorrhizol may induce caspase-independent apoptosis through ROS-mediated p38 MAPK and JNK activation in SCC-15 OSCC cells and prevent chemical-induced oral carcinogenesis. Therefore, xanthorrhizol seems to be a promising chemopreventive agent.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , MAP Kinase Signaling System/drug effects , Mouth Neoplasms/pathology , Phenols/pharmacology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Squamous Cell/metabolism , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Cell Survival , Cricetinae , Humans , Male , Mouth Neoplasms/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Reactive Oxygen Species/metabolism
19.
J Cell Biochem ; 114(5): 1115-23, 2013 May.
Article in English | MEDLINE | ID: mdl-23150431

ABSTRACT

Regulator of calcineurin 1 (RCAN1) is located on the Down syndrome critical region (DSCR) locus in human chromosome 21. In this study, we investigated the functional role of RCAN1 in the reactive oxygen species (ROS)-mediated neuronal death signaling. We found that RCAN1 was able to protect the cells from H(2)O(2) -induced cytotoxicity. The expression of RCAN1 caused an inhibition of the H(2)O(2) -induced activation of mitogen-activated protein kinases (MAPKs) and AP-1. In contrast, RCAN1 significantly enhanced the activity of cAMP response element-binding protein (CREB). Furthermore, RCAN1 induced the expression of the CREB target gene, Bcl-2. Consistently, knockdown of endogenous RCAN1 using shRNA down regulated the phosphorylation of CREB and the expression of Bcl-2, which protects the cells from H(2)O(2) -induced cytotoxicity. Our data provide a new mechanism for the cytoprotective function of RCAN1 in response to oxidant-induced apoptosis.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cytoprotection/drug effects , Hydrogen Peroxide/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Cell Death/drug effects , Enzyme Activation/drug effects , Gene Knockdown Techniques , Humans , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Neurons/enzymology , PC12 Cells , Rats , Transcription Factor AP-1/metabolism
20.
Free Radic Res ; 47(2): 95-103, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23153365

ABSTRACT

Staurosporine, a non-specific protein kinase inhibitor, has been shown to induce neurite outgrowth in PC12 cells, but the mechanism by which staurosporine induces neurite outgrowth is still obscure. In the present study, we investigated whether the activation of Rac1 was responsible for the neurite outgrowth triggered by staurosporine. Staurosporine caused rapid neurite outgrowth independent of the ERK signaling pathways. In contrast, neurite outgrowth in response to staurosporine was accompanied by activation of Rac1, and the Rac1 inhibitor NSC23766 attenuated the staurosporine-induced neurite outgrowth in a concentration-dependent manner. In addition, suppression of Rac1 activity by expression of the dominant negative mutant Rac1N17 also blocked the staurosporine-induced morphological differentiation of PC12 cells. Staurosporine caused an activation of NADPH oxidase and increased the production of reactive oxygen species (ROS), which was prevented by NSC23766 and diphenyleneiodonium (DPI), an NADPH oxidase inhibitor. Staurosporine-induced neurite outgrowth was attenuated by pretreatment with DPI and exogenous addition of sublethal concentration of H2O2 accelerated neurite outgrowth triggered by staurosporine. These results indicate that activation of Rac1, which leads to ROS generation, is required for neurite outgrowth induced by staurosporine in PC12 cells.


Subject(s)
Neurites/physiology , Staurosporine/pharmacology , rac1 GTP-Binding Protein/metabolism , Aminoquinolines/pharmacology , Animals , Cell Line , Enzyme Activation , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Peroxide/pharmacology , Mutation , NADPH Oxidases/metabolism , Neurites/drug effects , Onium Compounds/pharmacology , Oxidation-Reduction , PC12 Cells , Protein Kinase Inhibitors/metabolism , Pyrimidines/pharmacology , Rats , Reactive Oxygen Species/metabolism , Signal Transduction , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...