Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 270
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Cell Physiol ; 239(6): e31245, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38497504

ABSTRACT

Parathyroid hormone (PTH) serves dual roles in bone metabolism, exhibiting both anabolic and catabolic effects. The anabolic properties of PTH have been utilized in the treatment of osteoporosis with proven efficacy in preventing fractures. Despite these benefits, PTH can be administered therapeutically for up to 2 years, and its use in patients with underlying malignancies remains a subject of ongoing debate. These considerations underscore the need for a more comprehensive understanding of the underlying mechanisms. p21-activated kinase 4 (PAK4) is involved in bone resorption and cancer-associated osteolysis; however, its role in osteoblast function and PTH action remains unknown. Therefore, in this study, we aimed to clarify the role of PAK4 in osteoblast function and its effects on PTH-induced anabolic activity. PAK4 enhanced MC3T3-E1 osteoblast viability and proliferation and upregulated cyclin D1 expression. PAK4 also augmented osteoblast differentiation, as indicated by increased mineralization found by alkaline phosphatase and Alizarin Red staining. Treatment with PTH (1-34), an active PTH fragment, stimulated PAK4 expression and phosphorylation in a protein kinase A-dependent manner. In addition, bone morphogenetic protein-2 (which is known to promote bone formation) increased phosphorylated PAK4 (p-PAK4) and PAK4 levels. PAK4 regulated the expression of both phosphorylated and total ß-catenin, which are critical for osteoblast proliferation and differentiation. Moreover, p-PAK4 directly interacted with ß-catenin, and disruption of ß-catenin's binding to T-cell factor impaired PAK4- and PTH-induced osteoblast differentiation. Our findings elucidate the effect of PAK4 on enhancing bone formation in osteoblasts and its pivotal role in the anabolic activity of PTH mediated through its interaction with ß-catenin. These insights improve the understanding of the mechanisms underlying PTH activity and should inform the development of more effective and safer osteoporosis treatments.


Subject(s)
Cell Differentiation , Cell Proliferation , Osteoblasts , Parathyroid Hormone , beta Catenin , p21-Activated Kinases , Animals , Humans , Mice , beta Catenin/metabolism , beta Catenin/genetics , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin D1/genetics , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Parathyroid Hormone/pharmacology , Parathyroid Hormone/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Cells, Cultured
2.
Hepatology ; 76(2): 345-356, 2022 08.
Article in English | MEDLINE | ID: mdl-35108418

ABSTRACT

BACKGROUND AND AIMS: p21-activated kinase 4 (PAK4), an oncogenic protein, has emerged as a promising target for anticancer drug development. Its role in oxidative stress conditions, however, remains elusive. We investigated the effects of PAK4 signaling on hepatic ischemia/reperfusion (I/R) injury. APPROACH AND RESULTS: Hepatocyte- and myeloid-specific Pak4 knockout (KO) mice and their littermate controls were subjected to a partial hepatic I/R (HIR) injury. We manipulated the catalytic activity of PAK4, either through genetic engineering (gene knockout, overexpression of wild-type [WT] or dominant-negative kinase) or pharmacological inhibitor, coupled with a readout of nuclear factor erythroid 2-related factor 2 (Nrf2) activity, to test the potential function of PAK4 on HIR injury. PAK4 expression was markedly up-regulated in liver during HIR injury in mice and humans. Deletion of PAK4 in hepatocytes, but not in myeloid cells, ameliorated liver damages, as demonstrated in the decrease in hepatocellular necrosis and inflammatory responses. Conversely, the forced expression of WT PAK4 aggravated the pathological changes. PAK4 directly phosphorylated Nrf2 at T369, and it led to its nuclear export and proteasomal degradation, all of which impaired antioxidant responses in hepatocytes. Nrf2 silencing in liver abolished the protective effects of PAK4 deficiency. A PAK4 inhibitor protected mice from HIR injury. CONCLUSIONS: PAK4 phosphorylates Nrf2 and suppresses its transcriptional activity. Genetic or pharmacological suppression of PAK4 alleviates HIR injury. Thus, PAK4 inhibition may represent a promising intervention against I/R-induced liver injury.


Subject(s)
Liver Diseases , Reperfusion Injury , p21-Activated Kinases , Animals , Apoptosis , Humans , Ischemia/metabolism , Ischemia/pathology , Liver/pathology , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Diseases/prevention & control , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Phosphorylation , Reperfusion Injury/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
3.
Eur J Nutr ; 62(3): 1415-1425, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36629892

ABSTRACT

PURPOSE: In our previous study, we showed that Lycium chinense Miller fruit extract (LFE) exerted hepatoprotective effects in mice. In the current study, we examined the effect of LFE on liver enzyme levels in subjects with mild hepatic dysfunction. METHODS: A total of 90 subjects, aged 19 to 70 years old, with abnormal alanine aminotransferase (ALT) levels, were randomly placed into either an LFE (n = 45) treatment group or a placebo group (n = 45). During the 12-week clinical trial, subjects in each group received either LFE or placebo capsules, and were instructed to take four tablets per day (1760 mg/day). The primary outcome of the study was the changes of ALT and γ-glutamyltransferase (GGT) levels in each subject. The safety of LFE supplementation was assessed and adverse events were recorded. RESULTS: LFE supplementation for 12 weeks resulted in a significant reduction of ALT (P = 0.0498) and GGT (P = 0.0368) levels in comparison to the placebo. No clinically significant changes were observed in any safety parameters. CONCLUSION: These results suggest that LFE can be applied to subjects with mild hepatic dysfunction with no possible side effects. TRIAL REGISTRATION: This study was registered at the Clinical Research Information Service (CRIS) as no. KCT0003985.


Subject(s)
Liver Diseases , Lycium , Double-Blind Method , Fruit , Liver Diseases/drug therapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Humans , Adult , Middle Aged , Aged
4.
J Allergy Clin Immunol ; 149(1): 156-167.e7, 2022 01.
Article in English | MEDLINE | ID: mdl-34051221

ABSTRACT

BACKGROUND: Binding IgE to a cognate allergen causes aggregation of Fcε receptor I (FcεRI) in mast cells, resulting in activation of receptor-associated Src family tyrosine kinases, including Lyn and Syk. Protein tyrosine phosphatase, receptor type C (PTPRC), also known as CD45, has emerged as a positive regulator of FcεRI signaling by dephosphorylation of the inhibitory tyrosine of Lyn. OBJECTIVE: Sirtuin 6 (Sirt6), a NAD+-dependent deacetylase, exhibits an anti-inflammatory property. It remains to be determined, however, whether Sirt6 attenuates mast cell-associated diseases, including anaphylaxis. METHODS: FcεRI signaling and mast cell degranulation were measured after IgE cross-linking in murine bone marrow-derived mast cells (BMMCs) and human cord blood-derived mast cells. To investigate the function of Sirt6 in mast cell activation in vivo, we used mast cell-dependent animal models of passive systemic anaphylaxis (PSA) and passive cutaneous anaphylaxis (PCA). RESULTS: Sirt6-deficient BMMCs augmented IgE-FcεRI-mediated signaling and degranulation compared to wild-type BMMCs. Reconstitution of mast cell-deficient KitW-sh/W-sh mice with BMMCs received from Sirt6 knockout mice developed more severe PSA and PCA compared to mice engrafted with wild-type BMMCs. Similarly, genetic overexpression or pharmacologic activation of Sirt6 suppressed mast cell degranulation and blunted responses to PCA. Mechanistically, Sirt6 deficiency increased PTPRC transcription via acetylating histone H3, leading to enhanced aggregation of FcεRI in BMMCs. Finally, we recapitulated the Sirt6 regulation of PTPRC and FcεRI signaling in human mast cells. CONCLUSIONS: Sirt6 acts as a negative regulator of FcεRI signaling cascade in mast cells by suppressing PTPRC transcription. Activation of Sirt6 may therefore represent a promising and novel therapeutic strategy for anaphylaxis.


Subject(s)
Anaphylaxis/immunology , Mast Cells/immunology , Receptors, IgE/immunology , Sirtuins/immunology , Animals , Bone Marrow Cells/cytology , Fetal Blood/cytology , Humans , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Sirtuins/genetics
5.
J Enzyme Inhib Med Chem ; 37(1): 2133-2146, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35920284

ABSTRACT

p21-Activated kinase 4 (PAK4), one of the serine/threonine kinases activated by Rho-family GTPases, has been widely studied as an oncogenic protein that is overexpressed in many types of cancers. In our recent study, PAK4 upregulation was observed in mice exhibiting hepatic ischaemia-reperfusion (I/R) and in liver transplantation patients. Liver I/R injury was also attenuated in Pak4 KO mice. Herein, we report a novel series of pyrazolo[3,4-d]pyrimidine derivatives of type I ½ PAK4 inhibitors. The most potent compound SPA7012 was evaluated to determine the pharmacological potential of PAK4 inhibitor in I/R injury in mice. Mice with I/R injury showed typical patterns of liver damage, as demonstrated by increases in serum levels of aminotransferases and proinflammatory cytokines, hepatocellular necrosis and apoptosis, and inflammatory cell infiltration, relative to sham mice. Conversely, intraperitoneal administration of SPA7012 dramatically attenuated biochemical and histopathologic changes. Mechanistically, stabilisation of nuclear factor-erythroid 2-related factor 2 (Nrf2), a master regulator of anti-oxidative response, was observed following SPA7012 treatment. SPA7012 treatment in primary hepatocytes also attenuated hypoxia-reoxygenation-induced apoptotic cell death and inflammation. Together, these results provide experimental evidence supporting the use of PAK4 inhibitors for alleviation of I/R-induced liver damage.


Subject(s)
Reperfusion Injury , p21-Activated Kinases , Animals , Apoptosis , Liver/metabolism , Mice , Protein Serine-Threonine Kinases , Pyrimidines/pharmacology , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , p21-Activated Kinases/metabolism
6.
Int J Mol Sci ; 23(21)2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36362370

ABSTRACT

Sirtuin 1 (SIRT1) regulates cellular processes by deacetylating non-histone targets, including transcription factors and intracellular signalling mediators; thus, its abnormal activation is closely linked to the pathophysiology of several diseases. However, its function in Toxoplasma gondii infection is unclear. We found that SIRT1 contributes to autophagy activation via the AMP-activated protein kinase (AMPK) and PI3K/AKT signalling pathways, promoting anti-Toxoplasma responses. Myeloid-specific Sirt1-/- mice exhibited an increased cyst burden in brain tissue compared to wild-type mice following infection with the avirulent ME49 strain. Consistently, the intracellular survival of T. gondii was markedly increased in Sirt1-deficient bone-marrow-derived macrophages (BMDMs). In contrast, the activation of SIRT1 by resveratrol resulted in not only the induction of autophagy but also a significantly increased anti-Toxoplasma effect. Notably, SIRT1 regulates the FoxO-autophagy axis in several human diseases. Importantly, the T. gondii-induced phosphorylation, acetylation, and cytosolic translocation of FoxO1 was enhanced in Sirt1-deficient BMDMs and the pharmacological inhibition of PI3K/AKT signalling reduced the cytosolic translocation of FoxO1 in BMDMs infected with T. gondii. Further, the CaMKK2-dependent AMPK signalling pathway is responsible for the effect of SIRT1 on the FoxO3a-autophagy axis and for its anti-Toxoplasma activity. Collectively, our findings reveal a previously unappreciated role for SIRT1 in Toxoplasma infection.


Subject(s)
Toxoplasma , Animals , Humans , Mice , AMP-Activated Protein Kinases/metabolism , Autophagy , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 1/genetics , Toxoplasma/metabolism , Forkhead Transcription Factors/metabolism
7.
Anal Chem ; 93(2): 801-811, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33284604

ABSTRACT

An easily accessible colorimetric and fluorescence probe 4-((3-chloro-1,4-dioxo-1,4-dihydronaphthalen-2-yl)amino)benzenesulfonamide (4CBS) was successfully developed for the selective and sensitive detection of Sn2+ in an aqueous solution. The sensing mechanism involves reduction of -C═O into -C-OH groups in 4CBS upon the addition of Sn2+, which initiates the fluorescence turn-on mode. A better linear relationship was achieved between fluorescence intensity and Sn2+ concentration in the range of 0-62.5 µM, with a detection limit (LOD) of 0.115 µM. The binding mechanism of 4CBS for Sn2+ was confirmed by Fourier transform infrared analysis, NMR titrations, and mass (electrospray ionization) spectral analysis. Likewise, the proposed sensing mechanism was supported by quantum chemical calculations. Moreover, bioimaging studies demonstrated that the chemosensing probe 4CBS is an effective fluorescent marker for the detection of Sn2+ in living cells and zebrafish. Significantly, 4CBS was able to discriminate between Sn2+ in human cancer cells and Sn2+ in normal live cells.


Subject(s)
Colorimetry/methods , Sulfonamides/chemical synthesis , Tin/chemistry , Animals , Cell Line , Electrochemical Techniques , Humans , Larva , Mice , Models, Molecular , Molecular Structure , Optical Imaging , Sensitivity and Specificity , Sulfonamides/chemistry , Water , Zebrafish
8.
Cell Biol Toxicol ; 37(2): 193-207, 2021 04.
Article in English | MEDLINE | ID: mdl-32394328

ABSTRACT

Sirtuin 6 (Sirt6) is important for maintaining kidney homeostasis and function. Cd exposure increases the risk of developing kidney diseases. However, the role of Sirt6 in kidney disease mechanisms is unclear. Here, we evaluated the role of Sirt6 in Cd-induced kidney toxicity. After Cd exposure, p62/sequestosome-1 (SQSTM1), an autophagy substrate, accumulated in mouse kidney mesangial cells in monomeric and polyubiquitinated (polyUb) forms. Sirt6 accumulated in response to Cd treatment at concentrations below the half-maximal inhibitory concentration and decreased after 12 h of treatment. Sirt6 and p62 co-localized in the nucleus and redistributed to the cytosol after Cd treatment. Sirt6 was mainly present in nuclei-rich membrane fractions. Sirt6 interacted with p62. Ub, and microtubule-associated protein light chain 3 (LC3). Knockdown of p62 promoted Sirt6 nuclear accumulation and inhibited apoptosis. Sirt6 overexpression altered levels of polyUb-p62 and apoptosis. At earlier times during Cd treatment, polyubiquitination of p62 and apoptosis were reduced. Cytoplasmic translocation of Sirt6 occurred later, with increased polyubiquitination of p62 and apoptosis. Bafilomycin 1 (BaF1) treatment promoted cytosolic Sirt6 accumulation, increasing cell death. Silencing autophagy related 5 (Atg5) increased nuclear Sirt6 levels, reduced polyUb-p62, and inhibited cell death, indicating that autophagy was necessary for Sirt6 redistribution. Cd resistance was associated with reduced polyUb-p62 and persistent Sirt6 expression. Cd treatment in mice for 4 weeks promoted p62, Sirt6, and LC3-II accumulation, inducing apoptosis in kidney tissues. Overall, our findings show that polyUb-p62 targeted Sirt6 to autophagosomes, playing a crucial role in Cd-induced cell death and kidney damage.


Subject(s)
Cadmium/toxicity , Cytoplasm/metabolism , Kidney/pathology , Polyubiquitin/metabolism , Sequestosome-1 Protein/metabolism , Sirtuins/metabolism , Toxicity Tests , Ubiquitination , Animals , Apoptosis/drug effects , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Cell Line , Kidney/drug effects , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice, Inbred C57BL , Protein Binding/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Subcellular Fractions/metabolism , Ubiquitination/drug effects
9.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768915

ABSTRACT

Ursolic acid (UA), a pentacyclic triterpenoid extracted from various plants, inhibits cell growth, metastasis, and tumorigenesis in various cancers. Chemotherapy resistance and the side effects of paclitaxel (PTX), a traditional chemotherapy reagent, have limited the curative effect of PTX in esophageal cancer. In this study, we investigate whether UA promotes the anti-tumor effect of PTX and explore the underlying mechanism of their combined effect in esophageal squamous cell carcinoma (ESCC). Combination treatment with UA and PTX inhibited cell proliferation and cell growth more effectively than either treatment alone by inducing more significant apoptosis, as indicated by increased sub-G1 phase distribution and protein levels of cleaved-PARP and cleaved caspase-9. Similar to the cell growth suppressive effect, the combination of UA and PTX significantly inhibited cell migration by targeting uPA, MMP-9, and E-cadherin in ESCC cells. In addition, combination treatment with UA and PTX significantly activated p-GSK-3ß and suppressed the activation of Akt and FOXM1 in ESCC cells. Those effects were enhanced by the Akt inhibitor LY2940002 and inverted by the Akt agonist SC79. In an in vivo evaluation of a murine xenograft model of esophageal cancer, combination treatment with UA and PTX suppressed tumor growth significantly better than UA or PTX treatment alone. Thus, UA effectively potentiates the anti-tumor efficacy of PTX by targeting the Akt/FOXM1 cascade since combination treatment shows significantly more anti-tumor potential than PTX alone both in vitro and in vivo. Combination treatment with UA and PTX could be a new strategy for curing esophageal cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Paclitaxel/pharmacology , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cadherins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/immunology , Forkhead Box Protein M1/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Neoplasm Invasiveness/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Ursolic Acid
10.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073071

ABSTRACT

Gastric cancer is the fifth most common cancer and the third leading cause of cancer-related deaths worldwide. Histone deacetylase (HDAC) inhibitors are a new class of cytostatic agents available for the treatment of various cancers and diseases. Although numerous clinical and pre-clinical trials on the anticancer effects of panobinostat have been conducted, only a few reports have investigated its efficacy in gastric cancer. The present study aimed to investigate the effects of panobinostat in gastric cancer cells. Panobinostat significantly inhibited the cell viability and proliferation of the gastric cancer cell lines SNU484 and SNU638 in a dose-dependent manner; it reduced the colony-forming ability of these cells. Moreover, it induced apoptosis as indicated by increased protein levels of cleaved poly ADP-ribose polymerase and cleaved caspase-3. Panobinostat induced the G2/M cell cycle arrest in SNU484 and SNU638 cells and subsequently decreased the G2/M phase regulatory-associated protein expression of p-Wee1, Myt1, and Cdc2. Furthermore, panobinostat significantly inhibited the metastasis of SNU484 and SNU638 cells by regulating the expression of MMP-9 and E-cadherin. Further, it decreased the protein levels of p-Akt and forkhead box protein M1 (FOXM1). These effects were reversed by the Akt agonist SC79 and were accelerated by the Akt inhibitor LY2940002. Moreover, tumor growth in xenograft animal experiments was suppressed by panobinostat. These results indicated that panobinostat inhibits the proliferation, metastasis, and cell cycle progression of gastric cancer cells by promoting apoptosis and inactivating Akt/FOXM1 signaling. Cumulatively, our present study suggests that panobinostat is a potential drug for the treatment of gastric cancer.


Subject(s)
Antineoplastic Agents , Apoptosis/drug effects , Cell Survival/drug effects , Panobinostat , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Forkhead Box Protein M1/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Mice , Neoplasm Metastasis/drug therapy , Panobinostat/pharmacology , Panobinostat/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism
11.
Biochem Biophys Res Commun ; 531(4): 508-514, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32807498

ABSTRACT

Osteoporosis is a degenerative disease characterized by reduced bone mass, in which deregulated bone remodeling by osteoclasts and osteoblasts is a main pathogenesis. Although recently tussilagone, a major active component of flower buds of Tussilago farfara, has been shown to inhibit osteoclastogenesis, its effect on estrogen deficiency-induced osteoporosis remains unknown. This study examined the effect of tussilagone on bone loss in ovariectomized mice and further explored its impact on osteoclast apoptosis and osteoblast formation in addition to osteoclastogenesis. Tussilagone suppression of osteoclastogenesis was confirmed in bone marrow derived macrophages, which was observed with the 1/10 concentration of that of the previous study. As demonstrated by ApoPercentage dye staining and Western blotting, tussilagone enhanced apoptosis in differentiated osteoclasts by increasing estrogen receptor α and Fas ligand expression. On the contrary, either osteoblast differentiation or mineralization was not affected by tussilagone. Lastly, administering tussilagone to mice for 6 weeks prevented trabecular microarchitecture impairment in ovariectomized mice compared to vehicle control groups. These findings suggest that tussilagone or Tussilago farfara prevents osteoporotic bone loss by suppressing osteoclast differentiation and inducing osteoclast apoptosis, and that it may therefore offer a possible remedy against resorptive bone diseases.


Subject(s)
Osteoclasts/drug effects , Osteoporosis/drug therapy , Sesquiterpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Disease Models, Animal , Estrogens/deficiency , Female , Macrophages/drug effects , Mice, Inbred C57BL , Osteoclasts/pathology , Osteogenesis/drug effects , Osteoporosis/metabolism , Osteoporosis/pathology , Ovariectomy , Sesquiterpenes/isolation & purification , Tussilago/chemistry
12.
Cell Biol Toxicol ; 36(6): 609, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32827127

ABSTRACT

In the original publication the grant number is incorrectly published.

13.
J Immunol ; 201(10): 2986-2997, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30341186

ABSTRACT

Connexin 43 (Cx43) deficiency was found to increase mortality in a mouse model of bacterial peritonitis, and Cx43 is upregulated in macrophages by LPS treatment. In this study, we characterized a novel signaling pathway for LPS-induced Cx43 expression in RAW264.7 cells and thioglycolate-elicited peritoneal macrophages (TGEMs). LPS alone or LPS-containing conditioned medium (CM) upregulated Cx43. Overexpression or silencing of Cx43 led to the enhancement or inhibition, respectively, of CM-induced TGEM migration. This response involved the inducible NO synthase (iNOS)/focal adhesion kinase (FAK)/Src pathways. Moreover, CM-induced migration was compromised in TGEMs from Cx43+/- mice compared with TGEMs from Cx43+/+ littermates. Cx43 was upregulated by a serum/glucocorticoid-regulated kinase 1 (SGK) activator and downregulated, along with inhibition of CM-induced TGEM migration, by knockdown of the SGK gene or blockade of the SGK pathway. LPS-induced SGK activation was abrogated by Torin2, whereas LPS-induced Cx43 was downregulated by both Torin2 and rapamycin. Analysis of the effects of FK506 and methylprednisolone, common immunosuppressive agents following organ transplantation, suggested a link between these immunosuppressive drugs and impaired macrophage migration via the Cx43/iNOS/Src/FAK pathway. In a model of Escherichia coli infectious peritonitis, GSK650349-, an SGK inhibitor, or Torin2-treated mice showed less accumulation of F4/80+CD11b+ macrophages in the peritoneal cavity, with a delay in the elimination of bacteria. Furthermore, following pretreatment with Gap19, a selective Cx43 hemichannel blocker, the survival of model mice was significantly reduced. Taken together, our study suggested that Cx43 in macrophages was associated with macrophage migration, an important immune process in host defense to infection.


Subject(s)
Cell Movement/immunology , Connexin 43/biosynthesis , Macrophages/immunology , Signal Transduction/immunology , Animals , Connexin 43/immunology , Focal Adhesion Kinase 1/immunology , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation/immunology , Immediate-Early Proteins/immunology , Immediate-Early Proteins/metabolism , Lipopolysaccharides/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , RAW 264.7 Cells , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , src-Family Kinases/immunology , src-Family Kinases/metabolism
14.
Int J Mol Sci ; 21(24)2020 Dec 10.
Article in English | MEDLINE | ID: mdl-33321911

ABSTRACT

Ursolic acid (UA) possesses various pharmacological activities, such as antitumorigenic and anti-inflammatory effects. In the present study, we investigated the mechanisms underlying the effects of UA against esophageal squamous cell carcinoma (ESCC) (TE-8 cells and TE-12 cells). The cell viability assay showed that UA decreased the viability of ESCC in a dose-dependent manner. In the soft agar colony formation assay, the colony numbers and size were reduced in a dose-dependent manner after UA treatment. UA caused the accumulation of vacuoles and LC3 puncta, a marker of autophagosome, in a dose-dependent manner. Autophagy induction was confirmed by measuring the expression levels of LC3 and p62 protein in ESCC cells. UA increased LC3-II protein levels and decreased p62 levels in ESCC cells. When autophagy was hampered using 3-methyladenine (3-MA), the effect of UA on cell viability was reversed. UA also significantly inhibited protein kinase B (Akt) activation and increased p-Akt expression in a dose-dependent manner in ESCC cells. Accumulated LC3 puncta by UA was reversed after wortmannin treatment. LC3-II protein levels were also decreased after treatment with Akt inhibitor and wortmannin. Moreover, UA treatment increased cellular reactive oxygen species (ROS) levels in ESCC in a time- and dose-dependent manner. Diphenyleneiodonium (an ROS production inhibitor) blocked the ROS and UA induced accumulation of LC3-II levels in ESCC cells, suggesting that UA-induced cell death and autophagy are mediated by ROS. Therefore, our data indicate that UA inhibits the growth of ESCC cells by inducing ROS-dependent autophagy.


Subject(s)
Antineoplastic Agents/toxicity , Autophagy , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Triterpenes/toxicity , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Microtubule-Associated Proteins/metabolism , Ursolic Acid
15.
Int J Mol Sci ; 20(19)2019 Sep 23.
Article in English | MEDLINE | ID: mdl-31547587

ABSTRACT

The Hippo pathway is often dysregulated in many carcinomas, which results in various stages of tumor progression. Ursolic acid (UA), a natural compound that exists in many herbal plants, is known to obstruct cancer progression and exerts anti-carcinogenic effect on a number of human cancers. In this study, we aimed to examine the biological mechanisms of action of UA through the Hippo pathway in gastric cancer cells. MTT assay showed a decreased viability of gastric cancer cells after treatment with UA. Following treatment with UA, colony numbers and the sizes of gastric cancer cells were significantly diminished and apoptosis was observed in SNU484 and SNU638 cells. The invasion and migration rates of gastric cancer cells were suppressed by UA in a dose-dependent manner. To further determine the gene expression patterns that are related to the effects of UA, a microarray analysis was performed. Gene ontology analysis revealed that several genes, such as the Hippo pathway upstream target gene, ras association domain family (RASSF1), and its downstream target genes (MST1, MST2, and LATS1) were significantly upregulated by UA, while the expression of YAP1 gene, together with oncogenes (FOXM1, KRAS, and BATF), were significantly decreased. Similar to the gene expression profiling results, the protein levels of RASSF1, MST1, MST2, LATS1, and p-YAP were increased, whereas those of CTGF were decreased by UA in gastric cancer cells. The p-YAP expression induced in gastric cancer cells by UA was reversed with RASSF1 silencing. In addition, the protein levels in the Hippo pathway were increased in the UA-treated xenograft tumor tissues as compared with that in the control tumor tissues; thus, UA significantly inhibited the tumorigenesis of gastric cancer in vivo in xenograft animals. Collectively, UA diminishes the proliferation and metastasis of gastric cancer via the regulation of Hippo pathway through Rassf1, which suggests that UA can be used as a potential chemopreventive and therapeutic agent for gastric cancer.


Subject(s)
Carcinogenesis/drug effects , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Triterpenes/pharmacology , Tumor Suppressor Proteins/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Hippo Signaling Pathway , Humans , Mice , Protein Serine-Threonine Kinases/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays , Ursolic Acid
16.
Korean J Physiol Pharmacol ; 23(1): 47-54, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30627009

ABSTRACT

Estrogen withdrawal in post-menopausal women leads to overactivation of osteoclasts, which contributes to the development of osteoporosis. Inflammatory cytokines are known as one of mechanisms of osteoclast activation after estrogen deficiency. SPA0355 is a thiourea derivative that has been investigated for its antioxidant and anti-inflammatory activities. However, its efficacy in bone resorption has not been previously investigated. The aim of this study was to investigate the impact of SPA0355 on the development of osteoporosis and to explore its mode of action. In vitro experiments showed that SPA0355 inhibited receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis in primary bone marrow-derived macrophages. This effect appears to be independent of estrogen receptor activation as ICI 180,782 failed to abrogate its effects on osteoclasts. Further signaling studies revealed that SPA0355 suppressed activation of the MAPKs, Akt, and NF-κB pathways. SPA0355 also increased osteoblastic differentiation, as evidenced by its effects on alkaline phosphatase activity and mineralization nodule formation. Intraperitoneal administration of SPA0355 to ovariectomized mice prevented bone loss, as verified by three-dimensional images and bone morphometric parameters derived from µCT analysis. Noticeably, SPA0355 did not show hepatotoxicity and nephrotoxicity and also had little effect on hematological parameters. Taken together, the results indicate that SPA0355 may protect against bone loss in ovariectomized mice by stimulation of osteoblast differentiation and by inhibition of osteoclast resorption. Therefore, SPA0355 is a safe and potential candidate for management of postmenopausal osteoporosis.

17.
Hepatology ; 65(1): 225-236, 2017 01.
Article in English | MEDLINE | ID: mdl-27532371

ABSTRACT

Sirtuin 2 (Sirt2) is known to negatively regulate anoxia-reoxygenation injury in myoblasts. Because protein levels of Sirt2 are increased in ischemia-reperfusion (I/R)-injured liver tissues, we examined whether Sirt2 is protective or detrimental against hepatic I/R injury. We overexpressed Sirt2 in the liver of C57BL/6 mice using a Sirt2 adenovirus. Wild-type and Sirt2 knockout mice were subjected to a partial (70%) hepatic ischemia for 45 minutes, followed by various periods of reperfusion. In another set of experiments, wild-type mice were pretreated intraperitoneally with AGK2, a Sirt2 inhibitor. Isolated hepatocytes and Kupffer cells from wild-type and Sirt2 knockout mice were subjected to hypoxia-reoxygenation injury to determine the in vitro effects of Sirt2. Mice subjected to I/R injury showed typical patterns of hepatocellular damage. Prior injection with Sirt2 adenovirus aggravated liver injury, as demonstrated by increases in serum aminotransferases, prothrombin time, proinflammatory cytokines, hepatocellular necrosis and apoptosis, and neutrophil infiltration relative to control virus-injected mice. Pretreatment with AGK2 resulted in significant improvements in serum aminotransferase levels and histopathologic findings. Similarly, experiments with Sirt2 knockout mice also revealed reduced hepatocellular injury. The molecular mechanism of Sirt2's involvement in this aggravation of hepatic I/R injury includes the deacetylation and inhibition of mitogen-activated protein kinase phosphatase-1 and consequent activation of mitogen-activated protein kinases. CONCLUSION: Sirt2 is an aggravating factor during hepatic I/R injury. (Hepatology 2017;65:225-236).


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Liver Diseases/enzymology , Liver Diseases/etiology , Liver/blood supply , Reperfusion Injury/complications , Sirtuin 2/physiology , Acetylation , Animals , Disease Progression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
FASEB J ; 31(9): 3999-4010, 2017 09.
Article in English | MEDLINE | ID: mdl-28536120

ABSTRACT

Sirtuin (Sirt)6 has been implicated in negative regulation of inflammation and lipid metabolism, although its function in the progression from simple steatosis to nonalcoholic steatohepatitis (NASH) remains to be defined. To explore the role of hepatocyte Sirt6 in NASH development, we generated hepatocyte-specific Sirt6-knockout (KO) mice that were fed a high-fat and high-fructose (HFHF) diet for 16 wk. HFHF-fed KO mice had increased hepatic steatosis and inflammation and aggravated glucose intolerance and insulin resistance compared with wild-type mice. HFHF-induced liver fibrosis and oxidative stress and related gene expression were significantly elevated in KO mice. In the livers of KO mice, nuclear factor erythroid 2-related factor 2 (Nrf2) was down-regulated; conversely, BTB domain and CNC homolog 1 (Bach1), a nuclear repressor of Nrf2, were up-regulated. We discovered that Sirt6, which interacts with Bach1 under basal condition, induces its detachment from the antioxidant response element (ARE) region of heme oxygenase 1 promoter. Furthermore, we found that Sirt6 promotes Nrf2 binding to ARE in response to oxidative stimuli, which leads to the expression of phase II/antioxidant enzymes. Finally, we showed that HFHF-induced steatosis, inflammation, and fibrosis were ameliorated by adenoviral Sirt6 overexpression. Sirt6 may be a useful therapeutic target for amelioration of NASH by curbing inflammation and oxidative stress.-Ka, S.-O, Bang, I. H., Bae, E. J., Park, B.-H. Hepatocyte-specific sirtuin 6 deletion predisposes to nonalcoholic steatohepatitis by up-regulation of Bach1, an Nrf2 repressor.


Subject(s)
NF-E2-Related Factor 2/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Sirtuins/metabolism , Animals , Basic-Leucine Zipper Transcription Factors , Diet, High-Fat/adverse effects , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/adverse effects , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hep G2 Cells , Hepatocytes , Histones/genetics , Histones/metabolism , Humans , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Promoter Regions, Genetic , Sirtuins/genetics , Up-Regulation
19.
Am J Pathol ; 186(12): 3297-3315, 2016 12.
Article in English | MEDLINE | ID: mdl-27746184

ABSTRACT

Recently, the roles of sirtuins (SIRTs) in tumorigenesis have been of interest to oncologists, and protein kinase CK2 α1 (CSNK2A1) has been shown to be involved in tumorigenesis by phosphorylating various proteins, including SIRT1. Therefore, we evaluated the roles of CSNK2A1, SIRT6, and phosphorylated SIRT6 and their relationships in breast carcinoma. Nuclear expression of CSNK2A1 and SIRT6 predicted shorter overall survival and relapse-free survival by multivariate analysis. Inhibition of CSNK2A1 decreased the proliferative and invasive activity of cancer cells. In addition, CSNK2A1 was bound to SIRT6 and phosphorylated SIRT6; evidence for this is provided from immunofluorescence staining, co-immunoprecipitation of CSNK2A1 and SIRT6, a glutathione S-transferase pull-down assay, an in vitro kinase assay, and transfection of mutant CSNK2A1. Knockdown of SIRT6 decreased the proliferation and invasiveness of cancer cells. Overexpression of SIRT6 increased proliferation, but mutation at the Ser338 phosphorylation site of SIRT6 inhibited the proliferation of MCF7 cells. Moreover, both knockdown of SIRT6 and a mutation at the phosphorylation site of SIRT6 decreased expression of matrix metallopeptidase 9, ß-catenin, cyclin D1, and NF-κB. Especially, SIRT6 expression was associated with the nuclear localization of ß-catenin. This study demonstrates that CSNK2A1 and SIRT6 are indicators of poor prognosis for breast carcinomas and that CSNK2A1-mediated phosphorylation of SIRT6 might be involved in the progression of breast carcinoma.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Sirtuins/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Proliferation , Cyclin D1/metabolism , Disease Progression , Gene Expression , Humans , Mutation , NF-kappa B/metabolism , Phosphorylation , Prognosis , Sirtuins/metabolism , beta Catenin/metabolism
20.
Int Psychogeriatr ; 29(2): 269-279, 2017 02.
Article in English | MEDLINE | ID: mdl-27760581

ABSTRACT

BACKGROUND: Existing literature suggests that mental health literacy is positively associated with mental health services utilization. Despite an aging population that faces significant mental health concerns in Korea, the role of mental health literacy on mental health services utilization is not known among older adults in Korea. This study aimed to (1) identify whether mental health literacy mediates the association between population characteristics and mental health services utilization and (2) identify an optimal path model for mental health services utilization among Korean older adults. METHODS: Using a cross-sectional survey with a quota sampling strategy, we collected and analyzed responses from 596 community-dwelling individuals ages 65 years and older. We used structural equation modeling (SEM) to estimate the effect of mental health literacy as a mediator. RESULTS: When controlling for other relevant covariates in the optimal path model, mental health literacy mediated the relationships between three socio-demographic factors (education, general literacy, and health status) and mental health services utilization. The model fit index shows that the SEM fits very well (CFI = 0.92, NFI = 0.90, RMSEA = 0.07). CONCLUSIONS: Efforts to improve mental health literacy through community-based education programs may need to particularly target Korean older adults with the relevant socio-demographic characteristics to enhance their utilization of appropriate mental health services.


Subject(s)
Health Literacy , Mental Health Services/statistics & numerical data , Mental Health , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Health Status , Humans , Independent Living , Male , Regression Analysis , Republic of Korea/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL