Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(2)2022 Jan 15.
Article in English | MEDLINE | ID: mdl-35055132

ABSTRACT

Anterior gradient protein 2 homolog (AGR2), an endoplasmic reticulum protein, is secreted in the tumor microenvironment. AGR2 is a member of the disulfide isomerase family, is highly expressed in multiple cancers, and promotes cancer metastasis. In this study, we found that etravirine, which is a non-nucleoside reverse transcriptase inhibitor, could induce AGR2 degradation via autophagy. Moreover, etravirine diminished proliferation, migration, and invasion in vitro. Moreover, in an orthotopic xenograft mouse model, the combination of etravirine and paclitaxel significantly suppressed cancer progression and metastasis. This drug may be a promising therapeutic agent for the treatment of ovarian cancer.


Subject(s)
Mucoproteins/metabolism , Nitriles/administration & dosage , Oncogene Proteins/metabolism , Ovarian Neoplasms/drug therapy , Paclitaxel/administration & dosage , Pyrimidines/administration & dosage , Reverse Transcriptase Inhibitors/administration & dosage , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mucoproteins/genetics , Neoplasm Metastasis , Nitriles/pharmacology , Oncogene Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Proteolysis , Pyrimidines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
2.
FASEB J ; 36(1): e22068, 2022 01.
Article in English | MEDLINE | ID: mdl-34918396

ABSTRACT

Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) has been frequently overexpressed in many types of malignancy, suggesting its oncogenic function. It recognizes phosphorylated serine or threonine (pSer/Thr) of a target protein and isomerizes the adjacent proline (Pro) residue, thereby altering folding, subcellular localization, stability, and function of target proteins. The oncogenic transcription factor, Nrf2 harbors the pSer/Thr-Pro motif. This prompted us to investigate whether Pin1 could bind to Nrf2 and influence its stability and function in the context of implications for breast cancer development and progression. The correlation between Pin1 and Nrf2 in the triple-negative breast cancer cells was validated by RNASeq analysis as well as immunofluorescence staining. Interaction between Pin1 and Nrf2 was assessed by co-immunoprecipitation and an in situ proximity ligation assay. We found that mRNA and protein levels of Pin1 were highly increased in the tumor tissues of triple-negative breast cancer patients and the human breast cancer cell line. Genetic or pharmacologic inhibition of Pin1 enhanced the ubiquitination and degradation of Nrf2. In contrast, the overexpression of Pin1 resulted in the accumulation of Nrf2 in the nucleus, without affecting its transcription. Notably, the phosphorylation of Nrf2 at serine 215, 408, and 577 is essential for its interaction with Pin1. We also identified phosphorylated Ser104 and Thr277 residues in Keap1, a negative regulator of Nrf2, for Pin1 binding. Pin1 plays a role in breast cancer progression through stabilization and constitutive activation of Nrf2 by competing with Keap1 for Nrf2 binding.


Subject(s)
Breast Neoplasms/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neoplasm Proteins/metabolism , Animals , Breast Neoplasms/genetics , Female , HEK293 Cells , Humans , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Neoplasm Proteins/genetics , Protein Binding , Protein Stability , Proteolysis , Ubiquitination
3.
Prog Mol Biol Transl Sci ; 181: 271-287, 2021.
Article in English | MEDLINE | ID: mdl-34127196

ABSTRACT

Generation of proper models for studying human genetic diseases has been hindered until recently by the scarcity of primary cell samples from genetic disease patients and inefficient genetic modification tools. However, recent advances in clustered, regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology and human induced pluripotent stem cells (hiPSCs) have provided an opportunity to explore the function of pathogenic variants and obtain gene-corrected cells for autologous cell therapy. In this chapter, we address recent applications of CRISPR/Cas9 to hiPSCs in genetic diseases, including neurodegenerative, cardiovascular, and rare diseases.


Subject(s)
Gene Editing , Induced Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Humans
4.
Cells ; 10(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33807329

ABSTRACT

The presence of brown adipocytes within white adipose tissue is associated with phenotypes that exhibit improved metabolism and proper body weight maintenance. Therefore, a variety of dietary agents that facilitate the browning of white adipocytes have been investigated. In this study, we screened a natural product library comprising 133 compounds with the potential to promote the browning of white adipocytes, and found that D-mannitol induces the browning of 3T3-L1 adipocytes by enhancing the expression of brown fat-specific genes and proteins, and upregulating lipid metabolism markers. D-mannitol also increased the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase 1 (ACC), suggesting a possible role in lipolysis and fat oxidation. Moreover, an increase in the expression of genes associated with D-mannitol-induced browning was strongly correlated with the activation of the ß3-adrenergic receptor as well as AMPK, protein kinase A (PKA), and PPARγ coactivator 1α (PGC1α). D-mannitol effectively reduced the body weight of mice fed a high-fat diet, and increased the expression of ß1-oxidation and energy expenditure markers, such as Cidea, carnitine palmityl transferase 1 (CPT1), uncoupling protein 1 (UCP1), PGC1α, and acyl-coenzyme A oxidase (ACOX1) in the inguinal white adipose tissue. Our findings suggest that D-mannitol plays a dual regulatory role by inducing the generation of a brown fat-like phenotype and enhancing lipid metabolism. These results indicate that D-mannitol can function as an anti-obesity supplement.


Subject(s)
Adipose Tissue, Brown/metabolism , Mannitol/pharmacology , Receptors, Adrenergic, beta-3/metabolism , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Gene Expression Regulation/drug effects , Mannitol/chemistry , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Models, Animal , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phenotype , Signal Transduction/drug effects , Uncoupling Protein 1/metabolism
5.
Cells ; 10(4)2021 04 16.
Article in English | MEDLINE | ID: mdl-33923707

ABSTRACT

The overall five-year survival rate for late-stage patients of ovarian cancer is below 29% due to disease recurrence and drug resistance. Cancer stem cells (CSCs) are known as a major contributor to drug resistance and recurrence. Accordingly, therapies targeting ovarian CSCs are needed to overcome the limitations of present treatments. This study evaluated the effect of trimebutine maleate (TM) targeting ovarian CSCs, using A2780-SP cells acquired by a sphere culture of A2780 epithelial ovarian cancer cells. TM is indicated as a gastrointestinal motility modulator and is known to as a peripheral opioid receptor agonist and a blocker for various channels. The GI50 of TM was approximately 0.4 µM in A2780-SP cells but over 100 µM in A2780 cells, demonstrating CSCs specific growth inhibition. TM induced G0/G1 arrest and increased the AV+/PI+ dead cell population in the A2780-SP samples. Furthermore, TM treatment significantly reduced tumor growth in A2780-SP xenograft mice. Voltage gated calcium channels (VGCC) and calcium-activated potassium channels (BKCa) were overexpressed on ovarian CSCs and targeted by TM; inhibition of both channels reduced A2780-SP cells viability. TM reduced stemness-related protein expression; this tendency was reproduced by the simultaneous inhibition of VGCC and BKCa compared to single channel inhibition. In addition, TM suppressed the Wnt/ß-catenin, Notch, and Hedgehog pathways which contribute to many CSCs characteristics. Specifically, further suppression of the Wnt/ß-catenin pathway by simultaneous inhibition of BKCa and VGCC is necessary for the effective and selective action of TM. Taken together, TM is a potential therapeutic drug for preventing ovarian cancer recurrence and drug resistance.


Subject(s)
Drug Repositioning , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Trimebutine/therapeutic use , Calcium/metabolism , Calcium Channels/metabolism , Cell Death/drug effects , Cell Line, Tumor , Female , Humans , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Models, Biological , Neoplastic Stem Cells/drug effects , Protein Subunits/metabolism , Sodium/metabolism , Sodium Channels/metabolism , Transcription Factors/metabolism , Trimebutine/chemistry , Trimebutine/pharmacology , Wnt Signaling Pathway/drug effects
6.
Bone ; 142: 115707, 2021 01.
Article in English | MEDLINE | ID: mdl-33141068

ABSTRACT

Excessive bone resorption mediated by mature osteoclasts can cause osteoporosis, leading to fragility fractures. Therefore, an effective therapeutic strategy for anti-osteoporosis drugs is the reduction of osteoclast activity. In this study, the osteoclast inhibitory activity of a novel compound, N-phenyl-methylsulfonamido-acetamide (PMSA), was examined. PMSA treatment inhibited receptor activator of nuclear factor kappa B ligand (RNAKL)-induced osteoclast differentiation in bone marrow-derived macrophage cells (BMMs). We investigated two PMSAs, N-2-(3-acetylphenyl)-N-2-(methylsulfonyl)-N-1-[2-(phenylthio)phenyl] glycinamide (PMSA-3-Ac), and N-2-(5-chloro-2-methoxyphenyl)-N-2-(methylsulfonyl)-N-1-[2-(phenylthio)phenyl]glycinamide (PMSA-5-Cl), to determine their effects on osteoclast differentiation. PMSAs inhibited the signaling pathways at the early stage. PMSA-3-Ac inhibited tumor necrosis factor receptor-associated factor 6 (TRAF6) expression, whereas PMSA-5-Cl suppressed the mitogen-activated protein kinase (MAPK) signaling pathways. However, both PMSAs inhibited the master transcription factor, nuclear factor of activated T cell cytoplasmic-1 (NFATc1), by blocking nuclear localization. An in vivo study of PMSAs was performed in an ovariectomized (OVX) mouse model, and PMSA-5-Cl prevented bone loss in OVX mice. Therefore, our results suggested that PMSAs, specifically PMSA-5-Cl, may serve as a potential therapeutic agent for postmenopausal osteoporosis.


Subject(s)
Bone Resorption , Pharmaceutical Preparations , Acetamides , Animals , Bone Resorption/drug therapy , Cell Differentiation , Estrogens/pharmacology , Female , Humans , Mice , NFATC Transcription Factors , Osteoclasts , Osteogenesis , Ovariectomy , RANK Ligand
7.
Int J Mol Sci ; 21(24)2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33327533

ABSTRACT

The function of natural killer (NK) cell-derived interferon-γ (IFN-γ) expands to remove pathogens by increasing the ability of innate immune cells. Here, we identified the critical role of thioredoxin-interacting protein (TXNIP) in the production of IFN-γ in NK cells during bacterial infection. TXNIP inhibited the production of IFN-γ and the activation of transforming growth factor ß-activated kinase 1 (TAK1) activity in primary mouse and human NK cells. TXNIP directly interacted with TAK1 and inhibited TAK1 activity by interfering with the complex formation between TAK1 and TAK1 binding protein 1 (TAB1). Txnip-/- (KO) NK cells enhanced the activation of macrophages by inducing IFN-γ production during Pam3CSK4 stimulation or Staphylococcus aureus (S. aureus) infection and contributed to expedite the bacterial clearance. Our findings suggest that NK cell-derived IFN-γ is critical for host defense and that TXNIP plays an important role as an inhibitor of NK cell-mediated macrophage activation by inhibiting the production of IFN-γ during bacterial infection.


Subject(s)
Carrier Proteins/metabolism , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Thioredoxins/metabolism , Animals , Carrier Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Immunity, Innate/drug effects , Immunity, Innate/genetics , Killer Cells, Natural/immunology , Lipopeptides/pharmacology , Mice , Mice, Knockout , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcus aureus/pathogenicity , Thioredoxins/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
8.
Biochem Biophys Res Commun ; 533(4): 995-1003, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33012513

ABSTRACT

PIN1, the peptidyl-prolyl isomerase (PPIase), is an enzyme that changes the conformation of phosphoproteins. The conformational change induced by PIN1 alters the function and stability of the target proteins. PIN1 is overexpressed in many different types of malignancies, including breast, lung, cervical, brain and colorectal tumors. PIN1 overexpression has been associated with activation of multiple oncogenic signaling pathways during tumor development. Hypoxia-inducible factor 2α (HIF-2α), a transcription factor activated in hypoxia, plays a role in erythropoiesis, glycolysis, tissue invasion, metastasis and angiogenesis. In this study, we found the direct interaction between HIF-2α and PIN1 in colorectal cancer HCT116 cells. Notably, serine 16 and lysine 63 residues of PIN1 were critical for its interaction with HIF-2α. When PIN1 protein was silenced by transient transfection of PIN1 short interfering RNA, the expression of HIF-2α was attenuated under a hypoxic condition. Moreover, genetic and pharmacologic inhibition of PIN1 abrogated the expression of vascular endothelial growth factor and angiogenesis. The cycloheximide chase experiment revealed the stabilization of HIF-2α by PIN1. Both WW and PPIase domains of PIN1 appear to be critical for its interaction with HIF-2α.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neovascularization, Pathologic/etiology , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Chick Embryo , Female , HCT116 Cells , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , NIH 3T3 Cells , NIMA-Interacting Peptidylprolyl Isomerase/chemistry , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Interaction Domains and Motifs , Protein Stability , RNA Interference , RNA, Small Interfering/genetics , Tumor Hypoxia , Vascular Endothelial Growth Factor A/metabolism
9.
Molecules ; 25(20)2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33096734

ABSTRACT

Osteoclasts are large, multinucleated cells responsible for bone resorption and are induced in response to the regulatory activity of receptor activator of nuclear factor-kappa B ligand (RANKL). Excessive osteoclast activity causes pathological bone loss and destruction. Many studies have investigated molecules that specifically inhibit osteoclast activity by blocking RANKL signaling or bone resorption. In recent years, we screened compounds from commercial libraries to identify molecules capable of inhibiting RANKL-induced osteoclast differentiation. Consequently, we reported some compounds that are effective at attenuating osteoclast activity. In this study, we found that N-[2-(4-acetyl-1-piperazinyl)phenyl]-2-(3-methylphenoxy)acetamide (NAPMA) significantly inhibited the formation of multinucleated tartrate-resistant acid phosphatase (TRAP)-positive cells from bone marrow-derived macrophages in a dose-dependent manner, without cytotoxic effects. NAPMA downregulated the expression of osteoclast-specific markers, such as c-Fos, NFATc1, DC-STAMP, cathepsin K, and MMP-9, at the transcript and protein levels. Accordingly, bone resorption and actin ring formation were decreased in response to NAPMA treatment. Furthermore, we demonstrated the protective effect of NAPMA against ovariectomy-induced bone loss using micro-CT and histological analysis. Collectively, the results showed that NAPMA inhibited osteoclast differentiation and attenuated bone resorption. It is thus a potential drug candidate for the treatment of osteoporosis and other bone diseases associated with excessive bone resorption.


Subject(s)
Osteoclasts/drug effects , Osteoporosis , Ovariectomy , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Mice , Mice, Inbred C57BL , Osteoporosis/drug therapy , Osteoporosis/surgery , Structure-Activity Relationship
10.
Int J Mol Sci ; 21(19)2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33036254

ABSTRACT

Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy in women worldwide, with an overall 5 year survival rate below 30%. The low survival rate is associated with the persistence of cancer stem cells (CSCs) after chemotherapy. Therefore, CSC-targeting strategies are required for successful EOC treatment. Pan-human epidermal growth factor receptor 4 (HER4) and L-type calcium channels are highly expressed in ovarian CSCs, and treatment with the pan-HER inhibitor poziotinib or calcium channel blockers (CCBs) selectively inhibits the growth of ovarian CSCs via distinct molecular mechanisms. In this study, we tested the hypothesis that combination treatment with poziotinib and CCBs can synergistically inhibit the growth of ovarian CSCs. Combined treatment with poziotinib and manidipine (an L-type CCB) synergistically suppressed ovarian CSC sphere formation and viability compared with either drug alone. Moreover, combination treatment synergistically reduced the expression of stemness markers, including CD133, KLF4, and NANOG, and stemness-related signaling molecules, such as phospho-STAT5, phospho-AKT, phospho-ERK, and Wnt/ß-catenin. Moreover, poziotinib with manidipine dramatically induced apoptosis in ovarian CSCs. Our results suggest that the combinatorial use of poziotinib with a CCB can effectively inhibit ovarian CSC survival and function.


Subject(s)
Carcinoma, Ovarian Epithelial/drug therapy , Dihydropyridines/therapeutic use , Neoplastic Stem Cells/drug effects , Nitrobenzenes/therapeutic use , Ovarian Neoplasms/drug therapy , Piperazines/therapeutic use , Quinazolines/therapeutic use , AC133 Antigen , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/physiopathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Nanog Homeobox Protein , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/physiopathology , STAT5 Transcription Factor , Treatment Outcome , Tumor Suppressor Proteins , Wnt Signaling Pathway
11.
Sci Rep ; 10(1): 12089, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32694555

ABSTRACT

Histone modifications, including histone lysine methylation, regulate gene expression in the vasculature, and targeting tumor blood vessels through histone modification decreases tumor growth. SETD8, a methyltransferase that catalyzes the mono-methylation of histone H4 lysine 20 is known to promote tumorigenesis in various cancers and its high levels of expression are related to poor prognosis. However, the detailed mechanisms by which SETD8 stimulates tumor progression and angiogenesis are still not well understood. Recent studies have demonstrated that, in vitro, BVT-948 efficiently and selectively suppresses SETD8 activity and histone methylation levels. In this study, we showed that BVT-948-mediated SETD8 inhibition in HUVECs results in an inhibition of angiogenesis. Inhibition of SETD8 not only inhibited angiogenesis but also disrupted actin stress fiber formation and induced cell cycle arrest at S phase. These effects were accompanied by increased HES-1 expression levels, decreased osteopontin levels, and a decreased differentiation of human induced pluripotent stem cells into endothelial cells. Interestingly, BVT-948 treatment reduced pathological angiogenesis in mouse OIR model. These data illustrate the mechanisms by which SETD8 regulates angiogenesis and may enable the use of a SETD8 inhibitor to treat various pathological conditions that are known to be associated with excessive angiogenesis, including and tumor growth.


Subject(s)
Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic/genetics , Transcription Factor HES-1/genetics , Actins/metabolism , Biomarkers , Cell Line, Tumor , Cell Proliferation , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism
12.
Biology (Basel) ; 9(5)2020 May 02.
Article in English | MEDLINE | ID: mdl-32370249

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a leading form of chronic liver disease, with few biomarkers and treatment options currently available. Non-alcoholic steatohepatitis (NASH), a progressive disease of NAFLD, may lead to fibrosis, cirrhosis, and hepatocellular carcinoma. Epigenetic modification can contribute to the progression of NAFLD causing non-alcoholic steatohepatitis (NASH), in which the exact role of epigenetics remains poorly understood. To identify potential therapeutics for NASH, we tested small-molecule inhibitors of the epigenetic target histone methyltransferase EZH2, Tazemetostat (EPZ-6438), and UNC1999 in STAM NASH mice. The results demonstrate that treatment with EZH2 inhibitors decreased serum TNF-alpha in NASH. In this study, we investigated that inhibition of EZH2 reduced mRNA expression of inflammatory cytokines and fibrosis markers in NASH mice. In conclusion, these results suggest that EZH2 may present a promising therapeutic target in the treatment of NASH.

13.
Int J Mol Sci ; 21(7)2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32230901

ABSTRACT

Drug resistance in epithelial ovarian cancer (EOC) is reportedly attributed to the existence of cancer stem cells (CSC), because in most cancers, CSCs still remain after chemotherapy. To overcome this limitation, novel therapeutic strategies are required to prevent cancer recurrence and chemotherapy-resistant cancers by targeting cancer stem cells (CSCs). We screened an FDA-approved compound library and found four voltage-gated calcium channel blockers (manidipine, lacidipine, benidipine, and lomerizine) that target ovarian CSCs. Four calcium channel blockers (CCBs) decreased sphere formation, viability, and proliferation, and induced apoptosis in ovarian CSCs. CCBs destroyed stemness and inhibited the AKT and ERK signaling pathway in ovarian CSCs. Among calcium channel subunit genes, three L- and T-type calcium channel genes were overexpressed in ovarian CSCs, and downregulation of calcium channel genes reduced the stem-cell-like properties of ovarian CSCs. Expressions of these three genes are negatively correlated with the survival rate of patient groups. In combination therapy with cisplatin, synergistic effect was shown in inhibiting the viability and proliferation of ovarian CSCs. Moreover, combinatorial usage of manidipine and paclitaxel showed enhanced effect in ovarian CSCs xenograft mouse models. Our results suggested that four CCBs may be potential therapeutic drugs for preventing ovarian cancer recurrence.


Subject(s)
Antihypertensive Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Neoplastic Stem Cells/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Dihydropyridines/pharmacology , Drug Repositioning , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred BALB C , Neoplastic Stem Cells/drug effects , Nitrobenzenes , Ovarian Neoplasms , Paclitaxel/pharmacology , Piperazines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Microenvironment/drug effects
14.
Front Cell Dev Biol ; 8: 120, 2020.
Article in English | MEDLINE | ID: mdl-32258027

ABSTRACT

Peptidyl-prolyl isomerase (PIN1) specifically binds and isomerizes the phosphorylated serine/threonine-proline (pSer/Thr-Pro) motif, which results in the alteration of protein structure, function, and stability. The altered structure and function of these phosphorylated proteins regulated by PIN1 are closely related to cancer development. PIN1 is highly expressed in human cancers and promotes cancer as well as cancer stem cells by breaking the balance of oncogenes and tumor suppressors. In this review, we discuss the roles of PIN1 in cancer and PIN1-targeted small-molecule compounds.

15.
Biochem Biophys Res Commun ; 526(1): 158-164, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32201081

ABSTRACT

Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy, with an overall 5-year survival rate of only 30%. EOC is associated with drug resistance, frequent recurrence, and poor prognosis. A major contributor toward drug resistance might be cancer stem cells (CSCs), which may remain after chemotherapy. Here, we aimed to find therapeutic agents that target ovarian CSCs. We performed a high-throughput screening using the Clinical Compound Library with a sphere culture of A2780 EOCs. Poziotinib, a pan-human epidermal growth factor receptor (HER) inhibitor, decreased sphere formation, viability, and proliferation, and induced G1 cell cycle arrest and apoptosis in ovarian CSCs. In addition, poziotinib suppressed stemness and disrupted downstream signaling of Wnt/ß-catenin, Notch, and Hedgehog pathways, which contribute to many characteristics of CSCs. Interestingly, HER4 was overexpressed in ovarian CSCs and Poziotinib reduced the phosphorylation of STAT5, AKT, and ERK, which are regulated by HER4. Our results suggest that HER4 may be a promising therapeutic target for ovarian CSCs, and that poziotinib may be an effective therapeutic option for the prevention of ovarian cancer recurrence.


Subject(s)
Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Quinazolines/pharmacology , Receptor, ErbB-4/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , G1 Phase/drug effects , G1 Phase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/metabolism , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/genetics , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , beta Catenin/metabolism
16.
Stem Cell Res ; 42: 101676, 2020 01.
Article in English | MEDLINE | ID: mdl-31841972

ABSTRACT

Fabry disease is an X-linked inherited disease caused by a mutation in the galactosidase alpha (GLA) gene. Here, we generated a GLA knock-out cell line (GLA-KO hiPSCs) from normal human-induced pluripotent stem cells (hFSiPS1) using the CRISPR-Cas9 genome-editing tool. The GLA-KO hiPSCs maintained normal morphology, karyotypes, expression of stemness markers, and trilineage differentiation potential. Furthermore, the GLA-KO hiPSCs exhibited dissipation of GLA activity and abnormal Globotriaosylceramide (Gb3) accumulation. Our GLA-KO hiPSC line represents a valuable tool for studying the mechanisms involved in Fabry disease and the development of novel therapeutic alternatives to treat this rare condition.


Subject(s)
CRISPR-Cas Systems/genetics , Fabry Disease/genetics , Induced Pluripotent Stem Cells/metabolism , alpha-Galactosidase/metabolism , Animals , Cell Line , Fabry Disease/pathology , Humans , Mutation
17.
Food Chem Toxicol ; 131: 110582, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31220535

ABSTRACT

Alcoholism is a serious addiction that can lead to various health complications such as liver fibrosis, steatosis, and cirrhosis. Carvacrol is present in many plant-based essential oils and used as a preservative in the food industry. In this study, we have investigated the hepatoprotective role of carvacrol against ethanol-induced liver toxicity in mice. To determine the effect of carvacrol on liver injury parameters, 5 doses of 50% ethanol (10 mL/kg body weight) were orally administered every 12 h for inducing the hepatotoxicity in experimental mice. Interestingly, carvacrol pre-treatment (50 and 100 mg/kg) reversed the ethanol-induced effects on liver function, antioxidant markers, matrix metalloproteinases activities, and histological changes. Moreover, carvacrol binds to the active pocket of cytochrome P450 (Cyt P450) and inhibits its expression. Thus, our finding suggests carvacrol can be used as an adjuvant for the amelioration of alcohol-induced hepatotoxicity.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/therapeutic use , Fatty Liver, Alcoholic/prevention & control , Monoterpenes/therapeutic use , Protective Agents/therapeutic use , Animals , Autophagy/drug effects , Binge Drinking , Catalytic Domain , Cymenes , Cytochrome P-450 Enzyme Inhibitors/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Fatty Liver, Alcoholic/pathology , Liver/pathology , MAP Kinase Signaling System/drug effects , Male , Matrix Metalloproteinases/metabolism , Mice, Inbred ICR , Molecular Docking Simulation , Monoterpenes/metabolism , Oxidative Stress/drug effects , Protective Agents/metabolism , Protein Binding
18.
Sci Total Environ ; 679: 365-377, 2019 Aug 20.
Article in English | MEDLINE | ID: mdl-31085416

ABSTRACT

Human health and environment have been continuously getting exposure to toxic chemicals including nanomaterial; therefore, nontoxicity has recently attracted huge amount of attention. In this study, RU-AgNPs were synthesized by a green synthesis procedure and evaluated for their toxicity in human umbilical vein endothelial cells (HUVECs) as well as on zebrafish embryos via apoptotic pathway. The synthesized RU-AgNPs were average in size (20-25 nm) with a negative surface charge of -13.43 mV. As a result, RU-AgNPs potentiated the formation of reactive oxygen species (ROS) in HUVECs as confirmed by the results of immunoblotting analysis using apoptotic markers, such as Bax, Bcl2, and cytochrome C. Moreover, the induction of apoptosis in HUVECs was also authenticated in a dose-dependent manner after the treatment with RU-AgNPs by the Incucyte analysis. In vivo trials conducted on zebrafish visualized the mortality, malformation, and imbalanced in the heart rate, and cell death of the whole embryo, including severe morphological changes in the yolk sac and the tail of zebrafish. Furthermore, the results of western blot analysis demonstrated the increasing intensity of apoptotic biomarkers such as Bax, Bcl2, and Cyto C, including enhanced production of ROS, validating the cell death in zebrafish larvae. In addition, chemically functionalized silver nanoparticles found to be more cytotoxic than biogenic functionalized silver nanoparticles. Above-mentioned findings clearly demonstrate that Ru-AgNPs cause the toxicity via ROS-induced apoptotic pathway. Therefore, it is necessary to decide RU-AgNPs toxicity levels before being used in any biomedical application.


Subject(s)
Apoptosis , Embryo, Nonmammalian/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Metal Nanoparticles/toxicity , Oxidative Stress , Silver/toxicity , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Rumex/metabolism , Zebrafish
19.
Oncogenesis ; 8(3): 12, 2019 Feb 19.
Article in English | MEDLINE | ID: mdl-30783083

ABSTRACT

In cancer, activation of X-box binding protein (XBP1) has a critical role in tumorigenesis and cancer progression. Transcriptional regulatory mechanism of XBP1 in cancer development has been well known, however, regulation of ubiquitination and degradation of XBP1 has not been elucidated yet. Here we show that Fbw7, a substrate recognition component of the SKP1-Cullin-F-box-type E3 ligase, interacts with XBP1 in a phosphorylation-dependent manner, and facilitates XBP1 ubiquitination and protein degradation. Moreover, Fbw7 inhibits oncogenic pathways including NF-κB, AP1, and Myc induced by XBP1. Interestingly, XBP1 negatively regulates transcription of Fbw7 via a feedback mechanism through NF-κB/E2F-1 axis signaling pathway, suggesting that overexpression of XBP1s may contribute to low level of Fbw7 expression in human cancers. Therefore, a negative feedback loop between Fbw7 and XBP1 contributes to the regulation of tumor development and can be an attractive target for novel therapy in cancers.

20.
Molecules ; 23(12)2018 Nov 29.
Article in English | MEDLINE | ID: mdl-30501117

ABSTRACT

Osteoclasts are large multinucleated cells which are induced by the regulation of the receptor activator of nuclear factor kappa-Β ligand (RANKL), which is important in bone resorption. Excessive osteoclast differentiation can cause pathologic bone loss and destruction. Numerous studies have targeted molecules inhibiting RANKL signaling or bone resorption activity. In this study, 11 compounds from commercial libraries were examined for their effect on RANKL-induced osteoclast differentiation. Of these compounds, only 2-(3-(2-fluoro-4-methoxyphenyl)-6-oxo-1(6H)-pyridazinyl)-N-1H-indol-5-ylacetamide (2N1HIA) caused a significant decrease in multinucleated tartrate-resistant acid phosphatase (TRAP)-positive cell formation in a dose-dependent manner, without inducing cytotoxicity. The 2N1HIA compound neither affected the expression of osteoclast-specific gene markers such as TRAF6, NFATc1, RANK, OC-STAMP, and DC-STAMP, nor the RANKL signaling pathways, including p38, ERK, JNK, and NF-κB. However, 2N1HIA exhibited a significant impact on the expression levels of CD47 and cathepsin K, the early fusion marker and critical protease for bone resorption, respectively. The activity of matrix metalloprotease-9 (MMP-9) decreased due to 2N1HIA treatment. Accordingly, bone resorption activity and actin ring formation decreased in the presence of 2N1HIA. Taken together, 2N1HIA acts as an inhibitor of osteoclast differentiation by attenuating bone resorption activity and may serve as a potential candidate in preventing and/or treating osteoporosis, or other bone diseases associated with excessive bone resorption.


Subject(s)
Acetamides/pharmacology , Cathepsin K/metabolism , Cell Differentiation/drug effects , Osteoclasts/cytology , Osteoclasts/enzymology , Actins/metabolism , Animals , Bone Resorption/genetics , Bone Resorption/pathology , CD47 Antigen/metabolism , Cattle , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteogenesis/drug effects , RANK Ligand/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...