Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Cell Death Discov ; 10(1): 185, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649679

ABSTRACT

Distant metastasis is a significant hallmark affecting to the high death rate of patients with triple-negative breast cancer (TNBC). Thus, it is crucial to identify and develop new therapeutic strategies to hinder cancer metastasis. While emerging studies have hinted a pivotal role of glucose-regulated protein 94 (GRP94) in tumorigenesis, the exact biological functions and molecular mechanisms of GRP94 in modulating cancer metastasis remain to be elucidated. Our study demonstrated an increased expression of GRP94 in TNBC correlated with metastatic progression and unfavorable prognosis in patients. Functionally, we identified that GRP94 depletion significantly diminished TNBC tumorigenesis and subsequent lung metastasis. In contrast, GRP94 overexpression exacerbated the invasiveness, migration, and lung metastasis of non-TNBC cells. Mechanistically, we found that casein kinase 2 alpha (CK2α) active in advanced breast cancer phosphorylated GRP94 at a conserved serine 306 (S306) residue. This phosphorylation increased the stability of GRP94 and enhanced its interaction with LRP6, leading to activation of canonical Wnt signaling. From a therapeutic standpoint, we found that benzamidine, a novel CK2α inhibitor, effectively suppressed GRP94 phosphorylation, LRP6 stabilization, and metastasis of TNBC. Our results point to the critical role of CK2α-mediated GRP94 phosphorylation in TNBC metastasis through activation of Wnt signaling, highlighting GRP94 as a therapeutic target to impede TNBC metastasis.

2.
Molecules ; 28(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38138587

ABSTRACT

Inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease, is a complex gastrointestinal disorder with a multifactorial etiology, including environmental triggers, autoimmune mechanisms, and genetic predisposition. Despite advancements in therapeutic strategies for IBD, its associated mortality rate continues to rise, which is often attributed to unforeseen side effects of conventional treatments. In this context, we explored the potential of Ecklonia cava extract (ECE), derived from an edible marine alga known for its anti-inflammatory and antioxidant properties, in mitigating IBD. This study investigated the effectiveness of ECE as a preventive agent in a murine model of dextran sulfate sodium (DSS)-induced colitis. Our findings revealed that pretreatment with ECE significantly ameliorated colitis severity, as evidenced by increased colon length, reduced spleen weight, and histological improvements demonstrated by immunohistochemical analysis. Furthermore, ECE significantly attenuated the upregulation of inflammatory cytokines and mediators and the infiltration of immune cells known to be prominent features of colitis in mice. Notably, ECE alleviated dysbiosis of intestinal microflora and aided in the recovery of damaged intestinal mucosa. Mechanistically, ECE exhibited protective effects against pathogenic colitis by inhibiting the NLRP3/NF-κB pathways known to be pivotal regulators in the inflammatory signaling cascade. These compelling results suggest that ECE holds promise as a potential candidate for IBD prevention. It might be developed into a functional food for promoting gastrointestinal health. This research sheds light on the preventive potential of natural compounds like ECE in the management of IBD, offering a safer and more effective approach to combating this challenging disease.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Mice , Intestinal Barrier Function , Disease Models, Animal , Colitis/chemically induced , Colitis/drug therapy , Inflammation , Inflammatory Bowel Diseases/pathology , Dextran Sulfate/toxicity , Mice, Inbred C57BL , Colon/pathology
3.
Mol Oncol ; 17(11): 2380-2395, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37341064

ABSTRACT

The incidence of colitis-associated colorectal cancer (CAC) has increased due to a high-nutrient diet, increased environmental stimuli and inherited gene mutations. To adequately treat CAC, drugs should be developed by identifying novel therapeutic targets. E3 ubiquitin-protein ligase pellino homolog 3 (pellino 3; Peli3) is a RING-type E3 ubiquitin ligase involved in inflammatory signalling; however, its role in the development and progression of CAC has not been elucidated. In this study, we studied Peli3-deficient mice in an azoxymethane/dextran sulphate sodium-induced CAC model. We observed that Peli3 promotes colorectal carcinogenesis with increased tumour burden and oncogenic signalling pathways. Ablation of Peli3 reduced inflammatory signalling activation at the early stage of carcinogenesis. Mechanistic studies indicate that Peli3 enhances toll-like receptor 4 (TLR4)-mediated inflammation through ubiquitination-dependent degradation of interferon regulatory factor 4, a negative regulator of TLR4 in macrophages. Our study suggests an important molecular link between Peli3 and colonic inflammation-mediated carcinogenesis. Furthermore, Peli3 can be a therapeutic target in the prevention and treatment of CAC.


Subject(s)
Colitis-Associated Neoplasms , Toll-Like Receptor 4 , Animals , Mice , Carcinogenesis/genetics , Colitis-Associated Neoplasms/genetics , Inflammation/complications , Interferon Regulatory Factors/metabolism , Mice, Inbred C57BL , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
4.
Exp Mol Med ; 55(6): 1218-1231, 2023 06.
Article in English | MEDLINE | ID: mdl-37258579

ABSTRACT

The signaling pathways governing acetaminophen (APAP)-induced liver injury have been extensively studied. However, little is known about the ubiquitin-modifying enzymes needed for the regulation of APAP-induced liver injury. Here, we examined whether the Pellino3 protein, which has E3 ligase activity, is needed for APAP-induced liver injury and subsequently explored its molecular mechanism. Whole-body Peli3-/- knockout (KO) and adenovirus-mediated Peli3 knockdown (KD) mice showed reduced levels of centrilobular cell death, infiltration of immune cells, and biomarkers of liver injury, such as alanine aminotransferase (ALT) and aspartate aminotransferase (AST), upon APAP treatment compared to wild-type (WT) mice. Peli3 deficiency in primary hepatocytes decreased mitochondrial and lysosomal damage and reduced the mitochondrial reactive oxygen species (ROS) levels. In addition, the levels of phosphorylation at serine 9 in the cytoplasm and mitochondrial translocation of GSK3ß were decreased in primary hepatocytes obtained from Peli3-/- KO mice, and these reductions were accompanied by decreases in JNK phosphorylation and mitochondrial translocation. Pellino3 bound more strongly to GSK3ß compared with JNK1 and JNK2 and induced the lysine 63 (K63)-mediated polyubiquitination of GSK3ß. In rescue experiments, the ectopic expression of wild-type Pellino3 in Peli3-/- KO hepatocytes restored the mitochondrial translocation of GSK3ß, but this restoration was not obtained with expression of a catalytically inactive mutant of Pellino3. These findings are the first to suggest a mechanistic link between Pellino3 and APAP-induced liver injury through the modulation of GSK3ß polyubiquitination.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury, Chronic , Animals , Mice , Acetaminophen/adverse effects , Phosphorylation , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Liver/metabolism , Hepatocytes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Mice, Inbred C57BL
5.
Int J Mol Sci ; 23(23)2022 Nov 29.
Article in English | MEDLINE | ID: mdl-36499297

ABSTRACT

Heat shock proteins (HSPs) are highly conserved molecular chaperones with diverse cellular activities, including protein folding, assembly or disassembly of protein complexes, and maturation process under diverse stress conditions. HSPs also play essential roles in tumorigenesis, metastasis, and therapeutic resistance across cancers. Among them, HSP40s are widely accepted as regulators of HSP70/HSP90 chaperones and an accumulating number of biological functions as molecular chaperones dependent or independent of either of these chaperones. Despite large numbers of HSP40s, little is known about their physiologic roles, specifically in cancer progression. This article summarizes the multi-faceted role of DNAJB proteins as one subclass of the HSP40 family in cancer development and metastasis. Regulation and deregulation of DNAJB proteins at transcriptional, post-transcriptional, and post-translational levels contribute to tumor progression, particularly cancer metastasis. Furthermore, understanding differences in function and regulating mechanism between DNAJB proteins offers a new perspective on tumorigenesis and metastasis to improve therapeutic opportunities for malignant diseases.


Subject(s)
HSP40 Heat-Shock Proteins , Neoplasms , Humans , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Protein Folding , HSP90 Heat-Shock Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism
6.
Int J Oncol ; 59(5)2021 Nov.
Article in English | MEDLINE | ID: mdl-34558638

ABSTRACT

Cold­inducible RNA­binding protein (CIRBP) is a cold­shock protein comprised of an RNA­binding motif that is induced by several stressors, such as cold shock, UV radiation, nutrient deprivation, reactive oxygen species and hypoxia. CIRBP can modulate post­transcriptional regulation of target mRNA, which is required to control DNA repair, circadian rhythms, cell growth, telomere integrity and cardiac physiology. In addition, the crucial function of CIRBP in various human diseases, including cancers and inflammatory disease, has been reported. Although CIRBP is primarily considered to be an oncogene, it may also serve a role in tumor suppression. In the present study, the controversial roles of CIRBP in various human cancers is summarized, with a focus on the interconnectivity between CIRBP and its target mRNAs involved in tumorigenesis. CIRBP may represent an important prognostic marker and therapeutic target for cancer therapy.


Subject(s)
Neoplasms/etiology , RNA-Binding Proteins/physiology , Apoptosis , Humans , Inflammation/etiology , Neoplasm Invasiveness , Neoplasms/drug therapy , Prognosis , RNA-Binding Proteins/antagonists & inhibitors
7.
Int J Oncol ; 59(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34278454

ABSTRACT

Following the publication of the above review article, the authors have realized that they overlooked including the funding information in the Declarations section. Therefore, the following text should also have been included with the review: Funding: The present review was supported by the National Research Foundation of Korea grant funded by the Korean government (grant no. 2020R1F1A1061122) and Gachon University Research fund of 2018 (GCU-2018-0670) to SH. The authors regret their oversight, apologize to the funding bodies concerned, and regret any inconvenience caused. [the original article was published in International Journal of Oncology 58: 344­358, 2021; DOI: 10.3892/ijo.2021.5175].

8.
Cell Death Dis ; 12(5): 461, 2021 05 08.
Article in English | MEDLINE | ID: mdl-33966034

ABSTRACT

DNAJB9, a member of the heat shock protein 40 family, acts as a multifunctional player involved in the maintenance of their client proteins and cellular homeostasis. However, the mechanistic action of DNAJB9 in human malignancies is yet to be fully understood. In this study, we found that ectopic restoration of DNAJB9 inhibits the migration, invasion, in vivo metastasis, and lung colonization of triple-negative breast cancer (TNBC) cells. Mechanistically, DNAJB9 stabilizes FBXO45 protein by suppressing self-ubiquitination and reduces the abundance of ZEB1 by Lys48-linked polyubiquitination to inhibit the epithelial-mesenchymal transition (EMT) and metastasis. Clinically, the reduction of DNAJB9 expression, concomitant with decreased FBXO45 abundance in breast cancer tissues, correlates with poorer clinical outcomes of patients with breast cancer. Taken together, our results provide a novel insight into the metastasis of TNBC and define a promising therapeutic strategy for cancers with overactive ZEB1 by regulating the DNAJB9-FBXO45 signaling axis.


Subject(s)
F-Box Proteins/metabolism , HSP40 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Triple Negative Breast Neoplasms/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Triple Negative Breast Neoplasms/pathology
9.
Int J Oncol ; 58(3): 344-358, 2021 03.
Article in English | MEDLINE | ID: mdl-33650664

ABSTRACT

Radiotherapy (RT) followed by radical surgery is an effective standard treatment strategy for various types of cancer, including rectal cancer. The response to RT varies among patients, and the radiosensitivity of cancer cells determines the clinical outcome of patients. However, the application of RT to patients with radioresistant tumors may result in radiation­induced toxicity without clinical benefits. Currently, there are no effective methods to predict the response to RT. The limitations of the methods currently used to evaluate tumor radiosensitivity, which are mainly based on clinical and radiological features, are low sensitivity and specificity. Non­coding RNAs (ncRNAs) have emerged as a class of biomarkers for predicting radiosensitivity. In particular, the expression pattern of ncRNAs can predict the response to RT in patients with rectal cancer. Thus, ncRNAs may be used as potential biomarkers and therapeutic targets to improve the diagnosis and treatment outcome of patients with rectal cancer. In the present review, the current knowledge on the limitations of RT for rectal cancer and the association between ncRNA expression and sensitivity of rectal cancer to RT are presented. Additionally, the potential of ncRNAs as predictive biomarkers and therapeutic targets to mitigate resistance of rectal cancer to RT is discussed.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic/radiation effects , RNA, Untranslated/metabolism , Radiation Tolerance/genetics , Rectal Neoplasms/therapy , Biomarkers, Tumor/analysis , Humans , Proctectomy , Prognosis , RNA, Untranslated/analysis , Radiotherapy, Adjuvant , Rectal Neoplasms/genetics , Rectal Neoplasms/pathology
10.
Neoplasia ; 21(9): 882-892, 2019 09.
Article in English | MEDLINE | ID: mdl-31387015

ABSTRACT

Radioresistance poses a major challenge in the treatment of advanced rectal cancer. Therefore, understanding the detailed mechanisms of radioresistance may improve patient response to irradiation and the survival rate. To identify the novel targets that modulate the radiosensitivity of rectal cancer, we performed small RNA sequencing with human rectal cancer cell lines. Through bioinformatics analysis, we selected microRNA-310a (miR-130a) as a promising candidate to elucidate radioresistance. miR-130a was dramatically upregulated in radiosensitive rectal cancer cells and overexpression of miR-130a promotes rectal cancer cell radiosensitivity. Mechanically, miR-130a reversed the epithelial-mesenchymal transition phenotype of rectal cancer cells following inhibition of cell invasion upon irradiation. Moreover, miR-130a also inhibited the repair of irradiation-induced DNA damage followed by cell death. We identified that SOX4 was a direct target of miR-130a. Overexpression of SOX4 reversed the promotion activity of miR-130a on radiosensitivity. Together, our findings suggest that miR-130a functions as a radiosensitizer in rectal cancer and reveals a potential therapeutic target and preoperative prognostic marker for radiotherapy.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA Interference , Radiation Tolerance/genetics , Rectal Neoplasms/genetics , SOXC Transcription Factors/genetics , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Computational Biology/methods , DNA Damage , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Genes, Reporter , Humans , Mice , Models, Biological , Rectal Neoplasms/pathology , Rectal Neoplasms/radiotherapy , Xenograft Model Antitumor Assays
11.
Sci Rep ; 9(1): 9457, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31263239

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer, and is associated with a high percentage of cancer-related death globally. Furthermore, the success rate of therapeutic treatment for CRC patients mainly depends on the status of metastasis. Therefore, novel drugs or therapeutic techniques should be discovered for the treatment of metastatic CRC. In this study, we selected Astaxanthin (AXT), one of the most common carotenoids, as a novel metastasis inhibitor through high-throughput drug screening based on invadopodia staining, and confirmed the anti-migratory and anti-invasive activity of AXT. We demonstrated that AXT increases miR-29a-3p and miR-200a expression, and thereby suppresses the expression of MMP2 and ZEB1, respectively. As a result, AXT represses the epithelial-mesenchymal transition (EMT) of CRC cells. Through the mechanistic study, we identified that AXT shows anti-metastatic activity through the transcriptional repression of MYC transcription factor. Finally, we also confirmed that AXT suppresses the in vivo metastatic capacity of colon cancer cell using mouse model. Collectively, we uncovered the novel function of AXT in the inhibition of EMT and invadopodia formation, implicating the novel therapeutic potential for AXT in metastatic CRC patients.


Subject(s)
Down-Regulation/drug effects , MicroRNAs/metabolism , Proto-Oncogene Proteins c-myc/metabolism , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Movement/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness/prevention & control , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA, Small Interfering/metabolism , Transplantation, Heterologous , Xanthophylls/pharmacology , Xanthophylls/therapeutic use , Zinc Finger E-box-Binding Homeobox 1/metabolism
12.
Biomed Pharmacother ; 116: 108987, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31112870

ABSTRACT

BP10A is a novel two-herb medicine formula, consisting of Descurainiae sophia Semen and Peucedani praeruptorum Radix. This study was done to evaluate the antitumor efficacy of BP10A and its effect on the efficacy of the anticancer drugs oxaliplatin and irinotecan (CPT-11) in a colon tumor xenograft model. Chemical constituents from the ethanol extracts of BP10A were characterized with the ultra-performance liquid chromatography (UPLC) and each constituent was quantified with the UPLC-diode array detector method. Our study showed that BP10A exerted the cytotoxic effects in two colorectal cancer cell lines and its combination treatments with oxaliplatin or CPT-11 remarkably increased the in vitro cytotoxicity of each cancer drug assessed by the Ez-cytox assay. The in vivo antitumor activity of BP10A was evaluated in three colon cancer patient-derived tumor xenograft (PDTX) models with different genetic backgrounds. Oral administration with BP10A (250 and 500 mg/kg, daily) delayed tumor growth by 34-70% in the all PDTX models. Similarly, intraperitoneal injection of oxaliplatin (6 mg/kg) or CPT-11 (20 mg/kg) also suppressed tumor growth by 31.8-60.5% or by 24.3-50.4%, respectively. Furthermore, the combination treatment of BP10A with oxaliplatin or CPT-11 remarkably enhanced the antitumor activity of each anti-cancer drug and delayed tumor growth by 47.1-74.6% or by 74.4-82.9%, respectively. In accordance with the antitumor activity, the Ki-67 expression for tumor cell proliferation and the CD31 for angiogenesis were decreased, and TUNEL staining for tumor cell apoptosis was remarkably increased by the co-treatment of BP10A and the anticancer drugs as well as by each treatment of BP10A, oxaliplatin or CPT-11. Conclusively, BP10A has a strong tumor inhibitory effect against colon cancer and a synergistic effect with anticancer drugs, suggesting that BP10A could be considered as a good therapeutic candidate for the treatment of colon cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Drugs, Chinese Herbal/therapeutic use , Irinotecan/therapeutic use , Oxaliplatin/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Drugs, Chinese Herbal/pharmacology , Humans , Irinotecan/chemistry , Irinotecan/pharmacology , Male , Mice, Inbred BALB C , Mice, Nude , Oxaliplatin/chemistry , Oxaliplatin/pharmacology , Phytotherapy , Xenograft Model Antitumor Assays
13.
Carcinogenesis ; 40(1): 112-120, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30219864

ABSTRACT

SMA- and MAD-related protein 7 (SMAD7) is a general inhibitor of transforming growth factor-ß (TGF-ß) signaling that acts through interaction and degradation of TGF-ß receptors. SMAD7 has been demonstrated to be transcriptionally upregulated in chemical-induced skin tumors and TGF-ß-treated normal keratinocytes. To evaluate the function of SMAD7 in skin carcinogenesis in vivo, Smad7 transgenic mice that specifically express either wild-type (WT) SMAD7 (TG-Smad7-WT) or mutant SMAD7 (TG-Smad7-MT) in keratinocytes, as well as Smad7 keratinocyte-specific knockout (Smad72f/2f-K14Cre) mice, were subjected to chemical-induced skin carcinogenesis. WT-SMAD7-expressing transgenic mice showed significantly greater papilloma formation than did non-TG control and Smad7-MT mice. The expression of WT-SMAD7 attenuated DNA damage-induced apoptosis in epidermal keratinocytes by stimulating the ATM-dependent DNA repair pathway. Nonetheless, overexpression of WT-SMAD7 caused a susceptibility to 12-O-tetradecanoylphorbol-13-acetate-induced epidermal hyperproliferation through activation of epidermal growth factor (EGF) signaling. In agreement with the transgenic mouse data, keratinocyte-specific deletion of SMAD7 markedly suppressed the tumor formation by inhibiting ATM and epidermal growth factor receptor (EGFR) signaling. Moreover, specific inhibition of EGFR signaling attenuated the hyperproliferation and tumor formation in TG-Smad7-WT mice. Taken together, these data support a novel role for SMAD7 as a tumor promoter in skin carcinogenesis where SMAD7 stimulates the DNA repair pathway and EGFR signaling activation.


Subject(s)
DNA Repair , ErbB Receptors/physiology , Keratinocytes/physiology , Skin Neoplasms/etiology , Smad7 Protein/physiology , Animals , Ataxia Telangiectasia Mutated Proteins/physiology , Cell Proliferation , Mice , Mice, Inbred C57BL , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
14.
J Am Heart Assoc ; 7(21): e009514, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30571376

ABSTRACT

Background Hereditary hemorrhagic telangiectasia ( HHT ) is a rare genetic vascular disorder caused by mutations in endoglin ( ENG ), activin receptor-like kinase 1 ( ACVRL 1; ALK 1), or SMAD 4. Major clinical symptoms of HHT are arteriovenous malformations ( AVM s) found in the brain, lungs, visceral organs, and mucosal surface. Animal models harboring mutations in Eng or Alk1 recapitulate all of these HHT clinical symptoms and have been useful resources for studying mechanisms and testing potential drugs. However, animal models representing SMAD 4 mutations have been lacking. The goal of this study is to evaluate Smad4-inducible knockout ( iKO ) mice as an animal model of HHT and compare the phenotypes with other established HHT animal models. Methods and Results Global Smad4 deletion was induced at neonatal and adult stages, and hemoglobin levels, gastrointestinal hemorrhage, and presence of aberrant arteriovenous connections were examined. Neonatal Smad4- iKO mice exhibited signs of gastrointestinal bleeding and AVM s in the brain, intestine, nose, and retina. The radial expansion was decreased, and AVM s were detected on both distal and proximal retinal vasculature of Smad4- iKO s. Aberrant smooth muscle actin staining was observed in the initial stage AVM s and their connecting veins, indicating abnormal arterial flow to veins. In adult mice, Smad4 deficiency caused gastrointestinal bleeding and AVM s along the gastrointestinal tract and wounded skin. HHT -related phenotypes of Smad4- iKO s appeared to be comparable with those found in Alk1- iKO and Eng- iKO mice. Conclusions These data further confirm that SMAD signaling is crucial for normal arteriovenous network formation, and Smad4- iKO will be an alternative animal model of AVM research associated with HHT .


Subject(s)
Arteriovenous Malformations/genetics , Disease Models, Animal , Mice , Smad4 Protein/deficiency , Smad4 Protein/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Age Factors , Animals , Animals, Newborn , Mice, Knockout , Phenotype
15.
Article in English | MEDLINE | ID: mdl-29725354

ABSTRACT

Celtis choseniana is the traditional plant used at Korea as a herbal medicine to ameliorate inflammatory responses. Although Celtis choseniana has been traditionally used as a herbal medicine at Korea, no systemic research has been conducted on its anti-inflammatory activity. Therefore, the present study explored an anti-inflammatory effect and its underlying molecular mechanism using Celtis choseniana methanol extract (Cc-ME) in macrophage-mediated inflammatory responses. In vitro anti-inflammatory activity of Cc-ME was evaluated using RAW264.7 cells and peritoneal macrophages stimulated by lipopolysaccharide (LPS), pam3CSK4 (Pam3), or poly(I:C). In vivo anti-inflammatory activity of Cc-ME was investigated using acute inflammatory disease mouse models, such as LPS-induced peritonitis and HCl/EtOH-induced gastritis. The molecular mechanism of Cc-ME-mediated anti-inflammatory activity was examined by Western blot analysis and immunoprecipitation using whole cell and nuclear fraction prepared from the LPS-stimulated RAW264.7 cells and HEK293 cells. Cc-ME inhibited NO production and mRNA expression of inducible nitric oxide synthase (iNOS), cyclooxygenase (COX-2), and tumor necrosis factor-alpha (TNF-α) in the RAW264.7 cells and peritoneal macrophages induced by LPS, pam3, or poly(I:C) without cytotoxicity. High-performance liquid chromatography (HPLC) analysis showed that Cc-ME contained anti-inflammatory flavonoids quercetin, luteolin, and kaempferol. Among those, the content of luteolin, which showed an inhibitory effect on NO production, was highest. Cc-ME suppressed the NF-κB signaling pathway by targeting Src and interrupting molecular interactions between Src and p85, its downstream kinase. Moreover, Cc-ME ameliorated the morphological finding of peritonitis and gastritis in the mouse disease models. Therefore, these results suggest that Cc-ME exerted in vitro and in vivo anti-inflammatory activity in LPS-stimulated macrophages and mouse models of acute inflammatory diseases. This anti-inflammatory activity of Cc-ME was dominantly mediated by targeting Src in NF-κB signaling pathway during macrophage-mediated inflammatory responses.

16.
Int J Mol Sci ; 19(3)2018 Mar 19.
Article in English | MEDLINE | ID: mdl-29562668

ABSTRACT

Hydroquinone (HQ, 1,4-benzenediol) is a hydroxylated benzene metabolite with various biological activities, including anti-oxidative, neuroprotective, immunomodulatory, and anti-inflammatory functions. However, the anti-cancer activity of HQ is not well understood. In this study, the in vitro and in vivo anti-cancer activity of HQ was investigated in various cancer cells and tumor-bearing mouse models. HQ significantly induced the death of A431, SYF, B16F10, and MDA-MB-231 cells and also showed a synergistic effect on A431 cell death with other anti-cancer agents, such as adenosine-2',3'-dialdehyde and buthionine sulfoximine. In addition, HQ suppressed angiogenesis in fertilized chicken embryos. Moreover, HQ prevented lung metastasis of melanoma cells in mice in a dose-dependent manner without toxicity and adverse effects. HQ (10 mg/kg) also suppressed the generation of colon and reduced the thickness of colon tissues in azoxymethane/dextran sodium sulfate-injected mice. This study strongly suggests that HQ possesses in vitro and in vivo anti-cancer activity and provides evidence that HQ could be developed as an effective and safe anti-cancer drug.


Subject(s)
Antineoplastic Agents/pharmacology , Hydroquinones/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Azoxymethane , Benzoquinones/chemistry , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Chickens , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Dextran Sulfate , Hydroquinones/chemistry , Hydroquinones/therapeutic use , Inhibitory Concentration 50 , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Melanoma, Experimental/pathology , Mice, Inbred C57BL
17.
Oncotarget ; 9(2): 1705-1716, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29416724

ABSTRACT

The potential of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in inducing apoptosis is a hallmark in cancer therapeutics, after which its selective ability to achieve cell death pathways against cancer cells led to hope for recombinant TRAIL in cancer therapeutics. The present data from azoxymethane-initiated, dextran sulfate sodium-promoted colitis associated cancer (CAC) model strongly indicate the potential of rTRAIL in cancer prevention rather than in cancer therapeutics. Early treatment of rTRAIL significantly reduced colitis and CAC by inhibiting the recruitment of macrophages into the damaged mucosa and activating the scavenger activity with efferocytosis and the production of several growth factors. In contrast, late administration of rTRAIL as for anti-cancer effect did not decrease the initiation and development of CAC at all. Significant cancer preventing mechanisms of rTRAIL were identified. In the CAC model, anti-inflammation, regeneration, and efferocytosis was induced by treatment of TRAIL for 6 days, significant inhibitory activity was evident at 4 weeks and anti-oxidative and anti-inflammatory induction were noted at 12 weeks. Most importantly, TRAIL promoted tissue regeneration by enhancing the resolution of pathological inflammation through the activation of the NLRP3 inflammasome pathway. The results indicate that TRAIL reduces the induction of colitis and the initiation of CAC by inhibiting pro-inflammatory signaling and promoting tissue repair to maintain intestinal homeostasis through activation of the NLRP3 inflammasome. Therefore, TRAIL can be used as a chemopreventive agent against CAC, rather than as a therapeutic drug endowing apoptosis.

18.
Mol Med Rep ; 17(3): 4540-4546, 2018 03.
Article in English | MEDLINE | ID: mdl-29363730

ABSTRACT

TRIO and F-actin-binding protein (TrioBP), which was initially discovered as a binding partner of Trio and F­actin, is a critical factor associated with hearing loss in humans. However, the function of TrioBP in cancer has not been investigated. In the present study, TrioBP expression was indicated to be highly elevated in U87­MG and U343­MG cells. Furthermore, the TrioBP mRNA expression level was markedly increased in U87­MG and U251­MG cells compared with that in cerebral cortex cells, as determined by deep sequencing. Comprehensive analysis of a public TCGA dataset confirmed that TrioBP expression is elevated in patients with glioblastoma. In summary, the present data indicate that TrioBP expression is increased in glioblastoma cell lines and in patients with glioma, suggesting that TrioBP has potential as a diagnostic marker or therapeutic agent for glioma.


Subject(s)
Brain Neoplasms/pathology , Carrier Proteins/metabolism , Glioblastoma/pathology , Guanine Nucleotide Exchange Factors/metabolism , Microfilament Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Brain Neoplasms/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Glioblastoma/metabolism , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Microfilament Proteins/genetics , Microscopy, Confocal , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Sequence Analysis, RNA , Up-Regulation
19.
Cancer Lett ; 415: 30-39, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29217458

ABSTRACT

Triple-negative breast cancer (TNBC) is one of the most aggressive malignancies and is associated with high mortality rates due to the lack of effective therapeutic targets. In this study, we demonstrated that insulin-like growth factor-II mRNA-binding protein 2 and 3 (IMP2 and IMP3) are specifically overexpressed in TNBC and cooperate to promote cell migration and invasion. Downregulation of both IMP2 and IMP3 in TNBC cells was found to produce a synergistic effect in suppressing cell invasion and invadopodia formation, whereas overexpression of IMP2 and IMP3 in luminal subtype cells enhanced epithelial-mesenchymal transition and metastasis. We also showed that IMP2 and IMP3 are direct targets of microRNA-200a (miR-200a), which is downregulated in TNBC. Conversely, IMP2 and IMP3 suppressed the transcription of miR-200a by destabilizing progesterone receptor (PR) mRNA through recruitment of the CCR4-NOT transcription complex subunit 1 (CNOT1) complex. Together, our findings suggest that IMP2 and IMP3 partially determine the characteristic phenotype and synergistically promote the metastasis of TNBC by downregulating PR. The identified IMP2/3-miR-200a-PR axis represents a novel double-negative feedback loop and serves as a new potential therapeutic target for the treatment of TNBC.


Subject(s)
RNA-Binding Proteins/genetics , Receptors, Progesterone/genetics , Triple Negative Breast Neoplasms/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Metastasis , RNA Interference , RNA-Binding Proteins/metabolism , Receptors, Progesterone/metabolism , Sequence Homology, Nucleic Acid , Transplantation, Heterologous , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
20.
Anticancer Res ; 37(10): 5415-5423, 2017 10.
Article in English | MEDLINE | ID: mdl-28982851

ABSTRACT

Unopposed phosphoinositide 3-kinase (PI3K) activity and 3-phosphoinositide production in Jurkat cells, due to a mutation in the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor-suppressor protein, results in deregulation of PH domain-containing proteins including the serine/threonine kinase PKB. In Jurkat cells, PKB is constitutively active and phosphorylated at the activation-loop residue (Thr308). 3-Phosphoinositide-dependent protein kinase-1 (PDK1), an enzyme that also contains a PH domain, catalyses Thr308 phosphorylation of PKB in addition to other kinase families such as PKC isoforms. It is unknown, however, whether the loss of PTEN in Jurkat cells also results in unregulated PDK1 activity and whether such loss has an impact on activation-loop phosphorylation of other PDK1 substrates e.g. PKC. In this study, we addressed whether loss of PTEN in Jurkat cells affects PDK1 catalytic activity and intracellular localization. We demonstrated that reducing the level of 3-phosphoinositides in Jurkat cells with pharmacological inhibitors of PI3K or expression of PTEN does not affect PDK1 activity or its intracellular localization. We conclude, therefore, that although Jurkat cells lack PTEN expression, only a subset of pathways downstream of PDK1 are perturbed as a consequence of PTEN loss.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Leukemia, T-Cell/enzymology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Catalysis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...