Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 46
1.
Exp Biol Med (Maywood) ; 248(23): 2249-2261, 2023 Dec.
Article En | MEDLINE | ID: mdl-38158808

Chemokines critically orchestrate the tumorigenesis, metastasis, and stemness features of cancer cells that lead to poor outcomes. High plasma levels of transforming growth factor-ß1 (TGFß1) correlate with poor prognostic features in advanced lung cancer patients, thus suggesting the importance of TGFß1 in the lung tumor microenvironment. However, the role of chemokines in TGFß1-induced tumor stemness features remains unclear. Here, we clarify the previously undocumented role of CXCL1 in TGFß1-induced lung cancer stemness features. CXCL1 and its receptor CXCR2 were significantly upregulated in TGFß1-induced lung cancer stem cells (CSCs). CXCL1 silencing (shCXCL1) suppressed stemness gene expression, tumorsphere formation, colony formation, drug resistance, and in vivo tumorigenicity in TGFß1-induced lung tumorspheres. Immunohistochemistry staining showed that patients with stage II/III lung cancer had higher expression levels of CXCL1. The levels of CXCL1 were positively associated with lymph node metastasis and correlated with the expression of the CSC transcription factor Oct-4. Furthermore, online database analysis revealed that CXCL1 expression was negatively correlated with lung cancer survival in patients. Patients with high TGFß1/CXCL1/CD44 co-expression had a worse survival rate. We suggest that CXCL1 serves as a crucial factor in TGFß1-induced stemness features of lung cancer.


Lung Neoplasms , Humans , Lung Neoplasms/metabolism , Transforming Growth Factor beta1/metabolism , Cell Line, Tumor , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Neoplastic Stem Cells/metabolism , Tumor Microenvironment
2.
Cancer Metab ; 10(1): 23, 2022 Dec 06.
Article En | MEDLINE | ID: mdl-36474273

BACKGROUND: Resistance to chemotherapeutic drugs is a key factor for cancer recurrence and metastases in head and neck cancer (HNC). Cancer stem cells (CSCs) in tumors have self-renewal, differentiation, and higher drug resistance capabilities, resulting in a poor prognosis for patients. In glucose metabolism, pyruvate dehydrogenase kinase (PDK) inhibits pyruvate dehydrogenase and impedes pyruvate from being metabolized into acetyl-CoA and entering the tricarboxylic acid cycle to generate energy. Studies have reported that PDK1 and PDK2 inhibition suppresses the growth, motility, and drug resistance of cancer cells. Furthermore, while TGFß1 levels are persistently elevated in HNC patients with poor prognosis, the role of PDK isoforms in the TGFß1-promoted progression and stem-like properties of HNC is unclear. METHODS: Levels of PDK1 and PDK2 were evaluated in HNC tissue microarrays by immunohistochemistry to explore potential clinical relevance. PDK1 and PDK2 were knocked down by the lentivirus shRNA system to investigate their role in TGFß1-promoted tumor progression in vitro. RESULTS: We found that PDK2 levels were increased in the later stage of HNC tissues compared to constant PDK1 expression. After PDK1 and PDK2 knockdown, we discovered increased ATP production and decreased lactate production in TGFß1-treated and untreated HNC cells. However, only PDK2 silencing significantly inhibited the clonogenic ability of HNC cells. We subsequently found that TGFß1-promoted migration and invasion capabilities were decreased in PDK1 and PDK2 knockdown cells. The tumor spheroid-forming capability, motility, CSC genes, and multidrug-resistant genes were downregulated in PDK1 and PDK2 silencing CSCs. PDK1 and PDK2 inhibition reversed cisplatin and gemcitabine resistance of CSCs, but not paclitaxel resistance. CONCLUSION: The results demonstrated that the PDK1- and PDK2-mediated Warburg effect contributes to the TGFß1-enhanced stemness properties of HNC. Therefore, PDK1 and PDK2 may serve as molecular targets for the combination therapy of HNC.

3.
Int J Biol Macromol ; 221: 623-633, 2022 Nov 30.
Article En | MEDLINE | ID: mdl-36099992

A variety of anticancer activities have been established for fucoidan from brown algae, whereas whether cancer stem cells (CSCs) are inhibited by sulfated polysaccharides is unexplored. In this study, fucoidan extracted from Sargassum hemiphyllum was showed heat stable and might tolerate 140 °C treatment. Fucoidan did not exhibit cytotoxicity in 5637 and T24 bladder cancer cells. After fucoidan treatment, the stress fibers were aggregated into thick and abundant underneath the plasma membrane and getting around the cells, and the structure of F-actin showed a remarkable change in the filopodial protrusion in T24 and 5637 cells. Using culture inserts, transwell assays and time lapse recordings showed that fucoidan inhibited cell migration. In the epithelial-mesenchymal transition (EMT), fucoidan downregulated the expression of vimentin, a mesenchymal marker, and upregulated the expression of E-cadherin, an epithelial marker. Additionally, the transcription levels of Snail, Slug, Twist1, Twist2, MMP2 and MMP9 were significantly decreased by fucoidan, indicating EMT suppression. CSCs are implicated in tumor initiation, metastatic spread, drug resistance and tumor recurrence. Our results showed that fucoidan inhibited stemness gene expression and sphere formation in bladder CSCs. For the first time, our findings demonstrated that fucoidan inhibits CSC formation and provides evidence as potential anticancer therapy.


Sargassum , Urinary Bladder Neoplasms , Humans , Epithelial-Mesenchymal Transition , Sargassum/chemistry , Urinary Bladder Neoplasms/metabolism , Urinary Bladder , Cell Line, Tumor , Neoplastic Stem Cells/pathology , Polysaccharides/pharmacology , Polysaccharides/metabolism
4.
J Oral Pathol Med ; 51(9): 791-800, 2022 Oct.
Article En | MEDLINE | ID: mdl-35998229

BACKGROUND: The chemokine network orchestrates the cancer stem-like property and consequently participates in cancer progression. CXCR3 contributes cancer progressive property and immunomodulation in the tumor microenvironment. The two major isoforms of CXCR3 are scrutinized and the divergence is showed that CXCR3A promotes cancer cell growth and motility while CXCR3B functions contrarily in many studies. However, rare studies illustrate the role of CXCR3 isoforms in cancer stem-like property and chemoresistance, especially in head and neck cancer (HNC). METHODS: Levels of CXCR3, CXCR3B, and Sox2 were determined in HNC tissue microarray by immunohistochemistry staining to explore potential clinical relevance. Lentivirus-mediated CXCR3-isoform overexpression with MTS assay, clonogenic assay, transwell migration, sphere formation, and chemo-drug susceptibility were implemented to investigate the role of CXCR3-isofoms in HNC. RESULTS: High levels of CXCR3 were significantly associated with advanced stage (p < 0.01), regional lymph node metastasis (p < 0.05), and poor differentiation (p < 0.005) and further correlated with worse survival rate in oral cancer patients (p = 0.036). Higher levels of CXCR3B were found in regional lymphatic invasion of HNC and progressive stage of squamous cell carcinoma. Elevated Sox2 expression was significantly associated with the advanced stage of HNC in the oral cavity, and demonstrated a co-expression pattern with CXCR3B. Furthermore, lentivirus-mediated overexpression of CXCR3A and CXCR3B in SAS human oral cancer cells promoted cell mobility. CXCR3A overexpression enhanced sphere-forming ability and chemoresistance of CSCs by upregulating stemness-related genes. CONCLUSION: This study first provides a novel insight of CXCR3 isoform A in HNC cancer progression via regulating cancer stem-like properties and chemoresistance, suggesting that CXCR3A may be a prognostic marker and novel target for HNC therapy.


Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Carcinoma, Squamous Cell/metabolism , Drug Resistance, Neoplasm/genetics , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Protein Isoforms , Receptors, CXCR3/genetics , Tumor Microenvironment
5.
Front Cell Dev Biol ; 8: 586807, 2020.
Article En | MEDLINE | ID: mdl-33195241

The death receptor Fas can induce cell death through the extrinsic pathway of apoptosis in a variety of cells, including developing thymocytes. Although Fas-induced cell death has been researched and modeled extensively, most of the studies have been done in vitro because of the lethality of Fas triggering in vivo. Thus, little is known about the time line of this type of cell death in vivo, specifically, how does the presence of macrophages and pro-survival cytokines affect apoptosis progression. In addition, although the sequence and timing of events during intrinsic pathway activation in thymocytes in situ have been described, no corresponding data for the extrinsic pathway are available. To address this gap in our knowledge, we established a novel system to study Fas-induced thymocyte cell death using tissue explants. We found that within 1 h of Fas ligation, caspase 3 was activated, within 2 h phosphatidylserine was externalized to serve as an "eat-me" signal, and at the same time, we observed signs of cell loss, likely due to efferocytosis. Both caspase 3 activation and phosphatidylserine exposure were critical for cell loss. Although Fas ligand (FasL) was delivered simultaneously to all cells, we observed significant variation in the entry into the cell death pathway. This model also allowed us to revisit the role of Fas in negative selection, and we ruled out an essential part for it in the deletion of autoreactive thymocytes. Our work provides a timeline for the apoptosis-associated events following Fas triggering in situ and confirms the lack of involvement of Fas in the negative selection of thymocytes.

6.
Sleep Med ; 72: 20-27, 2020 08.
Article En | MEDLINE | ID: mdl-32540633

OBJECTIVE: Due to the complexity of obstructive sleep apnea syndrome (OSAS), engaging patients in the right treatment poses a constant challenge. A novel oral pressure therapy device, the intermittent negative air pressure Sleep Therapy System (iNAP), has proven to ameliorate respiratory events for OSAS patients. However, the mode of action and the characteristics of its responders are not yet fully understood. Therefore, we have first disclosed the mechanism and provided systemic models to predict the treatment response. METHODS: Series of imaging studies were carried out to differentiate the anatomical features of iNAP responders versus non-responders. Compatible electroencephalography was used to evaluate sleep status during magnetic resonance imaging (MRI) assessments. RESULTS: The upper airway volume was statistically widened under the iNAP treatment while patients were naturally asleep (p < 0.05). Negative predictors included several parameters related to oral-tissue redundancy, enlarged middle pharyngeal space, and longer distance of hyoidale to mandibular plane. Positive predictors included larger angulation of sella-articulate-gonion, longer distance of anterior nasal spine to posterior nasal spine, and elongated tongue, which could correspond to the fact that the iNAP had a greater ability to widen the retropalatal region. Furthermore, algorithms developed by these predictors were built to predict treatment response. CONCLUSIONS: We were able to confirm the effect of the iNAP in widening the upper airway. Anatomic features that can be visually observed or obtained through X-ray films, accompanied with the resulting algorithms, were provided to facilitate physicians' ability to predict patients' treatment response to the iNAP with greater sensitivity and efficiency.


Sleep Apnea, Obstructive , Air Pressure , Cephalometry , Humans , Pharynx , Polysomnography , Sleep Apnea, Obstructive/therapy , Tongue
7.
Biomed Res Int ; 2020: 8959726, 2020.
Article En | MEDLINE | ID: mdl-32462028

In both mouse models and clinical patients with lupus, autophagy levels were significantly elevated and correlated with disease activity. Furthermore, autophagy can promote the survival of B and T cells, plasma cell differentiation, and antibody production. These results suggest that autophagy may promote the progression of lupus by regulating the survival of autoreactive immune cells. Therefore, we aimed at studying whether suppressing autophagy can modulate lupus progression in vivo. First, we found that the autophagy levels in splenocytes and lymphocytes of peripheral blood (PB) were elevated and positively correlated with disease severity in lupus-prone mice. The shAtg5-lentivirus, which effectively inhibits autophagy in vitro, was then injected into the lupus-prone mice. Autophagy levels in lymph node cells and PB lymphocytes were reduced following Atg5 suppression. We also found that lymphadenopathy and the numbers of plasma cells, CD4-CD8-, and CD4+ T cells decreased in mice treated with the shAtg5-lentivirus. The mice treated with shAtg5-lentivirus exhibited lower levels of proteinuria, serum anti-dsDNA antibody, B-cell activating factor (BAFF), and glomerular immune complex deposition. Therefore, targeting autophagy to moderate overactivated autophagy in immune cells seems to be a novel strategy for combination therapy of lupus.


Autophagy-Related Protein 5 , Autophagy/genetics , Lupus Erythematosus, Systemic , RNA, Small Interfering , Animals , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spleen/cytology
8.
J Cell Mol Med ; 24(2): 1993-2003, 2020 01.
Article En | MEDLINE | ID: mdl-31856355

These days, cancer can still not be effectively cured because cancer cells readily develop resistance to anticancer drugs. Therefore, an effective combination of drugs with different mechanisms to prevent drug resistance has become a very important issue. Furthermore, the BH3-only protein BNIP3 is involved in both apoptotic and autophagic cell death. In this study, lung cancer cells were treated with a chemotherapy drug alone or in combination to identify the role of BNIP3 and autophagy in combination chemotherapy for treating cancer. Our data revealed that various combinational treatments of two drugs could increase cancer cell death and cisplatin in combination with rapamycin or LBH589, which triggered the cell cycle arrest at the S phase. Cells with autophagosome and pEGFP-LC3 puncta increased when treated with drugs. To confirm the role of autophagy, cancer cells were pre-treated with the autophagy inhibitor 3-methyladenine (3-MA). 3-MA sensitized cancer cells to chemotherapy drug treatments. These results suggest that autophagy may be responsible for cell survival in combination chemotherapy for lung cancer. Moreover, BNIP3 was induced and localized in mitochondria when cells were treated with drugs. The transfection of a dominant negative transmembrane deletion construct of BNIP3 (BNIP3ΔTM) and treatment of a reactive oxygen species (ROS) inhibitor suppressed chemo drug-induced cell death. These results indicate that BNIP3 and ROS may be involved in combination chemo drug-induced cell death. However, chemo drug-induced autophagy may protect cancer cells from drug cytotoxicity. As a result, inhibiting autophagy may improve the effects of combination chemotherapy when treating lung cancer.


Autophagy , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Membrane Proteins/metabolism , Platinum/therapeutic use , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , A549 Cells , Adenine/analogs & derivatives , Adenine/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Cell Death/drug effects , Drug Therapy, Combination , Humans , Lung Neoplasms/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Models, Biological , Platinum/pharmacology , Protein Transport/drug effects
9.
Microb Biotechnol ; 12(5): 920-931, 2019 09.
Article En | MEDLINE | ID: mdl-31199579

Polyketides are important secondary metabolites, many of which exhibit potent pharmacological applications. Biosynthesis of polyketides is carried out by a single polyketide synthase (PKS) or multiple PKSs in successive elongations of enzyme-bound intermediates related to fatty acid biosynthesis. The polyketide gene PKS306 from Pseudallescheria boydii NTOU2362 containing domains of ketosynthase (KS), acyltransferase (AT), dehydratase (DH), acyl carrier protein (ACP) and methyltransferase (MT) was cloned in an attempt to produce novel chemical compounds, and this PKS harbouring green fluorescent protein (GFP) was expressed in Saccharomyces cerevisiae. Although fluorescence of GFP and fusion protein analysed by anti-GFP antibody were observed, no novel compound was detected. 6-methylsalicylic acid synthase (6MSAS) was then used as a template and engineered with PKS306 by combinatorial fusion. The chimeric PKS containing domains of KS, AT, DH and ketoreductase (KR) from 6MSAS with ACP and MT from PKS306 demonstrated biosynthesis of a novel compound. The compound was identified with a deduced chemical formula of C7 H10 O3 , and the chemical structure was named as 2-hydroxy-2-(propan-2-yl) cyclobutane-1,3-dione. The novel compound synthesized by the chimeric PKS in this study demonstrates the feasibility of combinatorial fusion of PKS genes to produce novel polyketides.


Acyl Carrier Protein/metabolism , Acyltransferases/metabolism , Ligases/metabolism , Methyltransferases/metabolism , Multienzyme Complexes/metabolism , Oxidoreductases/metabolism , Polyketide Synthases/metabolism , Polyketides/metabolism , Pseudallescheria/enzymology , Recombinant Fusion Proteins/metabolism , Acyl Carrier Protein/genetics , Acyltransferases/genetics , Cloning, Molecular , Gene Expression , Ligases/genetics , Methyltransferases/genetics , Multienzyme Complexes/genetics , Oxidoreductases/genetics , Polyketide Synthases/genetics , Pseudallescheria/genetics , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
10.
Sleep Breath ; 23(3): 849-856, 2019 Sep.
Article En | MEDLINE | ID: mdl-30690676

PURPOSE: Patients with obstructive sleep apnea syndrome (OSAS) have difficulties in compliance with continuous positive airway pressure (CPAP) and the treatment outcome is heterogeneous. We proposed a proof-of-concept study of a novel intermittent negative air pressure (iNAP®) device for physicians to apply on patients who have failed or refused to use CPAP. METHODS: The iNAP® device retains the tongue and the soft palate in a forward position to decrease airway obstruction. A full nightly usage with the device was evaluated with polysomnography. Subgrouping by baseline apnea-hypopnea index (AHI) and body mass index (BMI) with different treatment response criteria was applied to characterize the responder group of this novel device. RESULTS: Thirty-five patients were enrolled: age 41.9 ± 12.2 years (mean ± standard deviation), BMI 26.6 ± 4.3 kg/m2, AHI 41.4 ± 24.3 events/h, and oxygen desaturation index (ODI) 40.9 ± 24.4 events/h at baseline. AHI and ODI were significantly decreased (p < 0.001) by the device. Patients with moderate OSAS, with baseline AHI between 15 to 30 events/h, achieved 64% response rate; and non-obese patients, with BMI below 25 kg/m2, achieved 57% response rate, with response rate defined as 50% reduction in AHI from baseline and treated AHI lower than 20. There were minimal side effects reported. CONCLUSIONS: In a proof-of-concept study, the device attained response to treatment as defined, in more than half of the moderate and non-obese OSAS patients, with minimal side effects.


Patient Compliance , Sleep Apnea, Obstructive/therapy , Ventilators, Negative-Pressure/statistics & numerical data , Adult , Continuous Positive Airway Pressure , Female , Humans , Male , Middle Aged , Polysomnography , Sleep Apnea, Obstructive/prevention & control , Treatment Outcome
11.
J Cell Mol Med ; 22(12): 5909-5918, 2018 12.
Article En | MEDLINE | ID: mdl-30246456

Although targeted therapy is usually the first-line treatment for advanced renal cell carcinoma (RCC), some patients can experience drug resistance. Cancer stem cells are tumour-initiating cells that play a vital role in drug resistance, metastasis and cancer relapse, while galectins (Gal) participate in tumour progression and drug resistance. However, the exact role of galectins in RCC stemness is yet unknown. In this study, we grew a subpopulation of RCC cells as tumour spheres with higher levels of stemness-related genes, such as Oct4, Sox2 and Nanog. Among the Gal family, Gal-3 in particular was highly expressed in RCC tumour spheres. To further investigate Gal-3's role in the stemness of RCC, lentivirus-mediated knockdown and overexpression of Gal-3 in RCC cells were used to examine both in vitro and in vivo tumorigenicity. We further assessed Gal-3 expression in RCC tissue microarray using immunohistochemistry. Upon suppressing Gal-3 in parental RCC cells, invasion, colony formation, sphere-forming ability, drug resistance and stemness-related gene expression were all significantly decreased. Furthermore, CXCL6, CXCL7 and CXCR2 were down-regulated in Gal-3-knockdown tumour spheres, while CXCR2 overexpression in Gal-3-knockdown RCC restored the ability of sphere formation. Gal-3 overexpression in RCC promoted both in vitro and in vivo tumorigenicity, and its expression was correlated with CXCR2 expression and tumour progression in clinical tissues. RCC patients with higher co-expressions of Gal-3 and CXCR2 demonstrated a worse survival rate. These results indicate that highly expressed Gal-3 may up-regulate CXCR2 to augment RCC stemness. Gal-3 may be a prognostic and innovative target of combined therapy for treating RCC.


Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Galectin 3/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, Interleukin-8B/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement , Cell Self Renewal , Chemokines/metabolism , Disease Progression , Down-Regulation , Drug Resistance, Neoplasm , Humans , Male , Mice, Inbred NOD , Mice, SCID , Prognosis , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Stem Cell Assay
12.
J Autoimmun ; 78: 92-100, 2017 03.
Article En | MEDLINE | ID: mdl-28089248

Sensing of nucleic acids by pattern recognition receptors is the key for the initiation and development of systemic lupus erythematosus (SLE). Triggering receptor expressed on myeloid cells-1 (TREM-1) is a novel innate immune receptor, which can amplify Toll-like receptor (TLR)-induced inflammatory responses. Although patients with lupus exhibit increased serum levels of soluble TREM-1 (sTREM-1), the role of TREM-1 in SLE remains unknown. In current study, we found serum sTREM-1 levels were significantly increased in lupus patients and positively correlated with disease activity. Additionally, diseased B6.lpr mice had elevated TREM-1 in the serum, spleen, and lymph nodes. To investigate the role of TREM-1 in lupus, we established Trem-1-/-.lpr mice. Trem-1-/-.lpr mice exhibited lower survival rates and more severe lupus symptoms, including elevated proteinuria, serum anti-dsDNA antibody levels, renal immune complex depositions and lymphocyte subpopulation expansions in both the spleen and lymph nodes. Besides, Trem-1-/-.lpr mice expressed higher serum B cell-activating factor (BAFF) levels and lymph node dendritic cells (DCs) were the major source of increased BAFF. Activation of membrane-bound TREM-1 could suppress TLR9-induced BAFF expression in bone marrow-derived DCs of B6.lpr mice. Moreover, levels of sTREM-1, which could act as an antagonist of membrane-bound TREM-1, were positively correlated with levels of BAFF in the sera of lupus patients. Our findings suggest a novel modulatory role of TREM-1 in the pathogenesis of SLE. sTREM-1 production is a useful diagnostic marker and a molecular target for combination therapy of lupus.


B-Cell Activating Factor/biosynthesis , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/deficiency , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation , Humans , Lupus Erythematosus, Systemic/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Knockout , Middle Aged , Mutation , Organ Specificity , Severity of Illness Index , Triggering Receptor Expressed on Myeloid Cells-1/blood , Triggering Receptor Expressed on Myeloid Cells-1/genetics , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Young Adult
13.
J Cell Mol Med ; 20(11): 2020-2028, 2016 11.
Article En | MEDLINE | ID: mdl-27297979

Within the tumour microenvironment, a complex network of chemokines and their receptors affects the initiation and progression of tumours. The higher levels of tumour necrosis factor-alpha (TNF-α) are associated with tumour progression and an anti-TNF-α monoclonal antibody has been used successfully to treat patients with renal cell carcinoma (RCC). However, the role of chemokines and their receptors in the TNF-α-promoted progression of RCC remains unclear. In this study, TNF-α was found to enhance the migration, invasion and epithelial-mesenchymal transition (EMT) of RCC cells. To further investigate the molecular mechanism of TNF-α on the progression of RCC, reverse transcription and quantitative PCR was used to screen chemokines and chemokine receptors that were associated with tumorigenesis. The results showed that TNF-α significantly increased the expressions of CXCR2 and CXCR3 and their related ligands in RCC cells. Subsequently, we used a lentiviral shRNA system to knockdown the expression of CXCR2 and/or CXCR3 in RCC cells. CXCR2 and CXCR3 silencing inhibited the induction of Slug and ZEB-1 with TNF-α treatment of RCC cells. In addition, the knockdown of both CXCR2 and CXCR3 resulted in a greater decrease in cell migration, invasion and clonogenic ability compared with either CXCR2 or CXCR3 knockdown alone. Moreover, CXCR2 and CXCR3 silencing significantly reduced the sphere-forming ability of RCC cells. High expression levels of CXCR2 and CXCR3 in cancer tissues correlated with tumour progression of renal cell carcinoma. These findings suggest that TNF-α augments CXCR2 and CXCR3 to promote the progression of renal cell carcinoma leading to a poor prognosis.


Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Disease Progression , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Receptors, CXCR3/metabolism , Receptors, CXCR4/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Chemokines/metabolism , Clone Cells , Epithelial-Mesenchymal Transition/drug effects , Gene Knockdown Techniques , Gene Silencing/drug effects , Humans , Neoplasm Invasiveness , Reproducibility of Results , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
14.
Neurosci Lett ; 628: 186-93, 2016 08 15.
Article En | MEDLINE | ID: mdl-27345388

Proinflammatory responses eliciting the microglial production of cytokines and nitric oxide (NO) have been reported to play a crucial role in the acute and chronic pathogenic effects of neurodegeneration. Chemical inhibitors of cyclin-dependent kinases (CDKs) may prevent the progression of neurodegeneration by both limiting cell proliferation and reducing cell death. However, the mechanism underlying the protective effect of CDK inhibitors on microglia remains unexplored. In this study, we found that olomoucine, a CDK inhibitor, alleviated lipopolysaccharide (LPS)-induced BV2 microglial cell death by reducing the generation of NO and inhibiting the gene expression of proinflammatory cytokines. In addition, olomoucine reduced inducible NO synthase promoter activity and alleviated NF-κB- and E2F-mediated transcriptional activation. NO-induced cell death involved mitochondrial disruptions such as cytochrome c release and loss of mitochondrial membrane potential, and pretreatment with olomoucine prior to NO exposure reduced these disruptions. Microarray analysis revealed that olomoucine treatment induced prominent down-regulation of Bcl2/adenovirus E1B 19-kDa-interacting protein 3 (BNIP3), a pro-apoptotic Bcl-2 family protein that is involved in mitochondrial disruption. As BNIP3 knock-down significantly increased the viability of LPS- and NO-treated BV2 cells, we conclude that olomoucine may protect cells by limiting proinflammatory responses, thereby reducing NO generation. Simultaneously, down-regulation of BNIP3 prevents NO stimulation from inducing mitochondrial disruption.


Apoptosis/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Inflammation/metabolism , Kinetin/administration & dosage , Membrane Proteins/metabolism , Microglia/drug effects , Mitochondrial Proteins/metabolism , Nitric Oxide/metabolism , Animals , Cell Line , Cell Proliferation , Down-Regulation , E2F Transcription Factors/metabolism , Enzyme Inhibitors/administration & dosage , Inflammation/chemically induced , Inflammation Mediators/metabolism , Lipopolysaccharides , Mice , Microglia/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism
15.
Oncotarget ; 6(7): 4936-52, 2015 Mar 10.
Article En | MEDLINE | ID: mdl-25669973

Cancer stem cells (CSCs) are comprised of a rare sub-population of cells in tumors that have been proposed to be responsible for high recurrence rates and resistance to chemotherapy. Galectins are highly expressed in cancers that correlate with the aggressiveness of tumors. Galectins may also promote the resistance of cancer cells to chemotherapy. However, the role of galectins in CSCs remains unknown. In this study, sphere formation was used to enrich H1299 human lung CSCs that had self-renewal ability, advanced tumorigenic potential, and that highly expressed stem/progenitor cell markers such as Oct4, Sox2, Nanog, and CD133. A novel candidate molecule, galectin-3, for stemness was found in lung CSCs. The expression of galectin-3 robustly increased in lung cancer spheres over serial passages, but its suppression in the H1299 monolayer or spheres resulted in reduced expression of stemness-related genes, sphere-forming ability, tumorigenicity, chemoresistance, and tumor initiation in mice. Notably, the overexpression of galectin-3 in A549 lung cancer cells, which have low capability to grow as tumor spheres, promoted CSC formation. ß-catenin activity was increased in H1299 spheres and counteracted by galectin-3 suppression. Thus, galectin-3 may act as a cofactor by interacting with ß-catenin to augment the transcriptional activities of stemness-related genes. Furthermore, galectin-3 expression correlated with tumor progression and expressions of ß-catenin and CSC marker CD133 in lung cancer tissues. Targeting galectin-3 signaling may provide a new strategy for lung cancer treatment by inhibiting stem-like properties.


Galectin 3/metabolism , Lung Neoplasms/metabolism , beta Catenin/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogens/analysis , Carcinogens/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Galectin 3/analysis , HEK293 Cells , Heterografts , Humans , Immunohistochemistry , Lung Neoplasms/chemistry , Lung Neoplasms/pathology , Mice, Inbred NOD , Mice, SCID , Tissue Array Analysis , beta Catenin/analysis
16.
Exp Biol Med (Maywood) ; 240(4): 498-507, 2015 Apr.
Article En | MEDLINE | ID: mdl-25519430

A feature of allergic airway disease is the observed increase of nitric oxide (NO) in exhaled breath. Gram-negative bacterial infections have also been linked with asthma exacerbations. However, the role of NO in asthma exacerbations with gram-negative bacterial infections is still unclear. In this study, we examined the role of NO in lipopolysaccharide (LPS)-induced inflammation in an ovalbumin (OVA)-challenged mouse asthma model. To determine whether NO affected the LPS-induced response, a NO donor (S-nitroso-N-acetylpenicillamine, SNAP) or a selective inhibitor of NO synthase (1400W) was injected intraperitoneally into the mice before the LPS stimulation. Decreased levels of proinflammatory cytokines were demonstrated in the bronchoalveolar lavage fluid from mice treated with SNAP, whereas increased levels of cytokines were found in the 1400W-treated mice. To further explore the molecular mechanism of NO-mediated inhibition of proinflammatory responses in macrophages, RAW 264.7 cells were treated with 1400W or SNAP before LPS stimulation. LPS-induced inflammation in the cells was attenuated by the presence of NO. The LPS-induced IκB kinase (IKK) activation and the expression of IKK were reduced by NO through attenuation of the interaction between Hsp90 and IKK in the cells. The IKK decrease in the lung immunohistopathology was verified in SNAP-treated asthma mice, whereas IKK increased in the 1400W-treated group. We report for the first time that NO attenuates the interaction between Hsp90 and IKK, decreasing the stability of IKK and causing the down-regulation of the proinflammatory response. Furthermore, the results suggest that NO may repress LPS-stimulated innate immunity to promote pulmonary bacterial infection in asthma patients.


Asthma/prevention & control , HSP90 Heat-Shock Proteins/physiology , I-kappa B Kinase/physiology , Inflammation/prevention & control , Nitric Oxide/pharmacology , Nitric Oxide/therapeutic use , Signal Transduction/physiology , Animals , Asthma/chemically induced , Asthma/physiopathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Imines/pharmacology , Imines/therapeutic use , Inflammation/chemically induced , Inflammation/physiopathology , Lipopolysaccharides/adverse effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase/antagonists & inhibitors , Ovalbumin/adverse effects , S-Nitroso-N-Acetylpenicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine/therapeutic use , Signal Transduction/drug effects
17.
Mol Carcinog ; 54(3): 167-77, 2015 Mar.
Article En | MEDLINE | ID: mdl-24115089

The deregulation of epigenetics was involved in early and subsequent carcinogenic events. Reversing cancer epigenetics to restore a normal epigenetic condition could be a rational approach for cancer treatment and specialized prevention. In the present study, we found that the expression levels of two epigenetic markers, histone H3K27 trimethylation (H3K27me3), was low but histone H3S10 phosphorylation (pH3Ser10) was high in human bladder cancer tissues, which showed opposite expression patterns in their normal counterparts. Thus, we investigated whether a natural product, emodin, has the ability to reverse these two epigenetic modifications and inhibit bladder cancer cell growth. Emodin significantly inhibited the cell growth of four bladder cancer cell lines in a dose- and time-dependent manner. Emodin treatment did not induce specific cell cycle arrest, but it altered epigenetic modifications. Emodin treatment resulted in the suppression of pH3Ser10 and increased H3K27me3, contributing to gene silencing in bladder cancer cells. Microarray analysis demonstrated that oncogenic genes including fatty acid binding protein 4 (FABP4) and fibroblast growth factor binding protein 1 (HBP17), RGS4, tissue inhibitor of metalloproteinase 3 (TIMP3), WNT5b, URB, and collagen, type VIII, alpha 1 (COL8A1) responsible for proliferation, survival, inflammation, and carcinogenesis were significantly repressed by emodin. The ChIP assays also showed that emodin increased H3K27me3 but decreased pH3Ser10 modifications on the promoters of repressed genes, which indicate that emodin reverses the cancer epigenetics towards normal epigenetic situations. In conclusion, our work demonstrates the significant anti-neoplastic activity of emodin on bladder cancer cells and elucidates the novel mechanisms of emodin-mediated epigenetic modulation of target genes. Our study warrants further investigation of emodin as an effective therapeutic or preventive agent for bladder cancer.


Emodin/pharmacology , Epigenesis, Genetic/drug effects , Protein Kinase Inhibitors/pharmacology , Urinary Bladder Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , DNA Methylation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histones/genetics , Histones/metabolism , Humans , Phosphorylation , Promoter Regions, Genetic/drug effects , Tumor Cells, Cultured , Urothelium/cytology , Urothelium/metabolism
18.
Exp Biol Med (Maywood) ; 239(6): 715-23, 2014 Jun.
Article En | MEDLINE | ID: mdl-24719374

The hallmark of systemic lupus erythematosus (SLE) is the presence of high levels of anti-double-stranded DNA autoantibody (anti-dsDNA) in sera. In addition, pathogen infections coincide frequently with the occurrence of lupus. Our study was designed to investigate the contribution of anti-dsDNA, extracellular and intracellular Toll-like receptors (TLRs), a family of pattern-recognition receptors for sensing invading pathogens, in the pathogenesis of lupus. Although cell surface-expressed TLR4 may promote lupus progression, intracellular nucleic acid-sensing TLR9 plays either stimulatory or protective roles in different murine lupus models. To examine the role of TLR4, TLR9, and anti-dsDNA in SLE, we generated transgenic mice carrying anti-dsDNA antibody transgene and challenged the mice with TLR4- and TLR9-agonists, lipopolysaccharides (LPS), and CpG oligodeoxynucleotide (CpG ODN1826 and 2216), respectively. Splenocytes from these mice were found to secrete higher levels of interleukin-10 (IL-10) and anti-dsDNA when treated with a combination of TLR4 and TLR9 agonists (LPS + CpG). In addition, the transgenic mice were intraperitoneally administered with CpG or combined CpG and LPS to determine whether extracellular TLR4 and intracellular TLR9 activations could affect lupus progression in vivo. It was found that serum levels of anti-dsDNA antibodies and interferon-alpha were higher in CpG + LPS-treated transgenic mice than those in non-transgenic mice. Besides, elevated levels of proteinuria, blood urine nitrogen, and immune complex depositions in kidney were found in treated transgenic mice. Anti-dsDNA and simultaneous activation of surface-expressed TLR4 and endosomal TLR9 are crucial to promote the lupus progression.


Antibodies, Antinuclear/metabolism , Endosomes/metabolism , Gene Expression Regulation , Lupus Erythematosus, Systemic/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Animals , Antibodies, Antinuclear/genetics , Endosomes/genetics , Lipopolysaccharides/pharmacology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Transgenic , Oligodeoxyribonucleotides/pharmacology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics
19.
J Am Soc Nephrol ; 25(7): 1486-95, 2014 Jul.
Article En | MEDLINE | ID: mdl-24511119

Galectin-1, a ß-galactoside-binding lectin, is involved in many physiologic and pathologic processes, including cell adhesion, differentiation, angiogenesis, and tumor progression. However, the role of galectin-1 in kidney cancer remains elusive. This study evaluated the role of galectin-1 in the progression and clinical prognosis of renal cell carcinoma. We found significant overexpression of galectin-1 in both kidney cancer cell lines and metastatic tissue specimens from patients with renal cell carcinoma. Knockdown of galectin-1 gene expression in renal cancer cell lines reduced cell invasion, clonogenic ability, and epithelial-mesenchymal transition in vitro; reduced tumor outgrowth in vivo; and inhibited the angiogenesis-inducing activity of these cells in vitro and in vivo. Galectin-1 knockdown decreased CXCR4 expression levels in kidney cancer cells, and restoration of CXCR4 expression in galectin-1-silenced cells rescued cell motility and clonogenic ability. Additional studies suggested that galectin-1 induced CXCR4 expression through activation of nuclear factor-κB (NF-κB). Analysis of patient specimens confirmed the clinical significance and positive correlation between galectin-1 and CXCR4 expression levels and revealed concomitant overexpression of galectin-1 and CXCR4 associated adversely with overall and disease-free survival. Our findings suggest that galectin-1 promotes tumor progression through upregulation of CXCR4 via NF-κB. The coordinated upregulation of galectin-1 and CXCR4 may be a novel prognostic factor for survival in patients with renal cell carcinoma and the galectin-1-CXCR4 axis may serve as a therapeutic target in this disease.


Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Galectin 1/physiology , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Receptors, CXCR4/physiology , Up-Regulation , Disease Progression , Humans , Prognosis , Survival Rate , Tumor Cells, Cultured
20.
PLoS One ; 8(9): e73401, 2013.
Article En | MEDLINE | ID: mdl-24023871

Histone deacetylase inhibitors (HDACIs) have potent anti-cancer activity in a variety of cancer models. Understanding the molecular mechanisms involved in the therapeutic responsiveness of HDACI is needed before its clinical application. This study aimed to determine if a potent HDACI, LBH589 (Panobinostat), had differential therapeutic responsiveness towards LNCaP and PC-3 prostate cancer (PCa) cells. The former showed prometaphase arrest with subsequent apoptosis upon LBH589 treatment, while the latter was less sensitive and had late G2 arrest. The LBH589 treatment down-regulated HDAC6 and sustained ERK activation, and contributed to prometaphase arrest. Mechanistically, LBH589 inhibited HDAC6 activity, caused its dissociation from protein phosphatase PP1α, and increased 14-3-3ζ acetylation. Acetylated 14-3-3ζ released its mask effect on serine 259 of c-Raf and serine 216 of Cdc25C subsequent to de-phosphorylation by PP1α, which contributed to ERK activation. Enhanced ERK activity by LBH589 further down-regulated HDAC6 protein levels and sustained ERK activation by free-forward regulation. The sustained Cdc25C and ERK activation resulted in early M-phase (prometaphase) arrest and subsequent apoptosis in the most sensitive LNCaP cells but not in PC-3 cells. This study provides pre-clinical evidence that HDAC6 may serve as a sensitive therapeutic target in the treatment of prostate cancer with HDACI LBH589 for clinical translation. This study also posits a novel mechanism of HDAC6 participation in regulating the c-Raf-PP1-ERK signaling pathway and contributing to M phase cell-cycle transition.


Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Indoles/pharmacology , M Phase Cell Cycle Checkpoints/drug effects , Prostatic Neoplasms/pathology , 14-3-3 Proteins/metabolism , Acetylation/drug effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Panobinostat , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction/drug effects , cdc25 Phosphatases/metabolism
...