Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
J Biomed Sci ; 31(1): 46, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38725007

ABSTRACT

BACKGROUND: Cathepsin S (CTSS) is a cysteine protease that played diverse roles in immunity, tumor metastasis, aging and other pathological alterations. At the cellular level, increased CTSS levels have been associated with the secretion of pro-inflammatory cytokines and disrupted the homeostasis of Ca2+ flux. Once CTSS was suppressed, elevated levels of anti-inflammatory cytokines and changes of Ca2+ influx were observed. These findings have inspired us to explore the potential role of CTSS on cognitive functions. METHODS: We conducted classic Y-maze and Barnes Maze tests to assess the spatial and working memory of Ctss-/- mice, Ctss+/+ mice and Ctss+/+ mice injected with the CTSS inhibitor (RJW-58). Ex vivo analyses including long-term potentiation (LTP), Golgi staining, immunofluorescence staining of sectioned whole brain tissues obtained from experimental animals were conducted. Furthermore, molecular studies were carried out using cultured HT-22 cell line and primary cortical neurons that treated with RJW-58 to comprehensively assess the gene and protein expressions. RESULTS: Our findings reported that targeting cathepsin S (CTSS) yields improvements in cognitive function, enhancing both working and spatial memory in behavior models. Ex vivo studies showed elevated levels of long-term potentiation levels and increased synaptic complexity. Microarray analysis demonstrated that brain-derived neurotrophic factor (BDNF) was upregulated when CTSS was knocked down by using siRNA. Moreover, the pharmacological blockade of the CTSS enzymatic activity promoted BDNF expression in a dose- and time-dependent manner. Notably, the inhibition of CTSS was associated with increased neurogenesis in the murine dentate gyrus. These results suggested a promising role of CTSS modulation in cognitive enhancement and neurogenesis. CONCLUSION: Our findings suggest a critical role of CTSS in the regulation of cognitive function by modulating the Ca2+ influx, leading to enhanced activation of the BDNF/TrkB axis. Our study may provide a novel strategy for improving cognitive function by targeting CTSS.


Subject(s)
Brain-Derived Neurotrophic Factor , Cathepsins , Cognition , Animals , Male , Mice , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Cathepsins/drug effects , Cathepsins/genetics , Cathepsins/metabolism , Cognition/drug effects , Cognition/physiology , Mice, Knockout , Receptor, trkB/metabolism , Receptor, trkB/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
2.
Pharmaceuticals (Basel) ; 17(1)2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38256960

ABSTRACT

The use of tocilizumab against the interleukin-6 receptor (IL-6R) has been demonstrated as inhibiting the progression of diverse cancers in vitro and in vivo. Nonetheless, evidence regarding the anti-tumor effects of tocilizumab on human colorectal carcinoma (CRC) corresponding to IL-6R expression levels remains scarce. To investigate the influence of IL-6R expression, SW480 and HT-29 cells inoculated subcutaneously into NU/NU mice were used as human CRC xenograft models with anti-IL-6R antibody (tocilizumab) therapy. The IL-6R expression levels, histology of CRC growth/invasiveness, and tumor growth-related signaling pathway were estimated by H&E and immunohistochemical staining. SW480 tumor cells with higher IL-6R expression levels showed better responsiveness in tocilizumab therapy than in the treated HT-29 group. Likewise, therapeutic effects of tocilizumab on the proliferative ability with mitotic index and Ki-67 expressions, invasiveness with MMP-9 proteinase expressions, and ERK 1/2 and STAT3 signaling transduction in the SW480 treatment group were superior to the HT-29 treatment group. In light of our results, IL-6R is the key indicator for the efficacy of tocilizumab treatment in CRC xenografts. From the perspective of precision medicine, tumor response to anti-IL-6R antibody therapy could be predicted on the basis of IL-6R expression levels. In this manner, tocilizumab may serve as a targeted and promising anti-CRC therapy.

4.
Curr Issues Mol Biol ; 45(6): 4908-4922, 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37367061

ABSTRACT

BACKGROUND/AIM: Colorectal cancer (CRC) is the third most common cancer with a high mortality rate worldwide. Although gallic acid and hesperidin exert anticancer activity, synergistic effects of gallic acid and hesperidin against CRC remain elusive. This study aims to investigate the therapeutic mechanism of a novel combination of gallic acid and hesperidin against CRC cell growth, including cell viability, cell-cycle-associated proteins, spheroid formation, and stemness. METHODS: Gallic acid and hesperidin derived from Hakka pomelo tea (HPT) were detected by colorimetric methods and high-performance liquid chromatography using ethyl acetate as an extraction medium. CRC cell lines (HT-29 and HCT-116) treated with the combined extract were investigated in our study for cell viability (trypan blue or soft agar colony formation assay), cell cycle (propidium iodide staining), cell-cycle-associated proteins (immunoblotting), and stem cell markers (immunohistochemistry staining). RESULTS: Compared with other extraction methods, HPT extraction using an ethyl acetate medium exerts the most potent effect on inhibiting HT-29 cell growth in a dose-dependent manner. Furthermore, the treatment with combined extract had a higher inhibitory effect on CRC cell viability than gallic acid or hesperidin alone. The underlying mechanism was involved in G1-phase arrest and Cip1/p21 upregulation that could attenuate HCT-116 cell proliferation (Ki-67), stemness (CD-133), and spheroid growth in a 3D formation assay mimicking in vivo tumorigenesis. CONCLUSION: Gallic acid and hesperidin exert synergistic effects on cell growth, spheroids, and stemness of CRC and may serve as a potential chemopreventive agent. Further testing for the safety and effectiveness of the combined extract in large-scale randomized trials is required.

5.
Cells ; 11(18)2022 09 09.
Article in English | MEDLINE | ID: mdl-36139397

ABSTRACT

On arrested neutrophils a focal adhesive cluster of ~200 high affinity (HA) ß2-integrin bonds under tension is sufficient to trigger Ca2+ flux that signals an increase in activation in direct proportion to increments in shear stress. We reasoned that a threshold tension acting on individual ß2-integrin bonds provides a mechanical means of transducing the magnitude of fluid drag force into signals that enhance the efficiency of neutrophil recruitment and effector function. Tension gauge tethers (TGT) are a duplex of DNA nucleotides that rupture at a precise shear force, which increases with the extent of nucleotide overlap, ranging from a tolerance of 54pN to 12pN. TGT annealed to a substrate captures neutrophils via allosteric antibodies that stabilize LFA-1 in a high- or low-affinity conformation. Neutrophils sheared on TGT substrates were recorded in real time to form HA ß2-integrin bonds and flux cytosolic Ca2+, which elicited shape change and downstream production of reactive oxygen species. A threshold force of 33pN triggered consolidation of HA ß2-integrin bonds and triggered membrane influx of Ca2+, whereas an optimum tension of 54pN efficiently transduced activation at a level equivalent to chemotactic stimulation on ICAM-1. We conclude that neutrophils sense the level of fluid drag transduced through individual ß2-integrin bonds, providing an intrinsic means to modulate inflammatory response in the microcirculation.


Subject(s)
CD18 Antigens , Lymphocyte Function-Associated Antigen-1 , Adhesives , Calcium , Intercellular Adhesion Molecule-1 , Neutrophils , Nucleotides , Reactive Oxygen Species
6.
Am J Respir Cell Mol Biol ; 64(6): 734-746, 2021 06.
Article in English | MEDLINE | ID: mdl-33730527

ABSTRACT

Tobacco smoking is a well-known risk factor for both fibrogenesis and fibrotic progression; however, the mechanisms behind these processes remain enigmatic. RTKs (receptor tyrosine kinases) have recently been reported to drive profibrotic phenotypes in fibroblasts during pulmonary fibrosis (PF). Using a phospho-RTK array screen, we identified the RTK AXL as a top upregulated RTK in response to smoke. Both expression and signaling activity of AXL were indeed elevated in lung fibroblasts exposed to tobacco smoke, whereas no significant change to the levels of a canonical AXL ligand, Gas6 (growth arrest-specific 6), was seen upon smoke treatment. Notably, we found that smoke-exposed human lung fibroblasts exhibited highly proliferative and invasive activities and were capable of inducing fibrotic lung lesions in mice. Conversely, genetic suppression of AXL in smoke-exposed fibroblasts cells led to suppression of AXL downstream pathways and aggressive phenotypes. We further demonstrated that AXL interacted with MARCKS (myristoylated alanine-rich C kinase substrate) and cooperated with MARCKS in regulating downstream signaling activity and fibroblast invasiveness. Pharmacological inhibition of AXL with AXL-specific inhibitor R428 showed selectivity for smoke-exposed fibroblasts. In all, our data suggest that AXL is a potential marker for smoke-associated PF and that targeting of the AXL pathway is a potential therapeutic strategy in treating tobacco smoking-related PF.


Subject(s)
Proto-Oncogene Proteins/metabolism , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Smoking/adverse effects , Animals , Cell Survival , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lung/pathology , Mice, Inbred C57BL , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Phenotype , Pulmonary Fibrosis/pathology , Signal Transduction , Up-Regulation/genetics , Axl Receptor Tyrosine Kinase
7.
Theranostics ; 11(10): 4672-4687, 2021.
Article in English | MEDLINE | ID: mdl-33754020

ABSTRACT

Rationale: Oxaliplatin-induced peripheral neuropathy (OIPN) is a common adverse effect that causes delayed treatment and poor prognosis among colorectal cancer (CRC) patients. However, its mechanism remains elusive, and no effective treatment is available. Methods: We employed a prospective cohort study of adult patients with pathologically confirmed stage III CRC receiving adjuvant chemotherapy with an oxaliplatin-based regimen for investigating OIPN. To further validate the clinical manifestations and identify a potential therapeutic strategy, animal models, and in vitro studies on the mechanism of OIPN were applied. Results: Our work found that (1) consistent with clinical findings, OIPN was observed in animal models. Targeting the enzymatic activity of cathepsin S (CTSS) by pharmacological blockade and gene deficiency strategy alleviates the manifestations of OIPN. (2) Oxaliplatin treatment increases CTSS expression by enhancing cytosol translocation of interferon response factor 1 (IRF1), which then facilitates STIM-dependent store-operated Ca2+ entry homeostasis. (3) The cytokine array demonstrated an increase in anti-inflammatory cytokines and suppression of proinflammatory cytokines in mice treated with RJW-58. (4) Mechanistically, inhibiting CTSS facilitated olfactory receptors transcription factor 1 release from P300/CBP binding, which enhanced binding to the interleukin-10 (IL-10) promoter region, driving IL-10 downstream signaling pathway. (5) Serum CTSS expression is increased in CRC patients with oxaliplatin-induced neurotoxicity. Conclusions: We highlighted the critical role of CTSS in OIPN, which provides a therapeutic strategy for the common adverse side effects of oxaliplatin.


Subject(s)
Cathepsins/genetics , Neurons/metabolism , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cathepsins/antagonists & inhibitors , Cathepsins/drug effects , Chemotherapy, Adjuvant , Cohort Studies , Colorectal Neoplasms/drug therapy , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors , Female , Fluorouracil/therapeutic use , Ganglia, Spinal , Humans , In Vitro Techniques , Leucovorin/therapeutic use , Male , Mice , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Molecular Targeted Therapy , Neural Conduction , Neurons/drug effects , Organoplatinum Compounds/therapeutic use , Oxaliplatin/adverse effects , Oxaliplatin/pharmacology , Peripheral Nervous System Diseases/chemically induced , Prospective Studies
8.
Theranostics ; 11(9): 4122-4136, 2021.
Article in English | MEDLINE | ID: mdl-33754052

ABSTRACT

Rationale: Cigarette smoking is a major risk factor for lung cancer development and progression; however, the mechanism of how cigarette smoke activates signaling pathways in promoting cancer malignancy remains to be established. Herein, we aimed to determine the contribution of a signaling protein, myristoylated alanine-rich C kinase substrate (MARCKS), in smoke-mediated lung cancer. Methods: We firstly examined the levels of phosphorylated MARCKS (phospho-MARCKS) in smoke-exposed human lung cancer cells and specimens as well as non-human primate airway epithelium. Next, the MARCKS-interactome and its gene networks were identified. We also used genetic and pharmacological approaches to verify the functionality and molecular mechanism of smoke-induced phospho-MARCKS. Results: We observed that MARCKS becomes activated in airway epithelium and lung cancer cells in response to cigarette smoke. Functional proteomics revealed MARCKS protein directly binds to NF-κB-activating protein (NKAP). Following MARCKS phosphorylation at ser159 and ser163, the MARCKS-NKAP interaction was inhibited, leading to the activation of NF-κB signaling. In a screen of two cohorts of lung cancer patients, we confirmed that phospho-MARCKS is positively correlated with phospho-NF-κB (phospho-p65), and poor survival. Surprisingly, smoke-induced phospho-MARCKS upregulated the expression of pro-inflammatory cytokines, epithelial-mesenchymal transition, and stem-like properties. Conversely, targeting of MARCKS phosphorylation with MPS peptide, a specific MARCKS phosphorylation inhibitor, suppressed smoke-mediated NF-κB signaling activity, pro-inflammatory cytokines expression, aggressiveness and stemness of lung cancer cells. Conclusion: Our results suggest that phospho-MARCKS is a novel NF-kB activator in smoke-mediated lung cancer progression and provide a promising molecular model for developing new anticancer strategies.


Subject(s)
Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , NF-kappa B/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Smoke/adverse effects , A549 Cells , Animals , Cell Line, Tumor , Cigarette Smoking/adverse effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Humans , Phosphorylation/drug effects , Phosphorylation/physiology , Primates , Signal Transduction/drug effects
9.
J Chin Med Assoc ; 83(8): 774-778, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32433346

ABSTRACT

BACKGROUND: The advancements in neonatal critical care have not only improved the outcomes of extreme prematurity but also prolonged the process of death in terminally ill neonates. This study analyzed the characteristics of neonates who died at a single tertiary center in Taiwan. The utilization of neonatal hospice care before and after the legalization of life-sustaining treatment (LST) withdrawal in Taiwan in 2013 was also compared. METHODS: This study enrolled the neonatal mortality cases in the Taipei Veterans General Hospital during January 2008 to December 2017 through chart review. Data on birth history, primary diagnosis, complications, and death circumstances were recorded and analyzed. RESULTS: In total, 105 neonatal deaths were analyzed. The circumstances of death were as follows: 22 (21%) cases of full LST and cardiopulmonary resuscitation (CPR) performed until death; 63 (60%) cases of LST initiated but no more CPR after do-not-resuscitate (DNR) consents signed; 8 (7.6%) cases of LST withdrawn; 4 (3.8%) cases of DNR signed without LST initiation; 3 (2.9%) cases of CPR not performed, although no DNR signed; and 5 (4.8%) cases of discharge against medical advice under critical condition. The incidence of written DNR consents (57.9% in 2008-2009 vs 93.8% in 2016-2017; p = 0.02) showed an increasing trend. Regarding the incidence of comorbidities, renal failure rate was higher in the DNR group than in the non-DNR group (p = 0.002). CONCLUSION: There was an increasing trend for written DNR consent and the utilization of neonatal hospice care. Renal failure, as a comorbidity, was significantly associated with the written DNR consent in the neonates. Further studies to evaluate the factors associated with neonatal hospice care utilization are suggested.


Subject(s)
Hospice Care , Resuscitation Orders/legislation & jurisprudence , Female , Humans , Infant, Newborn , Male , Retrospective Studies , Taiwan , Tertiary Care Centers
10.
Inflammation ; 43(2): 518-531, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31776889

ABSTRACT

Pinolenic acid (PNA) is a rare n-6 polyunsaturated fatty acid (n-6 PUFA) originally identified in pine seeds. Previous studies demonstrated that PNA and its elongation metabolite, Δ7-eicosatrienoic acid (Δ7-ETrA), exerted an anti-inflammatory effect in cultured cells by suppressing prostaglandin E2 (PGE2) production. The objective of this study was to further examine the in vivo anti-inflammatory properties of PNA. Using human THP-1 macrophage, we first confirmed that incorporation of PNA into cellular phospholipids suppressed the production of interleukin-6 (IL-6) (by 46%), tumor necrosis factor-α (TNF-α) (by 18%), and prostaglandin E2 (PGE2) (by 87%), and the expression of type-2 cyclooxygenase (COX-2) (by 27%). Furthermore, we demonstrated that injection of PNA or Δ7-ETrA suppressed 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced mouse ear edema, as measured by ear thickness (by 15%) and biopsy weight (by up to 29%). Both PUFA also lowered proportions of infiltrated leukocytes, neutrophils, and macrophages using flow cytometric analysis. Topical application of PNA or Δ7-ETrA on mouse back skin suppressed TPA-induced pro-inflammatory mediator production, including IL-1ß, IL-6, TNF-α, and PGE2, as well as the phosphorylation of p38- and JNK-mitogen-activated protein kinase (MAPK), but not that of ERK-MAPK. That no PNA or Δ7-ETrA was detected in the ear disc after the PUFA injection suggests that their anti-inflammatory effect might not be due to fatty acid incorporation, but to modulation of cell signaling. In conclusion, PNA and Δ7-ETrA exerted the in vivo anti-inflammatory effect by suppressing mouse ear edema and dorsal skin inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation Mediators/antagonists & inhibitors , Linolenic Acids/pharmacology , Macrophages/drug effects , Skin/drug effects , THP-1 Cells/drug effects , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Humans , Inflammation Mediators/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred ICR , Skin/metabolism , THP-1 Cells/metabolism
11.
Nutr Cancer ; 72(2): 209-217, 2020.
Article in English | MEDLINE | ID: mdl-31179754

ABSTRACT

To investigate the influence of longan flower extract (LFE) on the sensitization of colorectal cancer (CRC) cells to 5-fluorouracil (5-FU) treatment, HT-29, Colo 320DM and SW480 cells were treated with LFE and 5-FU alone and in combination, and the cell viability was then assessed by trypan blue exclusion, the cell cycle by propidium iodide staining, the mitochondria membrane potential by rhodamine 123 staining, and the expression levels of associated genes by immunoblotting and quantitative real-time polymerase chain reaction. LFE and 5-FU synergistically inhibited cell proliferation of HT-29 and Colo 320DM cells. Combined treatment also elevated the level of loss of mitochondria membrane potential of these two CRC cells and arrested HT-29 cells in the S phase of the cell cycle, in association with down-regulation of cyclin A mRNA expression. LFE synergistically potentiated chemosensitivity to 5-FU in at least two CRC cell lines. The results indicated that LFE has potential as a novel agent for the sensitization of CRC cells to 5-FU.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colorectal Neoplasms/drug therapy , Drug Synergism , Flowers/chemistry , Fluorouracil/pharmacology , Plant Extracts/pharmacology , Sapindaceae/chemistry , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Plant Extracts/chemistry
12.
J Chin Med Assoc ; 83(2): 180-187, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31876795

ABSTRACT

BACKGROUND: Esophageal atresia (EA) and tracheoesophageal fistula (TEF) are serious congenital anomalies with high morbidity and mortality. Diagnostic and therapeutic fiberoptic endoscopy has been used in children to evaluate and manage trachea-esophageal anomalies. This study aimed to evaluate the prognostic factors and the role of fiberoptic bronchoesophagoscopy (FB) in managing children with EA and TEF. METHODS: From 2000 to 2017, hospitalized children with suspected EA and TEF were enrolled in the study. All associated medical records were retrospectively reviewed. Basic characteristics, diagnoses, age of surgical reconstruction, FB findings, associated anomalies, and survival durations were reviewed. Prognostic factors associated with the patients' mortality were analyzed. RESULTS: A total of 33 children were enrolled, and 91% of them were type C. The median age at the time of hospitalization was 26 days (range, birth to 9 years), including 20 (61%) low-birth-weight infants and 26 (79 %) referred patients. FB was performed in patients preoperatively (39%) and postoperatively (96.8%). Among them, 28 patients (85%) had associated anomalies, including 17 (52%) cardiac and 23 (70%) airway anomalies. The median age of 31 patients who underwent surgical reconstruction was 3 (range, 0-39) days. Esophageal anastomotic stricture (21/31, 67.7%) was the most common postsurgical complication. Twenty-three patients (74.2%) received postoperative FB-guided interventions, including balloon dilatation, laser therapy, and stent implantation. Among the 9 mortality cases, the median age at death was 270 (range, 4-3246) days. Significant factor associated with mortality was delayed (> 48 h old) or no surgical reconstruction (p = 0.030). CONCLUSION: Delayed (>48-hour old) or no surgical reconstruction was significantly related to mortality in children with congenital EA and TEF. Preoperative and postoperative FB evaluations helped to facilitate diagnoses and nonsurgical managements and resolve the patients' tracheoesophageal problems.


Subject(s)
Bronchoscopy , Esophageal Atresia/surgery , Esophagoscopy , Tracheoesophageal Fistula/surgery , Child , Child, Preschool , Esophageal Atresia/mortality , Female , Fiber Optic Technology , Humans , Infant , Infant, Newborn , Male , Prognosis , Retrospective Studies , Tracheoesophageal Fistula/mortality
13.
Biochim Biophys Acta Mol Cell Res ; 1866(12): 118517, 2019 12.
Article in English | MEDLINE | ID: mdl-31340164

ABSTRACT

Cathepsin S (CTSS), a lysosomal cysteine protease, has been reported to be associated with extracellular matrix (ECM) degradation, thus promoting cell migration and invasion, but whether CTSS regulates other intracellular mechanisms during metastasis remains unknown. The expression of CTSS was knocked down using siRNA transfection, and enzymatic activity was inhibited by the highly-selective CTSS inhibitor RJW-58. The results of in vitro functional assays, western blot analysis, and an in vivo colonization model demonstrated that CTSS was positively related to cellular adhesive ability. Moreover, both CTSS knockdown and inhibition significantly decreased Ca2+ influx via store-operated Ca2+ entry (SOCE) without changing STIM1 and Orai1 expression levels, while RJW-58 dose-dependently reduced the activation of the Ca2+-dependent downstream effectors, NFAT1 and Rac1. The results of immunoprecipitation assays demonstrated that CTSS could bind to STIM1, which was reversed by CTSS inhibition. In addition, confocal microscopy and super-resolution imaging showed that CTSS inhibition led to STIM1 puncta accumulation in the endoplasmic reticulum and reduced the interaction between active STIM1 and EB1. In conclusion, we have demonstrated for the first time that the lysosomal cysteine protease, CTSS, plays an important role in mediating Ca2+ homeostasis by regulating STIM1 trafficking, which leads to the suppression of cell migration and invasion.


Subject(s)
Calcium/metabolism , Cathepsins/metabolism , Cysteine Proteases/metabolism , Lysosomes/metabolism , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Female , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Wound Healing/drug effects
14.
Oncotarget ; 7(31): 49310-49321, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27384479

ABSTRACT

The development of resistance to platinum drugs in cancer cells severely reduces the efficacy of these drugs. Thus, the discovery of novel drugs or combined strategies to overcome drug resistance is imperative. In addition to our previous finding that combined D-penicillamine with platinum drugs exerts synergistic cytotoxicity, we recently identified a novel therapeutic strategy by combining an iron chelating agent desferal with platinum drugs to overcome platinum resistance in an oxaliplatin-resistant human cervical cancer cell line, S3. Further study demonstrated that the level of platinum-DNA adduct formation positively correlated with cell death in combination of desferal with platinums than that of each drug alone in S3 cells. Decrement of human copper transporter 1 (hCtr1) and transferrin receptor 1 (TfR1) expression involved in the development of platinum resistance in S3 cells. Moreover, desferal promoted the expression of hCtr1 through the upregulation of Sp1. The overexpression of Sp1 increased the expression of NF-κB and translocated it into the nucleus to bind to the TfR1 promoter region, which subsequently increased the expression of TfR1. Importantly, the cotreatment of oxaliplatin with desferal significantly potentiated the oxaliplatin-elicited antitumoral effect in the oxaliplatin-resistant xenograft animal model without any toxic effect observed. Taken together, these results demonstrated that the combination of desferal with oxaliplatin can overcome oxaliplatin resistance through the regulation of hCtr1 and TfR1, and may have beneficial effect for treatment of patient with oxaliplatin-refractory tumors.


Subject(s)
Antigens, CD/metabolism , Cation Transport Proteins/metabolism , Deferoxamine/administration & dosage , Organoplatinum Compounds/administration & dosage , Receptors, Transferrin/metabolism , Sp1 Transcription Factor/metabolism , Uterine Cervical Neoplasms/drug therapy , Active Transport, Cell Nucleus , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Nucleus/metabolism , Chelating Agents/chemistry , Copper Transporter 1 , DNA Adducts , Drug Resistance, Neoplasm , Female , Humans , Iron/chemistry , Male , Mice , Mice, Nude , Neoplasm Transplantation , Oxaliplatin , Promoter Regions, Genetic , Up-Regulation
15.
Inflammation ; 38(6): 2133-45, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26111478

ABSTRACT

Sciadonic acid (SCA), pinolenic acid (PNA), and Δ7-eicosatrienoic acid (Δ7-ETrA) are three non-methylene-interrupted fatty acids (NMIFA). Using murine microglial BV-2 cells, this study determined how NMIFA incorporation modulated phospholipid fatty acid composition and the production of pro-inflammatory mediators. Each NMIFA was rapidly taken up and incorporated in BV-2 cells, resulting in the differential redistribution of total lipids. The cellular phospholipid fatty acid compositions were altered, and a significant decrease in the proportions of total monounsaturated fatty acids (MUFA) was observed while the proportions of NMIFA and its metabolites accounted for 38% of the fatty acid total. Incubation of microglial cells with NMIFA suppressed production of LPS-stimulated pro-inflammatory mediators, including nitric oxide (NO), prostaglandin E2 (PGE2), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), as well as the over-expression of inducible nitric oxide synthase (iNOS) and type 2 cyclooxygenase (COX-2). These inhibitory effects could be accounted for, in part, by the inactivation of mitogen-activated protein kinases (MAPK) signaling. In conclusion, Δ7-ETrA, PNA, and SCA are anti-inflammatory NMIFA that may be useful in suppressing in vitro immune responses involved in neural inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arachidonic Acids/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Inflammation Mediators/metabolism , Linolenic Acids/pharmacology , Microglia/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phospholipids/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Arachidonic Acids/metabolism , Cell Line , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Activation , Fatty Acids, Monounsaturated/metabolism , Inflammation Mediators/immunology , Interleukin-6/metabolism , Linolenic Acids/metabolism , MAP Kinase Signaling System/drug effects , Mice , Microglia/immunology , Microglia/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
Biochem Pharmacol ; 95(1): 28-37, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25801007

ABSTRACT

The platinum-based regimen is the front-line treatment of chemotherapy. However, development of platinum resistance often causes therapeutic failure in this disease. We previously have generated an oxaliplatin-resistant subline, named S3, from human cervical carcinoma SiHa cells, and its resistant phenotype was well-characterized. In the present study, we aimed to identify the novel therapeutic strategy by combining copper chelator D-penicillamine with oxaliplatin, and to elucidate the underlying mechanisms for overcoming oxaliplatin resistance. As the result, D-penicillamine exerted synergistic killing effects only in S3 cells when combined with oxaliplatin and cisplatin by using Chou-Talalay method. Further study showed that the amounts of platinum DNA adduct formed were positively correlated to the percentage of cell death in S3 cells when co-treated D-penicillamine with oxaliplatin and cisplatin. D-penicillamine promoted copper influx transporter hCtr1 expression through upregulation of Sp1. Sp1 overexpression induced p53 translocation from nucleus to cytosol and caused p53 degradation through ubiquitination, which subsequently suppressed the expression of the copper efflux transporter ATP7A. Importantly, co-treatment of cisplatin with D-penicillamine enhanced oxaliplatin-elicited antitumor effect in the oxalipatin-resistant S3 xenograft tumors, but not found in SiHa xenograft model. Notably, Mice received D-penicillamine alone or in combination of D-penicillamine ad oxalipatin, increased hCtrl protein level in S3 xenograft tumor, however, the protein level of ATP7A was decreased. Taken together, this study provides insight into that the co-manipulation of hCtrl and ATP7A by D-penicillamine could increase the therapeutic efficacy of platinum drugs in oxaliplatin resistant tumors, especially in resistant phenotype with downexpression of hCtrl and overexpression of ATP7A.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm/drug effects , Organoplatinum Compounds/administration & dosage , Penicillamine/administration & dosage , Uterine Cervical Neoplasms/drug therapy , Animals , Chelating Agents/administration & dosage , Cisplatin/administration & dosage , Drug Synergism , Drug Therapy, Combination , Female , Humans , Male , Mice , Mice, Nude , Oxaliplatin , Uterine Cervical Neoplasms/pathology
17.
Oncotarget ; 6(10): 8007-18, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25779665

ABSTRACT

When COS7 fibroblasts and other cells were exposed to UVC irradiation and cold shock at 4°C for 5 min, rapid upregulation and nuclear accumulation of NOS2, p53, WWOX, and TRAF2 occurred in 10-30 min. By time-lapse microscopy, an enlarging gas bubble containing nitric oxide (NO) was formed in the nucleus in each cell that finally popped out to cause "bubbling death". Bubbling occurred effectively at 4 and 22°C, whereas DNA fragmentation was markedly blocked at 4°C. When temperature was increased to 37°C, bubbling was retarded and DNA fragmentation occurred in 1 hr, suggesting that bubbling death is switched to apoptosis with increasing temperatures. Bubbling occurred prior to nuclear uptake of propidium iodide and DAPI stains. Arginine analog Nω-LAME inhibited NO synthase NOS2 and significantly suppressed the bubbling death. Unlike apoptosis, there were no caspase activation and flip-over of membrane phosphatidylserine (PS) during bubbling death. Bubbling death was significantly retarded in Wwox knockout MEF cells, as well as in cells overexpressing TRAF2 and dominant-negative p53. Together, UV/cold shock induces bubbling death at 4°C and the event is switched to apoptosis at 37°C. Presumably, proapoptotic WWOX and p53 block the protective TRAF2 to execute the bubbling death.


Subject(s)
Apoptosis/physiology , Cell Death/physiology , Fibroblasts/cytology , Fibroblasts/radiation effects , Animals , Apoptosis/radiation effects , COS Cells , Cell Death/radiation effects , Cell Line, Tumor , Chlorocebus aethiops , Cold Temperature , Fibroblasts/metabolism , HCT116 Cells , Humans , Melanoma, Experimental/pathology , Mice , Nitric Oxide/metabolism , Ultraviolet Rays
18.
Oncotarget ; 6(6): 3737-51, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25686832

ABSTRACT

Zfra is a 31-amino-acid zinc finger-like protein, which participates in the tumor necrosis factor signaling. Here, we determined that when nude mice and BALB/c mice were pre-injected with nanogram levels of a synthetic Zfra1-31 or truncated Zfra4-10 peptide via tail veins, these mice became resistant to the growth, metastasis and stemness of melanoma cells, and many malignant cancer cells. The synthetic peptides underwent self-polymerization in phosphate-buffered saline. Alteration of the Ser8 phosphorylation site to Gly8 abolished Zfra aggregation and its-mediated cancer suppression in vivo. Injected Zfra peptide autofluoresced due to polymerization and was trapped mainly in the spleen. Transfer of Zfra-stimulated spleen cells to naïve mice conferred resistance to cancer growth. Zfra-binding cells, designated Hyal-2+ CD3- CD19- Z cells, are approximately 25-30% in the normal spleen, but are significantly downregulated (near 0-3%) in tumor-growing mice. Zfra prevented the loss of Z cells caused by tumors. In vitro stimulation or education of naïve spleen cells with Zfra allowed generation of activated Z cells to confer a memory anticancer response in naïve or cancer-growing mice. In particular, Z cells are abundant in nude and NOD-SCID mice, and can be readily activated by Zfra to mount against cancer growth.


Subject(s)
Adaptor Proteins, Signal Transducing/pharmacology , Antigens, CD19/immunology , CD3 Complex/immunology , Cell Adhesion Molecules/immunology , Hyaluronoglucosaminidase/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Spleen/drug effects , Spleen/immunology , Amino Acid Sequence , Animals , Cell Adhesion Molecules/genetics , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Humans , Hyaluronoglucosaminidase/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Molecular Sequence Data , Neoplasm Metastasis , Neoplasms/pathology , Peptide Fragments/pharmacology , Spleen/pathology
19.
Inflammation ; 38(2): 534-45, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24993153

ABSTRACT

Eicosatrienoic acid (Δ11,14,17-20:3; ETrA) is a rare naturally occurring n-3 polyunsaturated fatty acid (PUFA). Using murine RAW264.7 cells, the objectives were to determine how ETrA modulated phospholipid fatty acid compositions and the production of pro-inflammatory mediators. Incubation cells with ETrA dose-dependently increased the proportions of phospholipid ETrA and its metabolites to 33 % of the fatty acid total. Incorporation of ETrA also reduced the proportions of total n-6 PUFA and monounsaturated fatty acids (MUFA) by 30 and 60 %, respectively. ETrA suppressed LPS-stimulated nuclear factor-kappa B (NF-κB)-mediated nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) expression. However, no such suppressive effect on the production of prostaglandin E2 (PGE2), cytokines, or expression of cyclooxygenase-2 (COX-2) was observed. As compared with ETrA, eicosapentaenoic acid (EPA) exerted a more potent anti-inflammatory effect. In conclusion, although ETrA suppresses significant NO synthesis and iNOS expression, this n-3 PUFA was a less potent anti-inflammatory agent than EPA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arachidonic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide/biosynthesis , Animals , Cell Line , Cyclooxygenase 2/biosynthesis , Dinoprostone/biosynthesis , Fatty Acids, Unsaturated/metabolism , Gene Expression/drug effects , Lipopolysaccharides , Mice , NF-kappa B/metabolism , Phospholipids/metabolism
20.
Transl Respir Med ; 1(1): 15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-27234396

ABSTRACT

It is generally agreed that the pro-inflammatory, pro-survival transcription factor NF-κB is a tumor promoter. Tumor necrosis factor alpha (TNF-α or TNF) mediates NF-κB activation. Tumor suppressor WWOX (FOR or WOX1) is a downstream effector of the TNF signaling. Thus, activation of both WWOX (FOR or WOX1) and NF-κB may occur during TNF signaling and/or under stress conditions. Indeed, the first WW domain of WWOX induces the activation of NF-κB-responsive promoter without TNF participation. It appears that WWOX counteracts with NF-κB in regulating cell survival and death. For example, WWOX becomes activated with Tyr33 phosphorylation and relocates together with NF-κB and many transcription factors to the nucleus to cause neuronal death in sciatic nerve-transected rats. While WWOX is frequently lost in lung cancer and many other cancers, NF-κB activation-induced cancer promotion probably requires WWOX-independent signaling networks to induce expression of pro-survival factors. The antagonistic role of WWOX and NF-κB in the regulation of lung cancer progression is discussed.

SELECTION OF CITATIONS
SEARCH DETAIL