Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 48
1.
Am J Chin Med ; 52(2): 565-581, 2024.
Article En | MEDLINE | ID: mdl-38480502

L48H37 is a synthetic curcumin analog that has anticancer potentials. Here, we further explored the anticancer effect of L48H37 on oral cancer cells and its mechanistic acts. Cell cycle distribution was assessed using flow cytometric analysis. Apoptosis was elucidated by staining with PI/Annexin V and activation of the caspase cascade. Cellular signaling was explored using apoptotic protein profiling, Western blotting, and specific inhibitors. Our findings showed that L48H37 significantly reduced the cell viability of SCC-9 and HSC-3 cells, resulting in sub-G1 phase accumulation and increased apoptotic cells. Apoptotic protein profiling revealed that L48H37 increased cleaved caspase-3, and downregulated cellular inhibitor of apoptosis protein 1 (cIAP1) and X-linked inhibitor of apoptosis protein (XIAP) in SCC-9 cells, and the downregulated cIAP1 and XIAP in both oral cancer cells were also demonstrated by Western blotting. Meanwhile, L48H37 triggered the activation of caspases and mitogen-activated protein kinases (MAPKs). The involvement of c-Jun N-terminal kinase (JNK) and p38 MAPK (p38) in the L48H37-triggered apoptotic cascade in oral cancer cells was also elucidated by specific inhibitors. Collectively, these findings indicate that L48H37 has potent anticancer activity against oral cancer cells, which may be attributed to JNK/p38-mediated caspase activation and the resulting apoptosis. This suggests a potential benefit for L48H37 for the treatment of oral cancer.


Curcumin , Mouth Neoplasms , Humans , Caspases/metabolism , Curcumin/pharmacology , Cell Line, Tumor , Apoptosis , p38 Mitogen-Activated Protein Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Caspase 3/metabolism , Mouth Neoplasms/drug therapy , Inhibitor of Apoptosis Proteins/pharmacology
2.
Methods ; 224: 35-46, 2024 Apr.
Article En | MEDLINE | ID: mdl-38373678

Bivalent Smac mimetics have been shown to possess binding affinity and pro-apoptotic activity similar to or more potent than that of native Smac, a protein dimer able to neutralize the anti-apoptotic activity of an inhibitor of caspase enzymes, XIAP, which endows cancer cells with resistance to anticancer drugs. We design five new bivalent Smac mimetics, which are formed by various linkers tethering two diazabicyclic cores being the IAP binding motifs. We built in silico models of the five mimetics by the TwistDock workflow and evaluated their conformational tendency, which suggests that compound 3, whose linker is n-hexylene, possess the highest binding potency among the five. After synthesis of these compounds, their ability in tumour cell growth inhibition and apoptosis induction displayed in experiments with SK-OV-3 and MDA-MB-231 cancer cell lines confirms our prediction. Among the five mimetics, compound 3 displays promising pro-apoptotic activity and deserves further optimization.


Antineoplastic Agents , Neoplasms , Humans , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism , X-Linked Inhibitor of Apoptosis Protein/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Molecular Conformation , Apoptosis , Cell Line, Tumor
4.
J Pharmacol Sci ; 154(1): 30-36, 2024 Jan.
Article En | MEDLINE | ID: mdl-38081681

Overexpression of inhibitor of apoptosis (IAP) proteins is associated with poor prognosis. In multiple myeloma (MM), the IAP inhibitors (IAPi), LCL161, have been evaluated in preclinical and clinical settings but are not fully effective. Among IAPs, XIAP has the strongest anti-apoptotic function with direct binding activity to caspases and cIAP1 and cIAP2 are positive regulator of NF-κB signaling. Prior IAPi such as LCL161 has high affinity to cIAP1 and cIAP2 resulting in inferior inhibiting activity against XIAP. A novel dimeric IAPi, AZD5582 (C58H78N8O8), have high binding potency to XIAP with EC50 dose of 15 nM, enabling to simultaneous inhibit XIAP and cIAP1/2. AZD5582 monotherapy showed cell growth inhibition for all MM cell lines, MM1S, RPMI8226, U266 and KMS-5 and induced apoptosis. AZD5582 further showed anti-proliferation effect under the IL-6 additional condition and inhibited JAK-STAT signaling triggered by IL-6. AZD5582 combined with carfilzomib therapy showed a synergistic effect. Enhanced apoptosis was also observed in combination therapy. Synergistic effect was further observed with other conventional therapeutics. Simultaneous XIAP and cIAP1/2 inhibition by the dimeric IAPi AZD5582 is promising. This study provides a rationale of AZD5582 as a new treatment strategy in monotherapy and in combination therapy.


Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Interleukin-6 , Cell Line, Tumor , Apoptosis , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism , X-Linked Inhibitor of Apoptosis Protein/pharmacology
5.
Cell Immunol ; 384: 104674, 2023 02.
Article En | MEDLINE | ID: mdl-36706656

Second mitochondria-derived activator of caspases (SMAC) mimetics are small molecule drugs that mimic the activity of the endogenous SMAC protein. SMAC and SMAC mimetics antagonize inhibitors of apoptosis proteins (IAPs), thereby sensitizing cells to apoptosis. As such, SMAC mimetics are being tested in numerous clinical trials for cancer. In addition to their direct anti-cancer effect, it has been suggested that SMAC mimetics may activate T cells, thereby promoting anti-tumor immunity. Here, we tested the effect of three clinically relevant SMAC mimetics on activation of primary human T cells. As previously reported, SMAC mimetics killed tumor cells and activated non-canonical NF-κB in T cells at clinically relevant doses. Surprisingly, none of the SMAC mimetics augmented T cell responses. Rather, SMAC mimetics impaired T cell proliferation and decreased the proportion of IFNγ/TNFα double-producing T cells. These results question the assumption that SMAC mimetics are likely to boost anti-tumor immunity in cancer patients.


Caspases , Neoplasms , Humans , Caspases/pharmacology , Caspases/therapeutic use , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , Inhibitor of Apoptosis Proteins/therapeutic use , Cytokines , Neoplasms/drug therapy , Apoptosis , Mitochondria/metabolism , Cell Proliferation , Mitochondrial Proteins/metabolism , Cell Line, Tumor
6.
Stem Cell Res Ther ; 13(1): 417, 2022 08 13.
Article En | MEDLINE | ID: mdl-35964136

BACKGROUND: Periodontal ligament stem cells (PDLSCs) are the ideal seed cells for periodontal tissue regeneration. It is well established that persistent inflammation significantly impairs the osteogenic differentiation capability of PDLSCs. Therefore, maintaining PDLSC osteogenic potential under the inflammatory microenvironment is important for treating bone loss in periodontitis. The aim of our study was to explore the potential role of circular RNA BIRC6 (circBIRC6) in regulating osteogenic differentiation of PDLSCs in the inflammatory conditions. METHODS: Alkaline phosphatase staining, Alizarin Red staining, quantitative real-time polymerase chain reaction, western blotting and immunofluorescence staining were used to evaluated the effects of circBIRC6 on the osteogenic differentiation of PDLSCs. RNA pull-down and luciferase assays were performed to explore the interaction between circBIRC6 and miR-543. Then, the downstream signaling pathway affected by circBIRC6/miR-543 axis was further investigated. RESULTS: The expression level of circBIRC6 was higher in PDLSCs exposed to inflammatory stimulus and in periodontitis tissues compared to the respective controls. Downregulation of circBIRC6 enhanced the osteogenic potential of PDLSCs under the inflammatory conditions, and upregulation of circBIRC6 led to opposite findings. Mechanistically, we found that circBIRC6 modulated PDLSC osteogenic differentiation through sponging miR-543. More importantly, we have demonstrated that circBIRC6/miR-543 axis regulated the mineralization capacity of PDLSCs via PTEN/PI3K/AKT/mTOR signaling pathway in the inflammatory microenvironment. CONCLUSIONS: In summary, the expression of miR-543 is significantly increased following circBIRC6 downregulation, leading to inhibition of PTEN and subsequently activation of PI3K/AKT/mTOR signaling pathway. Therefore, targeting circBIRC6 might represent a potential therapeutic strategy for improving bone loss in periodontitis.


MicroRNAs , Periodontitis , Humans , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Osteogenesis , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Periodontal Ligament , Periodontitis/genetics , Periodontitis/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Circular/genetics , Signal Transduction , Stem Cells/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
7.
Clin Exp Immunol ; 207(3): 340-350, 2022 05 12.
Article En | MEDLINE | ID: mdl-35553628

Cellular inhibitors of apoptosis proteins 1 (cIAP1) and 2 (cIAP2) are involved in signaling pathways mediated by Toll-like receptors (TLRs) and tumor necrosis factor (TNF)-α. Excessive activation of TLRs and TNF-α underlies the immunopathogenesis of Crohn's disease (CD) and ulcerative colitis (UC). However, the roles played by cIAP1 and cIAP2 in the development of CD and UC remain poorly understood. In this study, we attempted to clarify the molecular link between cIAP1/cIAP2 and colonic inflammation. Human monocyte-derived dendritic cells (DCs) treated with siRNAs specific for cIAP1 or cIAP2 exhibited reduced pro-inflammatory cytokine responses upon stimulation with TLR ligands. Expression of cIAP1 and cIAP2 in human DCs was suppressed in the presence of interferon regulatory factor 4 (IRF4). This effect was associated with inhibition of cIAP1 and cIAP2 polyubiquitination. To verify these in vitro findings, we created mice overexpressing IRF4 in DCs and showed that these mice were resistant to trinitrobenzene sulfonic acid-induced colitis as compared with wild-type mice; these effects were accompanied by reduced expression levels of cIAP1 and cIAP2. Pro-inflammatory cytokine production by mesenteric lymph node cells upon stimulation with TLR ligands was reduced in mice with DC-specific IRF4 overexpression as compared with that in wild-type mice. Finally, in clinical samples of the colonic mucosa from patients with CD, there was a negative relationship between the percentage of IRF4+ DCs and percentages of cIAP1+ or cIAP2+ lamina propria mononuclear cells. These data suggest that the colitogenic roles of cIAP1 and cIAP2 are negatively regulated by IRF4.


Cytokines , Inhibitor of Apoptosis Proteins , Interferon Regulatory Factors/metabolism , Animals , Apoptosis , Cytokines/metabolism , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , Interferon Regulatory Factors/genetics , Ligands , Mice , Tumor Necrosis Factor-alpha/metabolism
8.
Clin Transl Sci ; 15(1): 55-62, 2022 01.
Article En | MEDLINE | ID: mdl-33742767

Inhibitor of apoptosis proteins (IAPs) regulate apoptosis and modulate NF-κB signaling thereby driving expression of genes involved in immune/inflammatory responses. The orally available IAP antagonist Debio 1143 has potential to enhance tumor response to chemoradiotherapy and/or immunotherapy. Patients with pre-operative squamous cell carcinomas of the head and neck (SCCHN) received: Debio 1143 monotherapy (200 mg/day [D]1-15 +/- 2); Debio 1143 (200 mg/day D1-15 +/- 2) plus cisplatin (40 mg/m2 D 1 and 8); cisplatin alone (40 mg/m2 D 1 and 8; EudraCT: 2014-004655-31). Pharmacokinetic/pharmacodynamic effects were assessed in plasma and resected tumors. Primary end point; effect of Debio 1143 on cellular IAP-1 (cIAP-1). Levels of cIAP-1/-2, X-linked inhibitor of apoptosis protein (XIAP), tumor infiltrating lymphocytes (TILs), including CD8+ T cells, programmed cell death protein 1 (PD-1), PD-ligand 1 (PD-L1), and gene expression were also analyzed. Twenty-three of 26 patients completed treatment. In the Debio 1143 monotherapy cohort (n = 13), mean tumor concentrations of Debio 1143 were 18-fold (maximum 55.2-fold) greater than in plasma, exceeding the half-maximal inhibitory concentration for cIAPs and XIAP by 100 to 1000-fold, with significant engagement/degradation of cIAP-1 (p < 0.05). Overall, levels of CD8+ TILs, PD-1, and PD-L1 positive immune cells increased significantly (p < 0.05) following Debio 1143 treatment. Changes were observed in the expression of genes related to NF-κB signaling. Treatments were well-tolerated. Debio 1143 penetrated SCCHN tumors, engaged cIAP-1, and induced immune inflammatory changes in the tumor microenvironment. Based on the mode of action demonstrated here and in previous studies, these data support future combinations of Debio 1143 with immune-checkpoint agents.


Inhibitor of Apoptosis Proteins/pharmacology , Inhibitor of Apoptosis Proteins/pharmacokinetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Tumor Microenvironment/drug effects , Clinical Trials as Topic , Cohort Studies , Humans , Inhibitor of Apoptosis Proteins/administration & dosage , Pharmacogenetics
9.
Cells ; 9(4)2020 04 18.
Article En | MEDLINE | ID: mdl-32325691

Aiming to promote cancer cell apoptosis is a mainstream strategy of cancer therapy. The second mitochondria-derived activator of caspase (SMAC)/direct inhibitor of apoptosis protein (IAP)-binding protein with low pI (DIABLO) protein is an essential and endogenous antagonist of inhibitor of apoptosis proteins (IAPs). SMAC mimetics (SMs) are a series of synthetically chemical compounds. Via database analysis and literature searching, we summarize the potential mechanisms of endogenous SMAC inefficiency, degradation, mutation, releasing blockage, and depression. We review the development of SMs, as well as preclinical and clinical outcomes of SMs in solid tumor treatment, and we analyze their strengths, weaknesses, opportunities, and threats from our point of view. We also highlight several questions in need of further investigation.


Apoptosis Regulatory Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Neoplasms/drug therapy , Animals , Apoptosis Regulatory Proteins/drug effects , Caspases/drug effects , Caspases/metabolism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/drug effects , Neoplasms/metabolism
10.
Biotech Histochem ; 93(7): 512-518, 2018.
Article En | MEDLINE | ID: mdl-29926741

Although the precise mechanism is unknown, neuron apoptosis is believed to participate in neuropathy caused by acrylamide (ACR). Telomerase reverse transcriptase (TERT) exhibits an anti-apoptotic function, but its contribution to the pathogenesis of ACR neurotoxicity is unclear. We investigated adult male rats that were given 30, 40 and 50 mg/kg ACR three times/week for 4 weeks. We found that ACR treatment caused significant deficits in sensory/motor function as measured by gait score, landing foot spread distance, movement initiation test and tail immersion test. Histological examination showed that the cerebral cortex in all ACR treated animals exhibited fewer neurons and more condensed nuclei than normal cortex. A significant increase in apoptosis was found in the cerebral cortex of rat brains subjected to ACR treatment in a dose-dependent manner. The expression of TERT in the brain was significantly reduced by ACR treatment. The pro-apoptotic cleaved caspase-3 protein level was increased, while the anti-apoptotic Bcl-2 protein level was decreased by 30 - 50 mg/kg ACR. Our findings indicate that TERT and its downstream regulators of neuron apoptosis, including Bcl-2 and cleaved caspase-3, were involved in ACR neurotoxicity.


Telomerase/metabolism , Acrylamide/toxicity , Animals , Apoptosis , Disease Models, Animal , Down-Regulation , Inhibitor of Apoptosis Proteins/pharmacology , Models, Neurological , Nervous System Diseases/chemically induced , Rats , Telomerase/drug effects , Telomerase/genetics
11.
Am J Med Sci ; 354(5): 506-512, 2017 11.
Article En | MEDLINE | ID: mdl-29173363

BACKGROUND: The aim of the present study was to investigate the antitumor effect of our novel survivin-targeted small interfering RNA (siRNA) nanoliposomes on xenograft mouse models with human cervical carcinoma HeLa cells, and to evaluate pharmacokinetics. MATERIALS AND METHODS: siRNA nanoliposome was prepared and transfected into xenograft mouse models. Tumor growth in mice was determined and survivin expression was analyzed by using histologic and immunohischemical staining. Furthermore, low, moderate and high doses of survivin siRNA nanoliposomes were injected in 3 groups, and plasma concentrations were detected at various time points by reverse transcription quantitative polymerase chain reaction. Biodistribution of siRNA in tumor and other important organs were also determined. RESULTS: Survivin expression was significantly downregulated by survivin siRNA delivery mediated by nanoliposome, along with significant suppression of cell growth. Peak concentrations were obtained at 15 minutes after injection in each group, with 1,042,538.00, 6,837,099.54 and 14,631,333.15pg/mL, respectively, and the plasma concentration decreased significantly after 24 hours. The half-time life of survivin siRNA nanoliposomes in each group was 3.60, 2.64 and 2.80 hours, respectively. The area under curve values were 952,190.88, 6,800,687.79 and 13,803,680.96h/pg/mL, and the total drug clearance were 1,050.12, 441.13 and 434.67mL/h/kg. A significant accumulation of Cy5-labeled siRNA was found in the tumor, and a nonspecific accumulation was reduced significantly in lung. CONCLUSIONS: Our findings revealed that survivin suppression by siRNA may contribute to tumor inhibition through both proliferation inhibition and apoptosis promotion effect, and the pharmacokinetic characteristics serve as a fundamental role for further studies on its applicability for cancer therapy.


Inhibitor of Apoptosis Proteins/pharmacology , Liposomes/pharmacology , Nanoparticles/administration & dosage , RNA, Small Interfering/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Down-Regulation/drug effects , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/pharmacokinetics , Liposomes/pharmacokinetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacokinetics , Survivin , Tissue Distribution , Xenograft Model Antitumor Assays
12.
Immunology ; 152(3): 462-471, 2017 11.
Article En | MEDLINE | ID: mdl-28664991

Peptide recognition through the MHC class I molecule by cytotoxic T lymphocytes (CTLs) leads to the killing of cancer cells. A potential challenge for T-cell immunotherapy is that dendritic cells (DCs) are exposed to the MHC class I-peptide complex for an insufficient amount of time. To improve tumour antigen presentation to T cells and thereby initiate a more effective T-cell response, we generated artificial antigen-presenting cells (aAPCs) by incubating human immature DCs (imDCs) with poly(lactic-co-glycolic) acid nanoparticles (PLGA-NPs) encapsulating tumour antigenic peptides, followed by maturation with lipopolysaccharide. Tumour antigen-specific CTLs were then induced using either peptide-loaded mature DCs (mDCs) or aAPCs, and their activities were analysed using both ELISpot and cytotoxicity assays. We found that the aAPCs induced significantly stronger tumour antigen-specific CTL responses than the controls, which included both mDCs and aAPCs loaded with empty nanoparticles. Moreover, frozen CTLs that were generated by exposure to aAPCs retained the capability to eradicate HLA-A2-positive tumour antigen-bearing cancer cells. These results indicated that aAPCs are superior to DCs when inducing the CTL response because the former are capable of continuously presenting tumour antigens to T cells in a sustained manner. The development of aAPCs with PLGA-NPs encapsulating tumour antigenic peptides is a promising approach for the generation of effective CTL responses in vitro and warrants further assessments in clinical trials.


Antigen Presentation , Cancer Vaccines/pharmacology , Cytotoxicity, Immunologic/drug effects , Dendritic Cells/drug effects , Inhibitor of Apoptosis Proteins/pharmacology , Lactic Acid/chemistry , Lipopolysaccharides/pharmacology , MART-1 Antigen/pharmacology , Nanoparticles , Neoplasms/therapy , Peptide Fragments/pharmacology , Polyglycolic Acid/chemistry , T-Lymphocytes, Cytotoxic/drug effects , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cell Survival/drug effects , Delayed-Action Preparations , Dendritic Cells/immunology , Dendritic Cells/metabolism , Drug Compounding , Drug Liberation , Humans , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/immunology , Kinetics , Lipopolysaccharides/immunology , MART-1 Antigen/chemistry , MART-1 Antigen/immunology , MCF-7 Cells , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Polylactic Acid-Polyglycolic Acid Copolymer , Solubility , Survivin , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
13.
Acta Ophthalmol ; 95(6): e495-e502, 2017 Sep.
Article En | MEDLINE | ID: mdl-28650568

OBJECTIVE: On the basis of the functional roles of the embryonic stem cell niche (ESCN) in the human limbal stem cells (LSCs), we proposed to explore the potential roles of microRNAs in regulating the self-renewal and differentiation of LSCs cultured in the ESCN. METHODS: The LSCs were cultured in different media, either in CnT-20 media or in CnT-20 + 20% ES culture supernatant (ESC-CM). The LSCs cultured in ESC-CM were then transfected with microRNA-31 (miR-31) mimic or antago-31. The colony-forming efficiency (CFE) was analysed. Cell cycle, apoptosis, mitochondrial potential and reactive oxygen species were analysed by flow cytometry, and quantitative real-time PCR was used to determine the expression levels of FIH-1, P21, P63, ABCG2, CK3, microRNA-31, microRNA-143, microRNA-145 and microRNA-184. Indirect immunostaining was employed to detect the expression of P63, ABCG2, survivin, connexin-43 and CK3. Western blot was employed to detect the expression of FIH-1, P63, P21, CK3, caspase 3, Tcf4, ß-catenin, survivin, GSK3ß and pGSK3ß. RESULTS: Compared with cells grown in CnT-20, the level of miR-31 in cells grown in ESC-CM was lower. We investigated the roles that miR-31 and FIH-1 play in regulating the functional properties of LSCs. We used antagomirs (antago) to reduce the level of miR-31 in LSCs. Antago-31 increased FIH-1 levels and significantly reduced P21 expressional level in LSCs compared to irrelevant-antago (Ir-antago) treatment. The downregulation of miR-31 in LSCs promotes the maintenance of stemness. CONCLUSION: ES culture supernatant (ESC-CM) regulates the fate of LSCs in part by inhibiting the miR-31/FIH-1/P21 axis. This study may have a high impact on the expansion of LSCs in regenerative medicine, especially for ocular surface reconstruction.


Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Embryonic Stem Cells/cytology , Epithelium, Corneal/cytology , Inhibitor of Apoptosis Proteins/pharmacology , Limbus Corneae/cytology , MicroRNAs/antagonists & inhibitors , Mixed Function Oxygenases/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Apoptosis , Blotting, Western , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Embryonic Stem Cells/metabolism , Epithelium, Corneal/metabolism , Flow Cytometry , Gene Expression Regulation , Humans , Limbus Corneae/metabolism , MicroRNAs/biosynthesis , MicroRNAs/genetics , Mixed Function Oxygenases/biosynthesis , Mixed Function Oxygenases/genetics , RNA/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
14.
J Neurooncol ; 132(2): 231-238, 2017 04.
Article En | MEDLINE | ID: mdl-28283800

Approximately half of surgically-treated patients with low-grade-glioma (LGG) suffer recurrence or metastasis. Currently there is no effective drug treatment. While the selective COX-2 inhibitor celecoxib showed anti-neoplastic activity against several malignant tumors, its effects against LGG remain to be elucidated. Ours is the first report that the expression level of COX-2 in brain tissue samples from patients with LGG and in LGG cell lines is higher than in the non-neoplastic region and in normal brain cells. We found that celecoxib attenuated LGG cell proliferation in a dose-dependent manner. It inhibited the generation of prostaglandin E2 and induced apoptosis and cell-cycle arrest. We also show that celecoxib hampered the activation of the Akt/survivin- and the Akt/ID3 pathway in LGGs. These findings suggest that celecoxib may have a promising therapeutic potential and that the early treatment of LGG patients with the drug may be beneficial.


Apoptosis/physiology , Brain Neoplasms/pathology , Brain/pathology , Cell Proliferation/physiology , Cyclooxygenase 2/metabolism , Glioma/pathology , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Brain/metabolism , Celecoxib/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/physiology , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , Inhibitor of Differentiation Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Survivin
15.
Exp Cell Res ; 350(1): 19-31, 2017 Jan 01.
Article En | MEDLINE | ID: mdl-27816606

Survivin, as an anti-apoptotic protein and a cell cycle regulator, is recently gaining importance for its regenerative potential in salvaging injured hypoxic cells of vital organs such as heart. Different strategies are being employed to upregulate survivin expression in dying hypoxic cardiomyocytes. We investigated the cardioprotective potential of a cell permeable survivin mutant protein SurR9C84A, for the management of hypoxia mediated cardiomyocyte apoptosis, in a novel and clinically relevant model employing primary human cardiomyocytes (HCM). The aim of this research work was to study the efficacy and mechanism of SurR9C84A facilitated cardioprotection and regeneration in hypoxic HCM. To mimic hypoxic microenvironment in vitro, well characterized HCM were treated with 100µm (48h) cobalt chloride to induce hypoxia. Hypoxia induced (HI) HCM were further treated with SurR9C84A (1µg/mL) in order to analyse its cardioprotective efficacy. Confocal microscopy showed rapid internalization of SurR9C84A and scanning electron microscopy revealed the reinstatement of cytoskeleton projections in HI HCM. SurR9C84A treatment increased cell viability, reduced cell death via, apoptosis (Annexin-V assay), and downregulated free cardiac troponin T and MMP-9 expression. SurR9C84A also upregulated the expression of proliferation markers (PCNA and Ki-67) and downregulated mitochondrial depolarization and ROS levels thereby, impeding cell death. Human Apoptosis Array further revealed that SurR9C84A downregulated expression of pro-apoptotic markers and augmented expression of HSPs and HTRA2/Omi. SurR9C84A treatment led to enhanced levels of survivin, VEGF, PI3K and pAkt. SurR9C84A proved non-toxic to normoxic HCM, as validated through unaltered cell proliferation and other marker levels. Its pre-treatment exhibited lesser susceptibility to hypoxia/damage. SurR9C84A holds a promising clinical potential for human cardiomyocyte survival and proliferation following hypoxic injury.


Apoptosis/drug effects , Inhibitor of Apoptosis Proteins/pharmacology , Membrane Potential, Mitochondrial/drug effects , Myocytes, Cardiac/drug effects , Cell Hypoxia/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , Inhibitor of Apoptosis Proteins/genetics , Myocytes, Cardiac/metabolism , Survivin , Up-Regulation
16.
Biomater Sci ; 4(4): 614-26, 2016 Apr.
Article En | MEDLINE | ID: mdl-26845086

Survivin belongs to the family of inhibitor of apoptosis proteins (IAP) and is present in most cancers while being below detection limits in most terminally differentiated adult tissues, making it an attractive protein to target for diagnostic and, potentially, therapeutic roles. Sub-100 nm poly(propargyl acrylate) (PA) particles were surface modified through the copper-catalyzed azide/alkyne cycloaddition of an azide-terminated survivin ligand derivative (azTM) originally proposed by Abbott Laboratories and speculated to bind directly to survivin (protein) at its dimer interface. Using affinity pull-down studies, it was determined that the PA/azTM nanoparticles selectively bind survivin and the particles can enhance apoptotic cell death in glioblastoma cell lines and other survivin over-expressing cell lines such as A549 and MCF7 relative to cells incubated with the original Abbott-derived small molecule inhibitor.


Acrylates/chemistry , Apoptosis , Azides/chemistry , Inhibitor of Apoptosis Proteins/chemistry , Nanoparticles/chemistry , Neoplasm Proteins/chemistry , Polymers/chemistry , Apoptosis/physiology , Azides/pharmacology , Catalysis , Cell Line, Tumor , Copper/chemistry , Cycloaddition Reaction , Humans , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , Ligands , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism
17.
Cancer Sci ; 107(4): 452-60, 2016 Apr.
Article En | MEDLINE | ID: mdl-26825848

Several molecular targeting drugs are being evaluated for endometrial cancer; selecting patients whose cancers are sensitive to these agents is of paramount importance. Previously, we developed the cancer tissue-originated spheroid method for primary cancer cells taken from patients' tumors as well as patient-derived xenografts. In this study, we successfully prepared and cultured cancer tissue-originated spheroids from endometrial cancers. Characteristics of the original tumors were well retained in cancer tissue-originated spheroids including morphology and expression of p53 or neuroendocrine markers. We screened 79 molecular targeting drugs using two cancer tissue-originated spheroid lines derived from endometrioid adenocarcinoma grade 3 and serous adenocarcinoma. Among several hits, we focused on everolimus, a mammalian target of rapamycin complex 1 inhibitor, and YM155, a survivin inhibitor. When sensitivity to everolimus or YM155 was assessed in 12 or 11 cancer tissue-originated spheroids, respectively, from different endometrial cancer patients, the sensitivity varied substantially. The cancer tissue-originated spheroids sensitive to everolimus showed remarkable suppression of proliferation. The phosphorylation status of the mammalian target of rapamycin complex 1 downstream molecules before and after everolimus treatment did not predict the effect of the drug. In contrast, the cancer tissue-originated spheroids sensitive to YM155 showed remarkable cell death. The effect of YM155 was also confirmed in vivo. The histological type correlated with YM155 sensitivity; non-endometrioid adenocarcinomas were sensitive and endometrioid adenocarcinomas were resistant. Non-canonical autophagic cell death was the most likely cause of cell death in a sensitive cancer tissue-originated spheroid. Thus, sensitivity assays using cancer tissue-originated spheroids from endometrial cancers may be useful for screening drugs and finding biomarkers.


Drug Evaluation, Preclinical , Endometrial Neoplasms/drug therapy , Everolimus/pharmacology , Imidazoles/pharmacology , Molecular Targeted Therapy , Naphthoquinones/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/antagonists & inhibitors , Primary Cell Culture , Spheroids, Cellular/drug effects , Survivin , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
18.
Cell Death Dis ; 6: e1893, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26426685

The discovery of cancer cell-selective tumour necrosis factor-related apoptosis inducing ligand (TRAIL)-induced apoptosis generated broad excitement and development of TRAIL receptor agonists (TRA) as potential cancer therapy. Studies demonstrating the synergistic combination effect of SMAC mimetics and TRA further suggested potentially effective treatment in multiple tumour settings. However, predictive biomarkers allowing identification of patients that could respond to treatment are lacking. Here, we described a high throughput combination screen conducted across a panel of 31 breast cancer cell lines in which we observed highly synergistic activity between TRAIL and the inhibitors of apoptosis proteins (IAP) inhibitor (IAPi) AZD5582 in ~30% of cell lines. We detected no difference in the expression levels of the IAPi or TRAIL-targeted proteins or common modulators of the apoptotic pathway between the sensitive and resistant cell lines. Synergistic combination effect of AZD5582 and TRAIL correlated with sensitivity to TRAIL, but not to AZD5582 as a single agent. TRAIL treatment led to significantly greater activity of Caspase-8 in sensitive than in resistant cell lines (P=0.002). The majority (12/14) of AZD5582+TRAIL-resistant cell lines retained a functional cell death pathway, as they were sensitive to AZD5582+TNFα combination treatment. This suggested that failure of the TRAIL receptor complex to transduce the death signal to Caspase-8 underlies AZD5582+TRAIL resistance. We developed a 3D spheroid assay and demonstrated its suitability for the ex vivo analysis of the Caspase-8 activity as a predictive biomarker. Altogether, our study demonstrated a link between the functionality of the TRAIL receptor pathway and the synergistic activity of the IAPi+TRA combination treatment. It also provided a rationale for development of the Caspase-8 activity assay as a functional predictive biomarker that could allow better prediction of the response to IAPi+TRA-based therapies than the analysis of expression levels of protein biomarkers.


Alkynes/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Caspase 8/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , Oligopeptides/pharmacology , Alkynes/administration & dosage , Animals , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Enzyme Activation/drug effects , Female , HT29 Cells , Humans , Inhibitor of Apoptosis Proteins/administration & dosage , Mice , Mice, Nude , Oligopeptides/administration & dosage , TNF-Related Apoptosis-Inducing Ligand , Xenograft Model Antitumor Assays
19.
Life Sci ; 137: 81-8, 2015 Sep 15.
Article En | MEDLINE | ID: mdl-26188594

AIMS: Shear stress-induced apoptosis is one of the leading problems in seeding cells of tissue-engineered blood vessels (TEBVs). We aim to determine the human bone mesenchymal stem cell (hBMSC) apoptosis under shear stress and its possible mechanism. MAIN METHODS: hBMSCs were subjected to 3-, 10-, and 30-dyn/cm(2) shear stress in vitro. Cell multiplication and apoptosis were analyzed by flow cytometry. Apoptosis-related genes were screened by a microarray and evidenced by real-time polymerase chain reaction (RT-PCR). hBMSCs were treated with the human recombinant cell inhibitor of apoptosis protein 1 (cIAP1) and its inhibitor, direct IAP-binding protein with low pl (DIABLO), and then cell apoptosis was analyzed. KEY FINDINGS: Exposure to shear stress (3dyn/cm(2) for >6h) activated apoptosis progress of hBMSCs. However, the same degree of shear stress (3dyn/cm(2) for 6h) did not induce apoptosis. Microarray screening and RT-PCR revealed that Bcl-2-related ovarian killer (BOK) and apoptotic protease-activating factor 1 (APAF1), key molecules of the mitochondrial apoptosis pathway, were markedly upregulated under 3-dyn/cm(2) shear stress. Then, we observed that cIAP1, a Caspase 9 inhibitor, was elevated under 3dyn/cm(2) at short-time exposure (2 or 6h), and it was reduced at long-time exposure (24h). When treated with human recombinant cIAP1, Caspase 3 activity and LDH release of hBMSCs were decreased, and vice versa when treated with DIABLO. SIGNIFICANCE: cIAP1 attenuates hBMSC apoptosis when cells were exposed to shear stress through the regulation of the BOK-APAF1-Caspase 9-Caspase 3 pathway. It may present a pharmacological target to enhance hBMSC biological function in the application of TEBVs.


Apoptosis/drug effects , Blood Vessel Prosthesis , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/drug effects , Mitochondria/drug effects , Mitochondrial Proteins/pharmacology , Stress, Mechanical , Tissue Engineering , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/pharmacology , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured , Gene Expression/drug effects , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Recombinant Proteins/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors
20.
Biol Pharm Bull ; 38(6): 827-35, 2015.
Article En | MEDLINE | ID: mdl-25787895

Dendritic cell (DC)-based vaccines are a promising therapeutic modality for cancer. Results from recent trials and approval of the first DC vaccine by the U.S. Food and Drugs Administration for prostate cancer have paved the way for DC-based vaccines. A total of 21 hormone refractory prostate cancer (HRPC) patients with a life expectancy >3 months were randomised into two groups. DC loaded with recombinant Prostate Specific Membrane Antigen (rPSMA) and recombinant Survivin (rSurvivin) peptides was administered as an subcutaneous (s.c.) injection (5×10(6) cells). Docetaxel (75 mg/m(2) intravenous (i.v.)) and prednisone (5 mg, bis in die (b.i.d.)) served as control. Clinical and immunological responses were evaluated. Primary endpoints were safety and feasibility; secondary endpoint was overall survival. Responses were evaluated on day 15, day 30, day 60, and day 90. DC vaccination was well tolerated with no signs of grade 2 toxicity. DC vaccination induced delayed-type hypersensitivity reactivity and an immune response in all patients. Objective Response Rate (ORR) by Response Evaluation Criteria in Solid Tumours (RECIST) was 72.7% (8/11) versus 45.4 (5/11) in the docetaxel arm and immune related response criteria (irRC) was 54.5% (6/11) compared with 27.2% (3/11) in the control arm. The DC arm showed stable disease (SD) in 6 patients, progressive disease (PD) in 3 patients, and partial remission (PR) in two patients compared to SD in 5 patients, PD in 6 patients, and PR in none in the docetaxel arm. There was a cellular response, disease stabilization, no adverse events, and partial remission with the rPSMA and rSurvivin primed DC vaccine.


Antineoplastic Agents/therapeutic use , Cancer Vaccines , Dendritic Cells , Inhibitor of Apoptosis Proteins/therapeutic use , Prostate-Specific Antigen/therapeutic use , Prostatic Neoplasms/drug therapy , Vaccination , Aged , Aged, 80 and over , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Docetaxel , Humans , Hypersensitivity, Delayed/immunology , Inhibitor of Apoptosis Proteins/pharmacology , Male , Middle Aged , Neoplasm Proteins , Prostate-Specific Antigen/pharmacology , Prostatic Neoplasms/immunology , Survivin , Taxoids/pharmacology , Taxoids/therapeutic use
...