1
|
Li F, Mao Q, Wang J, Zhang X, Lv X, Wu B, Yan T, Jia Y. Salidroside inhibited cerebral ischemia/reperfusion-induced oxidative stress and apoptosis via Nrf2/Trx1 signaling pathway. Metab Brain Dis 2022; 37:2965-2978. [PMID: 35976554 DOI: 10.1007/s11011-022-01061-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/29/2022] [Indexed: 11/28/2022]
Abstract
Cerebral ischemia reperfusion injury (CIRI) is still a serious problem threatening human health. Salidroside (SAL) is a natural phenylpropanoid glycoside compound with antioxidant, anti-inflammatory, and anti-ischemic properties. This study investigated the protective mechanism of SAL on middle cerebral artery occlusion (MCAO)- and oxygen-glucose deprivation/reoxygenation (OGD/R) model-induced CIRI via regulating the nuclear factor erythroid 2-related factor 2 (Nrf2)/thioredoxin 1 (Trx1) axis. The results indicated that SAL (50 mg/kg or 100 mg/kg, intraperitoneal injection) not only effectively alleviated infarction rate, improved histopathological changes, relieved apoptosis by strengthening the suppression of cleaved caspase-3 and Bax/Bcl-2 proteins and decreased malondialdehyde (MDA) formation, but also increased superoxide dismutase (SOD) and catalase (CAT) activities and upregulated the expressions of Nrf2 and Trx1 on MCAO-induced CIRI rats. SAL also efficiently inhibited apoptosis and decreased oxidative stress in OGD/R-stimulated PC12 cells. Furthermore, blocking the Nrf2/Trx1 pathway using tretinoin, an Nrf2 inhibitor, significantly reversed the protective effect of SAL on OGD/R-induced oxidative stress. Moreover, SAL reduced the expression of apoptosis signal-regulating kinase-1 (ASK1) and mitogen-activated protein kinase (MAPK) family proteins. These results demonstrated that SAL inhibited oxidative stress through Nrf2/Trx1 signaling pathway, and subsequently reduced CIRI-induced apoptosis by inhibiting ASK1/MAPK.
Collapse
Affiliation(s)
- Fuyuan Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Qianqian Mao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinyu Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, China
| | - Xiaoying Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinyan Lv
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Wu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, China
| | - Tingxu Yan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, China.
| | - Ying Jia
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, China.
| |
Collapse
|
2
|
Kaur H, Ghosh S, Kumar P, Basu B, Nagpal K. Ellagic acid-loaded, tween 80-coated, chitosan nanoparticles as a promising therapeutic approach against breast cancer: In-vitro and in-vivo study. Life Sci 2021; 284:119927. [PMID: 34492262 DOI: 10.1016/j.lfs.2021.119927] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 01/23/2023]
Abstract
AIMS Among polyphenolic phytoconstituents with anticancer properties, Ellagic acid (EA) is widely reported for its translational potential in vitro but efficient in vivo delivery of EA has been a challenge. We, for the first time, used a tween 80 coated nano delivery of Ellagic acid to evaluate its preclinical efficacy in vitro and in vivo for breast cancer. MAIN METHODS To overcome the challenges of in vivo delivery, two batches of chitosan-based nanoformulations of EA (with and without tween 80 coating) were prepared by the ionotropic gelation method. The nanoformulations were characterized and further evaluated in vitro against breast cancer cells (MCF7) and in vivo with EAC tumor-bearing mice for establishing their anticancer efficacy compared to Ellagic acid alone. A quantitative simulation study was undertaken to understand if the observed antitumor efficacy is due to the synergistic efficacy of the Chitosan-Ellagic acid combination. KEY FINDINGS Results revealed that nanoformulations consist of good nano-sized encapsulation of EA and showed good drug entrapment-release capacity. Nano-encapsulated EA is biocompatible and exhibited higher cytotoxicity in vitro compared to EA alone. Similarly, significantly higher tumor regression was observed in nano-EA treated mice compared to EA alone, and best efficacy was observed with the nanoformulation with tween 80 coating. Furthermore, nanoformulations showed higher apoptosis in tumor tissues with no significant tissue toxicity in vital organs. SIGNIFICANCE We report synergism of Chitosan-Ellagic acid combination in the tween 80 coated nanoparticles of Ellagic acid resulting in enhanced anti-breast tumor efficacy that may be of translational value for other tumor types, too.
Collapse
Affiliation(s)
- Harsheen Kaur
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, AUUP 201303, India
| | - Sandip Ghosh
- Department of Neuroendocrinology & Experimental Hematology, Chittaranjan National Cancer Institute, Kolkata 700026, India
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Biswarup Basu
- Department of Neuroendocrinology & Experimental Hematology, Chittaranjan National Cancer Institute, Kolkata 700026, India.
| | - Kalpana Nagpal
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, AUUP 201303, India.
| |
Collapse
|
3
|
Chung KS, Yoo CB, Lee JH, Lee HH, Park SE, Han HS, Lee SY, Kwon BM, Choi JH, Lee KT. Regulation of ROS-Dependent JNK Pathway by 2'-Hydroxycinnamaldehyde Inducing Apoptosis in Human Promyelocytic HL-60 Leukemia Cells. Pharmaceutics 2021; 13:pharmaceutics13111794. [PMID: 34834209 PMCID: PMC8618870 DOI: 10.3390/pharmaceutics13111794] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/28/2022] Open
Abstract
The present study demonstrated that 2'-hydroxycinnamaldehyde (2'-HCA) induced apoptosis in human promyelocytic leukemia HL-60 cells through the activation of mitochondrial pathways including (1) translocation of Bim and Bax from the cytosol to mitochondria, (2) downregulation of Bcl-2 protein expression, (3) cytochrome c release into the cytosol, (4) loss of mitochondrial membrane potential (ΔΨm), and (5) caspase activation. 2'-HCA also induced the activation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase1/2 (ERK1/2) in HL-60 cells. The pharmacological and genetic inhibition of JNK effectively prevented 2'-HCA-induced apoptosis and activator protein-1 (AP-1)-DNA binding. In addition, 2'-HCA resulted in the accumulation of reactive oxygen species (ROS) and depletion of intracellular glutathione (GSH) and protein thiols (PSH) in HL-60 cells. NAC treatment abrogated 2'-HCA-induced JNK phosphorylation, AP-1-DNA binding, and Bim mitochondrial translocation, suggesting that oxidative stress may be required for 2'-HCA-induced intrinsic apoptosis. Xenograft mice inoculated with HL-60 leukemia cells demonstrated that the intraperitoneal administration of 2'-HCA inhibited tumor growth by increasing of TUNEL staining, the expression levels of nitrotyrosine and pro-apoptotic proteins, but reducing of PCNA protein expression. Taken together, our findings suggest that 2'-HCA induces apoptosis via the ROS-dependent JNK pathway and could be considered as a potential therapeutic agent for leukemia.
Collapse
Affiliation(s)
- Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
| | - Chae-Bin Yoo
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
| | - Jeong-Hun Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea;
| | - Hwi-Ho Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
| | - Sang-Eun Park
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmarcy, Kyung Hee University, Seoul 02447, Korea
| | - Hee-Soo Han
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea;
| | - Su-Yeon Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmarcy, Kyung Hee University, Seoul 02447, Korea
| | - Byoung-Mok Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea;
| | - Jung-Hye Choi
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea;
- Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea;
- Correspondence: ; Tel.: +82-2-961-0860
| |
Collapse
|
4
|
Guesmi F, Prasad S, Ali MB, Ismail IA, Landoulsi A. Thymus hirtus sp. algeriensis Boiss. and Reut. volatile oil enhances TRAIL/Apo2L induced apoptosis and inhibits colon carcinogenesis through upregulation of death receptor pathway. Aging (Albany NY) 2021; 13:21975-21990. [PMID: 34543231 PMCID: PMC8507293 DOI: 10.18632/aging.203552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/11/2021] [Indexed: 11/25/2022]
Abstract
Background: The aim of the study is to determine the anticancer activity of Thymus algeriensis (TS) and its underlying mechanisms using in vitro and in animal models. Methods: HCT116 cells were treated with TS essential oil alone or with TRAIL, and then its anticancer effect was determined by using MTT assay, live dead assay, caspase activation and PARP cleavage. Further mechanisms of its anticancer effects was determined by analyzing expression of death receptor signaling pathway using Western blotting. A mouse model was also used to assess the antitumor potential of thyme essential oil. Results: TS oily fraction showed tumor growth inhibitory effect even at lower concentration. TS induces apoptotic cell death as indicated by cleavage of PARP, and activation of the initiator and effector caspases (caspase-3, -8 and -9). Further, results showed that TS increases the expression of death receptors (DRs) and reduces the expression of TRAIL decoy receptors (DcRs). In addition, upregulation of signaling molecules of MAPK pathway (p38 kinase, ERK, JNK), down-regulation of c-FLIP, and overexpression of SP1 and CHOP were observed by TS. Further in animal model, intragastric administration of TS (12.5 mg/ml and 50 mg/ml) prevented colorectal carcinogenesis by blocking multi-steps in carcinoma. Conclusion: Overall, these results indicate that thymus essential oil promotes apoptosis in HCT116 cells and impedes tumorigenesis in animal model. Moreover, thyme potentiates TRAIL-induced cell death through upregulation of DRs, CHOP and SP1 as well as downregulation of antiapoptotic proteins in HCT116 cells. However, therapeutic potential of TS needs to be further explored.
Collapse
Affiliation(s)
- Fatma Guesmi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Laboratory of Biochemistry and Molecular Biology, University of Carthage, Faculty of Sciences of Bizerte, Zarzouna, Bizerte 7021, Tunisia
| | - Sahdeo Prasad
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Research and Development, Noble Pharma LLC, Menomonie, WI 54751, USA
| | - Manel Ben Ali
- Department of Biology, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Ismail A Ismail
- Department of Biology, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Ahmed Landoulsi
- Laboratory of Biochemistry and Molecular Biology, University of Carthage, Faculty of Sciences of Bizerte, Zarzouna, Bizerte 7021, Tunisia
| |
Collapse
|
5
|
Retraction: Novel Insights into the Synergistic Interaction of a Thioredoxin Reductase Inhibitor and TRAIL: The Activation of the ASK1-ERK-Sp1 Pathway. PLoS One 2020; 15:e0231103. [PMID: 32210462 PMCID: PMC7094841 DOI: 10.1371/journal.pone.0231103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
|
6
|
Zhou Q, Wu X, Wang X, Yu Z, Pan T, Li Z, Chang X, Jin Z, Li J, Zhu Z, Liu B, Su L. The reciprocal interaction between tumor cells and activated fibroblasts mediated by TNF-α/IL-33/ST2L signaling promotes gastric cancer metastasis. Oncogene 2020; 39:1414-1428. [PMID: 31659258 PMCID: PMC7018661 DOI: 10.1038/s41388-019-1078-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/13/2019] [Accepted: 10/15/2019] [Indexed: 01/25/2023]
Abstract
Gastric cancer (GC) is characterized by extensive local invasion, distant metastasis and poor prognosis. In most cases, GC progression is associated with aberrant expression of cytokines or activation of signaling cascades mediated by tumor-stroma interactions. However, the mechanisms by which these interactions contribute to GC progression are poorly understood. In this study, we find that IL-33 and its receptor ST2L are upregulated in the human GC and served as prognostic markers for poor survival of GC patients. In a co-culture model with GC cells and cancer-associated fibroblasts (CAFs), we further demonstrate that CAFs-derived IL-33 enhances the migration and invasion of GC cells by inducing the epithelial-mesenchymal transition (EMT) through activation of the ERK1/2-SP1-ZEB2 pathway in a ST2L-dependent manner. Furthermore, the secretion of IL-33 by CAFs can be induced by the proinflammatory cytokines TNF-α that is released by GC cells via TNFR2-NF-κB-IRF-1 pathway. Additionally, silencing of IL-33 expression in CAFs or ST2L expression in GC cells inhibits the peritoneal dissemination and metastatic potential of GC cells in nude mice. Taken together, these results characterize a critical role of the interaction between epithelial-stroma mediated by the TNF-α/IL-33/ST2L signaling in GC progression, and provide a rationale for targeting this pathway to treat GC metastasis.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
- Department of Urology, Center for Organ Transplantation, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Xiongyan Wu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Xiaofeng Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Zhenjia Yu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Tao Pan
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Zhen Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Xinyu Chang
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Zhijian Jin
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Jianfang Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Zhenggang Zhu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China
| | - Bingya Liu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China.
| | - Liping Su
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, People's Republic of China.
| |
Collapse
|
7
|
Paschoalin T, Martens AA, Omori ÁT, Pereira FV, Juliano L, Travassos LR, Machado-Santelli GM, Cunha RLOR. Antitumor effect of chiral organotelluranes elicited in a murine melanoma model. Bioorg Med Chem 2019; 27:2537-2545. [PMID: 30962115 DOI: 10.1016/j.bmc.2019.03.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023]
Abstract
Protease roles in cancer progression have been demonstrated and their inhibitors display antitumor effects. Cathepsins are lysosomal cysteine proteases that have increased expression in tumor cells, and tellurium compounds were described as potent cysteine protease inhibitors and also assayed in several animal models. In this work, the two enantiomeric forms of 1-[Butyl(dichloro)-λ4-tellanyl]-2-[1S-methoxyethyl]benzene (organotelluranes RF-13R and RF-13S) were evaluated as inhibitors of cathepsins B and L, showing significant enantiodiscrimination. We observed their cytotoxic effects on a murine melanoma model, effectively inhibiting tumor progression in vivo. The enantiomers were able to inhibit melanoma cell viability, migration and invasion in vitro. Besides, RF-13S and RF-13R were able to inhibit endothelial cell angiogenesis using a tube formation assay in vitro, in a stereodependent manner. These organotelluranes affected cell morphology, showing disassembling of the actin cytoskeleton. These results suggest organotelluranes as potential antitumor agents, acting directly on tumor cell proliferation, migration and invasion, and on endothelial cells, disrupting angiogenesis, showing low toxicity and high efficiency. Taken together our results suggest that this class of compounds should be further studied to reveal their potential as antitumoral agents.
Collapse
Affiliation(s)
- Thaysa Paschoalin
- Departamento de Microbiologia, Imunologia e Parasitologia, Unidade de Oncologia Experimental (UNONEX), Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil.
| | - Adam A Martens
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Álvaro T Omori
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, Brazil
| | - Felipe V Pereira
- Departamento de Microbiologia, Imunologia e Parasitologia, Unidade de Oncologia Experimental (UNONEX), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luiz Juliano
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luiz R Travassos
- Departamento de Microbiologia, Imunologia e Parasitologia, Unidade de Oncologia Experimental (UNONEX), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Glaucia M Machado-Santelli
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Rodrigo L O R Cunha
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, Brazil.
| |
Collapse
|
8
|
Xu T, Luo Y, Kong F, Lv B, Zhao S, Chen J, Liu W, Cheng L, Zhou Z, Zhou Z, Huang Y, Li L, Zhao X, Qian D, Fan J, Yin G. GIT1 is critical for formation of the CD31 hiEmcn hi vessel subtype in coupling osteogenesis with angiogenesis via modulating preosteoclasts secretion of PDGF-BB. Bone 2019; 122:218-230. [PMID: 30853660 DOI: 10.1016/j.bone.2019.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/06/2019] [Accepted: 03/06/2019] [Indexed: 01/08/2023]
Abstract
G protein-coupled receptor kinase 2 interacting protein-1 (GIT1) is a scaffold protein that plays a vital role in bone modeling and remodeling during osteogenesis coupled with angiogenesis. Recent studies have shown that a specialized subset of vascular endothelium strongly positive for CD31 and Endomucin (CD31hiEmcnhi) is coupled with anabolic bone formation. Based on our previous finding that GIT1 knockout (GIT1 KO) mice have impaired angiogenesis and bone formation, we hypothesized that GIT1 affects formation of the CD31hiEmcnhi vessel subtype. In the current study, GIT1 knockout (GIT1 KO) mice displayed a significant decrease in trabecular bone mass and CD31hiEmcnhi vessel number, compared to their wild-type counterparts. In the fracture healing mouse model, GIT1 KO mice contained a lower number of CD31hiEmcnhi vessels in fracture callus at days 7 and 14. However, no significant differences in the number of preosteoclasts in bone marrow, trabecular bone and callus in GIT1 KO mice were observed, compared with wild-type mice. Notably, concentrations of serum platelet-derived growth factor-BB(PDGF-BB) secreted by preosteoclasts associated with CD31hiEmcnhi vessel formation were lower in GIT1 KO mice. In addition, PDGF-BB-associated expression of phosphorylated extracellular signal-regulated kinase- 1/2 (ERK1/2) and specificity protein 1 (SP1) was significantly decreased in preosteoclasts of GIT1 KO mice. These results collectively suggest that GIT1 is a critical participant in formation of the CD31hiEmcnhi vessel subtype, highlighting a novel biologic function of this scaffold protein in preosteoclasts.
Collapse
Affiliation(s)
- Tao Xu
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - YongJun Luo
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - FanQi Kong
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Bin Lv
- Department of Orthopedics, The Affiliated People's Hospital with Jiangsu University, Zhenjiang, Jiangsu Province 212000, China
| | - ShuJie Zhao
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Jian Chen
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Wei Liu
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Lin Cheng
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Zheng Zhou
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - ZhiMin Zhou
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - YiFan Huang
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - LinWei Li
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Xuan Zhao
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - DingFei Qian
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China
| | - Jin Fan
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China.
| | - GuoYong Yin
- Department of Orthopedic, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd., Nanjing 210029, China.
| |
Collapse
|
9
|
Elucidation for modulation of death receptor (DR) 5 to strengthen apoptotic signals in cancer cells. Arch Pharm Res 2019; 42:88-100. [DOI: 10.1007/s12272-018-01103-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
|
10
|
Guesmi F, Ben Hmed M, Prasad S, Tyagi AK, Landoulsi A. In vivo pathogenesis of colon carcinoma and its suppression by hydrophilic fractions of Clematis flammula via activation of TRAIL death machinery (DRs) expression. Biomed Pharmacother 2018; 109:2182-2191. [PMID: 30551475 DOI: 10.1016/j.biopha.2018.11.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 11/08/2018] [Accepted: 11/10/2018] [Indexed: 02/06/2023] Open
Abstract
This work focused on characterizing hydrophilic fractions of Clematis flammula (CFl). The data here clearly demonstrated that hydrolate fractions act as a free radical scavengers and inhibited proliferation of different cell lines in a time- and concentration-dependent manner, transwell, and with a significant cytotoxic effect. Treating cells with CFl had the effect of suppressing cell growth attenuated by ROS generation in colonic carcinoma. Moreover, CFl in HCT116 cells suppressed survival, proliferation, invasion, angiogenesis and metastasis in vitro by inhibiting gene expression. Following CFl treatment, caspases and PARP cleavage were detected. The up- and down-regulated genes obtained from the WBA of the effect of CFl showed that several biological processes were associated with apoptosis and induction of G1 cell cycle arrest. CFl synergizes the effect of TRAIL by down-regulating the expression of cell survival proteins involved in apoptosis compared to cells treated with CFl or TRAIL alone. Our findings showed that CFl sensitizes apoptosis in TRAIL-resistant cells by activating MAPKs, SP1, and CHOP, that induced DR5 expression. Overall, our data showed that CFl is a promising antitumor agent through kinases and transcription factor induction, both of which are required to activate TRAIL receptors. Colon inflammation induced by LPS was inhibited by CFl hydrolate.
Collapse
Affiliation(s)
- Fatma Guesmi
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA; Laboratory of Biochemistry and Molecular Biology, Faculty of Sciences of Bizerte, University of Carthage, Tunisia.
| | - Marwa Ben Hmed
- Research Unit of Macromolecular Biochemistry and Genetics, Faculty of Sciences of Gafsa, 2112, Tunisia
| | - Sahdeo Prasad
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| | - Amit K Tyagi
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| | - Ahmed Landoulsi
- Laboratory of Biochemistry and Molecular Biology, Faculty of Sciences of Bizerte, University of Carthage, Tunisia
| |
Collapse
|
11
|
Zhang J, Zhang B, Li X, Han X, Liu R, Fang J. Small molecule inhibitors of mammalian thioredoxin reductase as potential anticancer agents: An update. Med Res Rev 2018; 39:5-39. [DOI: 10.1002/med.21507] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering; Lanzhou University; Lanzhou China
- School of Pharmacy; Lanzhou University; Lanzhou China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering; Lanzhou University; Lanzhou China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering; Lanzhou University; Lanzhou China
| | - Xiao Han
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering; Lanzhou University; Lanzhou China
| | - Ruijuan Liu
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering; Lanzhou University; Lanzhou China
- School of Pharmacy; Lanzhou University; Lanzhou China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering; Lanzhou University; Lanzhou China
| |
Collapse
|
12
|
Fluoxetine induces apoptosis through endoplasmic reticulum stress via mitogen-activated protein kinase activation and histone hyperacetylation in SK-N-BE(2)-M17 human neuroblastoma cells. Apoptosis 2017. [PMID: 28647884 DOI: 10.1007/s10495-017-1390-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
13
|
Vij P, Hardej D. Alterations in antioxidant/oxidant gene expression and proteins following treatment of transformed and normal colon cells with tellurium compounds. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 43:216-224. [PMID: 27017383 DOI: 10.1016/j.etap.2016.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 03/07/2016] [Accepted: 03/10/2016] [Indexed: 06/05/2023]
Abstract
The current study evaluated the potential of TeCl4 and DPDT to accumulate within cells and cause oxidative stress. HO-1, antioxidant gene expression and protein alterations were studied.
Collapse
Affiliation(s)
- Puneet Vij
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, NY 11439, USA
| | - Diane Hardej
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, NY 11439, USA.
| |
Collapse
|
14
|
Fassl A, Tagscherer KE, Richter J, De-Castro Arce J, Savini C, Rösl F, Roth W. Inhibition of Notch1 signaling overcomes resistance to the death ligand Trail by specificity protein 1-dependent upregulation of death receptor 5. Cell Death Dis 2015; 6:e1921. [PMID: 26469969 PMCID: PMC4632291 DOI: 10.1038/cddis.2015.261] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 11/28/2022]
Abstract
The Notch1 signaling pathway contributes to tumorigenesis by influencing differentiation, proliferation and apoptosis. Here, we demonstrate that inhibition of the Notch1 signaling pathway sensitizes glioblastoma cell lines and glioblastoma initiating cells to apoptosis induced by the death ligand TRAIL. This sensitization occurs through transcriptional upregulation of the death receptor 5 (DR5, TRAIL-R2). The increase in DR5 expression is abrogated by concomitant repression of the transcription factor Sp1, which directly binds to the DR5 promoter in the absence of Notch1 as revealed by chromatin immunoprecipitation. Consistent with these findings, Notch1 inhibition resulted in increased DR5 promoter activity, which was impaired by mutation of one out of two Sp1-binding sites within the proximal DR5 promoter. Moreover, we demonstrate that JNK signaling contributes to the regulation of DR5 expression by Notch1. Taken together, our results identify Notch1 as key driver for TRAIL resistance and suggest Notch1 as a promising target for anti-glioblastoma therapy.
Collapse
Affiliation(s)
- A Fassl
- Molecular Tumor Pathology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, and Institute of Pathology, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - K E Tagscherer
- Molecular Tumor Pathology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, and Institute of Pathology, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - J Richter
- Molecular Tumor Pathology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, and Institute of Pathology, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - J De-Castro Arce
- Division of Viral Transformation Mechanisms, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - C Savini
- Division of Viral Transformation Mechanisms, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - F Rösl
- Division of Viral Transformation Mechanisms, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - W Roth
- Molecular Tumor Pathology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, and Institute of Pathology, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| |
Collapse
|
15
|
Yuan K, Yong S, Xu F, Zhou T, McDonald JM, Chen Y. Calmodulin antagonists promote TRA-8 therapy of resistant pancreatic cancer. Oncotarget 2015; 6:25308-19. [PMID: 26320171 PMCID: PMC4694833 DOI: 10.18632/oncotarget.4490] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/30/2015] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer is highly malignant with limited therapy and a poor prognosis. TRAIL-activating therapy has been promising, however, clinical trials have shown resistance and limited responses of pancreatic cancers. We investigated the effects of calmodulin(CaM) antagonists, trifluoperazine(TFP) and tamoxifen(TMX), on TRA-8-induced apoptosis and tumorigenesis of TRA-8-resistant pancreatic cancer cells, and underlying mechanisms. TFP or TMX alone did not induce apoptosis of resistant PANC-1 cells, while they dose-dependently enhanced TRA-8-induced apoptosis. TMX treatment enhanced efficacy of TRA-8 therapy on tumorigenesis in vivo. Analysis of TRA-8-induced death-inducing-signaling-complex (DISC) identified recruitment of survival signals, CaM/Src, into DR5-associated DISC, which was inhibited by TMX/TFP. In contrast, TMX/TFP increased TRA-8-induced DISC recruitment/activation of caspase-8. Consistently, caspase-8 inhibition blocked the effects of TFP/TMX on TRA-8-induced apoptosis. Moreover, TFP/TMX induced DR5 expression. With a series of deletion/point mutants, we identified CaM antagonist-responsive region in the putative Sp1-binding domain between -295 to -300 base pairs of DR5 gene. Altogether, we have demonstrated that CaM antagonists enhance TRA-8-induced apoptosis of TRA-8-resistant pancreatic cancer cells by increasing DR5 expression and enhancing recruitment of apoptotic signal while decreasing survival signals in DR5-associated DISC. Our studies support the use of these readily available CaM antagonists combined with TRAIL-activating agents for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Kaiyu Yuan
- Department of Pathology, University of Alabama at Birmingham, Alabama 35294, Birmingham, USA
| | - Sun Yong
- Department of Pathology, University of Alabama at Birmingham, Alabama 35294, Birmingham, USA
| | - Fei Xu
- Department of Pathology, University of Alabama at Birmingham, Alabama 35294, Birmingham, USA
| | - Tong Zhou
- Department of Medicine, University of Alabama at Birmingham, Alabama 35294, Birmingham, USA
| | - Jay M McDonald
- Department of Pathology, University of Alabama at Birmingham, Alabama 35294, Birmingham, USA
- Birmingham Veterans Affairs Medical Center, Alabama 35294, Birmingham, USA
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, Alabama 35294, Birmingham, USA
- Birmingham Veterans Affairs Medical Center, Alabama 35294, Birmingham, USA
| |
Collapse
|
16
|
Karpel-Massler G, Pareja F, Aimé P, Shu C, Chau L, Westhoff MA, Halatsch ME, Crary JF, Canoll P, Siegelin MD. PARP inhibition restores extrinsic apoptotic sensitivity in glioblastoma. PLoS One 2014; 9:e114583. [PMID: 25531448 PMCID: PMC4273972 DOI: 10.1371/journal.pone.0114583] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/11/2014] [Indexed: 01/23/2023] Open
Abstract
Background Resistance to apoptosis is a paramount issue in the treatment of Glioblastoma (GBM). We show that targeting PARP by the small molecule inhibitors, Olaparib (AZD-2281) or PJ34, reduces proliferation and lowers the apoptotic threshold of GBM cells in vitro and in vivo. Methods The sensitizing effects of PARP inhibition on TRAIL-mediated apoptosis and potential toxicity were analyzed using viability assays and flow cytometry in established GBM cell lines, low-passage neurospheres and astrocytes in vitro. Molecular analyses included western blots and gene silencing. In vivo, effects on tumor growth were examined in a murine subcutaneous xenograft model. Results The combination treatment of PARP inhibitors and TRAIL led to an increased cell death with activation of caspases and inhibition of formation of neurospheres when compared to single-agent treatment. Mechanistically, pharmacological PARP inhibition elicited a nuclear stress response with up-regulation of down-stream DNA-stress response proteins, e.g., CCAAT enhancer binding protein (C/EBP) homology protein (CHOP). Furthermore, Olaparib and PJ34 increased protein levels of DR5 in a concentration and time-dependent manner. In turn, siRNA-mediated suppression of DR5 mitigated the effects of TRAIL/PARP inhibitor-mediated apoptosis. In addition, suppression of PARP-1 levels enhanced TRAIL-mediated apoptosis in malignant glioma cells. Treatment of human astrocytes with the combination of TRAIL/PARP inhibitors did not cause toxicity. Finally, the combination treatment of TRAIL and PJ34 significantly reduced tumor growth in vivo when compared to treatment with each agent alone. Conclusions PARP inhibition represents a promising avenue to overcome apoptotic resistance in GBM.
Collapse
Affiliation(s)
- Georg Karpel-Massler
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Fresia Pareja
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Pascaline Aimé
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Chang Shu
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Lily Chau
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | | | - John F Crary
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Peter Canoll
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Markus D Siegelin
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| |
Collapse
|
17
|
van Roosmalen IAM, Quax WJ, Kruyt FAE. Two death-inducing human TRAIL receptors to target in cancer: similar or distinct regulation and function? Biochem Pharmacol 2014; 91:447-56. [PMID: 25150214 DOI: 10.1016/j.bcp.2014.08.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/11/2014] [Accepted: 08/11/2014] [Indexed: 12/11/2022]
Abstract
The emergence during evolution of two tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptors, receptor-1/DR4 and -2/DR5, able to induce apoptosis has raised the question whether they differ in function and regulation, which is of key importance for selecting either DR4 or DR5 selective pro-apoptotic agents for cancer treatment. In this review we found practically no information regarding possible differences in DR4 and DR5 function based on structural differences. On the other hand, a panel of different DR4 or DR5 selective pro-apoptotic agonists have been developed that were explored for efficacy in different tumour types in a large number of studies. Leukemic cells appear mainly sensitive for DR4-induced apoptosis, contrasting the situation in other tumour types that show heterogeneity in receptor preference and, in some cases, a slight overall preference for DR5. Both receptors were found to mediate intracellular stress-induced apoptosis, although this is most frequently reported for DR5. Interestingly, DR5 was also found to transmit non-apoptotic signalling in resistant tumour cells and recently nuclear localization and a role in microRNA maturation has been described. DR4 expression is most heavily regulated by promoter methylation, intracellular trafficking and post-translational modifications. DR5 expression is predominantly regulated at the transcriptional level, which may reflect its ability to respond to cellular stressors. It will be important to further increase our understanding of the mechanisms determining TRAIL receptor preference in order to select the appropriate TRAIL receptor selective agonists for therapy, and to develop novel strategies to enhance apoptosis activation in tumours.
Collapse
Affiliation(s)
- Ingrid A M van Roosmalen
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Wim J Quax
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Frank A E Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
18
|
Farooqi AA, Yaylim I, Ozkan NE, Zaman F, Halim TA, Chang HW. Restoring TRAIL mediated signaling in ovarian cancer cells. Arch Immunol Ther Exp (Warsz) 2014; 62:459-74. [PMID: 25030086 DOI: 10.1007/s00005-014-0307-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 06/26/2014] [Indexed: 02/08/2023]
Abstract
Ovarian cancer has emerged as a multifaceted and genomically complex disease. Genetic/epigenetic mutations, suppression of tumor suppressors, overexpression of oncogenes, rewiring of intracellular signaling cascades and loss of apoptosis are some of the deeply studied mechanisms. In vitro and in vivo studies have highlighted different molecular mechanisms that regulate tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mediated apoptosis in ovarian cancer. In this review, we bring to limelight, expansion in understanding systematical characterization of ovarian cancer cells has led to the rapid development of new drugs and treatments to target negative regulators of TRAIL mediated signaling pathway. Wide ranging synthetic and natural agents have been shown to stimulate mRNA and protein expression of death receptors. This review is compartmentalized into programmed cell death protein 4, platelet-derived growth factor signaling and miRNA control of TRAIL mediated signaling to ovarian cancer. Mapatumumab and PRO95780 have been tested for efficacy against ovarian cancer. Use of high-throughput screening assays will aid in dissecting the heterogeneity of this disease and increasing a long-term survival which might be achieved by translating rapidly accumulating information obtained from molecular and cellular studies to clinic researches.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, RLMC, 35 km Ferozepur Road, Lahore, Pakistan,
| | | | | | | | | | | |
Collapse
|
19
|
Apoptosis signal-regulating kinase 1 is involved in brain-derived neurotrophic factor (BDNF)-enhanced cell motility and matrix metalloproteinase 1 expression in human chondrosarcoma cells. Int J Mol Sci 2013; 14:15459-78. [PMID: 23892595 PMCID: PMC3759868 DOI: 10.3390/ijms140815459] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/15/2013] [Accepted: 07/15/2013] [Indexed: 12/11/2022] Open
Abstract
Chondrosarcoma is the primary malignancy of bone that is characterized by a potent capacity to invade locally and cause distant metastasis, and is therefore associated with poor prognoses. Chondrosarcoma further shows a predilection for metastasis to the lungs. The brain-derived neurotrophic factor (BDNF) is a small molecule in the neurotrophin family of growth factors that is associated with the disease status and outcome of cancers. However, the effect of BDNF on cell motility in human chondrosarcoma cells is mostly unknown. Here, we found that human chondrosarcoma cell lines had significantly higher cell motility and BDNF expression compared to normal chondrocytes. We also found that BDNF increased cell motility and expression of matrix metalloproteinase-1 (MMP-1) in human chondrosarcoma cells. BDNF-mediated cell motility and MMP-1 up-regulation were attenuated by Trk inhibitor (K252a), ASK1 inhibitor (thioredoxin), JNK inhibitor (SP600125), and p38 inhibitor (SB203580). Furthermore, BDNF also promoted Sp1 activation. Our results indicate that BDNF enhances the migration and invasion activity of chondrosarcoma cells by increasing MMP-1 expression through a signal transduction pathway that involves the TrkB receptor, ASK1, JNK/p38, and Sp1. BDNF thus represents a promising new target for treating chondrosarcoma metastasis.
Collapse
|