1
|
Shukla A, Sharma A, Gupta S, Mishra A, Singh A. Antitumor potential of ivermectin against T-cell lymphoma-bearing hosts. Med Oncol 2025; 42:169. [PMID: 40257544 DOI: 10.1007/s12032-025-02726-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Ivermectin, a broad-spectrum antiparasitic agent from the ivermectin family, has shown promising anticancer potential. Originally developed for veterinary and human use against parasitic infections, ivermectin demonstrated significant antitumor effects in our study against tumor cells (Dalton's lymphoma cells). A dose-dependent decrease in tumor cell viability was observed following 24-h treatment with ivermectin, with an IC₅₀ value calculated at 10.55 µg/mL. In comparison, the standard anticancer drug cisplatin exhibited a slightly higher cytotoxic potency, with an IC₅₀ of 8.32 µg/mL under the same treatment duration. Flow cytometric analysis revealed that ivermectin induced cell cycle arrest in the G0-G1 phase. Apoptotic tumor cell death was confirmed via Annexin V/PI staining, further supported by nuclear condensation, a hallmark of apoptosis, visualized through both confocal microscopy and flow cytometry. The apoptosis was determined to be mitochondrial-dependent, as evidenced by a decline in mitochondrial membrane potential (ΔΨm) observed through JC-1 assay. The treatment increased DAPI-positive cells and exhibited severe chromatin condensation. Additionally, cell death was validated using Acridine Orange and Propidium Iodide staining, which highlighted increased cell membrane rupture and death through apoptosis and necrosis. Mitochondrial dependent apoptosis further supported by increased ROS production upon ivermectin treatment. Moreover, In vivo, ivermectin treatment led to a substantial reduction in tumor size in tumor-bearing mice, along with normalization of spleen size, body weight, and improvement histopathology of liver. These findings collectively support the therapeutic potential of ivermectin as a repurposed anticancer agent, acting through multiple mechanisms including cell cycle arrest, ROS generation, mitochondrial dysfunction, and apoptosis.
Collapse
Affiliation(s)
- Alok Shukla
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Arpit Sharma
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
- Department of Pharmacology, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India
| | - Shivani Gupta
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Abha Mishra
- Biomolecular Engineering Laboratory, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, India.
| | - Amit Singh
- Department of Pharmacology, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India.
- Baba Kinaram Medical College, Chandauli, 232104, India.
| |
Collapse
|
2
|
Khalef L, Lydia R, Filicia K, Moussa B. Cell viability and cytotoxicity assays: Biochemical elements and cellular compartments. Cell Biochem Funct 2024; 42:e4007. [PMID: 38593323 DOI: 10.1002/cbf.4007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/01/2024] [Accepted: 03/29/2024] [Indexed: 04/11/2024]
Abstract
Cell viability and cytotoxicity assays play a crucial role in drug screening and evaluating the cytotoxic effects of various chemicals. The quantification of cell viability and proliferation serves as the cornerstone for numerous in vitro assays that assess cellular responses to external factors. In the last decade, several studies have developed guidelines for defining and interpreting cell viability and cytotoxicity based on morphological, biochemical, and functional perspectives. As this domain continues to experience ongoing growth, revealing new mechanisms orchestrating diverse cell cytotoxicity pathways, we suggest a revised classification for multiple assays employed in evaluating cell viability and cell death. This classification is rooted in the cellular compartment and/or biochemical element involved, with a specific focus on mechanistic and essential aspects of the process. The assays are founded on diverse cell functions, encompassing metabolic activity, enzyme activity, cell membrane permeability and integrity, adenosine 5'-triphosphate content, cell adherence, reduction equivalents, dye inclusion or exclusion, constitutive protease activity, colony formation, DNA fragmentation and nuclear splitting. These assays present straightforward, reliable, sensitive, reproducible, cost-effective, and high-throughput approaches for appraising the effects of newly formulated chemotherapeutic biomolecules on the cell survival during the drug development process.
Collapse
Affiliation(s)
- Lefsih Khalef
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| | - Radja Lydia
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| | - Khettar Filicia
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| | - Berkoud Moussa
- Département de Biochimie et Microbiologie, Laboratoire d'Ecologie, Biotechnologie et Santé, Université Mouloud Mammeri de Tizi ouzou, Tizi Ouzou, Algeria
| |
Collapse
|
3
|
Gregory CD. Hijacking homeostasis: Regulation of the tumor microenvironment by apoptosis. Immunol Rev 2023; 319:100-127. [PMID: 37553811 PMCID: PMC10952466 DOI: 10.1111/imr.13259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023]
Abstract
Cancers are genetically driven, rogue tissues which generate dysfunctional, obdurate organs by hijacking normal, homeostatic programs. Apoptosis is an evolutionarily conserved regulated cell death program and a profoundly important homeostatic mechanism that is common (alongside tumor cell proliferation) in actively growing cancers, as well as in tumors responding to cytotoxic anti-cancer therapies. Although well known for its cell-autonomous tumor-suppressive qualities, apoptosis harbors pro-oncogenic properties which are deployed through non-cell-autonomous mechanisms and which generally remain poorly defined. Here, the roles of apoptosis in tumor biology are reviewed, with particular focus on the secreted and fragmentation products of apoptotic tumor cells and their effects on tumor-associated macrophages, key supportive cells in the aberrant homeostasis of the tumor microenvironment. Historical aspects of cell loss in tumor growth kinetics are considered and the impact (and potential impact) on tumor growth of apoptotic-cell clearance (efferocytosis) as well as released soluble and extracellular vesicle-associated factors are discussed from the perspectives of inflammation, tissue repair, and regeneration programs. An "apoptosis-centric" view is proposed in which dying tumor cells provide an important platform for intricate intercellular communication networks in growing cancers. The perspective has implications for future research and for improving cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Christopher D. Gregory
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarterEdinburghUK
| |
Collapse
|
4
|
Stearns-Reider KM, Hicks MR, Hammond KG, Reynolds JC, Maity A, Kurmangaliyev YZ, Chin J, Stieg AZ, Geisse NA, Hohlbauch S, Kaemmer S, Schmitt LR, Pham TT, Yamauchi K, Novitch BG, Wollman R, Hansen KC, Pyle AD, Crosbie RH. Myoscaffolds reveal laminin scarring is detrimental for stem cell function while sarcospan induces compensatory fibrosis. NPJ Regen Med 2023; 8:16. [PMID: 36922514 PMCID: PMC10017766 DOI: 10.1038/s41536-023-00287-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
We developed an on-slide decellularization approach to generate acellular extracellular matrix (ECM) myoscaffolds that can be repopulated with various cell types to interrogate cell-ECM interactions. Using this platform, we investigated whether fibrotic ECM scarring affected human skeletal muscle progenitor cell (SMPC) functions that are essential for myoregeneration. SMPCs exhibited robust adhesion, motility, and differentiation on healthy muscle-derived myoscaffolds. All SPMC interactions with fibrotic myoscaffolds from dystrophic muscle were severely blunted including reduced motility rate and migration. Furthermore, SMPCs were unable to remodel laminin dense fibrotic scars within diseased myoscaffolds. Proteomics and structural analysis revealed that excessive collagen deposition alone is not pathological, and can be compensatory, as revealed by overexpression of sarcospan and its associated ECM receptors in dystrophic muscle. Our in vivo data also supported that ECM remodeling is important for SMPC engraftment and that fibrotic scars may represent one barrier to efficient cell therapy.
Collapse
Affiliation(s)
- Kristen M Stearns-Reider
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Michael R Hicks
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Katherine G Hammond
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Joseph C Reynolds
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Alok Maity
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yerbol Z Kurmangaliyev
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jesse Chin
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Adam Z Stieg
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | | | - Sophia Hohlbauch
- Asylum Research, An Oxford Instruments Company, Santa Barbara, CA, 93117, USA
| | - Stefan Kaemmer
- Park Systems, 3040 Olcott St, Santa Clara, CA, 95054, USA
| | - Lauren R Schmitt
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, 80045, USA
| | - Thanh T Pham
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, 80045, USA
| | - Ken Yamauchi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Bennett G Novitch
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Roy Wollman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, 80045, USA
| | - April D Pyle
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
5
|
Hu CY, Su BH, Lee YC, Wang CT, Yang ML, Shen WT, Fu JT, Chen SY, Huang WY, Ou CH, Tsai YS, Kuo FC, Shiau AL, Shieh GS, Wu CL. Interruption of the long non-coding RNA HOTAIR signaling axis ameliorates chemotherapy-induced cachexia in bladder cancer. J Biomed Sci 2022; 29:104. [PMID: 36471329 PMCID: PMC9724340 DOI: 10.1186/s12929-022-00887-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cisplatin-based chemotherapy is the first line of treatment for bladder cancer. However, cisplatin induces muscle wasting associated with NF-κB and cancer cachexia. HOTAIR, an oncogenic long non-coding RNA (lncRNA), promotes cancer progression in different cancers. Crosstalk between HOTAIR and NF-κB is documented. Prothymosin α (ProT) plays important roles in cancer progression and inflammation. However, the potential link between HOTAIR, ProT, and cisplatin-induced cancer cachexia remains unexplored. Here, we investigated the contribution of HOTAIR in cisplatin-induced cancer cachexia and dissected the potential signaling cascade involving the epidermal growth factor receptor (EGFR), ProT, NF-κB, and HOTAIR. MATERIALS AND METHODS Expression of ProT and HOTAIR transcripts and their correlations in tumor tissues of bladder cancer patients and bladder cancer cell lines were determined by RT-qPCR. Next, levels of phospho-EGFR, EGFR, phospho-NF-κB, and NF-κB were examined by immunoblot analysis in human bladder cancer cells treated with cisplatin. Expression of HOTAIR in cisplatin-treated cells was also assessed by RT-qPCR. Pharmacological inhibitors and overexpression and knockdown approaches were exploited to decipher the signaling pathway. The murine C2C12 myoblasts were used as an in vitro muscle atrophy model. The syngeneic murine MBT-2 bladder tumor was used to investigate the role of mouse Hotair in cisplatin-induced cancer cachexia. RESULTS Expression of ProT and HOTAIR was higher in bladder tumors than in normal adjacent tissues. There were positive correlations between ProT and HOTAIR expression in clinical bladder tumors and bladder cancer cell lines. Cisplatin treatment increased EGFR and NF-κB activation and upregulated ProT and HOTAIR expression in bladder cancer cells. ProT overexpression increased, whereas ProT knockdown decreased, HOTAIR expression. Notably, cisplatin-induced HOTAIR upregulation was abrogated by EGFR inhibitors or ProT knockdown. ProT-induced HOTAIR overexpression was diminished by NF-κB inhibitors. HOTAIR overexpression enhanced, whereas its knockdown reduced, cell proliferation, cachexia-associated pro-inflammatory cytokine expression, and muscle atrophy. Cachexia-associated symptoms were ameliorated in mice bearing Hotair-knockdown bladder tumors undergoing cisplatin treatment. CONCLUSIONS We demonstrate for the first time a critical role for HOTAIR and identify the involvement of the EGFR-ProT-NF-κB-HOTAIR signaling axis in cisplatin-induced cachexia in bladder cancer and likely other cancers. Our findings also provide therapeutic targets for this disease.
Collapse
Affiliation(s)
- Che-Yuan Hu
- grid.64523.360000 0004 0532 3255Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan ,grid.64523.360000 0004 0532 3255Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138, Sheng Li Road, Tainan, 704302 Taiwan
| | - Bing-Hua Su
- grid.412896.00000 0000 9337 0481School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ya-Che Lee
- grid.413878.10000 0004 0572 9327Department of Urology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Chung-Teng Wang
- grid.64523.360000 0004 0532 3255Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, 1 University Road, Tainan, 701401 Taiwan
| | - Mei-Lin Yang
- grid.64523.360000 0004 0532 3255Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, 1 University Road, Tainan, 701401 Taiwan ,grid.413878.10000 0004 0572 9327Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Wan-Ting Shen
- grid.64523.360000 0004 0532 3255Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, 1 University Road, Tainan, 701401 Taiwan
| | - Jing-Ting Fu
- grid.64523.360000 0004 0532 3255Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, 1 University Road, Tainan, 701401 Taiwan
| | - Shih-Yao Chen
- grid.411636.70000 0004 0634 2167Department of Nursing, College of Nursing, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Wei-Yun Huang
- grid.64523.360000 0004 0532 3255Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, 1 University Road, Tainan, 701401 Taiwan
| | - Chien-Hui Ou
- grid.64523.360000 0004 0532 3255Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138, Sheng Li Road, Tainan, 704302 Taiwan
| | - Yuh-Shyan Tsai
- grid.64523.360000 0004 0532 3255Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138, Sheng Li Road, Tainan, 704302 Taiwan
| | - Feng-Chih Kuo
- grid.260565.20000 0004 0634 0356Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ai-Li Shiau
- grid.64523.360000 0004 0532 3255Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, 1 University Road, Tainan, 701401 Taiwan ,grid.413878.10000 0004 0572 9327Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Gia-Shing Shieh
- grid.64523.360000 0004 0532 3255Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138, Sheng Li Road, Tainan, 704302 Taiwan ,grid.454740.6Department of Urology, Tainan Hospital, Ministry of Health and Welfare, Executive Yuan, Tainan, Taiwan
| | - Chao-Liang Wu
- grid.413878.10000 0004 0572 9327Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan ,grid.64523.360000 0004 0532 3255Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, 1 University Road, Tainan, 701401 Taiwan
| |
Collapse
|
6
|
Wang JL, Chen WG, Zhang JJ, Xu CJ. Nogo-A-Δ20/EphA4 interaction antagonizes apoptosis of neural stem cells by integrating p38 and JNK MAPK signaling. J Mol Histol 2021; 52:521-537. [PMID: 33555537 DOI: 10.1007/s10735-021-09960-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/25/2021] [Indexed: 11/26/2022]
Abstract
Nogo-A protein consists of two main extracellular domains: Nogo-66 (rat amino acid [aa] 1019-1083) and Nogo-A-Δ20 (extracellular, active 180 amino acid Nogo-A region), which serve as strong inhibitors of axon regeneration in the adult CNS (Central Nervous System). Although receptors S1PR2 and HSPGs have been identified as Nogo-A-Δ20 binding proteins, it remains at present elusive whether other receptors directly interacting with Nogo-A-Δ20 exist, and decrease cell death. On the other hand, the key roles of EphA4 in the regulation of glioblastoma, axon regeneration and NSCs (Neural Stem Cells) proliferation or differentiation are well understood, but little is known the relationship between EphA4 and Nogo-A-Δ20 in NSCs apoptosis. Thus, we aim to determine whether Nogo-A-Δ20 can bind to EphA4 and affect survival of NSCs. Here, we discover that EphA4, belonging to a member of erythropoietin-producing hepatocellular (Eph) receptors family, could be acting as a high affinity ligand for Nogo-A-Δ20. Trans-membrane protein of EphA4 is needed for Nogo-A-Δ20-triggered inhibition of NSCs apoptosis, which are mediated by balancing p38 inactivation and JNK MAPK pathway activation. Finally, we predict at the atomic level that essential residues Lys-205, Ile-190, Pro-194 in Nogo-A-Δ20 and EphA4 residues Gln-390, Asn-425, Pro-426 might play critical roles in Nogo-A-Δ20/EphA4 binding via molecular docking.
Collapse
Affiliation(s)
- Jun-Ling Wang
- Center for Reproductive Medicine, Affiliated Hospital 1 of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Wei-Guang Chen
- Department of Histology & Embryology, School of Basic Medical Science, Wenzhou Medical University, Cha Shan University Town, No.1 Central North Road, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Jia-Jia Zhang
- School of 1St Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Chao-Jin Xu
- Department of Histology & Embryology, School of Basic Medical Science, Wenzhou Medical University, Cha Shan University Town, No.1 Central North Road, Wenzhou, 325035, Zhejiang, People's Republic of China.
| |
Collapse
|
7
|
Joo MK, Shin S, Ye DJ, An HG, Kwon TU, Baek HS, Kwon YJ, Chun YJ. Combined treatment with auranofin and trametinib induces synergistic apoptosis in breast cancer cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2021; 84:84-94. [PMID: 33103613 DOI: 10.1080/15287394.2020.1835762] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Auranofin is a gold complex used as an anti-rheumatic agent and may act as a potent anticancer drug against breast tumors. Trametinib is a specific mitogen-activated protein kinase inhibitor, approved for the treatment of metastatic melanoma. The aim of this study was to examine the synergistic effects of auranofin and trametinib on apoptosis in MCF-7 human breast cancer cells. The combination treatment inhibited cancer cell proliferation and induced cell cycle arrest at the sub-G1 phase and apoptosis via poly (ADP-ribose) polymerase cleavage and caspase-3/7 activation. It is noteworthy that this treatment significantly increased p38 mitogen-activated protein kinase (MAPK) phosphorylation to induce mitochondrial stress, subsequently promoting cancer cell apoptosis through release of apoptosis-inducing factor. Further data demonstrated that combined treatment significantly induced increase in nuclear translocation of AIF. These results indicated that activation of the p38 MAPK signaling pathway and mitochondrial apoptosis may contribute to the synergistic consequences in MCF-7 cells. Collectively, our data demonstrated that combined treatment with auranofin and trametinib exhibited synergistic breast cancer cell death and this combination might be utilized as a novel therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Min-Kyung Joo
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Sangyun Shin
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Dong-Jin Ye
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Hong-Gyu An
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Tae-Uk Kwon
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Hyoung-Seok Baek
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Yeo-Jung Kwon
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Young-Jin Chun
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| |
Collapse
|
8
|
Kumar R, Saneja A, Panda AK. An Annexin V-FITC-Propidium Iodide-Based Method for Detecting Apoptosis in a Non-Small Cell Lung Cancer Cell Line. Methods Mol Biol 2021; 2279:213-223. [PMID: 33683697 DOI: 10.1007/978-1-0716-1278-1_17] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Annexin V and propidium iodide staining is widely used for determining the cellular death through apoptosis. In the presence of Ca2+ ions, annexin V has a strong binding affinity for phosphatidylserine, a membrane phospholipid that during apoptosis is translocated from the inner side of the cell membrane to its outer side. On the other hand, propidium iodide has ability for DNA binding and it can only enter into necrotic or late apoptotic cells. This chapter describes a commonly used method for detection of apoptosis in a non-small cell lung cancer cell line using annexin V and propidium iodide dye. We describe the detection of different stages of apoptosis in the A549 lung cancer cell line treated with dihydroartemisinin (DHA). This apoptosis detection method can be used to determine the efficacy of different kinds of drugs on cultured cancer cell lines.
Collapse
Affiliation(s)
- Robin Kumar
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Ankit Saneja
- Product Development Cell, National Institute of Immunology, New Delhi, India.
| | - Amulya K Panda
- Product Development Cell, National Institute of Immunology, New Delhi, India.
| |
Collapse
|
9
|
Biswas S, Ghosh S, Samanta A, Das S, Mukherjee U, Maitra S. Bisphenol A impairs reproductive fitness in zebrafish ovary: Potential involvement of oxidative/nitrosative stress, inflammatory and apoptotic mediators. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 267:115692. [PMID: 33254711 DOI: 10.1016/j.envpol.2020.115692] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) is a highly pervasive chemical in consumer products with its ascribed endocrine-disrupting properties. Several studies have shown the cytotoxic, genotoxic, and carcinogenic property of BPA over a multitude of tissues. Although BPA exposure has earlier been implicated in female infertility, the underlying molecular mechanisms explaining the toxicity of BPA in the ovary remains less understood. In the present study, a plausible correlation between redox balance or inflammatory signaling and reproductive fitness upon BPA exposure has been examined in zebrafish (Danio rerio) ovary. Congruent with significant alteration of major antioxidant enzymes (SOD1, SOD2, catalase, GPx1α, GSTα1) at the transcript level, 30 d BPA exposure at environmentally relevant concentrations (1, 10 and 100 μg L-1) promotes ovarian ROS/RNS synthesis, lipid peroxidation but attenuates catalase activity indicating elevated stress response. BPA promotes a sharp increase in ovarian p38 MAPK, NF-κB phosphorylation (activation), inducible nitric oxide synthase (Nos2a), and pro-inflammatory cytokines (TNF-α and IL-1β) expression, the reliable markers for inflammatory response. Congruent to an increased number of atretic follicles, BPA-exposed zebrafish ovary reveals elevated Bax/Bcl2 ratio, activation of caspase-8, -3 and DNA breakdown suggesting heightened cell death. Importantly, significant alteration in nuclear estrogen receptor (ER) transcripts (esr1, esr2a, and esr2b) and proteins (ERα, ERβ), gonadotropin receptors, and markers associated with steroidogenesis and growth factor gene expression in BPA-exposed ovary correlates well with impaired ovarian functions and maturational response. Collectively, elevated oxidative/nitrosative stress-mediated inflammatory response and altered ER expression can influence ovarian health and reproductive fitness in organisms exposed to BPA environment.
Collapse
Affiliation(s)
- Subhasri Biswas
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Soumyajyoti Ghosh
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Anwesha Samanta
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Sriparna Das
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Urmi Mukherjee
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Sudipta Maitra
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India.
| |
Collapse
|
10
|
TAK1 is involved in sodium L-lactate-stimulated p38 signaling and promotes apoptosis. Mol Cell Biochem 2020; 476:873-882. [PMID: 33111211 DOI: 10.1007/s11010-020-03952-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022]
Abstract
In the present study, we found that the phosphorylation of p38 mitogen-activated protein kinase (p38) was significantly increased in L-lactate-treated HeLa cells, which is under concentration- and time-dependent manner. The protein level of Bcl-2 was significantly reduced and Bax and C-caspase3 were significantly increased in L-lactate-treated cells. qRT-PCR analysis suggested that the expression level of apoptosis-related genes Bax, C-myc, and FasL were significantly upregulated by L-lactate treatment. In addition, p38 inhibitor SB203580 blocked the L-lactate-stimulated phosphorylation of p38 (p-p38) and apoptosis, which suggested that L-lactate-stimulated apoptosis may be related to the activation of p38. Moreover, TAK1 inhibitor Takinib reduced L-lactate-triggered phosphorylation of p38 and also apoptosis; however, ASK1 inhibitor NQDI-1 did not. Cells transfected with siRNA of TAK1(siTAK1) showed similar results with Takinib inhibitor. These results suggested that the L-lactate treatment elevated activation of p38 and apoptosis was related to TAK1. In this study, we suggested that TAK1 plays an important role in L-lactate-stimulated activation of p38 affecting apoptosis in HeLa cells.
Collapse
|
11
|
Huang YK, Chen SY, Wong MY, Chiu CH, Chu C. Pathogenicity differences of Salmonella enterica serovars Typhimurium, Enteritidis, and Choleraesuis-specific virulence plasmids and clinical S. Choleraesuis strains with large plasmids to the human THP-1 cell death. Microb Pathog 2018; 128:69-74. [PMID: 30583022 DOI: 10.1016/j.micpath.2018.12.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 11/17/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022]
Abstract
Salmonella is a common foodborne and zoonotic pathogen. Only a few serovars carry a virulence plasmid (pSV), which enhances the pathogenicity of the host. Here, we investigated the pathogenicity roles of the pSVs among wild-type, plasmid-less, and complemented S. Typhimurium, S. Enteritidis S. Choleraesuis in invasion, phagocytosis, and intracellular bacterial survival in human THP-1 cells and cell death patterns by flow cytometry and difference in cell death patterns between pig and human S. Choleraesuis isolates with large pSCVs. Virulence plasmid (pSTV) led to slightly increasing cellular apoptosis for S. Typhimurium; virulence plasmid (pSEV) enhanced apoptosis and necrosis significantly for S. Enteritidis; and pSCV reduced apoptosis significantly for S. Choleraesuis. After complementation, pSTV increased the intracellular survival of pSCV-less Choleraesuis and the cytotoxicity against human THP-1 cells. Using the Cytochalasin D to differentiate the invasion of S. Choleraaesuis and phagocytosis of THP-1 cells determined that pSCV were responsible for invasion and phagocytosis at 0 h and inhibited intracellular replication in THP-1 cells, and pSTV were responsible for invasion and increased intracellular survival for S. Choleraesuis in THP-1 cells. The human isolates with large pSCV induced more cellular apoptosis and necrosis than the pig isolates. In conclusion, human S. Choleraesuis isolates carrying large pSCVs were more adapted to human THP-1 cells for more cell death than pig isolates with large pSCV. The role of pSVs in invasion, phagocytosis, intracellular survival and apoptosis differed among hosted serovars.
Collapse
Affiliation(s)
- Yao-Kuang Huang
- Division of Thoracic and Cardiovascular Surgery, Chiayi Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Sheng-Ya Chen
- Division of Thoracic and Cardiovascular Surgery, Chiayi Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan; Department of Microbiology, Immunology, and Biopharmaceuticals, National Chiayi University, Chiayi, Taiwan
| | - Min Yi Wong
- Division of Thoracic and Cardiovascular Surgery, Chiayi Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Children's Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chishih Chu
- Department of Microbiology, Immunology, and Biopharmaceuticals, National Chiayi University, Chiayi, Taiwan.
| |
Collapse
|
12
|
Dakic A, DiVito K, Fang S, Suprynowicz F, Gaur A, Li X, Palechor-Ceron N, Simic V, Choudhury S, Yu S, Simbulan-Rosenthal CM, Rosenthal D, Schlegel R, Liu X. ROCK inhibitor reduces Myc-induced apoptosis and mediates immortalization of human keratinocytes. Oncotarget 2018; 7:66740-66753. [PMID: 27556514 PMCID: PMC5341834 DOI: 10.18632/oncotarget.11458] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/11/2016] [Indexed: 01/06/2023] Open
Abstract
The Myc/Max/Mad network plays a critical role in cell proliferation, differentiation and apoptosis and c-Myc is overexpressed in many cancers, including HPV-positive cervical cancer cell lines. Despite the tolerance of cervical cancer keratinocytes to high Myc expression, we found that the solitary transduction of the Myc gene into primary cervical and foreskin keratinocytes induced rapid cell death. These findings suggested that the anti-apoptotic activity of E7 in cervical cancer cells might be responsible for negating the apoptotic activity of over-expressed Myc. Indeed, our earlier in vitro studies demonstrated that Myc and E7 synergize in the immortalization of keratinocytes. Since we previously postulated that E7 and the ROCK inhibitor, Y-27632, were members of the same functional pathway in cell immortalization, we tested whether Y-27632 would inhibit apoptosis induced by the over-expression of Myc. Our findings indicate that Y-27632 rapidly inhibited Myc-induced membrane blebbing and cellular apoptosis and, more generally, functioned as an inhibitor of extrinsic and intrinsic pathways of cell death. Most important, Y-27632 cooperated with Myc to immortalize keratinocytes efficiently, indicating that apoptosis is a major barrier to Myc-induced immortalization of keratinocytes. The anti-apoptotic activity of Y-27632 correlated with a reduction in p53 serine 15 phosphorylation and the consequent reduction in the expression of downstream target genes p21 and DAPK1, two genes involved in the induction of cell death.
Collapse
Affiliation(s)
- Aleksandra Dakic
- Department of Pathology, Georgetown University Medical School, Washington, DC 20057, USA.,Center for Cell Reprogramming, Georgetown University Medical School, Washington, DC 20057, USA
| | - Kyle DiVito
- Department of Molecular and Cell Biology and Biochemistry, Georgetown University Medical School, Washington, DC 20057, USA
| | - Shuang Fang
- Department of Pathology, Georgetown University Medical School, Washington, DC 20057, USA.,Center for Cell Reprogramming, Georgetown University Medical School, Washington, DC 20057, USA
| | - Frank Suprynowicz
- Department of Pathology, Georgetown University Medical School, Washington, DC 20057, USA.,Center for Cell Reprogramming, Georgetown University Medical School, Washington, DC 20057, USA
| | - Anirudh Gaur
- Department of Molecular and Cell Biology and Biochemistry, Georgetown University Medical School, Washington, DC 20057, USA
| | - Xin Li
- Department of Biostatistics, Bioinformatics, Georgetown University Medical School, Washington, DC 20057, USA
| | - Nancy Palechor-Ceron
- Department of Pathology, Georgetown University Medical School, Washington, DC 20057, USA.,Center for Cell Reprogramming, Georgetown University Medical School, Washington, DC 20057, USA
| | - Vera Simic
- Department of Pathology, Georgetown University Medical School, Washington, DC 20057, USA.,Center for Cell Reprogramming, Georgetown University Medical School, Washington, DC 20057, USA
| | - Sujata Choudhury
- Department of Pathology, Georgetown University Medical School, Washington, DC 20057, USA.,Center for Cell Reprogramming, Georgetown University Medical School, Washington, DC 20057, USA
| | - Songtao Yu
- Department of Pathology, Georgetown University Medical School, Washington, DC 20057, USA.,Center for Cell Reprogramming, Georgetown University Medical School, Washington, DC 20057, USA
| | - Cynthia M Simbulan-Rosenthal
- Department of Molecular and Cell Biology and Biochemistry, Georgetown University Medical School, Washington, DC 20057, USA
| | - Dean Rosenthal
- Department of Molecular and Cell Biology and Biochemistry, Georgetown University Medical School, Washington, DC 20057, USA
| | - Richard Schlegel
- Department of Pathology, Georgetown University Medical School, Washington, DC 20057, USA.,Center for Cell Reprogramming, Georgetown University Medical School, Washington, DC 20057, USA
| | - Xuefeng Liu
- Department of Pathology, Georgetown University Medical School, Washington, DC 20057, USA.,Center for Cell Reprogramming, Georgetown University Medical School, Washington, DC 20057, USA
| |
Collapse
|
13
|
Djiadeu P, Farmakovski N, Azzouz D, Kotra LP, Sweezey N, Palaniyar N. Surfactant protein D regulates caspase-8-mediated cascade of the intrinsic pathway of apoptosis while promoting bleb formation. Mol Immunol 2017; 92:190-198. [PMID: 29107869 DOI: 10.1016/j.molimm.2017.10.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/17/2017] [Accepted: 10/17/2017] [Indexed: 01/10/2023]
Abstract
Surfactant-associated protein D (SP-D) is a soluble innate immune collectin present on many mucosal surfaces. We recently showed that SP-D suppresses the extrinsic pathway of apoptosis by downregulating caspase-8 activation. However, the effects of SP-D on the intrinsic pathway of apoptosis are not clearly understood. In the intrinsic pathway, cytochrome c is released by mitochondria into the cytoplasm. Oxidation of cytochrome c by cytochrome c oxidase activates the apoptosome and caspase-9 cascade. Both caspase-8- and caspase-9-mediated branches are activated in the intrinsic pathway of apoptosis; however, little is known about the relevance of the caspase-8 pathway in this context. Here we studied the effects of SP-D on different branches of the intrinsic pathway of apoptosis using UV-irradiated Jurkat T-cells. We found that SP-D does not inhibit the caspase-9 branch of apoptosis and the relevance of the caspase-8-related branch became apparent when the caspase-9 pathway was inhibited by blocking cytochrome c oxidase. Under these conditions, SP-D reduces the activation of caspase-8, executioner caspase-3 and exposure of phosphatidylserine (PS) on the membranes of dying cells. By contrast, SP-D increases the formation of nuclear and membrane blebs. Inhibition of caspase-8 confirms the effect of SP-D is unique to the caspase-8 pathway. Overall, SP-D suppresses certain aspects of the intrinsic pathway of apoptosis via reduction of caspase-8 activation and PS flipping while at the same time increasing membrane and nuclear bleb formation. This novel regulatory aspect of SP-D could help to regulate intrinsic pathway of apoptosis to promote effective blebbing and breakdown of dying cells.
Collapse
Affiliation(s)
- Pascal Djiadeu
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada; Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
| | - Nicole Farmakovski
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada
| | - Dhia Azzouz
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada
| | - Lakshmi P Kotra
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada; Center for Molecular Design and Preformulations, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Neil Sweezey
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada; Departments of Paediatrics, Physiology and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Nades Palaniyar
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada; Departments of Laboratory Medicine and Pathobiology and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
14
|
Downregulation of MMP1 in MDS-derived mesenchymal stromal cells reduces the capacity to restrict MDS cell proliferation. Sci Rep 2017; 7:43849. [PMID: 28262842 PMCID: PMC5338350 DOI: 10.1038/srep43849] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/27/2017] [Indexed: 12/12/2022] Open
Abstract
The role of mesenchymal stromal cells (MSCs) in the pathogenesis of myelodysplastic syndromes (MDS) has been increasingly addressed, but has yet to be clearly elucidated. In this investigation, we found that MDS cells proliferated to a greater extent on MDS-derived MSCs compared to normal MSCs. Matrix metalloproteinase 1(MMP1), which was downregulated in MDS-MSCs, was identified as an inhibitory factor of MDS cell proliferation, given that treatment with an MMP1 inhibitor or knock-down of MMP1 in normal MSCs resulted in increased MDS cell proliferation. Further investigations indicated that MMP1 induced apoptosis of MDS cells by interacting with PAR1 and further activating the p38 MAPK pathway. Inhibition of either PAR1 or p38 MAPK can reverse the apoptosis-inducing effect of MMP1. Taken together, these data indicate that downregulation of MMP1 in MSCs of MDS patients may contribute to the reduced capacity of MSCs to restrict MDS cell proliferation, which may account for the malignant proliferation of MDS cells.
Collapse
|
15
|
Lee D, Lee DS, Jung K, Hwang GS, Lee HL, Yamabe N, Lee HJ, Eom DW, Kim KH, Kang KS. Protective effect of ginsenoside Rb1 against tacrolimus-induced apoptosis in renal proximal tubular LLC-PK1 cells. J Ginseng Res 2017; 42:75-80. [PMID: 29348725 PMCID: PMC5766699 DOI: 10.1016/j.jgr.2016.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/05/2016] [Accepted: 12/26/2016] [Indexed: 02/04/2023] Open
Abstract
Background The aim of the present study was to evaluate the potential protective effects of six ginsenosides (Rb1, Rb2, Rc, Rd, Rg1, and Rg3) isolated from Panax ginseng against tacrolimus (FK506)-induced apoptosis in renal proximal tubular LLC-PK1 cells. Methods LLC-PK1 cells were treated with FK506 and ginsenosides, and cell viability was measured. Protein expressions of mitogen-activated protein kinases, caspase-3, and kidney injury molecule-1 (KIM-1) were evaluated by Western blotting analyses. The number of apoptotic cells was measured using an image-based cytometric assay. Results Reduction in cell viability by 60μM FK506 was ameliorated significantly by cotreatment with ginsenosides Rg1 and Rb1. The phosphorylation of p38, extracellular signal-regulated kinases, and KIM-1, and cleavage of caspase-3, increased markedly in LLC-PK1 cells treated with FK506 and significantly decreased after cotreatment with ginsenoside Rb1. The number of apoptotic cells decreased by 6.0% after cotreatment with ginsenoside Rb1 (10μM and 50μM). Conclusion The antiapoptotic effects of ginsenoside Rb1 on FK506-induced apoptosis were mediated by the inhibition of mitogen-activated protein kinases and caspase activation.
Collapse
Affiliation(s)
- Dahae Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Dong-Soo Lee
- Institute of Human-Environment Interface Biology, Biomedical Research Institute, Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kiwon Jung
- College of Pharmacy, CHA University, Pocheon, Republic of Korea
| | - Gwi Seo Hwang
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| | - Hye Lim Lee
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| | - Noriko Yamabe
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| | - Hae-Jeong Lee
- Department of Food and Nutrition, Gachon University, Seongnam, Republic of Korea
| | - Dae-Woon Eom
- Department of Pathology, University of Ulsan College of Medicine, Gangneung, Republic of Korea
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam, Republic of Korea
| |
Collapse
|
16
|
Mitogen-activated protein kinases are involved in hepatocanalicular dysfunction and cholestasis induced by oxidative stress. Arch Toxicol 2016; 91:2391-2403. [PMID: 27913845 DOI: 10.1007/s00204-016-1898-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/24/2016] [Indexed: 12/22/2022]
Abstract
In previous studies, we showed that the pro-oxidant model agent tert-butyl hydroperoxide (tBuOOH) induces alterations in hepatocanalicular secretory function by activating Ca2+-dependent protein kinase C isoforms (cPKC), via F-actin disorganization followed by endocytic internalization of canalicular transporters relevant to bile formation (Mrp2, Bsep). Since mitogen-activated protein kinases (MAPKs) may be downstream effectors of cPKC, we investigated here the involvement of the MAPKs of the ERK1/2, JNK1/2, and p38MAPK types in these deleterious effects. tBuOOH (100 µM, 15 min) increased the proportion of the active, phosphorylated forms of ERK1/2, JNK1/2, and p38MAPK, and panspecific PKC inhibition with bisindolylmaleimide-1 (100 nM) or selective cPKC inhibition with Gö6976 (1 μM) prevented the latter two events. In isolated rat hepatocyte couplets, tBuOOH (100 µM, 15 min) decreased the canalicular vacuolar accumulation of the fluorescent Bsep and Mrp2 substrates, cholylglycylamido fluorescein, and glutathione-methylfluorescein, respectively, and selective inhibitors of ERK1/2 (PD098059), JNK1/2 (SP600125), and p38MAPK (SB203580) partially prevented these alterations. In in situ perfused rat livers, these three MAPK inhibitors prevented tBuOOH (75 µM)-induced impairment of bile flow and the decrease in the biliary output of the Bsep and Mrp2 substrates, taurocholate, and dinitrophenyl-S-glutathione, respectively. The changes in Bsep/Mrp2 and F-actin localization induced by tBuOOH, as assessed by (immuno)fluorescence staining followed by analysis of confocal images, were prevented total or partially by the MAPK inhibitors. We concluded that MAPKs of the ERK1/2, JNK1/2, and p38MAPK types are all involved in cholestasis induced by oxidative stress, by promoting F-actin rearrangement and further endocytic internalization of canalicular transporters critical for bile formation.
Collapse
|
17
|
Chen B, Tan Y, Liang Y, Li Y, Chen L, Wu S, Xu W, Wang Y, Zhao W, Wu J. Per2 participates in AKT-mediated drug resistance in A549/DDP lung adenocarcinoma cells. Oncol Lett 2016; 13:423-428. [PMID: 28123577 DOI: 10.3892/ol.2016.5430] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 04/15/2016] [Indexed: 01/29/2023] Open
Abstract
Period2 (Per2) is a key mammalian circadian clock protein, and additionally has a tumor suppressive function. The present study aimed to investigate its role in drug resistance in A549/cisplatin (DDP) lung adenocarcinoma cells. Per2 knockdown and overexpression in A549/DDP cells were used to compare cell proliferation (by MTT assay), apoptosis (active-caspase 3 western blot) and clone forming assay. The activation of AKT/mechanistic target of rapamycin (mTOR) was investigated by a western blot assay. The Per2 expression level was decreased in A549/DDP cells compared with A549 cells. Per2 knockdown by short hairpin RNA protects A549/DDP cells from apoptosis, and promotes proliferation and migration. Per2 knockdown results in increased activation of the phosphoinositide 3-kinase (PI3K)/AKT/mTOR signaling pathway. Overexpression of Per2 in A549/DDP cells may reduce the activity of the PI3K/AKT/mTOR signaling pathway, and promote apoptosis of A549 cells. The results of the present study suggest that Per2 participates in AKT-mediated drug resistance in A549/DDP lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Bo Chen
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yaoxi Tan
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Liang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Li
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lei Chen
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shuangshuang Wu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Xu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Wang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Weihong Zhao
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jianqing Wu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
18
|
Huang WC, Chen SH, Chiang WH, Huang CW, Lo CL, Chern CS, Chiu HC. Tumor Microenvironment-Responsive Nanoparticle Delivery of Chemotherapy for Enhanced Selective Cellular Uptake and Transportation within Tumor. Biomacromolecules 2016; 17:3883-3892. [DOI: 10.1021/acs.biomac.6b00956] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Wen-Chia Huang
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Shih-Hong Chen
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
- Department
of Anesthesiology, National Taiwan University Hospital−Hsinchu Branch, Hsinchu 300, Taiwan
| | - Wen-Hsuan Chiang
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Chu-Wei Huang
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Chun-Liang Lo
- Department
of Biomedical Engineering, National Yang-Ming University, Taipei 112, Taiwan
| | - Chorng-Shyan Chern
- Department
of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106, Taiwan
| | - Hsin-Cheng Chiu
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
19
|
Li J, Yang Z, Li Y, Xia J, Li D, Li H, Ren M, Liao Y, Yu S, Chen Y, Yang Y, Zhang Y. Cell apoptosis, autophagy and necroptosis in osteosarcoma treatment. Oncotarget 2016; 7:44763-44778. [PMID: 27007056 PMCID: PMC5190133 DOI: 10.18632/oncotarget.8206] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 03/07/2016] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma is the most common primary bone tumor in children and adolescents. Although combined therapy including surgery and multi-agent chemotherapy have resulted in great improvements in the overall survival of patients, chemoresistance remains an obstacle for the treatment of osteosarcoma. Molecular targets or effective agents that are actively involved in cell death including apoptosis, autophagy and necroptosis have been studied. We summarized how these agents (novel compounds, miRNAs, or proteins) regulate apoptotic, autophagic and necroptotic pathways; and discussed the current knowledge on the role of these new agents in chemotherapy resistance in osteosarcoma.
Collapse
Affiliation(s)
- Jing Li
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| | - Zuozhang Yang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Yi Li
- Department of Oncology, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan, China
| | - Junfeng Xia
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Dongqi Li
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Huiling Li
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Mingyan Ren
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Yedan Liao
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Shunling Yu
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Yanjin Chen
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Yihao Yang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| | - Ya Zhang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China
| |
Collapse
|
20
|
Lee WH, Choong LY, Mon NN, Lu S, Lin Q, Pang B, Yan B, Krishna VSR, Singh H, Tan TZ, Thiery JP, Lim CT, Tan PBO, Johansson M, Harteneck C, Lim YP. TRPV4 Regulates Breast Cancer Cell Extravasation, Stiffness and Actin Cortex. Sci Rep 2016; 6:27903. [PMID: 27291497 PMCID: PMC4904279 DOI: 10.1038/srep27903] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 05/26/2016] [Indexed: 12/28/2022] Open
Abstract
Metastasis is a significant health issue. The standard mode of care is combination of chemotherapy and targeted therapeutics but the 5-year survival rate remains low. New/better drug targets that can improve outcomes of patients with metastatic disease are needed. Metastasis is a complex process, with each step conferred by a set of genetic aberrations. Mapping the molecular changes associated with metastasis improves our understanding of the etiology of this disease and contributes to the pipeline of targeted therapeutics. Here, phosphoproteomics of a xenograft-derived in vitro model comprising 4 isogenic cell lines with increasing metastatic potential implicated Transient Receptor Potential Vanilloid subtype 4 in breast cancer metastasis. TRPV4 mRNA levels in breast, gastric and ovarian cancers correlated with poor clinical outcomes, suggesting a wide role of TRPV4 in human epithelial cancers. TRPV4 was shown to be required for breast cancer cell invasion and transendothelial migration but not growth/proliferation. Knockdown of Trpv4 significantly reduced the number of metastatic nodules in mouse xenografts leaving the size unaffected. Overexpression of TRPV4 promoted breast cancer cell softness, blebbing, and actin reorganization. The findings provide new insights into the role of TRPV4 in cancer extravasation putatively by reducing cell rigidity through controlling the cytoskeleton at the cell cortex.
Collapse
Affiliation(s)
- Wen Hsin Lee
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lee Yee Choong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Naing Naing Mon
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - SsuYi Lu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Brendan Pang
- Cancer Science Institute of Singapore, Singapore
| | - Benedict Yan
- National University Hospital, Department of Laboratory Medicine, Singapore
| | | | - Himanshu Singh
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, Singapore
| | - Jean Paul Thiery
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cancer Science Institute of Singapore, Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | | | | | - Christian Harteneck
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| | - Yoon Pin Lim
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
- National University Cancer Institute, National University Health System, Singapore
| |
Collapse
|
21
|
Wang Z, Luo S, Wan Z, Chen C, Zhang X, Li B, Huang G, Chen L, He Z, Huang Z. Glabridin arrests cell cycle and inhibits proliferation of hepatocellular carcinoma by suppressing braf/MEK signaling pathway. Tumour Biol 2016; 37:5837-5846. [PMID: 26586395 DOI: 10.1007/s13277-015-4177-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/01/2015] [Indexed: 12/19/2022] Open
Abstract
Glabridin, an isoflavone isolated from licorice, owns a variety of pharmacological effects. Several reports have demonstrated that glabridin could regulate multiple cellular signaling pathways to inhibit the progression of cancer. However, the target proteins have not been elucidated yet. We used shape screening and induced fit docking to screen the protein data bank against glabridin. Braf and MEK1/2, important intermediate molecules of the braf/MEK cascade, were identified as the potential targets of glabridin. The experimental data showed that glabridin could inhibit the phosphorylation of MEK1/2 and the phosphorylation levels of downstream molecules including ERK1/2 and transcription factors ATF1 and CREB, but had no effect on the phosphorylation of braf. In particular, the in vitro pull-down assay indicated that glabridin selectively bound to braf and MEK1/2. What is more, exposure to glabridin significantly suppressed the proliferation of hepatocellular carcinoma HepG2 cell line. In addition, glabridin might arrest cell cycle in G1 through downregulation of cyclinD3, CDK2, and CDK4. In conclusion, glabridin is a potential multi-molecule-targeting inhibitor in the field of clinical prevention or treatment of cancer.
Collapse
Affiliation(s)
- Ziyou Wang
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China
- Department of Pathophysiology, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Shengqun Luo
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China
- Department of Pathophysiology, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Zheng Wan
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China
| | - Chuyan Chen
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China
| | - Xiangning Zhang
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China
- Department of Pathophysiology, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Binbin Li
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China
- Department of Pathophysiology, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - GuoLiang Huang
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China
| | - Liyong Chen
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China
| | - Zhiwei He
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China.
- Department of Pathophysiology, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China.
| | - Zunnan Huang
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, China.
- Department of Pathophysiology, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China.
| |
Collapse
|
22
|
Agarwal T, Narayana SGH, Pal K, Pramanik K, Giri S, Banerjee I. Calcium alginate-carboxymethyl cellulose beads for colon-targeted drug delivery. Int J Biol Macromol 2015; 75:409-17. [DOI: 10.1016/j.ijbiomac.2014.12.052] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 02/08/2023]
|
23
|
Arrowsmith RL, Atkin AJ, Botchway SW, Fairlamb IJS, Lynam JM, Moir JWB, Pascu SI, Ward JS, Zhang WQ. Confocal and fluorescence lifetime imaging sheds light on the fate of a pyrene-tagged carbon monoxide-releasing Fischer carbene chromium complex. Dalton Trans 2015; 44:4957-62. [PMID: 25553721 DOI: 10.1039/c4dt03312d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The synthesis of a new pyrene-containing Fischer carbene complex is described. The complex has a broad absorbance spectrum between 300 and 400 nm and, on excitation at 345 nm in CH2Cl2 solution, emission is observed at 395 and 415 nm. Emission is also observed in PBS buffer, but in this case the resulting spectra are much broader. Confocal and fluorescence lifetime imaging indicate that emission occurs on treating HeLa cells with the complex and co-localisation studies demonstrate that this is from the mitochondria and lipid-rich regions of the cell.
Collapse
Affiliation(s)
- Rory L Arrowsmith
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Angiotensin II receptor type 1 autoantibodies promote endothelial microparticles formation through activating p38 MAPK pathway. J Hypertens 2014; 32:762-70. [PMID: 24609215 DOI: 10.1097/hjh.0000000000000083] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Endothelial microparticles (EMPs) are small vesicular structures that serve as a marker of endothelial function. Angiotensin II receptor type 1 autoantibody (AT1-AA) can cause endothelial dysfunction. However, whether AT1-AA promotes EMPs formation and the mechanism remains obscure. METHODS The titres of sera AT1-AA of 126 hypertensive patients and 30 normotensive individuals were evaluated by ELISA. EMPs in the sera and the supernatants of human umbilical vein endothelial cells (HUVECs) were measured by flow cytometry. The phosphorylation levels of mitogen-activated protein kinase (MAPK) pathways in HUVECs treated by AT1-AA were assessed and their correlation with microparticle formation was also analysed. Furthermore, the production of intracellular reactive oxygen species (ROS) and nitric oxide in HUVECs was examined after incubation with 'injured' endothelial microparticle (iEMPs) (EMPs derived from AT1-AA treated HUVECs). RESULTS The positive rate of AT1-AA in 126 hypertensive patients was 21.4% (27/126), and higher than that in normotensive individuals [3.3% (1/30), P < 0.01]. Circulating EMP (CD31+/CD42b-) levels were corresponding to the AT1-AA titres in hypertensive group (r(2) = 0.3661, P < 0.01). AT1-AA promoted EMPs generation from HUVECs in a time and dose-dependent manner than the vehicle or nonspecific IgG. Meanwhile, AT1-AA significantly elevated phosphorylation level of P38 and ERK in HUVECs. Lorsartan and P38 inhibitor could suppress the AT1-AA's stimulation effect on EMPs generation. Moreover, the iEMP greatly increased ROS production and reduced nitric oxide synthesis in HUVECs. CONCLUSION Our findings showed that circulating EMPs levels positively correlate to the serum AT1-AA titres in essential hypertension patients. The AT1-AA could promote EMPs generation in HUVECs through activation of P38 MAPK signalling pathway, and this effect could be effectively inhibited by losartan or p38 inhibitor. Angiotensin receptor blockers (ARBs) may be more suitable for AT1-AA(+) hypertensive patients on account of suppressing the AT1-AA's stimulation effect on EMPs generation.
Collapse
|
25
|
Yen SY, Tseng JK, Chuang SM, Chen SE, Ju JC. Expression and activation of mitogen-activated protein kinases in matured porcine oocytes under thermal stress. J Reprod Dev 2014; 60:388-94. [PMID: 25087868 PMCID: PMC4219997 DOI: 10.1262/jrd.2014-004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we determined the expression and activation of p38 MAPK in matured porcine oocytes subjected to heat shock
(HS). When MII oocytes were heated, only the phosphorylated p38 levels relative to the total p38 levels decreased (P <
0.01) after HS, but no clear relationship with HS treatments was observed in the ERK, JNK and p90rsk expressions
of matured oocytes. To confirm p38 activation in matured oocytes, immunocytochemical staining was performed to localize its
expression and distribution in the ooplasm, and the results were largely consistent with previous Western blot analyses.
Moreover, when matured oocytes were co-cultured with a P38 MAPK inhibitor, SB203580, for 4 h at 41.5 C, the activation of its
immediate downstream substrate MAPKAPK-2 was not inhibited within any of the treatment groups. It appears that the MAPKAPK2
levels increased only under prolonged culture (HS4h and C4h) compared with the control group. In conclusion, p38 activity in
porcine oocytes was decreased after exposure to HS and prolonged culture. These alterations of p38 and activation of MAPKAPK2
may be associated with porcine oocyte viability under HS conditions, and a potential cross-talk between p38 MAPK and other
signaling cascades may exist, which warrants additional investigation.
Collapse
Affiliation(s)
- Shih-Ying Yen
- Department of Animal Science, National Chung Hsing University, Taichung 402, Taiwan, ROC
| | | | | | | | | |
Collapse
|
26
|
Heo SK, Noh EK, Yoon DJ, Jo JC, Park JH, Kim H. Dasatinib accelerates valproic acid-induced acute myeloid leukemia cell death by regulation of differentiation capacity. PLoS One 2014; 9:e98859. [PMID: 24918603 PMCID: PMC4053340 DOI: 10.1371/journal.pone.0098859] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 05/07/2014] [Indexed: 11/19/2022] Open
Abstract
Dasatinib is a compound developed for chronic myeloid leukemia as a multi-targeted kinase inhibitor against wild-type BCR-ABL and SRC family kinases. Valproic acid (VPA) is an anti-epileptic drug that also acts as a class I histone deacetylase inhibitor. The aim of this research was to determine the anti-leukemic effects of dasatinib and VPA in combination and to identify their mechanism of action in acute myeloid leukemia (AML) cells. Dasatinib was found to exert potent synergistic inhibitory effects on VPA-treated AML cells in association with G1 phase cell cycle arrest and apoptosis induction involving the cleavage of poly (ADP-ribose) polymerase and caspase-3, -7 and -9. Dasatinib/VPA-induced cell death thus occurred via caspase-dependent apoptosis. Moreover, MEK/ERK and p38 MAPK inhibitors efficiently inhibited dasatinib/VPA-induced apoptosis. The combined effect of dasatinib and VPA on the differentiation capacity of AML cells was more powerful than the effect of each drug alone, being sufficiently strong to promote AML cell death through G1 cell cycle arrest and caspase-dependent apoptosis. MEK/ERK and p38 MAPK were found to control dasatinib/VPA-induced apoptosis as upstream regulators, and co-treatment with dasatinib and VPA to contribute to AML cell death through the regulation of differentiation capacity. Taken together, these results indicate that combined dasatinib and VPA treatment has a potential role in anti-leukemic therapy.
Collapse
Affiliation(s)
- Sook-Kyoung Heo
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Eui-Kyu Noh
- Division of Hematology and Hematological Malignancies, Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Dong-Joon Yoon
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Jae-Cheol Jo
- Division of Hematology and Hematological Malignancies, Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Jae-Hoo Park
- Division of Hematology and Hematological Malignancies, Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Hawk Kim
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
- Division of Hematology and Hematological Malignancies, Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
- * E-mail:
| |
Collapse
|
27
|
Signalling mechanisms involved in renal pathological changes during cisplatin-induced nephropathy. Eur J Clin Pharmacol 2013; 69:1863-74. [PMID: 23929259 DOI: 10.1007/s00228-013-1568-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/24/2013] [Indexed: 12/20/2022]
Abstract
CONTEXT Cisplatin, a coordination platinum complex, is used as a potential anti-neoplastic agent, having well recognized DNA-damaging property that triggers cell-cycle arrest and cell death in cancer therapy. Beneficial chemotherapeutic actions of cisplatin can be detrimental for kidneys. BACKGROUND Unbound cisplatin gets accumulated in renal tubular cells, leading to cell injury and death. This liable action of cisplatin on kidneys is mediated by altered intracellular signalling pathways such as mitogen-activated protein kinase (MAPK), extracellular regulated kinase (ERK), or C- Jun N terminal kinase/stress-activated protein kinase (JNK/SAPK). Further, these signalling alterations are responsible for release and activation of tumour necrosis factor (TNF-α), mitochondrial dysfunction, and apoptosis, which ultimately cause the renal pathogenic process. Cisplatin itself enhances the generation of reactive oxygen species (ROS) and activation of nuclear factor-κB (NF-κB), inflammation, and mitochondrial dysfunction, which further leads to renal apoptosis. Cisplatin-induced nephropathy is also mediated through the p53 and protein kinase-Cδ (PKCδ) signalling pathways. OBJECTIVE This review explores these signalling alterations and their possible role in the pathogenesis of cisplatin-induced renal injury.
Collapse
|
28
|
α-Mangostin induces mitochondrial dependent apoptosis in human hepatoma SK-Hep-1 cells through inhibition of p38 MAPK pathway. Apoptosis 2013; 18:1548-60. [DOI: 10.1007/s10495-013-0888-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
29
|
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, Lee SJ. Apigenin induces c-Myc-mediated apoptosis in FRO anaplastic thyroid carcinoma cells. Mol Cell Endocrinol 2013; 369:130-9. [PMID: 23376608 DOI: 10.1016/j.mce.2013.01.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 01/14/2013] [Accepted: 01/17/2013] [Indexed: 01/11/2023]
Abstract
Apigenin promotes apoptosis in cancer cells. We studied the effect of apigenin on cell survival and c-Myc expression in FRO anaplastic thyroid carcinoma (ATC) cells. Apigenin caused apoptosis via the elevation of c-Myc levels in conjunction with the phosphorylation of p38 and p53. In the c-Myc siRNA-transfected and apigenin-treated cells, compared with the apigenin-treated control cells, apoptosis and phosphorylation of p38 and p53 were ameliorated. In the presence of apigenin, diminution of p38 and p53 did not affect cell survival although apigenin activated the phosphorylation of p38 and p53 via increased c-Myc levels. In conclusion, our results indicate that apigenin induces apoptosis mediated via c-Myc with concomitant phosphorylation of p53 and p38 in FRO ATC cells. These findings suggest that augmented c-Myc acts as a core regulator and is necessary for apigenin-induced apoptosis in FRO ATC cells.
Collapse
Affiliation(s)
- Si Hyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon 200-704, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
30
|
Salicylate Selectively Kills Cochlear Spiral Ganglion Neurons by Paradoxically Up-regulating Superoxide. Neurotox Res 2013; 24:307-19. [DOI: 10.1007/s12640-013-9384-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 02/11/2013] [Accepted: 02/13/2013] [Indexed: 12/12/2022]
|
31
|
Dey H, Liu ZR. Phosphorylation of p68 RNA helicase by p38 MAP kinase contributes to colon cancer cells apoptosis induced by oxaliplatin. BMC Cell Biol 2012; 13:27. [PMID: 23110695 PMCID: PMC3519718 DOI: 10.1186/1471-2121-13-27] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 10/15/2012] [Indexed: 12/28/2022] Open
Abstract
Background We previously demonstrated that p68 phosphorylation at threonine residues correlates with cancer cell apoptosis under the treatments of TNF-α and TRAIL (Yang, L. Mol Cancer Res Vol 3, pp 355–63 2005). Results In this report, we characterized the role of p68 phosphorylation in apoptosis induction under the treatment of oxaliplatin in the colon cancer cells. Our data suggest that oxaliplatin treatment activates p38 MAP kinase, which subsequently phosphorylates p68 at T564 and/or T446. The phosphorylation of p68, at least partially, mediates the effects of the drug on apoptosis induction, as mutations at these two sites greatly reduce the cancer cell death. Conclusion Our studies reveal an important molecular mechanism that mediates the effects of anti-cancer drug, providing a potential strategy for improving cancer treatment.
Collapse
Affiliation(s)
- Heena Dey
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | | |
Collapse
|
32
|
Harnett CC, Guerin PJ, Furtak T, Gauthier ER. Control of late apoptotic events by the p38 stress kinase in L-glutamine-deprived mouse hybridoma cells. Cell Biochem Funct 2012; 31:417-26. [PMID: 23080342 DOI: 10.1002/cbf.2916] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 09/18/2012] [Accepted: 09/19/2012] [Indexed: 01/08/2023]
Abstract
L-Glutamine (Gln) starvation rapidly triggers apoptosis in Sp2/0-Ag14 (Sp2/0) murine hybridoma cells. Here, we report on the role played by the stress-activated kinase p38 mitogen-activated protein kinase (MAPK) in this process. p38 activation was detected 2 h after Gln withdrawal and, although treatment with the p38 inhibitor SB203580 did not prevent caspase activation in Gln-starved cells, it reduced the occurrence of both nuclear condensation/fragmentation and apoptotic body formation. Similarly, transfection of Sp2/0 cells with a dominant negative p38 MAPK reduced the incidence of nuclear pyknosis and apoptotic body formation following 2 h of Gln starvation. Gln withdrawal-induced apoptosis was blocked by the overexpression of the anti-apoptotic protein Bcl-xL or by the caspase inhibitor Z-VAD-fmk. Interestingly, Bcl-xL expression inhibited p38 activation, but Z-VAD-fmk treatment did not, indicating that activation of this MAPK occurs downstream of mitochondrial dysfunction and is independent of caspases. Moreover, the anti-oxidant N-acetyl-l-cysteine prevented p38 phosphorylation, showing that p38 activation is triggered by an oxidative stress. Altogether, our findings indicate that p38 MAPK does not contribute to the induction of apoptosis in Gln-starved Sp2/0 cells. Rather, Gln withdrawal leads to mitochondrial dysfunction, causing an oxidative stress and p38 activation, the latter contributing to the formation of late morphological features of apoptotic Sp2/0 cells.
Collapse
Affiliation(s)
- Curtis C Harnett
- Biomolecular Sciences Program, Laurentian University, Sudbury, ON, Canada
| | | | | | | |
Collapse
|
33
|
Sheikh AQ, Taghian T, Hemingway B, Cho H, Kogan AB, Narmoneva DA. Regulation of endothelial MAPK/ERK signalling and capillary morphogenesis by low-amplitude electric field. J R Soc Interface 2012; 10:20120548. [PMID: 22993248 DOI: 10.1098/rsif.2012.0548] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Low-amplitude electric field (EF) is an important component of wound-healing response and can promote vascular tissue repair; however, the mechanisms of action on endothelium remain unclear. We hypothesized that physiological amplitude EF regulates angiogenic response of microvascular endothelial cells via activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway. A custom set-up allowed non-thermal application of EF of high (7.5 GHz) and low (60 Hz) frequency. Cell responses following up to 24 h of EF exposure, including proliferation and apoptosis, capillary morphogenesis, vascular endothelial growth factor (VEGF) expression and MAPK pathways activation were quantified. A db/db mouse model of diabetic wound healing was used for in vivo validation. High-frequency EF enhanced capillary morphogenesis, VEGF release, MEK-cRaf complex formation, MEK and ERK phosphorylation, whereas no MAPK/JNK and MAPK/p38 pathways activation was observed. The endothelial response to EF did not require VEGF binding to VEGFR2 receptor. EF-induced MEK phosphorylation was reversed in the presence of MEK and Ca(2+) inhibitors, reduced by endothelial nitric oxide synthase inhibition, and did not depend on PI3K pathway activation. The results provide evidence for a novel intracellular mechanism for EF regulation of endothelial angiogenic response via frequency-sensitive MAPK/ERK pathway activation, with important implications for EF-based therapies for vascular tissue regeneration.
Collapse
Affiliation(s)
- Abdul Q Sheikh
- Biomedical Engineering, SEEBME, University of Cincinnati, 2901 Woodside Drive, Cincinnati, OH 45221-0012, USA
| | | | | | | | | | | |
Collapse
|
34
|
Han H, Qiu L, Wang X, Qiu F, Wong Y, Yao X. Physalins A and B inhibit androgen-independent prostate cancer cell growth through activation of cell apoptosis and downregulation of androgen receptor expression. Biol Pharm Bull 2012; 34:1584-8. [PMID: 21963499 DOI: 10.1248/bpb.34.1584] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Androgen deprivation therapy is a common treatment strategy for advanced prostate cancer. Though effective initially, the tumor often progresses to androgen independent stage in most patients eventually after a period of remission. One of the key factors of development of resistance is reflected in expression of androgen receptor (AR). In this study, we showed that two natural compounds, physalins A and B, both secosteriods from Physalisalkekengi var. franchetii, significantly inhibited the growth of two androgen-independent cell lines CWR22Rv1 and C42B, induced apoptosis via c-Jun N-terminal kinase (JNK) and/or extracellular signal-regulated kinase (ERK) activation, and decreased AR expression. In addition, physalins A and B down-regulated the expression of prostate specific antigen (PSA) in C42B cells which is a target gene of AR. Our results suggest that physalin A and B might be useful agents in preventing the growth of androgen-independent prostate cancer (AI-PCa).
Collapse
Affiliation(s)
- Huiying Han
- Extracellular Matrix Pathology Section, Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892–4605, USA
| | | | | | | | | | | |
Collapse
|
35
|
Ewen K, Jackson A, Wilhelm D, Koopman P. A Male-Specific Role for p38 Mitogen-Activated Protein Kinase in Germ Cell Sex Differentiation in Mice1. Biol Reprod 2010; 83:1005-14. [DOI: 10.1095/biolreprod.110.086801] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
36
|
Dabrowski MJ, Zolnerciks JK, Balogh LM, Greene RJ, Kavanagh TJ, Atkins WM. Stereoselective effects of 4-hydroxynonenal in cultured mouse hepatocytes. Chem Res Toxicol 2010; 23:1601-7. [PMID: 20873854 PMCID: PMC3017229 DOI: 10.1021/tx100190k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
4-Hydroxynonenal (HNE) is produced from arachidonic acid or linoleic acid during oxidative stress. Although HNE is formed in tissues as a racemate, enantiospecific HNE effects have not been widely documented, nor considered. Therefore, a panel of cellular responses was compared after treatment with (R)-HNE, (S)-HNE, or racemic HNE. The phosphorylation status of Jun kinase (JNK) or Akt increased 28-fold or 2-3-fold, respectively, after treatment with 100 μM (S)-HNE and racemic HNE compared to (R)-HNE. In contrast, the increase in phosphorylation of MAPK was greatest for (R)-HNE. Caspase-3-dependent cleavage of the glutamate cysteine ligase (GCL) catalytic subunit and focal adhesion kinase (FAK) were greater in cells treated with (S)-HNE at 48 h. (S)-HNE also caused a greater number of subG1 nuclei, a hallmark of apoptosis, at 30 h after treatment. Together, the results demonstrate different dose- and time-dependent responses to (R)-HNE and (S)-HNE. The results further suggest that HNE enantiomers could differentially contribute to the progression of different diseases or contribute by different mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | - William M. Atkins
- To whom correspondence should be addressed. W. M. Atkins, Box 357610, Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195-7610. Tel: (206) 685-0379. Fax: (206) 685-3252.
| |
Collapse
|
37
|
Iacono A, Gómez R, Sperry J, Conde J, Bianco G, Meli R, Gómez-Reino JJ, Smith AB, Gualillo O. Effect of oleocanthal and its derivatives on inflammatory response induced by lipopolysaccharide in a murine chondrocyte cell line. ACTA ACUST UNITED AC 2010; 62:1675-82. [PMID: 20201078 DOI: 10.1002/art.27437] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE In joint diseases, cartilage homeostasis is disrupted by mechanisms that are driven by combinations of biologic factors that vary according to the disease process. In osteoarthritis (OA), biomechanical stimuli predominate, with up-regulation of both catabolic and anabolic factors. Likewise, OA progression is characterized by increased nitric oxide (NO) production, which has been associated with cartilage degradation. Given the relevance of cartilage degenerative diseases in our society, the development of a novel pharmacologic intervention is a critically important public health goal. Recently, oleocanthal isolated from extra virgin olive oil was found to display nonsteroidal antiinflammatory drug activity similar to that of ibuprofen, a drug widely used in the therapeutic management of joint inflammatory diseases. We undertook this study to evaluate the effect of oleocanthal and its derivatives on the modulation of NO production in chondrocytes. METHODS Cultured ATDC-5 chondrocytes were tested with different doses of oleocanthal and its derivatives. Cell viability was evaluated using the MTT assay. Nitrite accumulation was determined in culture supernatant using the Griess reaction. Inducible NO synthase (NOS2) protein expression was examined using Western blotting analysis. RESULTS Oleocanthal and its derivatives decreased lipopolysaccharide-induced NOS2 synthesis in chondrocytes without significantly affecting cell viability at lower concentrations. Among the derivatives we examined, derivative 231 was the most interesting, since its inhibitory effect on NOS2 was devoid of cytotoxicity even at higher concentrations. CONCLUSION This class of molecules shows potential as a therapeutic weapon for the treatment of inflammatory degenerative joint diseases.
Collapse
Affiliation(s)
- Anna Iacono
- Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Dass CR, Tan ML, Galloway SJ, Choong PF. Dz13 Induces a Cytotoxic Stress Response with Upregulation of E2F1 in Tumor Cells Metastasizing to or from Bone. Oligonucleotides 2010; 20:79-91. [DOI: 10.1089/oli.2009.0224] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Crispin R. Dass
- Department of Orthopaedics, St. Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Mei Lin Tan
- Department of Orthopaedics, St. Vincent's Hospital Melbourne, Fitzroy, Australia
- Department of Medicine, University of Melbourne, Parkville, Australia
| | - Stuart J. Galloway
- Department of Pathology, St. Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Peter F.M. Choong
- Department of Orthopaedics, St. Vincent's Hospital Melbourne, Fitzroy, Australia
- Department of Surgery, University of Melbourne, Parkville, Australia
- Sarcoma Service, Peter MacCallum Cancer Institute, East Melbourne, Australia
| |
Collapse
|
39
|
de Oliveira DM, Pitanga BPS, Grangeiro MS, Lima RMF, Costa MFD, Costa SL, Clarêncio J, El-Bachá RS. Catechol cytotoxicity in vitro: Induction of glioblastoma cell death by apoptosis. Hum Exp Toxicol 2010; 29:199-212. [DOI: 10.1177/0960327109360364] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The exposure to benzene is a public health problem. Although the most well-known effect of benzene is hematopoietic toxicity, there is little information about the benzene and its metabolites effects on the central nervous system (CNS). This study examined the toxic effects of 1,2-dihydroxybenzene (catechol), a benzene metabolite, to human glioblastoma GL-15 cells. GL-15 cell cultures were used as a model to provide more information about the toxic effects of aromatic compounds to the CNS. Catechol induced time- and concentration-dependent cytotoxic effects. Morphological changes, such as the retraction of the cytoplasm and chromatin clumping, were seen in cells exposed to 200 μM catechol for 48 hours. In cells exposed to 600 μM catechol for 48 hours, 78.0% of them presented condensed nuclei, and the Comet assay showed DNA damage. The percentage of cells labeled with annexin V (apoptotic cells) was greater in the group exposed to catechol (20.7%) than in control cells (0.4%). Exposure to catechol at concentrations greater than 100 μM enhanced Bax levels, and a decrease in Bcl-2 level was observed after the exposure to 600 μM catechol for 48 hours. Furthermore, catechol depleted reduced glutathione. Hence, catechol induced cell death mainly by apoptosis.
Collapse
Affiliation(s)
- DM de Oliveira
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - BPS Pitanga
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - MS Grangeiro
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - RMF Lima
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - MFD Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - SL Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - J. Clarêncio
- Laboratory of Microbiology and Immunoregulation, Gonçalo Moniz Research Center, Salvador, Bahia, Brazil
| | - RS El-Bachá
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil,
| |
Collapse
|
40
|
Local application of olprinone for promotion of peripheral nerve regeneration. J Orthop Sci 2009; 14:801-10. [PMID: 19997829 DOI: 10.1007/s00776-009-1398-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 07/29/2009] [Indexed: 11/26/2022]
Abstract
BACKGROUND Olprinone was applied locally to the sciatic nerves of rats, and nerve regeneration activity was examined in relation to heat shock protein (HSP)27 expression. METHODS Adult rat sciatic nerves were locally exposed to olprinone, physiological saline, or a mixture of saline and HCl and underwent crush injury. HSP27 protein levels were assessed by Western blotting and immunohistochemistry. Nerve regeneration was assessed by sciatic function index (SFI) and by morphological evaluation. RESULTS Olprinone was detected in ipsilateral L4-L6 dorsal root ganglion (DRG) and sciatic nerve proximal to the site of crush injury. Western blot analysis of the DRG 24 h postsurgery and the sciatic nerve 7 days postsurgery showed a marked increase in HSP27 protein levels in the olprinone-treated group. Immunohistochemical analysis was consistent with the Western blot analysis. SFI was significantly better for the olprinone-treated group than for either the saline- or HCl-treated groups at 7, 22, and 25 days, respectively, after application. The packing density and the number of myelinated axons 21 days after the crush injury in the olprinone-treated group were significantly greater in comparison to the other groups. CONCLUSION The local application of olprinone to the sciatic nerve increases HSP27 protein levels in DRG in the early stage and in the sciatic nerve 1 week thereafter. The results from this crush model indicate that increasing levels of HSP27 appear to result in therapeutic effects.
Collapse
|
41
|
Janson V, Behnam-Motlagh P, Henriksson R, Hörstedt P, Engström KG, Grankvist K. PHASE-CONTRAST MICROSCOPY STUDIES OF EARLY CISPLATIN-INDUCED MORPHOLOGICAL CHANGES OF MALIGNANT MESOTHELIOMA CELLS AND THE CORRESPONDENCE TO INDUCED APOPTOSIS. Exp Lung Res 2009; 34:49-67. [DOI: 10.1080/01902140701884398] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
42
|
Zhang Y, Zhou J, Xu W, Li A, Zhou J, Xu S. JWA sensitizes P-glycoprotein-mediated drug-resistant choriocarcinoma cells to etoposide via JNK and mitochondrial-associated signal pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2009; 72:774-781. [PMID: 19492242 DOI: 10.1080/15287390902841649] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A major obstacle in cancer chemotherapy is the phenomenon of multidrug resistance (MDR), increased P-glycoprotein expression, and abnormal apoptotic processes that may contribute to MDR. Our previous studies demonstrated that JWA is a pro-apoptotic molecule and required for arsenic trioxide and all-trans-retinoic acid-induced cancer cell apoptosis. In this study, the role of JWA in mediating MDR during treatment of choriocarcinoma cells was examined. Data showed that JWA expression was reduced significantly by etoposide (VP16) in JAR MDR cells (JAR/VP16) compared to parent JAR cells. VP16-induced apoptosis in JAR cells was dependent upon the presence of JWA. Knockdown of JWA attenuated VP16-induced apoptosis, and was accompanied by significantly reduced caspase-9 activity and inhibition of JNK phosphorylation. Loss of mitochondrial transmembrane potential induced by VP16 was accompanied by higher JWA expression. JWA was also involved in downregulation of P-glycoprotein through JNK signal pathway. These results suggest that JWA may play an important role in the therapeutic responses to chemotherapeutic agents used to treat choriocarcinoma.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Molecular Cell Biology and Genetics, Cancer Centre, Nanjing Medical University, Nanjing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
43
|
Zheng G, Chen Y, Zhang X, Cai T, Liu M, Zhao F, Luo W, Chen J. Acute cold exposure and rewarming enhanced spatial memory and activated the MAPK cascades in the rat brain. Brain Res 2008; 1239:171-80. [PMID: 18789908 DOI: 10.1016/j.brainres.2008.08.057] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 08/03/2008] [Accepted: 08/20/2008] [Indexed: 01/06/2023]
Abstract
Cold is a common stressor that is likely to occur in everyday occupational or leisure time activities. Although there is substantial literature on the effects of stress on memory from behavioral and pharmacologic perspectives, the effects of cold stress on learning and memory were little addressed. The aims of the present work were to investigate the effects of acute cold exposure on Y-maze learning and the activation of cerebral MAPK cascades of rats. We found that the 2-hour cold exposure (-15 degrees C) and a subsequent 30-min rewarming significantly increased the performance of the rats in the Y-maze test. Serum corticosterone (CORT) level was increased after the cold exposure. After a transient reduction following the cold exposure, the P-ERK levels in the hippocampus and PFC drastically increased 30 min later. The levels of P-JNK increased gradually after the cold exposure in all the three brain regions we investigated, but the level of P-p38 only increased in the PFC. The levels of GABAA receptor alpha1 subunit remained unchanged after the cold exposure. Furthermore, the performance of rats treated with cold plus muscimol or bicuculline in the Y-maze test was similar to that of the rats treated with those GABAergic agents alone. These results demonstrated that acute cold exposure and the subsequent rewarming could result in enhanced performance of spatial learning and memory, and the activation of MAPKs in the brain. However, GABAA receptor may not be involved in the acute cold exposure-induced enhancement of memory.
Collapse
Affiliation(s)
- Gang Zheng
- Department of Occupational and Environmental Health, School of Public Health, Fourth Military Medical University, 17 Changlexi Road, Xi'an 710032, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Durmuş Tekir S, Yalçin Arga K, Ulgen KO. Drug targets for tumorigenesis: insights from structural analysis of EGFR signaling network. J Biomed Inform 2008; 42:228-36. [PMID: 18790083 DOI: 10.1016/j.jbi.2008.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 07/15/2008] [Accepted: 08/17/2008] [Indexed: 02/01/2023]
Abstract
Deciphering the complex network structure is crucial in drug target identification. This study presents a framework incorporating graph theoretic and network decomposition methods to analyze system-level properties of the comprehensive map of the epidermal growth factor receptor (EGFR) signaling, which is a good candidate model system to study the general mechanisms of signal transduction. The graph theoretic analysis of the EGFR network indicates that it has small-world characteristics with scale-free topology. The employment of network decomposition analysis enlightened the system-level properties, such as network cross-talk, specific molecules in each pathway and participation of molecules in the network. Participating in a significant fraction of the fundamental paths connecting the ligands to the phenotypes, cofactor GTP and complex Gbeta/Ggamma were identified as "housekeeping" molecules, through which all pathways of EGFR network are cross-talking. c-Src-Shc complex is identified as important due to its role in all fundamental paths through tumorigenesis and being specific to this phenotype. Inhibitors of this complex may be good anti-cancer agents having very little or no effect on other phenotypes.
Collapse
Affiliation(s)
- Saliha Durmuş Tekir
- Department of Chemical Engineering, Boğaziçi University, 34342 Bebek-Istanbul, Turkey.
| | | | | |
Collapse
|
45
|
Zhang X, Lan W, Deng Y, Ma Y, Liu K, Peng J, Li Y, Hong H. Highly passage of Spodoptera litura cell line causes its permissiveness to baculovirus infection. Cytotechnology 2008; 57:233-43. [PMID: 19003180 DOI: 10.1007/s10616-008-9158-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Accepted: 07/01/2008] [Indexed: 01/20/2023] Open
Abstract
It is well known that the characteristics of cell lines possibly alter when cell lines are at high-passage number because of the environmental selection. We do not know whether non-permissive or low-permissive cell lines could become permissive or more permissive to virus infection after over-high passage. In the present studies, the alteration of the permissiveness of Spodoptera litura cell line Sl-zsu-1 to three baculovirus infection was investigated after over-high passage, and the possible mechanisms are also investigated. Vigorous apoptosis in Sl-zsu-1 cells was induced by both the recombinant Autographa californica multiple nucleopolyhedrovirus AcMNPV-GFP-actin and the celery looper Anagrapha falcifera multiple nucleopolyhedrovirus AfMNPV, suggesting the replication of the two viruses was blocked by apoptosis. However, the cells infected by S. litura multicapsid nucleopolyhedrovirus SpltMNPV did not undergo apoptosis, but the SpltMNPV titre of the supernatant was not detectable, suggesting this cell line was low-permissive for this virus infection and other factor(s) involved in blockage of the virus replication except apoptosis. However, when Sl-zsu-1 cells had been subcultured continuously for more than 4 years (high-passage cell), which was named as Sl-HP cell line afterwards, no significant apoptosis was induced by the three baculovirus in Sl-HP cells, and many replicated virions or nucleocapsids were observed in the cells. But the permissiveness of Sl-HP cells to the three viruses was very different according to the titre of viruses in the cell cultures. Interestingly, the DNA extracted from SpltMNPV could induce vigorous apoptosis of Sl-HP cells. Altogether, Sl-zsu-1 cell line becomes more permissive to baculovirus infection after over-high passage and multiple paths can block the baculovirus infectivity.
Collapse
Affiliation(s)
- Xuping Zhang
- College of Life Science, Central China Normal University, Wuhan, 430079, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Chang HL, Wu YC, Su JH, Yeh YT, Yuan SSF. Protoapigenone, a novel flavonoid, induces apoptosis in human prostate cancer cells through activation of p38 mitogen-activated protein kinase and c-Jun NH2-terminal kinase 1/2. J Pharmacol Exp Ther 2008; 325:841-9. [PMID: 18337475 DOI: 10.1124/jpet.107.135442] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In this study, we investigated the anticancer effect of protoapigenone on human prostate cancer cells. Protoapigenone inhibited cell growth through arresting cancer cells at S and G(2)/M phases as well as inducing apoptosis. Blockade of cell cycle by protoapigenone was associated with an increase in the levels of inactivated phospho (p)-Cdc25C (Ser216) and a decrease in the levels of activated p-cyclin B1 (Ser147), cyclin B1, and cyclin-dependent kinase (Cdk) 2. Protoapigenone triggered apoptosis by increasing the levels of cleaved poly(ADP-ribose) polymerase and caspase-3. In addition, activation of p38 mitogen-activated protein kinase (MAPK) and c-Jun NH2-terminal kinase (JNK)1/2 was a critical mediator in protoapigenone-induced cell death. Inhibition of the expression of p38 MAPK and JNK1/2 by pharmacological inhibitors or specific small interfering RNA reversed the protoapigenone-induced apoptosis through decreasing the level of cleaved caspase-3. In contrast, p38 MAPK, but not JNK1/2, was involved in the protoapigenone-mediated S and G(2)/M arrest by modulating the levels of Cdk2 and p-Cdc25C (Ser216). Moreover, in vivo xenograft study showed that protoapigenone had a significant inhibition of prostate tumor growth without major side effects on the mice we tested. This inhibition was associated with induction of apoptosis and activation of p38 MAPK and JNK1/2 in protoapigenone-treated tumor tissues. In conclusion, our results demonstrated protoapigenone suppressed prostate cancer cell growth through the activation of p38 MAPK and JNK1/2, with the potential to be developed as a chemotherapeutic agent for prostate cancer.
Collapse
Affiliation(s)
- Hsueh-Ling Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|
47
|
Hiraishi Y, Wada T, Nakatani K, Tojyo I, Matsumoto T, Kiga N, Negoro K, Fujita S. EGFR inhibitor enhances cisplatin sensitivity of oral squamous cell carcinoma cell lines. Pathol Oncol Res 2008; 14:39-43. [PMID: 18347929 PMCID: PMC2413131 DOI: 10.1007/s12253-008-9020-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 02/20/2008] [Indexed: 12/03/2022]
Abstract
Epidermal growth factor receptor (EGFR) is involved in multiple aspects of cancer cell biology. EGFR has already been identified as an important target for cancer therapy, with various kinds of EGFR inhibitors currently used in treatment of several human cancers. Recently, EGFR and its downstream signaling pathways were identified as being associated with cisplatin sensitivity. In addition, EGFR inhibitors have shown significant promise for patients who failed cisplatin-based therapy. In this study, we investigated whether treatment with an EGFR inhibitor improves cisplatin sensitivity in oral squamous cell carcinoma (OSCC) cell lines. The effects of a combination of AG1478, a specific EGFR tyrosine kinase inhibitor, with cisplatin were evaluated in cultured OSCC cell lines and cisplatin-resistant sublines. Higher expression of EGFR and p-EGFR was found in the two cisplatin-resistant cell lines compared with the corresponding parental cell lines. In addition, augmented inhibition of OSCC cell growth by the combination of AG1478 with cisplatin was found in both cell lines. These results suggest that the combination of an EGFR inhibitor and cisplatin may be useful as a rational strategy for the treatment of patients with oral cancer with acquired cisplatin resistance.
Collapse
Affiliation(s)
- Yukihiro Hiraishi
- Department of Oral and Maxillofacial Surgery, Wakayama Medical University, 811-1 Kimiidera, 641-8509 Wakayama, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Protein phosphatase 2A regulates life and death decisions via Akt in a context-dependent manner. Proc Natl Acad Sci U S A 2007; 104:19011-6. [PMID: 18006659 DOI: 10.1073/pnas.0706696104] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Here, we show how targeting protein phosphatase 2A (PP2A), a key regulator of cellular protein phosphorylation, can either induce or prevent apoptosis depending on what other signals the cell is receiving. The oncoprotein polyoma small T interacts with PP2A to regulate survival. In the presence of growth factors, small T induces apoptosis. Akt activity, which usually promotes survival, is required for this death response, because inhibitors of Akt or PI3 kinase protect cells from death. The activation of Akt under these conditions is partial, characterized by T308 phosphorylation but not S473 phosphorylation. In the absence of growth factors, small T protects from cell death. Here, small T uses PP2A to promote phosphorylation of Akt on both T308 and S473. This effect results in a different pattern of phosphorylation of Akt substrates and shifts Akt from a proapoptotic (presence of growth factors) to an antiapoptotic mode (absence of growth factors). An intriguing possibility is that Akt phosphorylation could be therapeutically disregulated to decrease the survival of cancer cells.
Collapse
|
49
|
Nadeau SI, Landry J. Mechanisms of Activation and Regulation of the Heat Shock-Sensitive Signaling Pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 594:100-13. [PMID: 17205679 DOI: 10.1007/978-0-387-39975-1_10] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Heat shock (HS), like many other stresses, induces specific and highly regulated signaling cascades that promote cellular homeostasis. The three major mitogen-activated protein kinases (MAPK) and protein kinase B (PKB/Akt) are the most notable of these HS-stimulated pathways. Their activation occurs rapidly and sooner than the transcriptional upregulation of heat shock proteins (Hsp), which generate a transient state of extreme resistance against subsequent thermal stress. The direct connection of these signaling pathways to cellular death or survival mechanisms suggests that they contribute importantly to the HS response. Some of them may counteract early noxious effects of heat, while others may bolster key apoptosis events. The triggering events responsible for activating these pathways are unclear. Protein denaturation, specific and nonspecific receptor activation, membrane alteration and chromatin structure perturbation are potential initiating factors.
Collapse
Affiliation(s)
- Sébastien Ian Nadeau
- Centre de recherche en cancérologie de I'Université Laval, L'Hôtel-Dieu de Québec, 9, rue McMahon, Québec, Canada G1 R 2J6
| | | |
Collapse
|
50
|
Madison MN, Kleshchenko YY, Nde PN, Simmons KJ, Lima MF, Villalta F. Human defensin alpha-1 causes Trypanosoma cruzi membrane pore formation and induces DNA fragmentation, which leads to trypanosome destruction. Infect Immun 2007; 75:4780-91. [PMID: 17635867 PMCID: PMC2044533 DOI: 10.1128/iai.00557-07] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human defensins play a fundamental role in the initiation of innate immune responses to some microbial pathogens. Here we show that human defensin alpha-1 displays a trypanocidal role against Trypanosoma cruzi, the causative agent of Chagas' disease. The toxicity of human defensin alpha-1 against T. cruzi is mediated by membrane pore formation and the induction of nuclear and mitochondrial DNA fragmentation, leading to trypanosome destruction. Exposure of trypomastigote and amastigote forms of T. cruzi to defensin alpha-1 significantly reduced parasite viability in a peptide concentration-dependent and saturable manner. The toxicity of defensin alpha-1 against T. cruzi is blocked by anti-defensin alpha-1 immunoglobulin G. Electron microscopic analysis of trypomastigotes exposed to defensin alpha-1 revealed pore formation in the cellular and flagellar membranes, membrane disorganization, and blebbing as well as cytoplasmic vacuolization. Furthermore, human defensin alpha-1 enters the trypanosome when membrane pores are present and is associated with later intracellular damage. Trypanosome membrane depolarization abolished the toxicity of defensin alpha-1 against the parasite. Preincubation of trypomastigotes with defensin alpha-1 followed by exposure to human epithelial cells significantly reduced T. cruzi infection in these cells. Thus, human defensin alpha-1 is an innate immune molecule that causes severe toxicity to T. cruzi and plays an important role in reducing cellular infection. This is the first report showing that human defensin alpha-1 causes membrane pore formation in a human parasite, leading to trypanosome destruction.
Collapse
Affiliation(s)
- M Nia Madison
- Department of Microbial Pathogenesis and Immune Response, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | | | | | | | | | | |
Collapse
|