1
|
Yang M, Liu J, Li J, Wen S, Hu Y, Lu W, Liu J, Huang P, Liu P. The rheumatoid arthritis drug auranofin exerts potent anti-lymphoma effect by stimulating TXNRD-mediated ROS generation and inhibition of energy metabolism. Redox Biol 2024; 75:103245. [PMID: 38909408 PMCID: PMC11254835 DOI: 10.1016/j.redox.2024.103245] [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: 05/09/2024] [Revised: 06/08/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024] Open
Abstract
Since the survival of lymphoma patients who experience disease progression or relapse remains very poor, new therapeutic approaches and effective drugs are urgently needed. Here we show that auranofin (AF), an anti-rheumatoid drug thought to inhibit thioredoxin reductases (TXNRDs) as its mechanism of action, exhibited potent activity against multiple cancer types, especially effective against B cell lymphoma. Surprisingly, a knockdown of TXNRD1 and TXNRD2 did not cause significant cytotoxicity, suggesting that abrogation of TXNRD enzyme per se was insufficient to cause cancer cell death. Further mechanistic study showed that the interaction of AF with TXNRD could convert this antioxidant enzyme to a ROS-generating molecule via disrupting its electron transport, leading to a leak of electrons that interact with molecular oxygen to form superoxide. AF also suppressed energy metabolism by inhibiting both mitochondria complex II and the glycolytic enzyme GAPDH, leading to a significant depletion of ATP and inhibition of cancer growth in vitro and in vivo. Importantly, we found that the AF-mediated ROS stress could induce PD-L1 expression, revealing an unwanted effect of AF in causing immune suppression. We further showed that a combination of AF with anti-PD-1 antibody could enhance the anticancer activity in a syngeneic immune-competent mouse B-cell lymphoma model. Our study suggests that AF could be a potential drug for lymphoma treatment, and its combination with immune checkpoint inhibitors would be a logical strategy to increase the therapeutic activity.
Collapse
Affiliation(s)
- Mengqi Yang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Department of Radiation Oncology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Jiaxin Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jianan Li
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shijun Wen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yumin Hu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenhua Lu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jinyun Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Metabolic Innovation Center, Zhongshan School of Medicine, Platform of Metabolomics Center for Precision Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Peng Huang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Metabolic Innovation Center, Zhongshan School of Medicine, Platform of Metabolomics Center for Precision Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Panpan Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Dos Reis Oliveira C, Pereira JC, Barros Ibiapina A, Roseno Martins IR, de Castro E Sousa JM, Ferreira PMP, Carneiro da Silva FC. Buthionine sulfoximine and chemoresistance in cancer treatments: a systematic review with meta-analysis of preclinical studies. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2023; 26:417-441. [PMID: 37606035 DOI: 10.1080/10937404.2023.2246876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Buthionine sulfoximine (BSO) is a synthetic amino acid that blocks the biosynthesis of reduced glutathione (GSH), an endogenous antioxidant cellular component present in tumor cells. GSH levels have been associated with tumor cell resistance to chemotherapeutic drugs and platinum compounds. Consequently, by depleting GSH, BSO enhances the cytotoxicity of chemotherapeutic agents in drug-resistant tumors. Therefore, the aim of this study was to conduct a systematic review with meta-analysis of preclinical studies utilizing BSO in cancer treatments. The systematic search was carried out using the following databases: PubMed, Web of Science, Scopus, and EMBASE up until March 20, 2023, in order to collect preclinical studies that evaluated BSO, alone or in association, as a strategy for antineoplastic therapy. One hundred nine investigations were found to assess the cytotoxic potential of BSO alone or in combination with other compounds. Twenty-one of these met the criteria for performing the meta-analysis. The evidence gathered indicated that BSO alone exhibits cytotoxic activity. However, this compound is generally used in combination with other antineoplastic strategies, mainly chemotherapy ones, to improve cytotoxicity to carcinogenic cells and treatment efficacy. Finally, this review provides important considerations regarding BSO use in cancer treatment conditions, which might optimize future studies as a potential adjuvant antineoplastic therapeutic tool.
Collapse
Affiliation(s)
| | - Joedna Cavalcante Pereira
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | | | | | - João Marcelo de Castro E Sousa
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
- Laboratory of Toxicological Genetics (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | - Felipe Cavalcanti Carneiro da Silva
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina, Brazil
- Laboratory of Toxicological Genetics (Lapgenic), Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
3
|
Liu S, Huang B, Cao J, Wang Y, Xiao H, Zhu Y, Zhang H. ROS fine-tunes the function and fate of immune cells. Int Immunopharmacol 2023; 119:110069. [PMID: 37150014 DOI: 10.1016/j.intimp.2023.110069] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/11/2023] [Accepted: 03/20/2023] [Indexed: 05/09/2023]
Abstract
The redox state is essential to the process of cell life, which determines cell fate. As an important signaling molecule of the redox state, reactive oxygen species (ROS) are crucial for the homeostasis of immune cells and participate in the pathological processes of different diseases. We discuss the underlying mechanisms and possible signaling pathways of ROS to fine-tune the proliferation, differentiation, polarization and function of immune cells, including T cells, B cells, neutrophils, macrophages, myeloid-derived inhibitory cells (MDSCs) and dendritic cells (DCs). We further emphasize how excessive ROS lead to programmed immune cell death such as apoptosis, ferroptosis, pyroptosis, NETosis and necroptosis, providing valuable insights for future therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Shiyu Liu
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Benqi Huang
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Jingdong Cao
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Yifei Wang
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Hao Xiao
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Yaxi Zhu
- Sepsis Translational Medicine Key Lab of Hunan Province, Department of Pathophysiology, School of Basic Medical Sciences, Central South University, 410008 Changsha, China.
| | - Huali Zhang
- Sepsis Translational Medicine Key Lab of Hunan Province, Department of Pathophysiology, School of Basic Medical Sciences, Central South University, 410008 Changsha, China.
| |
Collapse
|
4
|
Sousa-Pimenta M, Estevinho MM, Sousa Dias M, Martins Â, Estevinho LM. Oxidative Stress and Inflammation in B-Cell Lymphomas. Antioxidants (Basel) 2023; 12:antiox12040936. [PMID: 37107311 PMCID: PMC10135850 DOI: 10.3390/antiox12040936] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Mature lymphoid neoplasms arise de novo or by the transformation of more indolent lymphomas in a process that relies on the stepwise accumulation of genomic and transcriptomic alterations. The microenvironment and neoplastic precursor cells are heavily influenced by pro-inflammatory signaling, regulated in part by oxidative stress and inflammation. Reactive oxygen species (ROSs) are by-products of cellular metabolism able to modulate cell signaling and fate. Moreover, they play a crucial role in the phagocyte system, which is responsible for antigen presentation and the selection of mature B and T cells under normal conditions. Imbalances in pro-oxidant and antioxidant signaling can lead to physiological dysfunction and disease development by disrupting metabolic processes and cell signaling. This narrative review aims to analyze the impact of reactive oxygen species on lymphomagenesis, specifically examining the regulation of microenvironmental players, as well as the response to therapy for B-cell-derived non-Hodgkin lymphomas. Further research is needed to investigate the involvement of ROS and inflammation in the development of lymphomas, which may unravel disease mechanisms and identify innovative therapeutic targets.
Collapse
Affiliation(s)
- Mário Sousa-Pimenta
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto (IPO-Porto), 4200-072 Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria Manuela Estevinho
- Department of Gastroenterology, Vila Nova de Gaia/Espinho Hospital Center, 4434-502 Vila Nova de Gaia, Portugal
- Department of Biomedicine, Unit of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Miguel Sousa Dias
- Mountain Research Center (CIMO), Polytechnic Institute of Bragança, 5300-252 Bragança, Portugal
- Department of Biology and Biotechnology, Agricultural College of Bragança, Polytechnic Institute of Bragança, 5300-252 Bragança, Portugal
| | - Ângelo Martins
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Letícia M Estevinho
- Mountain Research Center (CIMO), Polytechnic Institute of Bragança, 5300-252 Bragança, Portugal
- Department of Biology and Biotechnology, Agricultural College of Bragança, Polytechnic Institute of Bragança, 5300-252 Bragança, Portugal
| |
Collapse
|
5
|
Muri J, Kopf M. The thioredoxin system: Balancing redox responses in immune cells and tumors. Eur J Immunol 2023; 53:e2249948. [PMID: 36285367 PMCID: PMC10100330 DOI: 10.1002/eji.202249948] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 02/02/2023]
Abstract
The thioredoxin (TRX) system is an important contributor to cellular redox balance and regulates cell growth, apoptosis, gene expression, and antioxidant defense in nearly all living cells. Oxidative stress, the imbalance between reactive oxygen species (ROS) and antioxidants, can lead to cell death and tissue damage, thereby contributing to aging and to the development of several diseases, including cardiovascular and allergic diseases, diabetes, and neurological disorders. Targeting its activity is also considered as a promising strategy in the treatment of cancer. Over the past years, immunologists have established an essential function of TRX for activation, proliferation, and responses in T cells, B cells, and macrophages. Upon activation, immune cells rearrange their redox system and activate the TRX pathway to promote proliferation through sustainment of nucleotide biosynthesis, and to support inflammatory responses in myeloid cells by allowing NF-κB and NLRP3 inflammasome responses. Consequently, targeting the TRX system may therapeutically be exploited to inhibit immune responses in inflammatory conditions. In this review, we summarize recent insights revealing key roles of the TRX pathway in immune cells in health and disease, and lessons learnt for cancer therapy.
Collapse
Affiliation(s)
- Jonathan Muri
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
6
|
Falchetti M, Delgobo M, Zancanaro H, Almeida K, das Neves RN, Dos Santos B, Stefanes NM, Bishop A, Santos-Silva MC, Zanotto-Filho A. Omics-based identification of an NRF2-related auranofin resistance signature in cancer: Insights into drug repurposing. Comput Biol Med 2023; 152:106347. [PMID: 36493734 DOI: 10.1016/j.compbiomed.2022.106347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/04/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
Auranofin is a thioredoxin reductase-1 inhibitor originally approved for the treatment of rheumatoid arthritis. Recently, auranofin has been repurposed as an anticancer drug, with pharmacological activity reported in multiple cancer types. In this study, we characterized transcriptional and genetic alterations associated with auranofin response in cancer. By integrating data from an auranofin cytotoxicity screen with transcriptome profiling of lung cancer cell lines, we identified an auranofin resistance signature comprising 29 genes, most of which are classical targets of the transcription factor NRF2, such as genes involved in glutathione metabolism (GCLC, GSR, SLC7A11) and thioredoxin system (TXN, TXNRD1). Pan-cancer analysis revealed that mutations in NRF2 pathway genes, namely KEAP1 and NFE2L2, are strongly associated with overexpression of the auranofin resistance gene set. By clustering cancer types based on auranofin resistance signature expression, hepatocellular carcinoma, and a subset of non-small cell lung cancer, head-neck squamous cell carcinoma, and esophageal cancer carrying NFE2L2/KEAP1 mutations were predicted resistant, whereas leukemia, lymphoma, and multiple myeloma were predicted sensitive to auranofin. Cell viability assays in a panel of 20 cancer cell lines confirmed the augmented sensitivity of hematological cancers to auranofin; an effect associated with dependence upon glutathione and decreased expression of NRF2 target genes involved in GSH synthesis and recycling (GCLC, GCLM and GSR) in these cancer types. In summary, the omics-based identification of sensitive/resistant cancers and genetic alterations associated with these phenotypes may guide an appropriate repurposing of auranofin in cancer therapy.
Collapse
Affiliation(s)
- Marcelo Falchetti
- Laboratório de Farmacologia e Bioquímica do Câncer (LabCancer), Departamento de Farmacologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Marina Delgobo
- Laboratório de Farmacologia e Bioquímica do Câncer (LabCancer), Departamento de Farmacologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Helena Zancanaro
- Laboratório de Farmacologia e Bioquímica do Câncer (LabCancer), Departamento de Farmacologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Karoline Almeida
- Laboratório de Farmacologia e Bioquímica do Câncer (LabCancer), Departamento de Farmacologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Raquel Nascimento das Neves
- Laboratório de Farmacologia e Bioquímica do Câncer (LabCancer), Departamento de Farmacologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, 88040-900, Brazil; Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
| | - Barbara Dos Santos
- Laboratório de Farmacologia e Bioquímica do Câncer (LabCancer), Departamento de Farmacologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Natália Marcéli Stefanes
- Laboratório de Oncologia Experimental e Hemopatias (LOEH), Departamento de Análises Clínicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Alexander Bishop
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA; Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
| | - Maria Cláudia Santos-Silva
- Laboratório de Oncologia Experimental e Hemopatias (LOEH), Departamento de Análises Clínicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Alfeu Zanotto-Filho
- Laboratório de Farmacologia e Bioquímica do Câncer (LabCancer), Departamento de Farmacologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, 88040-900, Brazil. https://labcancer.paginas.ufsc.br
| |
Collapse
|
7
|
Renatino Canevarolo R, Pereira de Souza Melo C, Moreno Cury N, Luiz Artico L, Ronchi Corrêa J, Tonhasca Lau Y, Sousa Mariano S, Reddy Sudalagunta P, Regina Brandalise S, Carolina de Mattos Zeri A, Andrés Yunes J. Glutathione levels are associated with methotrexate resistance in acute lymphoblastic leukemia cell lines. Front Oncol 2022; 12:1032336. [PMID: 36531023 PMCID: PMC9751399 DOI: 10.3389/fonc.2022.1032336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction Methotrexate (MTX), a folic acid antagonist and nucleotide synthesis inhibitor, is a cornerstone drug used against acute lymphoblastic leukemia (ALL), but its mechanism of action and resistance continues to be unraveled even after decades of clinical use. Methods To better understand the mechanisms of this drug, we accessed the intracellular metabolic content of 13 ALL cell lines treated with MTX by 1H-NMR, and correlated metabolome data with cell proliferation and gene expression. Further, we validated these findings by inhibiting the cellular antioxidant system of the cells in vitro and in vivo in the presence of MTX. Results MTX altered the concentration of 31 out of 70 metabolites analyzed, suggesting inhibition of the glycine cleavage system, the pentose phosphate pathway, purine and pyrimidine synthesis, phospholipid metabolism, and bile acid uptake. We found that glutathione (GSH) levels were associated with MTX resistance in both treated and untreated cells, suggesting a new constitutive metabolic-based mechanism of resistance to the drug. Gene expression analyses showed that eight genes involved in GSH metabolism were correlated to GSH concentrations, 2 of which (gamma-glutamyltransferase 1 [GGT1] and thioredoxin reductase 3 [TXNRD3]) were also correlated to MTX resistance. Gene set enrichment analysis (GSEA) confirmed the association between GSH metabolism and MTX resistance. Pharmacological inhibition or stimulation of the main antioxidant systems of the cell, GSH and thioredoxin, confirmed their importance in MTX resistance. Arsenic trioxide (ATO), a thioredoxin inhibitor used against acute promyelocytic leukemia, potentiated MTX cytotoxicity in vitro in some of the ALL cell lines tested. Likewise, the ATO+MTX combination decreased tumor burden and extended the survival of NOD scid gamma (NSG) mice transplanted with patient-derived ALL xenograft, but only in one of four ALLs tested. Conclusion Altogether, our results show that the cellular antioxidant defense systems contribute to leukemia resistance to MTX, and targeting these pathways, especially the thioredoxin antioxidant system, may be a promising strategy for resensitizing ALL to MTX.
Collapse
Affiliation(s)
| | | | | | | | | | - Yanca Tonhasca Lau
- Centro de Pesquisa Boldrini, Centro Infantil Boldrini, Campinas, SP, Brazil
| | | | - Praneeth Reddy Sudalagunta
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | | | - Ana Carolina de Mattos Zeri
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - José Andrés Yunes
- Centro de Pesquisa Boldrini, Centro Infantil Boldrini, Campinas, SP, Brazil,Medical Genetics Department, Faculty of Medical Sciences, State University of Campinas, Campinas, SP, Brazil,*Correspondence: José Andrés Yunes,
| |
Collapse
|
8
|
Laoukili J, van Schelven S, Küçükköse E, Verheem A, Goey K, Koopman M, Borel Rinkes I, Kranenburg O. BRAF V600E in colorectal cancer reduces sensitivity to oxidative stress and promotes site-specific metastasis by stimulating glutathione synthesis. Cell Rep 2022; 41:111728. [PMID: 36450250 DOI: 10.1016/j.celrep.2022.111728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/08/2022] [Accepted: 11/03/2022] [Indexed: 12/02/2022] Open
Abstract
The presence of BRAFV600E in colorectal cancer (CRC) is associated with a higher chance of distant metastasis. Oxidative stress in disseminated tumor cells limits metastatic capacity. To study the relationship between BRAFV600E, sensitivity to oxidative stress, and metastatic capacity in CRC, we use patient-derived organoids (PDOs) and tissue samples. BRAFV600E tumors and PDOs express high levels of glutamate-cysteine ligase (GCL), the rate-limiting enzyme in glutathione synthesis. Deletion of GCL in BRAFV600E PDOs strongly reduces their capacity to form distant liver and lung metastases but does not affect peritoneal metastasis outgrowth. Vice versa, the glutathione precursor N-acetyl-cysteine promotes organ-site-specific metastasis in the liver and the lungs but not in the peritoneum. BRAFV600E confers resistance to pharmacologically induced oxidative stress in vitro, which is partially overcome by treatment with the BRAF-inhibitor vemurafenib. We conclude that GCL-driven glutathione synthesis protects BRAFV600E-expressing tumors from oxidative stress during distant metastasis to the liver and the lungs.
Collapse
Affiliation(s)
- Jamila Laoukili
- Lab Translational Oncology, University Medical Center Utrecht, G04-228, PO Box 85500, 3508GA Utrecht, the Netherlands.
| | - Susanne van Schelven
- Lab Translational Oncology, University Medical Center Utrecht, G04-228, PO Box 85500, 3508GA Utrecht, the Netherlands
| | - Emre Küçükköse
- Lab Translational Oncology, University Medical Center Utrecht, G04-228, PO Box 85500, 3508GA Utrecht, the Netherlands
| | - André Verheem
- Lab Translational Oncology, University Medical Center Utrecht, G04-228, PO Box 85500, 3508GA Utrecht, the Netherlands
| | - Kaitlyn Goey
- Department of Medical Oncology, University Medical Center, Utrecht University, Utrecht, the Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center, Utrecht University, Utrecht, the Netherlands
| | - Inne Borel Rinkes
- Lab Translational Oncology, University Medical Center Utrecht, G04-228, PO Box 85500, 3508GA Utrecht, the Netherlands
| | - Onno Kranenburg
- Lab Translational Oncology, University Medical Center Utrecht, G04-228, PO Box 85500, 3508GA Utrecht, the Netherlands; Utrecht Platform for Organoid Technology, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
9
|
Gamberi T, Chiappetta G, Fiaschi T, Modesti A, Sorbi F, Magherini F. Upgrade of an old drug: Auranofin in innovative cancer therapies to overcome drug resistance and to increase drug effectiveness. Med Res Rev 2022; 42:1111-1146. [PMID: 34850406 PMCID: PMC9299597 DOI: 10.1002/med.21872] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 12/20/2022]
Abstract
Auranofin is an oral gold(I) compound, initially developed for the treatment of rheumatoid arthritis. Currently, Auranofin is under investigation for oncological application within a drug repurposing plan due to the relevant antineoplastic activity observed both in vitro and in vivo tumor models. In this review, we analysed studies in which Auranofin was used as a single drug or in combination with other molecules to enhance their anticancer activity or to overcome chemoresistance. The analysis of different targets/pathways affected by this drug in different cancer types has allowed us to highlight several interesting targets and effects of Auranofin besides the already well-known inhibition of thioredoxin reductase. Among these targets, inhibitory-κB kinase, deubiquitinates, protein kinase C iota have been frequently suggested. To rationalize the effects of Auranofin by a system biology-like approach, we exploited transcriptomic data obtained from a wide range of cell models, extrapolating the data deposited in the Connectivity Maps website and we attempted to provide a general conclusion and discussed the major points that need further investigation.
Collapse
Affiliation(s)
- Tania Gamberi
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Giovanni Chiappetta
- Biological Mass Spectrometry and Proteomics GroupPlasticité du Cerveau UMR 8249 CNRSParisESPCI Paris‐PSLFrance
| | - Tania Fiaschi
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Flavia Sorbi
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Francesca Magherini
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| |
Collapse
|
10
|
Tao J, Li C, Zheng Y, Wang F, Zhang M, Wu X, Chen Y, Zeng Q, Chen F, Fei W. Biological protein mediated ferroptotic tumor nanotherapeutics. J Mater Chem B 2021; 9:9262-9284. [PMID: 34730601 DOI: 10.1039/d1tb01289d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ferroptosis, a cell death pathway involving iron-related generation of lipid hydroperoxides for achieving incredible tumor suppression, has reignited the hope of chemotherapy in tumor treatment in the past decade. With extensive research studies, various bioactive proteins and cellular pathways have been demonstrated to regulate the occurrence and development of ferroptosis. The gradually established ferroptotic regulatory network is conducive to find effective proteins from a holistic perspective and guides better designs for future ferroptotic tumor therapies. The first section of this review summarizes the recent advances in ferroptotic regulatory mechanisms of proteins and attempts to clarify their latent function in the ferroptotic regulatory network. Second, the existing protein-mediated ferroptotic tumor nanotherapeutic strategies were reviewed, including the protein-mediated iron supplement, cell membrane transporter inhibition, glutathione peroxidase 4 interference, glutathione depletion, bioenzyme-mediated reactive oxygen species generation, heat shock protein inhibition, and tumor-overexpressed protein-triggered drug release for ferroptotic therapy. Finally, the future expectations and challenges of ferroptotic tumor nanotherapeutics for clinical cancer therapy are highlighted.
Collapse
Affiliation(s)
- Jiaoyang Tao
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Chaoqun Li
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Yongquan Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Fengmei Wang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Xiaodong Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Qingquan Zeng
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Fengying Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
11
|
Jovanovic B, Eiermann N, Talwar D, Boulougouri M, Dick TP, Stoecklin G. Thioredoxin 1 is required for stress granule assembly upon arsenite-induced oxidative stress. Food Chem Toxicol 2021; 156:112508. [PMID: 34390821 DOI: 10.1016/j.fct.2021.112508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/11/2021] [Accepted: 08/09/2021] [Indexed: 10/25/2022]
Abstract
Arsenic is a major water pollutant and health hazard, leading to acute intoxication and, upon chronic exposure, several diseases including cancer development. Arsenic exerts its pronounced cellular toxicity through its trivalent oxide arsenite (ASN), which directly inhibits numerous proteins including Thioredoxin 1 (Trx1), and causes severe oxidative stress. Cells respond to arsenic by inhibition of protein synthesis and subsequent assembly of stress granules (SGs), cytoplasmic condensates of stalled mRNAs, translation factors and RNA-binding proteins. The biological role of SGs is diverse and not completely understood; they are important for regulation of cell signaling and survival under stress conditions, and for adapting de novo protein synthesis to the protein folding capacity during the recovery from stress. In this study, we identified Trx1 as a novel component of SGs. Trx1 is required for the assembly of ASN-induced SGs, but not for SGs induced by energy deprivation or heat shock. Importantly, our results show that Trx1 is essential for cell survival upon acute exposure to ASN, through a mechanism that is independent of translation inhibition.
Collapse
Affiliation(s)
- Bogdan Jovanovic
- Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany; Center for Human Molecular Genetics, Faculty of Biology, University of Belgrade, Belgrade, Serbia.
| | - Nina Eiermann
- Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Deepti Talwar
- Division of Redox Regulation, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Maria Boulougouri
- Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Tobias P Dick
- Division of Redox Regulation, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Georg Stoecklin
- Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
12
|
Zaher DM, Ramadan WS, El-Awady R, Omar HA, Hersi F, Srinivasulu V, Hachim IY, Al-Marzooq FI, Vazhappilly CG, Merali S, Merali C, Soares NC, Schilf P, Ibrahim SM, Al-Tel TH. A Novel Benzopyrane Derivative Targeting Cancer Cell Metabolic and Survival Pathways. Cancers (Basel) 2021; 13:cancers13112840. [PMID: 34200264 PMCID: PMC8201054 DOI: 10.3390/cancers13112840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 02/03/2023] Open
Abstract
(1) Background: Today, the discovery of novel anticancer agents with multitarget effects and high safety margins represents a high challenge. Drug discovery efforts indicated that benzopyrane scaffolds possess a wide range of pharmacological activities. This spurs on building a skeletally diverse library of benzopyranes to identify an anticancer lead drug candidate. Here, we aim to characterize the anticancer effect of a novel benzopyrane derivative, aiming to develop a promising clinical anticancer candidate. (2) Methods: The anticancer effect of SIMR1281 against a panel of cancer cell lines was tested. In vitro assays were performed to determine the effect of SIMR1281 on GSHR, TrxR, mitochondrial metabolism, DNA damage, cell cycle progression, and the induction of apoptosis. Additionally, SIMR1281 was evaluated in vivo for its safety and in a xenograft mice model. (3) Results: SIMR1281 strongly inhibits GSHR while it moderately inhibits TrxR and modulates the mitochondrial metabolism. SIMR1281 inhibits the cell proliferation of various cancers. The antiproliferative activity of SIMR1281 was mediated through the induction of DNA damage, perturbations in the cell cycle, and the inactivation of Ras/ERK and PI3K/Akt pathways. Furthermore, SIMR1281 induced apoptosis and attenuated cell survival machinery. In addition, SIMR1281 reduced the tumor volume in a xenograft model while maintaining a high in vivo safety profile at a high dose. (4) Conclusions: Our findings demonstrate the anticancer multitarget effect of SIMR1281, including the dual inhibition of glutathione and thioredoxin reductases. These findings support the development of SIMR1281 in preclinical and clinical settings, as it represents a potential lead compound for the treatment of cancer.
Collapse
Affiliation(s)
- Dana M. Zaher
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Wafaa S. Ramadan
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Raafat El-Awady
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hany A. Omar
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Fatema Hersi
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Vunnam Srinivasulu
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
| | - Ibrahim Y. Hachim
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Farah I. Al-Marzooq
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Cijo G. Vazhappilly
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- School of Arts and Sciences, American University of Ras Al Khaimah, P.O. Box 10021, Ras Al Khaimah 10021, United Arab Emirates
| | - Salim Merali
- School of Pharmacy, Temple University, 3307 N Broad Street, Room 552, Philadelphia, PA 19140, USA; (S.M.); (C.M.)
| | - Carmen Merali
- School of Pharmacy, Temple University, 3307 N Broad Street, Room 552, Philadelphia, PA 19140, USA; (S.M.); (C.M.)
| | - Nelson C. Soares
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Paul Schilf
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany;
| | - Saleh M. Ibrahim
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany;
| | - Taleb H. Al-Tel
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence: ; Tel.: +971-6505-7417
| |
Collapse
|
13
|
Taguchi T, Kurata M, Onishi I, Kinowaki Y, Sato Y, Shiono S, Ishibashi S, Ikeda M, Yamamoto M, Kitagawa M, Yamamoto K. SECISBP2 is a novel prognostic predictor that regulates selenoproteins in diffuse large B-cell lymphoma. J Transl Med 2021; 101:218-227. [PMID: 33077808 DOI: 10.1038/s41374-020-00495-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 12/31/2022] Open
Abstract
The overexpression of glutathione peroxidase 4 (GPX4; an enzyme that suppresses peroxidation of membrane phospholipids) is considered a poor prognostic predictor of diffuse large B-cell lymphoma (DLBCL). However, the mechanisms employed in GPX4 overexpression remain unknown. GPX4 is translated as a complete protein upon the binding of SECISBP2 to the selenocysteine insertion sequence (SECIS) on the 3'UTR of GPX4 mRNA. In this study, we investigated the expression of SECISBP2 and its subsequent regulation of GPX4 and TXNRD1 in DLBCL patients. Moreover, we determined the significance of the expression of these selenoproteins in vitro using MD901 and Raji cells. SECISBP2 was positive in 45.5% (75/165 cases) of DLBCL samples. The SECISBP2-positive group was associated with low overall survival (OS) as compared to the SECISBP2-negative group (P = 0.006). Similarly, the SECISBP2 and GPX4 or TXNRD1 double-positive groups (P < 0.001), as well as the SECISBP2, GPX4, and TXNRD1 triple-positive group correlated with poor OS (P = 0.001), suggesting that SECISBP2 may serve as an independent prognostic predictor for DLBCL (hazard ratio (HR): 2.693, P = 0.008). In addition, western blotting showed a decrease in GPX4 and TXNRD1 levels in SECISBP2-knockout (KO) MD901 and Raji cells. Oxidative stress increased the accumulation of reactive oxygen species in SECISBP2-KO cells (MD901; P < 0.001, Raji; P = 0.020), and reduced cell proliferation (MD901; P = 0.001, Raji; P = 0.030), suggesting that SECISBP2-KO suppressed resistance to oxidative stress. Doxorubicin treatment increased the rate of cell death in SECISBP2-KO cells (MD901; P < 0.001, Raji; P = 0.048). Removal of oxidative stress inhibited the altered cell death rate. Taken together, our results suggest that SECISBP2 may be a novel therapeutic target in DLBCL.
Collapse
MESH Headings
- Aged
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic/genetics
- Gene Knockout Techniques
- Humans
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/mortality
- Male
- Middle Aged
- Oxidative Stress/genetics
- Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Prognosis
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Selenoproteins/genetics
- Selenoproteins/metabolism
- Thioredoxin Reductase 1/genetics
- Thioredoxin Reductase 1/metabolism
Collapse
Affiliation(s)
- Towako Taguchi
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Iichiroh Onishi
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yuko Kinowaki
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yunosuke Sato
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Department of Anesthesiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Sayuri Shiono
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Sachiko Ishibashi
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Masumi Ikeda
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Masahide Yamamoto
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Masanobu Kitagawa
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
14
|
Abstract
Metabolic pathways and redox reactions are at the core of life. In the past decade(s), numerous discoveries have shed light on how metabolic pathways determine the cellular fate and function of lymphoid and myeloid cells, giving rise to an area of research referred to as immunometabolism. Upon activation, however, immune cells not only engage specific metabolic pathways but also rearrange their oxidation-reduction (redox) system, which in turn supports metabolic reprogramming. In fact, studies addressing the redox metabolism of immune cells are an emerging field in immunology. Here, we summarize recent insights revealing the role of reactive oxygen species (ROS) and the differential requirement of the main cellular antioxidant pathways, including the components of the thioredoxin (TRX) and glutathione (GSH) pathways, as well as their transcriptional regulator NF-E2-related factor 2 (NRF2), for proliferation, survival and function of T cells, B cells and macrophages.
Collapse
Affiliation(s)
- Jonathan Muri
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland.
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
15
|
Karatas F, Acat M, Sahin S, Inci F, Karatas G, Neselioglu S, Haskul I, Erel O. The prognostic and predictive significance of serum thiols and disulfide levels in advanced non-small cell lung cancer. Aging Male 2020; 23:619-628. [PMID: 30651017 DOI: 10.1080/13685538.2018.1559805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Serum Total Thiol (TT), Native Thiol (NT), and Disulfide (SS) levels were found significantly lower in benign proliferative pathologies and cancer disease compared to healthy subjects. We conducted this prospective study to investigate the possible predictive and prognostic significance of these markers in non-small cell lung cancer (NSCLC), which is one of the most associated cancers with oxidative stress. This was a non-randomized, prospective, and case-control study of 120 subjects, including 60 patients with metastatic or inoperable NSCLC at the time of diagnosis and 60 demographically-matched controls. Morning fasting venous blood serum samples from both NSCLC and control group were stored at -80 °C for equal periods and then TT, NT, and SS levels were measured spectrophotometrically. Serum TT, NT, and SS levels were compared between groups and their relationships with demographic features and survival of NSCLC patients were analyzed. In results, Serum TT, NT, and SS levels were significantly lower in NSCLC patients than those in control group, with a low SS level being an independent indicator of poor survival.
Collapse
Affiliation(s)
- Fatih Karatas
- Department of Medical Oncology, Karabuk University Faculty of Medicine, Karabuk, Turkey
| | - Murat Acat
- Department of Pulmonology, Karabuk University Faculty of Medicine, Karabuk, Turkey
| | - Suleyman Sahin
- Department of Medical Oncology, Van Training and Research Hospital, Van, Turkey
| | - Fatih Inci
- Department of Medical Oncology, Karabuk University Faculty of Medicine, Karabuk, Turkey
| | - Gulsah Karatas
- Department of Phsical Therapy and Rehabilitation, Karabuk University Faculty of Medicine, Karabuk, Turkey
| | - Salim Neselioglu
- Department of Biochemistry, Yildirim Beyazit University Faculty of Medicine, Ankara, Turkey
| | - Ismail Haskul
- Department of Biochemistry, Karabuk University Faculty of Medicine, Karabuk, Turkey
| | - Ozcan Erel
- Department of Biochemistry, Yildirim Beyazit University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
16
|
Valle AC, Yeh C, Huang Y. Near Infrared-Activatable Platinum-Decorated Gold Nanostars for Synergistic Photothermal/Ferroptotic Therapy in Combating Cancer Drug Resistance. Adv Healthc Mater 2020; 9:e2000864. [PMID: 32945131 DOI: 10.1002/adhm.202000864] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/15/2020] [Indexed: 12/15/2022]
Abstract
Ferroptotic cell death results from glutathione peroxidase 4 (GPX4) inactivation and/or glutathione (GSH) depletion. Elevated GSH levels are often found in multidrug-resistant (MDR) tumor cells, reducing their sensitivity to chemotherapeutic drugs and the efficacy of treatment. MDR cells also acquire a dependency on GPX4, reducing their oxidative stress and promoting their survival. Therefore, the depletion of GSH and inactivation of GPX4 has the potential to be a superior treatment strategy for MDR tumors. Platinum-decorated gold nanostars (Pt-AuNS) are presented as a novel metal nanoprodrug for ferroptotic therapy against MDR tumors. Under dark conditions, the synthesized Pt-AuNS exhibit negligible levels of toxicity. Upon exposure of the Pt-AuNS to near-infrared (NIR) light, active metallic (Pt and Au) species are released, subsequently inducing cytotoxicity. The mechanism of action is attributed to GSH depletion and GPX4 inactivation, accumulating lipid hydroperoxides, which in turn leads to ferroptosis. In in vivo xenograft, the MDR cancer model confirmed the NIR light-activation of Pt-AuNS prodrugs, resulting in efficient ferroptotic therapeutic action against MDR tumors without long-term side effects. The findings lay the groundwork for using Pt-AuNS prodrugs responsive to NIR light as ferroptosis-inducing agents in chemo-resistant cancer cells and demonstrate their potential for use in future clinical applications.
Collapse
Affiliation(s)
- Andrea C. Valle
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| | - Chih‐Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| | - Yu‐Fen Huang
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
- Institute of Analytical and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| |
Collapse
|
17
|
Jalali S, Shi J, Buko A, Ahsan N, Paludo J, Serres M, Wellik LE, Abeykoon J, Kim H, Tang X, Yang ZZ, Novak AJ, Witzig TE, Ansell SM. Increased glutathione utilization augments tumor cell proliferation in Waldenstrom Macroglobulinemia. Redox Biol 2020; 36:101657. [PMID: 32763516 PMCID: PMC7404570 DOI: 10.1016/j.redox.2020.101657] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/03/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer cells. In Waldenstrom Macroglobulinemia (WM), the infiltration of IgM-secreting lymphoplamacytic cells into the bone marrow (BM) could shift the homeostasis of proteins and metabolites towards a permissive niche for tumor growth. Here, we investigated whether alerted metabolic pathways contribute to the pathobiology of WM and whether the cytokine composition of the BM promotes such changes. Metabolomics analysis on WM patients and normal donors' serum samples revealed a total of 75 metabolites that were significantly altered between two groups. While these metabolites belonged to amino acids, glucose, glutathione and lipid metabolism pathways, the highest number of the differentially expressed metabolites belonged to glutathione metabolism. Proteomics analysis and immunohistochemical staining both confirmed the increased protein levels mediating glutathione metabolism, including GCLC, MT1X, QPCT and GPX3. Moreover, treatment with IL-6 and IL-21, cytokines that induce WM cell proliferation and IgM secretion, increased gene expression of the amino acid transporters mediating glutathione metabolism, including ASCT2, SLC7A11 and 4F2HC, indicating that cytokines in the WM BM could modulate glutathione metabolism. Glutathione synthesis inhibition using Buthionine sulphoximine (BSO) significantly reduced WM cells proliferation in vitro, accompanied with decreased NFκB-p65 and MAPK-p38 phosphorylation. Moreover, BSO treatment significantly reduced the tumor growth rate in a WM xenograft model, further highlighting the role of glutathione metabolism in promoting tumor growth and proliferation. In summary, our data highlight a central role for glutathione metabolism in WM pathobiology and indicate that intervening with the metabolic processes could be a potential therapy for WM patients.
Collapse
Affiliation(s)
- Shahrzad Jalali
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jie Shi
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA; Department of Hematology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Alex Buko
- Human Metabolome Technologies (HMT) America, Boston, MA, USA
| | - Nagib Ahsan
- COBRE Center for Cancer Research Development, Proteomics Core Facility, Rhode Island Hospital, Providence, RI, 02903, USA; Division of Biology and Medicine, Brown University, Providence, RI, 02903, USA
| | - Jonas Paludo
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Makayla Serres
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Linda E Wellik
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jithma Abeykoon
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - HyoJin Kim
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Xinyi Tang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Anne J Novak
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Thomas E Witzig
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Stephen M Ansell
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
Zhang C, Fortin PY, Barnoin G, Qin X, Wang X, Fernandez Alvarez A, Bijani C, Maddelein ML, Hemmert C, Cuvillier O, Gornitzka H. An Artemisinin-Derivative-(NHC)Gold(I) Hybrid with Enhanced Cytotoxicity through Inhibition of NRF2 Transcriptional Activity. Angew Chem Int Ed Engl 2020; 59:12062-12068. [PMID: 32304346 DOI: 10.1002/anie.202002992] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/14/2020] [Indexed: 01/19/2023]
Abstract
A family of hybrid complexes combining two biologically active motifs, an artemisinin derivative and a cationic bis(NHC)-gold(I) unit, has been synthesized. One of these complexes, 2 a, has been analyzed by single-crystal X-ray diffraction. 2 a shows strong anticancer activities on a large panel of human cancer cell models (prostate, breast, lung, liver, bladder, bone, acute and chronic myeloid leukemias) with GI50 values in the nm range, together with a high selectivity. An original and distinctive mechanism of action, that is, through inhibition of the redox antioxidant NRF2 transcription factor (strongly associated with aggressiveness and resistance to cancer therapies) has been evidenced. 2 a could remarkably sensitize to sorafenib in HepG2 liver cells, in which dysregulated NRF2 signaling is linked to primary and acquired drug resistance. 2 a also inhibited NF-κB and HIF transcriptional activities, which are also associated with progression and resistance in cancer. Our findings provide evidence that hybrid (NHC)gold(I) compounds represent a new class of organometallic hybrid molecules that may yield new therapeutic agents.
Collapse
Affiliation(s)
- Chen Zhang
- LCC-CNRS, Université de Toulouse, CNRS, UPS, Toulouse, France.,Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France.,Present address: Shanghai Key Laboratory of New Drug Design, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Pierre-Yves Fortin
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Xue Qin
- LCC-CNRS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Xing Wang
- LCC-CNRS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | | | - Marie-Lise Maddelein
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Olivier Cuvillier
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Heinz Gornitzka
- LCC-CNRS, Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
19
|
Zhang C, Fortin P, Barnoin G, Qin X, Wang X, Fernandez Alvarez A, Bijani C, Maddelein M, Hemmert C, Cuvillier O, Gornitzka H. An Artemisinin‐Derivative–(NHC)Gold(I) Hybrid with Enhanced Cytotoxicity through Inhibition of NRF2 Transcriptional Activity. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002992] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Chen Zhang
- LCC-CNRS Université de Toulouse CNRS, UPS Toulouse France
- Institut de Pharmacologie et de Biologie Structurale Université de Toulouse CNRS, UPS Toulouse France
- Present address: Shanghai Key Laboratory of New Drug Design State Key Laboratory of Bioreactor Engineering School of Pharmacy East China University of Science and Technology Shanghai 200237 China
| | - Pierre‐Yves Fortin
- Institut de Pharmacologie et de Biologie Structurale Université de Toulouse CNRS, UPS Toulouse France
| | | | - Xue Qin
- LCC-CNRS Université de Toulouse CNRS, UPS Toulouse France
| | - Xing Wang
- LCC-CNRS Université de Toulouse CNRS, UPS Toulouse France
| | | | | | - Marie‐Lise Maddelein
- Institut de Pharmacologie et de Biologie Structurale Université de Toulouse CNRS, UPS Toulouse France
| | | | - Olivier Cuvillier
- Institut de Pharmacologie et de Biologie Structurale Université de Toulouse CNRS, UPS Toulouse France
| | | |
Collapse
|
20
|
Zhang H, Wang L, Chu Y. Reactive oxygen species: The signal regulator of B cell. Free Radic Biol Med 2019; 142:16-22. [PMID: 31185253 DOI: 10.1016/j.freeradbiomed.2019.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 05/17/2019] [Accepted: 06/04/2019] [Indexed: 02/09/2023]
Abstract
Reactive oxygen species (ROS) are indispensable for determining the fate of immune cells in both physiological and pathogenic environments, thus stimulating the interest of immunologists and clinicians. B cells are essential in maintaining immune homeostasis, and studies have indicated that ROS affect the maturation, activation and differentiation of B cells by controlling the signaling molecules in various molecular pathways. In the present review, we aimed to summarize the biological properties of ROS and the mechanisms by which ROS regulate B cell signaling pathways. We propose that ROS and their mediated signal transduction can be a new approach for manipulating B cell immune functions.
Collapse
Affiliation(s)
- Hushan Zhang
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China; Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China; Biotherapy Research Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
21
|
Pereira C, Rodrigues IS, Pereira LMG, Lisboa J, Pinto RD, Araújo L, Oliveira P, Benz R, Dos Santos NMS, do Vale A. Role of AIP56 disulphide bond and its reduction by cytosolic redox systems for efficient intoxication. Cell Microbiol 2019; 22:e13109. [PMID: 31454143 DOI: 10.1111/cmi.13109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/08/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022]
Abstract
Apoptosis-inducing protein of 56 kDa (AIP56) is a major virulence factor of Photobacterium damselae subsp. piscicida, a gram-negative pathogen that infects warm water fish species worldwide and causes serious economic losses in aquacultures. AIP56 is a single-chain AB toxin composed by two domains connected by an unstructured linker peptide flanked by two cysteine residues that form a disulphide bond. The A domain comprises a zinc-metalloprotease moiety that cleaves the NF-kB p65, and the B domain is involved in binding and internalisation of the toxin into susceptible cells. Previous experiments suggested that disruption of AIP56 disulphide bond partially compromised toxicity, but conclusive evidences supporting the importance of that bond in intoxication were lacking. Here, we show that although the disulphide bond of AIP56 is dispensable for receptor recognition, endocytosis, and membrane interaction, it needs to be intact for efficient translocation of the toxin into the cytosol. We also show that the host cell thioredoxin reductase-thioredoxin system is involved in AIP56 intoxication by reducing the disulphide bond of the toxin at the cytosol. The present study contributes to a better understanding of the molecular mechanisms operating during AIP56 intoxication and reveals common features shared with other AB toxins.
Collapse
Affiliation(s)
- Cassilda Pereira
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Inês S Rodrigues
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Liliana M G Pereira
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Johnny Lisboa
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Rute D Pinto
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Leonor Araújo
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Pedro Oliveira
- EPIUnit, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Roland Benz
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
| | - Nuno M S Dos Santos
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana do Vale
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
22
|
Halvarsson C, Rörby E, Eliasson P, Lang S, Soneji S, Jönsson JI. Putative Role of Nuclear Factor-Kappa B But Not Hypoxia-Inducible Factor-1α in Hypoxia-Dependent Regulation of Oxidative Stress in Hematopoietic Stem and Progenitor Cells. Antioxid Redox Signal 2019; 31:211-226. [PMID: 30827134 PMCID: PMC6590716 DOI: 10.1089/ars.2018.7551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 12/14/2022]
Abstract
Aims: Adaptation to low oxygen of hematopoietic stem cells (HSCs) in the bone marrow has been demonstrated to depend on the activation of hypoxia-inducible factor (HIF)-1α as well as the limited production of reactive oxygen species (ROS). In this study, we aimed at determining whether HIF-1α is involved in protecting HSCs from ROS. Results: Oxidative stress was induced by DL-buthionine-(S,R)-sulfoximine (BSO)-treatment, which increases the mitochondrial ROS level. Hypoxia rescued Lineage-Sca-1+c-kit+ (LSK) cells from BSO-induced apoptosis, whereas cells succumbed to apoptosis in normoxia. Apoptosis in normoxia was inhibited with the antioxidant N-acetyl-L-cysteine or by overexpression of anti-apoptotic BCL-2. Moreover, stabilized expression of oxygen-insensitive HIFs could not protect LSK cells from oxidative stress-induced apoptosis at normoxia, neither could short hairpin RNA to Hif-1α inhibit the protective effects by hypoxia in LSK cells. Likewise, BSO treatment of LSK cells from Hif-1α knockout mice did not suppress the effects seen in hypoxia. Microarray analysis identified the nuclear factor-kappa B (NF-κB) pathway as a pathway induced by hypoxia. By using NF-κB lentiviral construct and DNA-binding assay, we found increased NF-κB activity in cells cultured in hypoxia compared with normoxia. Using an inhibitor against NF-κB activation, we could confirm the involvement of NF-κB signaling as BSO-mediated cell death was significantly increased in hypoxia after adding the inhibitor. Innovation: HIF-1α is not involved in protecting HSCs and progenitors to elevated levels of ROS on glutathione depletion during hypoxic conditions. Conclusion: The study proposes a putative role of NF-κB signaling as a hypoxia-induced regulator in early hematopoietic cells.
Collapse
Affiliation(s)
- Camilla Halvarsson
- Experimental Hematology Unit, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Emma Rörby
- Experimental Hematology Unit, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Pernilla Eliasson
- Experimental Hematology Unit, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Stefan Lang
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Shamit Soneji
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Jan-Ingvar Jönsson
- Experimental Hematology Unit, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
23
|
Bian M, Fan R, Zhao S, Liu W. Targeting the Thioredoxin System as a Strategy for Cancer Therapy. J Med Chem 2019; 62:7309-7321. [PMID: 30963763 DOI: 10.1021/acs.jmedchem.8b01595] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Thioredoxin reductase (TrxR) participates in the regulation of redox reactions in organisms. It works mainly via its substrate molecule, thioredoxin, to maintain the redox balance and regulate signal transduction, which controls cell proliferation, differentiation, death, and other important physiological processes. In recent years, increasing evidence has shown that the overactivation of TrxR is related to the development of tumors. The exploration of TrxR-targeted antitumor drugs has attracted wide attention and is expected to provide new therapies for cancer treatment. In this perspective, we highlight the specific relationship between TrxR and apoptotic signaling pathways. The cytoplasm and mitochondria both contain TrxR, resulting in the activation of apoptosis. TrxR activity influences reactive oxygen species (ROS) and further regulates the inflammatory signaling pathway. In addition, we discuss representative TrxR inhibitors with anticancer activity and analyze the challenges in developing TrxR inhibitors as anticancer drugs.
Collapse
Affiliation(s)
- Mianli Bian
- Institute of Chinese Medicine, School of Pharmacy , Nanjing University of Chinese Medicine , Nanjing 210023 , P. R. China
| | - Rong Fan
- Institute of Chinese Medicine, School of Pharmacy , Nanjing University of Chinese Medicine , Nanjing 210023 , P. R. China
| | - Sai Zhao
- Institute of Chinese Medicine, School of Pharmacy , Nanjing University of Chinese Medicine , Nanjing 210023 , P. R. China.,Institute of New Medicine Research , Nanjing Hicin Pharmaceutical Co. Ltd. , Nanjing 210046 , P. R. China
| | - Wukun Liu
- Institute of Chinese Medicine, School of Pharmacy , Nanjing University of Chinese Medicine , Nanjing 210023 , P. R. China.,State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing 210009 , P. R. China
| |
Collapse
|
24
|
Muri J, Thut H, Heer S, Krueger CC, Bornkamm GW, Bachmann MF, Kopf M. The thioredoxin-1 and glutathione/glutaredoxin-1 systems redundantly fuel murine B-cell development and responses. Eur J Immunol 2019; 49:709-723. [PMID: 30802940 DOI: 10.1002/eji.201848044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/22/2019] [Accepted: 02/22/2019] [Indexed: 11/10/2022]
Abstract
Antioxidant systems maintain cellular redox homeostasis. The thioredoxin-1 (Trx1) and the glutathione (GSH)/glutaredoxin-1 (Grx1) systems are key players in preserving cytosolic redox balance. In fact, T lymphocytes critically rely on reducing equivalents from the Trx1 system for DNA biosynthesis during metabolic reprogramming upon activation. We here show that the Trx1 system is also indispensable for development and functionality of marginal zone (MZ) B cells and B1 cells in mice. In contrast, development of conventional B cells, follicular B-cell homeostasis, germinal center reactions, and antibody responses are redundantly sustained by both antioxidant pathways. Proliferating B2 cells lacking Txnrd1 have increased glutathione (GSH) levels and upregulated cytosolic Grx1, which is barely detectable in expanding thymocytes. These results suggest that the redox capacity driving proliferation is more robust and flexible in B cells than in T cells, which may have profound implications for the therapy of B and T-cell neoplasms.
Collapse
Affiliation(s)
- Jonathan Muri
- Institute of Molecular Health Sciences, ETH Zurich, Zürich, Switzerland
| | - Helen Thut
- Institute of Molecular Health Sciences, ETH Zurich, Zürich, Switzerland
| | - Sebastian Heer
- Institute of Molecular Health Sciences, ETH Zurich, Zürich, Switzerland
| | - Caroline C Krueger
- Department of BioMedical Research, University of Bern, Department of Immunology RIA, University Hospital Bern, Bern, Switzerland
| | - Georg W Bornkamm
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Zentrum München, München, Germany
| | - Martin F Bachmann
- Department of BioMedical Research, University of Bern, Department of Immunology RIA, University Hospital Bern, Bern, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zurich, Zürich, Switzerland
| |
Collapse
|
25
|
Chen X, Mims J, Huang X, Singh N, Motea E, Planchon SM, Beg M, Tsang AW, Porosnicu M, Kemp ML, Boothman DA, Furdui CM. Modulators of Redox Metabolism in Head and Neck Cancer. Antioxid Redox Signal 2018; 29:1660-1690. [PMID: 29113454 PMCID: PMC6207163 DOI: 10.1089/ars.2017.7423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/04/2017] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Head and neck squamous cell cancer (HNSCC) is a complex disease characterized by high genetic and metabolic heterogeneity. Radiation therapy (RT) alone or combined with systemic chemotherapy is widely used for treatment of HNSCC as definitive treatment or as adjuvant treatment after surgery. Antibodies against epidermal growth factor receptor are used in definitive or palliative treatment. Recent Advances: Emerging targeted therapies against other proteins of interest as well as programmed cell death protein 1 and programmed death-ligand 1 immunotherapies are being explored in clinical trials. CRITICAL ISSUES The disease heterogeneity, invasiveness, and resistance to standard of care RT or chemoradiation therapy continue to constitute significant roadblocks for treatment and patients' quality of life (QOL) despite improvements in treatment modality and the emergence of new therapies over the past two decades. FUTURE DIRECTIONS As reviewed here, alterations in redox metabolism occur at all stages of HNSCC management, providing opportunities for improved prevention, early detection, response to therapies, and QOL. Bioinformatics and computational systems biology approaches are key to integrate redox effects with multiomics data from cells and clinical specimens and to identify redox modifiers or modifiable target proteins to achieve improved clinical outcomes. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Xiaofei Chen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jade Mims
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Xiumei Huang
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Naveen Singh
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Edward Motea
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | | | - Muhammad Beg
- Department of Internal Medicine, Division of Hematology-Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Allen W. Tsang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mercedes Porosnicu
- Department of Internal Medicine, Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Melissa L. Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - David A. Boothman
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Cristina M. Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
26
|
Parrales A, McDonald P, Ottomeyer M, Roy A, Shoenen FJ, Broward M, Bruns T, Thamm DH, Weir SJ, Neville KA, Iwakuma T, Fulbright JM. Comparative oncology approach to drug repurposing in osteosarcoma. PLoS One 2018; 13:e0194224. [PMID: 29579058 PMCID: PMC5868798 DOI: 10.1371/journal.pone.0194224] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 02/27/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Osteosarcoma is an orphan disease for which little improvement in survival has been made since the late 1980s. New drug discovery for orphan diseases is limited by the cost and time it takes to develop new drugs. Repurposing already approved FDA-drugs can help overcome this limitation. Another limitation of cancer drug discovery is the lack of preclinical models that accurately recapitulate what occurs in humans. For OS using dogs as a model can minimize this limitation as OS in canines develops spontaneously, is locally invasive and metastasizes to the lungs as it does in humans. METHODS In our present work we used high-throughput screens to identify drugs from a library of 2,286 FDA-approved drugs that demonstrated selective growth inhibition against both human and canine OS cell lines. The identified lead compound was then tested for synergy with 7 other drugs that have demonstrated activity against OS. These results were confirmed with in vitro assays and an in vivo murine model of OS. RESULTS We identified 13 drugs that demonstrated selective growth inhibition against both human and canine OS cell lines. Auranofin was selected for further in vitro combination drug screens. Auranofin showed synergistic effects with vorinostat and rapamycin on OS viability and apoptosis induction. Auranofin demonstrated single-agent growth inhibition in both human and canine OS xenografts, and cooperative growth inhibition was observed in combination with rapamycin or vorinostat. There was a significant decrease in Ki67-positive cells and an increase in cleaved caspase-3 levels in tumor tissues treated with a combination of auranofin and vorinostat or rapamycin. CONCLUSIONS Auranofin, alone or in combination with rapamycin or vorinostat, may be useful new treatment strategies for OS. Future studies may evaluate the efficacy of auranofin in dogs with OS as a prelude to human clinical evaluation.
Collapse
Affiliation(s)
- Alejandro Parrales
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Peter McDonald
- High Throughput Screening Laboratory, University of Kansas Cancer Center, University of Kansas, Lawrence, Kansas, United States of America
| | - Megan Ottomeyer
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Anuradha Roy
- High Throughput Screening Laboratory, University of Kansas Cancer Center, University of Kansas, Lawrence, Kansas, United States of America
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
| | - Frank J. Shoenen
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, Kansas, United States of America
| | - Melinda Broward
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Tyce Bruns
- Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Douglas H. Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, United States of America
- University of Colorado Comprehensive Cancer Center, Aurora, Colorado, United States of America
| | - Scott J. Weir
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Department of Pediatrics, University of Missouri Kansas City, Kansas City, Missouri, United States of America
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas, Kansas City, Kansas, United States of America
| | - Kathleen A. Neville
- Arkansas Children’s Hospital, Little Rock, Arkansas, United States of America
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Division of Hematology and Oncology, Children’s Mercy Hospital and Clinics, Kansas City, Missouri, United States of America
| | - Joy M. Fulbright
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- Department of Pediatrics, University of Missouri Kansas City, Kansas City, Missouri, United States of America
- Division of Hematology and Oncology, Children’s Mercy Hospital and Clinics, Kansas City, Missouri, United States of America
| |
Collapse
|
27
|
Rashmi R, Huang X, Floberg JM, Elhammali AE, McCormick ML, Patti GJ, Spitz DR, Schwarz JK. Radioresistant Cervical Cancers Are Sensitive to Inhibition of Glycolysis and Redox Metabolism. Cancer Res 2018; 78:1392-1403. [PMID: 29339540 DOI: 10.1158/0008-5472.can-17-2367] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/21/2017] [Accepted: 01/10/2018] [Indexed: 12/14/2022]
Abstract
Highly glycolytic cervical cancers largely resist treatment by cisplatin and coadministered pelvic irradiation as the present standard of care. In this study, we investigated the effects of inhibiting glycolysis and thiol redox metabolism to evaluate them as alternate treatment strategies in these cancers. In a panel of multiple cervical cancer cell lines, we evaluated sensitivity to inhibition of glycolysis (2-deoxyglucose, 2-DG) with or without simultaneous inhibition of glutathione and thioredoxin metabolism (BSO/AUR). Intracellular levels of total and oxidized glutathione, thioredoxin reductase activity, and indirect measures of intracellular reactive oxygen species were compared before and after treatment. Highly radioresistant cells were the most sensitive to 2-DG, whereas intermediate radioresistant cells were sensitive to 2-DG plus BSO/AUR. In response to 2-DG/BSO/AUR treatment, we observed increased levels of intracellular oxidized glutathione, redox-sensitive dye oxidation, and decreased glucose utilization via multiple metabolic pathways including the tricarboxylic acid cycle. 2-DG/BSO/AUR treatment delayed the growth of tumors composed of intermediate radioresistant cells and effectively radiosensitized these tumors at clinically relevant radiation doses both in vitro and in vivo Overall, our results support inhibition of glycolysis and intracellular redox metabolism as an effective alternative drug strategy for the treatment of highly glycolytic and radioresistant cervical cancers.Significance: This study suggests a simple metabolic approach to strike at an apparent Achilles' heel in highly glycolytic, radioresistant forms of cervical cancers, possibly with broader applications in cancer therapy. Cancer Res; 78(6); 1392-403. ©2018 AACR.
Collapse
Affiliation(s)
- Ramachandran Rashmi
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Xiaojing Huang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - John M Floberg
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Adnan E Elhammali
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Michael L McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Gary J Patti
- Departments of Chemistry and Medicine, Washington University, St. Louis, Missouri
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Julie K Schwarz
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri. .,Department of Cell Biology and Physiology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
28
|
Abstract
Purpose of review The goal of this review is to summarize recent advances in our understanding of the regulation of redox homeostasis and the subtype-specific role of antioxidant enzymes in B-cell-derived malignancies. Furthermore, it presents selected prooxidative therapeutic strategies against B-cell neoplasms. Recent findings Recent reports have shown that the disturbed redox homeostasis in B-cell malignancies is regulated by cancer-specific signaling pathways and therefore varies between the individual subtypes. For instance, in a subtype of diffuse large B-cell lymphoma with increased oxidative phosphorylation, elevated reactive oxygen species are accompanied by higher levels of thioredoxin and glutathione and inhibition of either of these systems is selectively toxic to this subtype. In addition, growing number of small molecule inhibitors targeting antioxidant enzymes, such as auranofin, SK053, adenanthin, or decreasing glutathione level, such as imexon, buthionine sulfoximine, and L-cysteinase, trigger specific cytotoxic effects against B-cell malignancies. Lastly, attention is drawn to recent reports of effective treatment modalities involving prooxidative agents and interfering with redox homeostasis provided by stromal cells. Summary Recent findings reveal important differences in redox homeostasis within the distinct subsets of B-cell-derived malignancies that can be therapeutically exploited to improve existing treatment and to overcome drug resistance.
Collapse
|
29
|
Roh JL, Jang H, Kim EH, Shin D. Targeting of the Glutathione, Thioredoxin, and Nrf2 Antioxidant Systems in Head and Neck Cancer. Antioxid Redox Signal 2017; 27:106-114. [PMID: 27733046 DOI: 10.1089/ars.2016.6841] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AIMS The glutathione (GSH), thioredoxin (Trx), and Nrf2 systems represent a major defense against reactive oxygen species (ROS), the cellular imbalance of which in cancer promotes growth and therapeutic resistance. This study investigated whether targeting the GSH, Trx, and Nrf2 antioxidant systems effectively eliminated head and neck cancer (HNC). RESULTS At high concentrations, auranofin, but not buthionine sulfoximine (BSO) alone, decreased the viability of HNC, whereas even at low concentrations, auranofin plus BSO synergized to kill HNC cells. Dual silencing of the genes for GCLM and TrxR1 induced GSH depletion, Trx activity inhibition, and ROS accumulation, synergistically killing HNC cells. Inhibition of the GSH and Trx systems resulted in activation of the Nrf2-antioxidant response element (ARE) pathway, which may result in suboptimal GSH and Trx inhibition where HNC is resistant. Genetic inhibition of Nrf2 and/or HO-1 or trigonelline enhanced growth suppression, ROS accumulation, and cell death from GSH and Trx inhibition. The in vivo effects of GSH, Trx, and Nrf2 system inhibition were confirmed in a mouse HNC xenograft model by achieving growth inhibition >60% compared with those of control. Innovations: This study is the first to show that triple inhibition of GSH, Trx, and Nrf2 pathways could be an effective method to overcome the resistance of HNC. CONCLUSIONS Inhibition of the Nrf2-ARE pathway in addition to dual inhibition of the GSH and Trx antioxidant systems can effectively eliminate resistant HNC. Antioxid. Redox Signal. 27, 106-114.
Collapse
Affiliation(s)
- Jong-Lyel Roh
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Republic of Korea
| | - Hyejin Jang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Republic of Korea
| | - Eun Hye Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Republic of Korea
| | - Daiha Shin
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Republic of Korea
| |
Collapse
|
30
|
Poprac P, Jomova K, Simunkova M, Kollar V, Rhodes CJ, Valko M. Targeting Free Radicals in Oxidative Stress-Related Human Diseases. Trends Pharmacol Sci 2017; 38:592-607. [PMID: 28551354 DOI: 10.1016/j.tips.2017.04.005] [Citation(s) in RCA: 702] [Impact Index Per Article: 87.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 12/14/2022]
Abstract
Cancer and Alzheimer's disease (AD) are characterized by (i) opposing biological mechanisms, (ii) an inverse correlation between their incidences, and (iii) oxidative stress being a common denominator of both diseases. Increased formation of reactive oxygen species (ROS) in cancer cells from oncogenic signaling and/or metabolic disturbances leads to upregulation of cellular antioxidant capacity to maintain ROS levels below a toxic threshold. Combining drugs that induce high levels of ROS with compounds that suppress cellular antioxidant capacity by depleting antioxidant systems [glutathione (GSH), superoxide dismutase (SOD), and thioredoxin (TRX)] and/or targeting glucose metabolism represents a potential anticancer strategy. In AD, free metals and/or Aβ:metal complexes may cause damage to biomolecules in the brain (via Fenton reaction), including DNA. Metal chelation, based on the application of selective metal chelators or metal delivery, may induce neuroprotective signaling and represents a promising therapeutic strategy. This review examines therapeutic strategies based on the modulation of oxidative stress in cancer and AD.
Collapse
Affiliation(s)
- Patrik Poprac
- Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, 812 37 Bratislava, Slovakia
| | - Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University, Trieda Andreja Hlinku 1, 949 74 Nitra, Slovakia
| | - Miriama Simunkova
- Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, 812 37 Bratislava, Slovakia
| | - Vojtech Kollar
- School of Economics and Management in Public Administration in Bratislava, Furdekova 16, 851 04 Bratislava, Slovakia
| | | | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, 812 37 Bratislava, Slovakia.
| |
Collapse
|
31
|
Transglutaminase 2 modulation of NF-κB signaling in astrocytes is independent of its ability to mediate astrocytic viability in ischemic injury. Brain Res 2017; 1668:1-11. [PMID: 28522262 DOI: 10.1016/j.brainres.2017.05.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/14/2017] [Accepted: 05/10/2017] [Indexed: 12/20/2022]
Abstract
Transglutaminase 2 (TG2) is a multifunctional protein that can contribute to cell death and cell survival processes in a variety of disease contexts. Within the brain, TG2 has been shown to promote cell death in ischemic injury when expressed in astrocytes (Colak and Johnson, 2012). However, the specific functions and characteristics of astrocytic TG2 that mediate this effect are largely unknown. Therefore, the goal of this study was to investigate the role of astrocytic TG2 in mediating cellular viability processes in the context of ischemic injury, with a specific focus on its contributions to intracellular signaling cascades. We show that, in response to oxygen/glucose deprivation (OGD), acute lentiviral-mediated knockdown of TG2, as well as inhibition with an irreversible TG2 inhibitor, enhances cell survival. We also show that TG2 depletion increases nuclear factor-κB (NF-κB) signaling, whereas inhibition reduces NF-κB activity. Despite its clear contribution to NF-κB signaling, however, TG2 modulation of NF-κB signaling is not likely to be a major contributor to its ability to mediate astrocytic viability in this context. Overall, the results of this study provide insight into the role of TG2 in astrocytes and suggest possible avenues for future study of the relationship between astrocytic TG2 and ischemic injury.
Collapse
|
32
|
Tessoulin B, Eveillard M, Lok A, Chiron D, Moreau P, Amiot M, Moreau-Aubry A, Le Gouill S, Pellat-Deceunynck C. p53 dysregulation in B-cell malignancies: More than a single gene in the pathway to hell. Blood Rev 2017; 31:251-259. [PMID: 28284458 DOI: 10.1016/j.blre.2017.03.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/02/2017] [Indexed: 12/31/2022]
Abstract
TP53 deletion or mutation is frequent in B-cell malignancies and is associated with a low response rate. We describe here the p53 landscape in B-cell malignancies, from B-Acute Lymphoblastic Leukemia to Plasma Cell Leukemia, by analyzing incidence of gain or loss of function of actors both upstream and within the p53 pathway, namely MYC, RAS, ARF, MDM2, ATM and TP53. Abnormalities are not equally distributed and their incidence is highly variable among malignancies. Deletion and mutation, usually associated, of ATM or TP53 are frequent in Diffuse Large B-Cell Lymphoma and Mantle Cell Lymphoma. MYC gain, absent in post-GC malignancies, is frequent in B-Prolymphocytic-Leukemia, Multiple Myeloma and Plasma Cell Leukemias. RAS mutations are rare except in MM and PCL. Multiple Factorial Analysis notes that MYC deregulation is closely related to TP53 status. Moreover, MYC gain, TP53 deletion and RAS mutations are inversely correlated with survival. Based on this landscape, we further propose targeted therapeutic approaches for the different B-cell malignancies.
Collapse
Affiliation(s)
- B Tessoulin
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Department of Hematology, Nantes University Hospital, Nantes, France.
| | - M Eveillard
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Hematology Biology Department, Nantes University Hospital, Nantes, France
| | - A Lok
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Department of Hematology, Nantes University Hospital, Nantes, France
| | - D Chiron
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - P Moreau
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Department of Hematology, Nantes University Hospital, Nantes, France
| | - M Amiot
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - A Moreau-Aubry
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - S Le Gouill
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Department of Hematology, Nantes University Hospital, Nantes, France
| | - C Pellat-Deceunynck
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.
| |
Collapse
|
33
|
Sharbeen G, Youkhana J, Mawson A, McCarroll J, Nunez A, Biankin A, Johns A, Goldstein D, Phillips P. MutY-Homolog (MYH) inhibition reduces pancreatic cancer cell growth and increases chemosensitivity. Oncotarget 2017; 8:9216-9229. [PMID: 27999205 PMCID: PMC5354726 DOI: 10.18632/oncotarget.13985] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 12/12/2016] [Indexed: 12/30/2022] Open
Abstract
Patients with pancreatic ductal adenocarcinoma (PC) have a poor prognosis due to metastases and chemoresistance. PC is characterized by extensive fibrosis, which creates a hypoxic microenvironment, and leads to increased chemoresistance and intracellular oxidative stress. Thus, proteins that protect against oxidative stress are potential therapeutic targets for PC. A key protein that maintains genomic integrity against oxidative damage is MutY-Homolog (MYH). No prior studies have investigated the function of MYH in PC cells. Using siRNA, we showed that knockdown of MYH in PC cells 1) reduced PC cell proliferation and increased apoptosis; 2) further decreased PC cell growth in the presence of oxidative stress and chemotherapy agents (gemcitabine, paclitaxel and vincristine); 3) reduced PC cell metastatic potential; and 4) decreased PC tumor growth in a subcutaneous mouse model in vivo. The results from this study suggest MYH may be a novel therapeutic target for PC that could potentially improve patient outcome by reducing PC cell survival, increasing the efficacy of existing drugs and reducing metastatic spread.
Collapse
Affiliation(s)
- George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Janet Youkhana
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Amanda Mawson
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Joshua McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, Australia
| | - Andrea Nunez
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Andrew Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow, Scotland, United Kingdom
- The Kinghorn Cancer Centre, Cancer Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Amber Johns
- The Kinghorn Cancer Centre, Cancer Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - David Goldstein
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Phoebe Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
34
|
Sewastianik T, Szydlowski M, Jablonska E, Bialopiotrowicz E, Kiliszek P, Gorniak P, Polak A, Prochorec-Sobieszek M, Szumera-Cieckiewicz A, Kaminski TS, Markowicz S, Nowak E, Grygorowicz MA, Warzocha K, Juszczynski P. FOXO1 is a TXN- and p300-dependent sensor and effector of oxidative stress in diffuse large B-cell lymphomas characterized by increased oxidative metabolism. Oncogene 2016; 35:5989-6000. [PMID: 27132507 DOI: 10.1038/onc.2016.126] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 02/04/2016] [Accepted: 03/07/2016] [Indexed: 12/19/2022]
Abstract
Molecular profiling has led to identification of subtypes of diffuse large B-cell lymphomas (DLBCLs) differing in terms of oncogenic signaling and metabolic programs. The OxPhos-DLBCL subtype is characterized by enhanced mitochondrial oxidative phosphorylation. As increased oxidative metabolism leads to overproduction of potentially toxic reactive oxygen species (ROS), we sought to identify mechanisms responsible for adaptation of OxPhos cells to these conditions. Herein, we describe a mechanism involving the FOXO1-TXN-p300 redox-dependent circuit protecting OxPhos-DLBCL cells from ROS toxicity. We identify a BCL6-dependent transcriptional mechanism leading to relative TXN overexpression in OxPhos cells. We found that OxPhos cells lacking TXN were uniformly more sensitive to ROS and doxorubicin than control cells. Consistent with this, the overall survival of patients with high TXN mRNA expression, treated with doxorubicin-containing regimens, is significantly shorter than of those with low TXN mRNA expression. TXN overexpression curtails p300-mediated FOXO1 acetylation and its nuclear translocation in response to oxidative stress, thus attenuating FOXO1 transcriptional activity toward genes involved in apoptosis and cell cycle inhibition. We also demonstrate that FOXO1 knockdown in cells with silenced TXN expression markedly reduces ROS-induced apoptosis, indicating that FOXO1 is the major sensor and effector of oxidative stress in OxPhos-DLBCLs. These data highlight dynamic, context-dependent modulation of FOXO1 tumor-suppressor functions via acetylation and reveal potentially targetable vulnerabilities in these DLBCLs.
Collapse
Affiliation(s)
- T Sewastianik
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - M Szydlowski
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - E Jablonska
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - E Bialopiotrowicz
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - P Kiliszek
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - P Gorniak
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - A Polak
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - M Prochorec-Sobieszek
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - A Szumera-Cieckiewicz
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - T S Kaminski
- Department of Soft Condensed Matter, Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - S Markowicz
- Department of Immunology, Maria Sklodowska-Curie Memorial Cancer Center-Institute of Oncology, Warsaw, Poland
| | - E Nowak
- Department of Immunology, Maria Sklodowska-Curie Memorial Cancer Center-Institute of Oncology, Warsaw, Poland
| | - M A Grygorowicz
- Department of Immunology, Maria Sklodowska-Curie Memorial Cancer Center-Institute of Oncology, Warsaw, Poland
| | - K Warzocha
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - P Juszczynski
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| |
Collapse
|
35
|
Benhar M, Shytaj IL, Stamler JS, Savarino A. Dual targeting of the thioredoxin and glutathione systems in cancer and HIV. J Clin Invest 2016; 126:1630-9. [PMID: 27135880 PMCID: PMC4855928 DOI: 10.1172/jci85339] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although the use of antioxidants for the treatment of cancer and HIV/AIDS has been proposed for decades, new insights gained from redox research have suggested a very different scenario. These new data show that the major cellular antioxidant systems, the thioredoxin (Trx) and glutathione (GSH) systems, actually promote cancer growth and HIV infection, while suppressing an effective immune response. Mechanistically, these systems control both the redox- and NO-based pathways (nitroso-redox homeostasis), which subserve innate and cellular immune defenses. Dual inhibition of the Trx and GSH systems synergistically kills neoplastic cells in vitro and in mice and decreases resistance to anticancer therapy. Similarly, the population of HIV reservoir cells that constitutes the major barrier to a cure for AIDS is exquisitely redox sensitive and could be selectively targeted by Trx and GSH inhibitors. Trx and GSH inhibition may lead to a reprogramming of the immune response, tilting the balance between the immune system and cancer or HIV in favor of the former, allowing elimination of diseased cells. Thus, therapies based on silencing of the Trx and GSH pathways represent a promising approach for the cure of both cancer and AIDS and warrant further investigation.
Collapse
Affiliation(s)
- Moran Benhar
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| | | | - Jonathan S. Stamler
- Institute for Transformative Molecular Medicine, Department of Medicine, and Harrington Discovery Institute, University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | | |
Collapse
|
36
|
Differences in Redox Regulatory Systems in Human Lung and Liver Tumors Suggest Different Avenues for Therapy. Cancers (Basel) 2015; 7:2262-76. [PMID: 26569310 PMCID: PMC4695889 DOI: 10.3390/cancers7040889] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/14/2015] [Accepted: 10/29/2015] [Indexed: 12/26/2022] Open
Abstract
A common characteristic of many cancer cells is that they suffer from oxidative stress. They, therefore, require effective redox regulatory systems to combat the higher levels of reactive oxygen species that accompany accelerated growth compared to the normal cells of origin. An elevated dependence on these systems in cancers suggests that targeting these systems may provide an avenue for retarding the malignancy process. Herein, we examined the redox regulatory systems in human liver and lung cancers by comparing human lung adenocarcinoma and liver carcinoma to their respective surrounding normal tissues. Significant differences were found in the two major redox systems, the thioredoxin and glutathione systems. Thioredoxin reductase 1 levels were elevated in both malignancies, but thioredoxin was highly upregulated in lung tumor and only slightly upregulated in liver tumor, while peroxiredoxin 1 was highly elevated in lung tumor, but downregulated in liver tumor. There were also major differences within the glutathione system between the malignancies and their normal tissues. The data suggest a greater dependence of liver on either the thioredoxin or glutathione system to drive the malignancy, while lung cancer appeared to depend primarily on the thioredoxin system.
Collapse
|
37
|
You BR, Park WH. Auranofin induces mesothelioma cell death through oxidative stress and GSH depletion. Oncol Rep 2015; 35:546-51. [PMID: 26530353 DOI: 10.3892/or.2015.4382] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/11/2015] [Indexed: 11/06/2022] Open
Abstract
Mesothelioma is an aggressive tumor associated with asbestos exposure. Auranofin as an inhibitor of thioredoxin reductase (TrxR) affects many biological processes such as inflammation and proliferation. In the present study, we investigated the cellular effects of auranofin on patient-derived mesothelioma cells in relation to reactive oxygen species (ROS) and glutathione (GSH) levels. Basal TrxR1 levels have no difference between mesothelial cells and certain mesothelioma cells. In particular, ADA, CON and Hmeso mesothelioma cells showed lower levels of TrxR1 expression. Auranofin inhibited the proliferation of mesothelioma cells in a dose-dependent manner. Among mesothelioma cells were ADA and CON cells sensitive to auranofin. This agent also induced caspase-independent apoptosis and necrosis in ADA cells. In addition, auranofin increased ROS levels including O2(•-) and induced GSH depletion in mesothelioma cells. While N-acetyl cysteine (NAC) prevented cell death and decreased ROS levels in auranofin-treated mesothelioma cells, L-buthionine sulfoximine (BSO) intensified apoptosis and GSH depletion in these cells. In conclusion, auranofin induced mesothelioma cell death through oxidative stress and the death was regulated by the status of GSH content.
Collapse
Affiliation(s)
- Bo Ra You
- Department of Physiology, Medical School, Institute for Medical Sciences, Chonbuk National University, Jeonju 561‑180, Republic of Korea
| | - Woo Hyun Park
- Department of Physiology, Medical School, Institute for Medical Sciences, Chonbuk National University, Jeonju 561‑180, Republic of Korea
| |
Collapse
|
38
|
Lennicke C, Rahn J, Lichtenfels R, Wessjohann LA, Seliger B. Hydrogen peroxide - production, fate and role in redox signaling of tumor cells. Cell Commun Signal 2015; 13:39. [PMID: 26369938 PMCID: PMC4570748 DOI: 10.1186/s12964-015-0118-6] [Citation(s) in RCA: 373] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023] Open
Abstract
Hydrogen peroxide (H2O2) is involved in various signal transduction pathways and cell fate decisions. The mechanism of the so called “redox signaling” includes the H2O2-mediated reversible oxidation of redox sensitive cysteine residues in enzymes and transcription factors thereby altering their activities. Depending on its intracellular concentration and localization, H2O2 exhibits either pro- or anti-apoptotic activities. In comparison to normal cells, cancer cells are characterized by an increased H2O2 production rate and an impaired redox balance thereby affecting the microenvironment as well as the anti-tumoral immune response. This article reviews the current knowledge about the intracellular production of H2O2 along with redox signaling pathways mediating either the growth or apoptosis of tumor cells. In addition it will be discussed how the targeting of H2O2-linked sources and/or signaling components involved in tumor progression and survival might lead to novel therapeutic targets.
Collapse
Affiliation(s)
- Claudia Lennicke
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle/Saale, Germany
| | - Jette Rahn
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle/Saale, Germany
| | - Rudolf Lichtenfels
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle/Saale, Germany
| | - Ludger A Wessjohann
- Leibniz-Institute of Plant Biochemistry, Weinberg 3, 06120, Halle /Saale, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle/Saale, Germany.
| |
Collapse
|
39
|
Kiebala M, Singh MV, Piepenbrink MS, Qiu X, Kobie JJ, Maggirwar SB. Platelet Activation in Human Immunodeficiency Virus Type-1 Patients Is Not Altered with Cocaine Abuse. PLoS One 2015; 10:e0130061. [PMID: 26076359 PMCID: PMC4467977 DOI: 10.1371/journal.pone.0130061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/15/2015] [Indexed: 12/30/2022] Open
Abstract
Recent work has indicated that platelets, which are anucleate blood cells, significantly contribute to inflammatory disorders. Importantly, platelets also likely contribute to various inflammatory secondary disorders that are increasingly associated with Human Immunodeficiency Virus Type-1 (HIV) infection including neurological impairments and cardiovascular complications. Indeed, HIV infection is often associated with increased levels of platelet activators. Additionally, cocaine, a drug commonly abused by HIV-infected individuals, leads to increased platelet activation in humans. Considering that orchestrated signaling mechanisms are essential for platelet activation, and that nuclear factor-kappa B (NF-κB) inhibitors can alter platelet function, the role of NF-κB signaling in platelet activation during HIV infection warrants further investigation. Here we tested the hypothesis that inhibitory kappa B kinase complex (IKK) activation would be central for platelet activation induced by HIV and cocaine. Whole blood from HIV-positive and HIV-negative individuals, with or without cocaine abuse was used to assess platelet activation via flow cytometry whereas IKK activation was analyzed by performing immunoblotting and in vitro kinase assays. We demonstrate that increased platelet activation in HIV patients, as measured by CD62P expression, is not altered with reported cocaine use. Furthermore, cocaine and HIV do not activate platelets in whole blood when treated ex vivo. Finally, HIV-induced platelet activation does not involve the NF-κB signaling intermediate, IKKβ. Platelet activation in HIV patients is not altered with cocaine abuse. These results support the notion that non-IKK targeting approaches will be better suited for the treatment of HIV-associated inflammatory disorders.
Collapse
Affiliation(s)
- Michelle Kiebala
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- * E-mail:
| | - Meera V. Singh
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Michael S. Piepenbrink
- Division of Infectious Diseases, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - James J. Kobie
- Division of Infectious Diseases, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Sanjay B. Maggirwar
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| |
Collapse
|