1
|
Wen X, Ma H. Cytoplasmic Vacuolization: A Fascinating Morphological Alteration From Cellular Stress to Cell Death. Cancer Sci 2025; 116:1181-1192. [PMID: 40017124 PMCID: PMC12044657 DOI: 10.1111/cas.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/19/2025] [Accepted: 01/28/2025] [Indexed: 03/01/2025] Open
Abstract
Cytoplasmic vacuolization is a cellular morphological alteration characterized by the presence of substantial vacuole-like structures originating from various cellular organelles. This phenomenon is often observed in various anticancer treatments, including chemotherapeutic drugs, and photodynamic therapy (PDT), and is frequently linked with cell death. Nevertheless, the precise mechanisms underlying cytoplasmic vacuolization and ensuing cell death remain ambiguous. Cytoplasmic vacuolization associated cell death (CVACD) is a complex process characterized by cellular stress, encompassing ER stress, heightened membrane permeability, ion imbalance, and mitochondrial dysfunction. The MAPK signaling pathway is closely associated with the activation of CVACD. This review provides a thorough examination of contemporary studies on cytoplasmic vacuolization in mammalian cells, elucidating its etiology, origins, and molecular pathways. Additionally, it highlights the potential of CVACD as an innovative therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Xiaoxu Wen
- School of StomatologyHenan UniversityKaifengChina
| | - Hongru Ma
- College of Chemical and Biological EngineeringZhejiang UniversityHangzhouChina
| |
Collapse
|
2
|
Chang LC, Chiang SK, Chen SE, Hung MC. Exploring paraptosis as a therapeutic approach in cancer treatment. J Biomed Sci 2024; 31:101. [PMID: 39497143 PMCID: PMC11533606 DOI: 10.1186/s12929-024-01089-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
A variety of cell death pathways play critical roles in the onset and progression of multiple diseases. Paraptosis, a unique form of programmed cell death, has gained significant attention in recent years. Unlike apoptosis and necrosis, paraptosis is characterized by cytoplasmic vacuolization, swelling of the endoplasmic reticulum and mitochondria, and the absence of caspase activation. Numerous natural products, synthetic compounds, and newly launched nanomedicines have been demonstrated to prime cell death through the paraptotic program and may offer novel therapeutic strategies for cancer treatment. This review summarizes recent findings, delineates the intricate network of signaling pathways underlying paraptosis, and discusses the potential therapeutic implications of targeting paraptosis in cancer treatment. The aim of this review is to expand our understanding of this unique cell death process and explore the potential therapeutic implications of targeting paraptosis in cancer treatment.
Collapse
Affiliation(s)
- Ling-Chu Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 406040, Taiwan.
- Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan.
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40402, Taiwan.
| | - Shih-Kai Chiang
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Shuen-Ei Chen
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan
- Innovation and Development Center of Sustainable Agriculture (IDCSA), National Chung Hsing University, Taichung, 40227, Taiwan
- i-Center for Advanced Science and Technology (iCAST), National Chung Hsing University, Taichung, 40227, Taiwan
| | - Mien-Chie Hung
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 406040, Taiwan.
- Research Center for Cancer Biology, China Medical University, Taichung, 406040, Taiwan.
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40402, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan.
| |
Collapse
|
3
|
Afzal A, Afzal Z, Bizink S, Davis A, Makahleh S, Mohamed Y, Coniglio SJ. Phagocytosis Checkpoints in Glioblastoma: CD47 and Beyond. Curr Issues Mol Biol 2024; 46:7795-7811. [PMID: 39194679 DOI: 10.3390/cimb46080462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/06/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the deadliest human cancers with very limited treatment options available. The malignant behavior of GBM is manifested in a tumor which is highly invasive, resistant to standard cytotoxic chemotherapy, and strongly immunosuppressive. Immune checkpoint inhibitors have recently been introduced in the clinic and have yielded promising results in certain cancers. GBM, however, is largely refractory to these treatments. The immune checkpoint CD47 has recently gained attention as a potential target for intervention as it conveys a "don't eat me" signal to tumor-associated macrophages (TAMs) via the inhibitory SIRP alpha protein. In preclinical models, the administration of anti-CD47 monoclonal antibodies has shown impressive results with GBM and other tumor models. Several well-characterized oncogenic pathways have recently been shown to regulate CD47 expression in GBM cells and glioma stem cells (GSCs) including Epidermal Growth Factor Receptor (EGFR) beta catenin. Other macrophage pathways involved in regulating phagocytosis including TREM2 and glycan binding proteins are discussed as well. Finally, chimeric antigen receptor macrophages (CAR-Ms) could be leveraged for greatly enhancing the phagocytosis of GBM and repolarization of the microenvironment in general. Here, we comprehensively review the mechanisms that regulate the macrophage phagocytosis of GBM cells.
Collapse
Affiliation(s)
- Amber Afzal
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Zobia Afzal
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Sophia Bizink
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Amanda Davis
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Sara Makahleh
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Yara Mohamed
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
| | - Salvatore J Coniglio
- School of Integrative Science and Technology, Kean University, Union, NJ 07083, USA
- Department of Biological Sciences, Kean University, Union, NJ 07083, USA
| |
Collapse
|
4
|
Xu CC, Lin YF, Huang MY, Zhang XL, Wang P, Huang MQ, Lu JJ. Paraptosis: a non-classical paradigm of cell death for cancer therapy. Acta Pharmacol Sin 2024; 45:223-237. [PMID: 37715003 PMCID: PMC10789732 DOI: 10.1038/s41401-023-01159-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/28/2023] [Indexed: 09/17/2023]
Abstract
Due to the sustained proliferative potential of cancer cells, inducing cell death is a potential strategy for cancer therapy. Paraptosis is a mode of cell death characterized by endoplasmic reticulum (ER) and/or mitochondrial swelling and cytoplasmic vacuolization, which is less investigated. Considerable evidence shows that paraptosis can be triggered by various chemical compounds, particularly in cancer cells, thus highlighting the potential application of this non-classical mode of cell death in cancer therapy. Despite these findings, there remain significant gaps in our understanding of the role of paraptosis in cancer. In this review, we summarize the current knowledge on chemical compound-induced paraptosis. The ER and mitochondria are the two major responding organelles in chemical compound-induced paraptosis, which can be triggered by the reduction of protein degradation, disruption of sulfhydryl homeostasis, overload of mitochondrial Ca2+, and increased generation of reactive oxygen species. We also discuss the stumbling blocks to the development of this field and the direction for further research. The rational use of paraptosis might help us develop a new paradigm for cancer therapy.
Collapse
Affiliation(s)
- Chun-Cao Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yi-Fan Lin
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Pei Wang
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Ming-Qing Huang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, University of Macau, Macao, China.
- Zhuhai UM Science & Technology Research Institute, Zhuhai, 519000, China.
| |
Collapse
|
5
|
Hanson S, Dharan A, P. V. J, Pal S, Nair BG, Kar R, Mishra N. Paraptosis: a unique cell death mode for targeting cancer. Front Pharmacol 2023; 14:1159409. [PMID: 37397502 PMCID: PMC10308048 DOI: 10.3389/fphar.2023.1159409] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/15/2023] [Indexed: 07/04/2023] Open
Abstract
Programmed cell death (PCD) is the universal process that maintains cellular homeostasis and regulates all living systems' development, health and disease. Out of all, apoptosis is one of the major PCDs that was found to play a crucial role in many disease conditions, including cancer. The cancer cells acquire the ability to escape apoptotic cell death, thereby increasing their resistance towards current therapies. This issue has led to the need to search for alternate forms of programmed cell death mechanisms. Paraptosis is an alternative cell death pathway characterized by vacuolation and damage to the endoplasmic reticulum and mitochondria. Many natural compounds and metallic complexes have been reported to induce paraptosis in cancer cell lines. Since the morphological and biochemical features of paraptosis are much different from apoptosis and other alternate PCDs, it is crucial to understand the different modulators governing it. In this review, we have highlighted the factors that trigger paraptosis and the role of specific modulators in mediating this alternative cell death pathway. Recent findings include the role of paraptosis in inducing anti-tumour T-cell immunity and other immunogenic responses against cancer. A significant role played by paraptosis in cancer has also scaled its importance in knowing its mechanism. The study of paraptosis in xenograft mice, zebrafish model, 3D cultures, and novel paraptosis-based prognostic model for low-grade glioma patients have led to the broad aspect and its potential involvement in the field of cancer therapy. The co-occurrence of different modes of cell death with photodynamic therapy and other combinatorial treatments in the tumour microenvironment are also summarized here. Finally, the growth, challenges, and future perspectives of paraptosis research in cancer are discussed in this review. Understanding this unique PCD pathway would help to develop potential therapy and combat chemo-resistance in various cancer.
Collapse
Affiliation(s)
- Sweata Hanson
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Aiswarya Dharan
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Jinsha P. V.
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Sanjay Pal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Bipin G. Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Rekha Kar
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, United States
| | - Nandita Mishra
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| |
Collapse
|
6
|
Paraptosis and tumor immunity. Int Immunopharmacol 2023; 114:109491. [PMID: 36462335 DOI: 10.1016/j.intimp.2022.109491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022]
Abstract
Paraptosis is the programmed cell death pathway that leads to cellular necrosis. Manystudies have shown that prolonged paraptosis activation improves tumorimmunogenicity; this treatment reproduces the vaccinating effects of mM-CSFtransduced cells. In this short communication, we want to highlight the paraptosisprocess as a valuable strategy for clinical immunotherapy against cancer.
Collapse
|
7
|
Qiu Y, Chen T, Hu R, Zhu R, Li C, Ruan Y, Xie X, Li Y. Next frontier in tumor immunotherapy: macrophage-mediated immune evasion. Biomark Res 2021; 9:72. [PMID: 34625124 PMCID: PMC8501632 DOI: 10.1186/s40364-021-00327-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor-associated macrophages (TAMs), at the core of immunosuppressive cells and cytokines networks, play a crucial role in tumor immune evasion. Increasing evidences suggest that potential mechanisms of macrophage-mediated tumor immune escape imply interpretation and breakthrough to bottleneck of current tumor immunotherapy. Therefore, it is pivotal to understand the interactions between macrophages and other immune cells and factors for enhancing existing anti-cancer treatments. In this review, we focus on the specific signaling pathways through which TAMs involve in tumor antigen recognition disorders, recruitment and function of immunosuppressive cells, secretion of immunosuppressive cytokines, crosstalk with immune checkpoints and formation of immune privileged sites. Furthermore, we summarize correlative pre-clinical and clinical studies to provide new ideas for immunotherapy. From our perspective, macrophage-targeted therapy is expected to be the next frontier of cancer immunotherapy.
Collapse
Affiliation(s)
- Yingqi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510280, P. R. China
| | - Tong Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510280, P. R. China.,The Second School of Clinical Medicine, Southern Medical University, No. 1838 GuangzhongDadaoBei, Guangzhou, Guangdong, 510515, P. R. China
| | - Rong Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510280, P. R. China
| | - Ruiyi Zhu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510280, P. R. China.,The Second School of Clinical Medicine, Southern Medical University, No. 1838 GuangzhongDadaoBei, Guangzhou, Guangdong, 510515, P. R. China
| | - Chujun Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510280, P. R. China.,The Second School of Clinical Medicine, Southern Medical University, No. 1838 GuangzhongDadaoBei, Guangzhou, Guangdong, 510515, P. R. China
| | - Yingchen Ruan
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510280, P. R. China.,The Second School of Clinical Medicine, Southern Medical University, No. 1838 GuangzhongDadaoBei, Guangzhou, Guangdong, 510515, P. R. China
| | - Xiaoling Xie
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, No. 253 GongyeDadaoZhong, Guangzhou, Guangdong, 510280, P. R. China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, P. R. China.
| |
Collapse
|
8
|
Zhang G, Ren G, Zhao X, Wang H. RNA Interference-Mediated Aurora Kinase A Gene Silencing Inhibits Human Glioma Cells Proliferation and Tumor Growth in Mice. J Cancer 2021; 12:3024-3032. [PMID: 33854602 PMCID: PMC8040878 DOI: 10.7150/jca.55791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
Objective: This study aims to explore the roles of Aurora Kinase A (Aurora A) in human glioma progression and relevant molecular mechanisms involved. Methods: RNA interference (RNAi) technology was performed to silence the Aurora A gene in human glioma cell line U251 and U87. Western blot and real-time PCR were used to determine the protein and mRNA expression levels of Aurora A. Flow cytometry was performed to analyze the cell cycle distribution and MTT was used to examine the cell viability. Annexin V/FITC double staining and Hoechst 33258 staining were carried out to examine cell apoptosis. Xenograft tumor model was established to examine the effect of Aurora A siRNA on tumor growth in vivo. Results: RNAi-mediated Aurora A gene silencing with specific short interfering RNA (siRNA) significantly decreased Aurora A protein and mRNA expression levels in human glioma cell line U251 and U87. Aurora A knockdown in glioma cells with siRNA strongly inhibited cell proliferation, along with the accumulation of cells in the G1, G2/M phase and decrease in S phase. Furthermore, the enhancement of cell apoptosis in vitro and the suppression of xenograft tumor growth in vivo were also observed after Aurora A silencing in U251 cell. In addition, Aurora A knockdown resulted in decreased expression of anti-apoptotic protein Bcl-2 and cell cycle protein Cyclin D1, while increased expression of pro-apoptotic factor caspase-3. Conclusion: Aurora A can be used as a candidate targeting gene and inhibition of Aurora A is a potentially promising therapy for glioblastoma.
Collapse
Affiliation(s)
- Ge Zhang
- Department of Neurology, Luoyang Dong Fang Hospital, The Third Affiliated Hospital of Henan University of Science & Technology, Luoyang, Henan 471003, PR China
| | - Guanghui Ren
- Department of Neurology, Luoyang Dong Fang Hospital, The Third Affiliated Hospital of Henan University of Science & Technology, Luoyang, Henan 471003, PR China
| | - Xin Zhao
- Department of Neurology, Luoyang Dong Fang Hospital, The Third Affiliated Hospital of Henan University of Science & Technology, Luoyang, Henan 471003, PR China
| | - Haibo Wang
- Institute of Cardiovascular Disease, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| |
Collapse
|
9
|
Liu Q, Zhang Y, Zhang J, Tao K, Hambly BD, Bao S. Inverse correlation between Interleukin-34 and gastric cancer, a potential biomarker for prognosis. Cell Biosci 2020; 10:94. [PMID: 32765828 PMCID: PMC7399616 DOI: 10.1186/s13578-020-00454-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/25/2020] [Indexed: 12/17/2022] Open
Abstract
Background Gastric cancer (GC) is a malignancy with high morbidity/mortality, partly due to a lack of reliable biomarkers for early diagnosis. It is important to develop reliable biomarker(s) with specificity, sensitivity and convenience for early diagnosis. The role of tumour-associated macrophages (TAMs) and survival of GC patients are controversial. Macrophage colony stimulating factor (MCSF) regulates monocytes/macrophages. Elevated MCSF is correlated with invasion, metastasis and poor survival of tumour patients. IL-34, a ligand of the M-CSF receptor, acts as a “twin” to M-CSF, demonstrating overlapping and complimentary actions. IL-34 involvement in tumours is controversial, possibly due to the levels of M-CSF receptors. While the IL-34/M-CSF/M-CSFR axis is very important for regulating macrophage differentiation, the specific interplay between these cytokines, macrophages and tumour development is unclear. Methods A multi-factorial evaluation could provide more objective utility, particularly for either prediction and/or prognosis of gastric cancer. Precision medicine requires molecular diagnosis to determine the specifically mutant function of tumours, and is becoming popular in the treatment of malignancy. Therefore, elucidating specific molecular signalling pathways in specific cancers facilitates the success of a precision medicine approach. Gastric cancer tissue arrays were generated from stomach samples with TNM stage, invasion depth and the demography of these patients (n = 185). Using immunohistochemistry/histopathology, M-CSF, IL-34 and macrophages were determined. Results We found that IL-34 may serve as a predictive biomarker, but not as an independent, prognostic factor in GC; M-CSF inversely correlated with survival of GC in TNM III–IV subtypes. Increased CD68+ TAMs were a good prognostic factor in some cases and could be used as an independent prognostic factor in male T3 stage GC. Conclusion Our data support the potency of IL-34, M-CSF, TAMs and the combination of IL-34/TAMs as novel biological markers for GC, and may provide new insight for both diagnosis and cellular therapy of GC.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221000 China.,Discipline of Pathology, Bosch Institute and School of Medical Sciences, Charles Perkins Center D17, Sydney Medical School, The University of Sydney, Sydney, NSW 2006 Australia
| | - Ying Zhang
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221000 China
| | - Jiwei Zhang
- Department of Surgery, Songjiang Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201600 China
| | - Kun Tao
- Tongren Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200336 China
| | - Brett D Hambly
- Discipline of Pathology, Bosch Institute and School of Medical Sciences, Charles Perkins Center D17, Sydney Medical School, The University of Sydney, Sydney, NSW 2006 Australia.,Tongren Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200336 China
| | - Shisan Bao
- Discipline of Pathology, Bosch Institute and School of Medical Sciences, Charles Perkins Center D17, Sydney Medical School, The University of Sydney, Sydney, NSW 2006 Australia.,Tongren Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200336 China
| |
Collapse
|
10
|
The emerging role of paraptosis in tumor cell biology: Perspectives for cancer prevention and therapy with natural compounds. Biochim Biophys Acta Rev Cancer 2020; 1873:188338. [PMID: 31904399 DOI: 10.1016/j.bbcan.2020.188338] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/06/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022]
Abstract
Standard anti-cancer therapies promote tumor growth suppression mainly via induction of apoptosis. However, in most cases cancer cells acquire the ability to escape apoptotic cell death, thus becoming resistant to current treatments. In this setting, the interest in alternative cell death modes has recently increased. Paraptosis is a new form of programmed cell death displaying endoplasmic reticulum (ER) and/or mitochondria dilation, generally due to proteostasis disruption or redox and ion homeostasis alteration. Recent studies have highlighted that several natural compounds can trigger paraptosis in different tumor cell lines. Here, we review the molecular mechanisms underlying paraptotic cell death, as well as the natural products inducing this kind of cell death program. A better understanding of paraptosis should facilitate the development of new therapeutic strategies for cancer prevention and treatment.
Collapse
|
11
|
Ciavatta ML, Lefranc F, Carbone M, Mollo E, Gavagnin M, Betancourt T, Dasari R, Kornienko A, Kiss R. Marine Mollusk-Derived Agents with Antiproliferative Activity as Promising Anticancer Agents to Overcome Chemotherapy Resistance. Med Res Rev 2017; 37:702-801. [PMID: 27925266 PMCID: PMC5484305 DOI: 10.1002/med.21423] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/20/2016] [Accepted: 09/23/2016] [Indexed: 12/18/2022]
Abstract
The chemical investigation of marine mollusks has led to the isolation of a wide variety of bioactive metabolites, which evolved in marine organisms as favorable adaptations to survive in different environments. Most of them are derived from food sources, but they can be also biosynthesized de novo by the mollusks themselves, or produced by symbionts. Consequently, the isolated compounds cannot be strictly considered as "chemotaxonomic markers" for the different molluscan species. However, the chemical investigation of this phylum has provided many compounds of interest as potential anticancer drugs that assume particular importance in the light of the growing literature on cancer biology and chemotherapy. The current review highlights the diversity of chemical structures, mechanisms of action, and, most importantly, the potential of mollusk-derived metabolites as anticancer agents, including those biosynthesized by mollusks and those of dietary origin. After the discussion of dolastatins and kahalalides, compounds previously studied in clinical trials, the review covers potentially promising anticancer agents, which are grouped based on their structural type and include terpenes, steroids, peptides, polyketides and nitrogen-containing compounds. The "promise" of a mollusk-derived natural product as an anticancer agent is evaluated on the basis of its ability to target biological characteristics of cancer cells responsible for poor treatment outcomes. These characteristics include high antiproliferative potency against cancer cells in vitro, preferential inhibition of the proliferation of cancer cells over normal ones, mechanism of action via nonapoptotic signaling pathways, circumvention of multidrug resistance phenotype, and high activity in vivo, among others. The review also includes sections on the targeted delivery of mollusk-derived anticancer agents and solutions to their procurement in quantity.
Collapse
Affiliation(s)
- Maria Letizia Ciavatta
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital ErasmeUniversité Libre de Bruxelles (ULB)1070BrusselsBelgium
| | - Marianna Carbone
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Ernesto Mollo
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Margherita Gavagnin
- Consiglio Nazionale delle Ricerche (CNR)Istituto di Chimica Biomolecolare (ICB)Via Campi Flegrei 3480078PozzuoliItaly
| | - Tania Betancourt
- Department of Chemistry and BiochemistryTexas State UniversitySan MarcosTX78666
| | - Ramesh Dasari
- Department of Chemistry and BiochemistryTexas State UniversitySan MarcosTX78666
| | - Alexander Kornienko
- Department of Chemistry and BiochemistryTexas State UniversitySan MarcosTX78666
| | - Robert Kiss
- Laboratoire de Cancérologie et de Toxicologie ExpérimentaleFaculté de Pharmacie, Université Libre de Bruxelles (ULB)1050BrusselsBelgium
| |
Collapse
|
12
|
Lee D, Kim IY, Saha S, Choi KS. Paraptosis in the anti-cancer arsenal of natural products. Pharmacol Ther 2016; 162:120-33. [DOI: 10.1016/j.pharmthera.2016.01.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
13
|
Chockalingam S, Ghosh SS. Macrophage colony-stimulating factor and cancer: a review. Tumour Biol 2014; 35:10635-44. [PMID: 25238879 DOI: 10.1007/s13277-014-2627-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/10/2014] [Indexed: 11/26/2022] Open
Abstract
Tumor growth is influenced by a wide variety of external and internal factors. One of the most important mediators of tumor development is our immune system. The nonstop surveillance of the immune system was originally expected to clear the transformed cells from the body and guard against the development of tumor. But contradictory evidences are reported to show the involvement of immune system in supporting the growth and spread of tumor. Tumor infiltrating immune cells, in addition to harboring immunosuppressive activities, also promote angiogenesis and metastasis of tumor. Many growth factors and cytokines are involved in shaping this complex immune microenvironment of the tumor. Macrophage colony-stimulating factor (MCSF) is one such growth factor which is overexpressed in many tumors. In this review, we summarize the basic biology of MCSF, its role in cancer and discuss the involvement of tumor-associated macrophages (TAMs) in tumor development.
Collapse
Affiliation(s)
- S Chockalingam
- Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India,
| | | |
Collapse
|
14
|
WANG YAO, XU KUN, ZHANG HONGBING, ZHAO JUNHONG, ZHU XIUPING, WANG YANGZHENG, WU RENYI. Retinal ganglion cell death is triggered by paraptosis via reactive oxygen species production: A brief literature review presenting a novel hypothesis in glaucoma pathology. Mol Med Rep 2014; 10:1179-83. [DOI: 10.3892/mmr.2014.2346] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 05/13/2014] [Indexed: 11/06/2022] Open
|
15
|
Chockalingam S, Ghosh SS. Amelioration of cancer stem cells in macrophage colony stimulating factor-expressing U87MG-human glioblastoma upon 5-fluorouracil therapy. PLoS One 2013; 8:e83877. [PMID: 24391839 PMCID: PMC3877109 DOI: 10.1371/journal.pone.0083877] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 11/08/2013] [Indexed: 12/31/2022] Open
Abstract
Macrophage colony stimulating factor (MCSF) regulates growth, proliferation and differentiation of haematopoietic cell lineages. Many cancers are known to secrete high level of MCSF, which recruit macrophages into the tumour micro-environment, supporting tumour growth. Herein, we report the cloning of MCSF and subsequent generation of U87MG expressing MCSF stable cell line (U87-MCSF). Cytotoxicity of anti-cancer drug 5-fluorouracil (5-FU) was evaluated on both U87MG and U87-MCSF cells. Interestingly, the proliferation of U87-MCSF cells was less (p<0.001) than that of U87MG cells alone, after treatment with 5-FU. Significant decrease in expression levels of cyclin E and A2 quantified by real time PCR analysis corroborated the reduced proliferation of 5-FU treated U87-MCSF cells. However, JC-1 staining did not reveal any apoptosis upon 5-FU treatment. Notch-1 upregulation induced a possible epithelial-mesenchymal transition in U87-MCSF cells, which accounted for an increase in the proportion of CD24high/CD44less cancer stem cells in U87-MCSF cells after 5-FU treatment. The elevated resistance of U87-MCSF cells towards 5-FU was due to the increase in the expressions (10.2 and 6 fold) of ABCB1 and mdm2, respectively. Furthermore, increase in expressions of ABCG1, mdm2 and CD24 was also observed in U87MG cells after prolonged incubation with 5-FU. Our studies provided mechanistic insights into drug resistance of U87MG cells and also described the pivotal role played by MCSF in augmenting the resistance of U87MG cells to 5-FU.
Collapse
Affiliation(s)
- S. Chockalingam
- Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
- * E-mail:
| |
Collapse
|
16
|
Korsnes MS, Espenes A, Hermansen LC, Loader JI, Miles CO. Cytotoxic responses in BC3H1 myoblast cell lines exposed to 1-desulfoyessotoxin. Toxicol In Vitro 2013; 27:1962-9. [DOI: 10.1016/j.tiv.2013.06.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 05/27/2013] [Accepted: 06/24/2013] [Indexed: 12/19/2022]
|
17
|
Li W, Graeber MB. The molecular profile of microglia under the influence of glioma. Neuro Oncol 2012; 14:958-78. [PMID: 22573310 DOI: 10.1093/neuonc/nos116] [Citation(s) in RCA: 266] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Microglia, which contribute substantially to the tumor mass of glioblastoma, have been shown to play an important role in glioma growth and invasion. While a large number of experimental studies on functional attributes of microglia in glioma provide evidence for their tumor-supporting roles, there also exist hints in support of their anti-tumor properties. Microglial activities during glioma progression seem multifaceted. They have been attributed to the receptors expressed on the microglia surface, to glioma-derived molecules that have an effect on microglia, and to the molecules released by microglia in response to their environment under glioma control, which can have autocrine effects. In this paper, the microglia and glioma literature is reviewed. We provide a synopsis of the molecular profile of microglia under the influence of glioma in order to help establish a rational basis for their potential therapeutic use. The ability of microglia precursors to cross the blood-brain barrier makes them an attractive target for the development of novel cell-based treatments of malignant glioma.
Collapse
Affiliation(s)
- Wei Li
- Brain Tumor Research Laboratories, The Brain and Mind Research Institute, University of Sydney, 94 Mallett St, Camperdown, Sydney, NSW 2050, Australia
| | | |
Collapse
|
18
|
Paraptosis-like cell death induced by yessotoxin. Toxicol In Vitro 2011; 25:1764-70. [DOI: 10.1016/j.tiv.2011.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 08/31/2011] [Accepted: 09/06/2011] [Indexed: 01/07/2023]
|
19
|
Sperandio S, Poksay KS, Schilling B, Crippen D, Gibson BW, Bredesen DE. Identification of new modulators and protein alterations in non-apoptotic programmed cell death. J Cell Biochem 2011; 111:1401-12. [PMID: 20830744 DOI: 10.1002/jcb.22870] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study describes the first proteomic analysis of paraptosis--a non-apoptotic form of programmed cell death. As with apoptosis, the first description of paraptosis was based on morphological criteria. Since there are no known markers for paraptosis, the purpose of this study was to dissect changes in the proteome profile occurring during paraptosis. Using one- and two-dimensional SDS-PAGE, Western analysis, and mass spectrometry, we show that during paraptosis, alterations occur mainly in cytoskeletal proteins, signal transduction proteins, mitochondrial proteins, and some metabolic proteins. We also report the identification of: (1) a paraptosis inhibitor, phosphatidylethanolamine binding protein (PEBP-1), and (2) a candidate mediator of paraptosis, prohibitin. Identification of specific paraptotic changes will ultimately lead to tools to detect this type of programmed cell death in in vivo systems and allow for its further characterization.
Collapse
|
20
|
Immunotherapy of brain cancers: the past, the present, and future directions. Clin Dev Immunol 2011; 2010:296453. [PMID: 21437175 PMCID: PMC3061456 DOI: 10.1155/2010/296453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 12/30/2010] [Indexed: 11/17/2022]
Abstract
Treatment of brain cancers, especially high grade gliomas (WHO stage III and IV) is slowly making progress, but not as fast as medical researchers and the patients would like. Immunotherapy offers the opportunity to allow the patient's own immune system a chance to help eliminate the cancer. Immunotherapy's strength is that it efficiently treats relatively small tumors in experimental animal models. For some patients, immunotherapy has worked for them while not showing long-term toxicity. In this paper, we will trace the history of immunotherapy for brain cancers. We will also highlight some of the possible directions that this field may be taking in the immediate future for improving this therapeutic option.
Collapse
|
21
|
Haque A, Banik NL, Ray SK. Molecular alterations in glioblastoma: potential targets for immunotherapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 98:187-234. [PMID: 21199773 DOI: 10.1016/b978-0-12-385506-0.00005-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glioblastoma is the most common and deadly brain tumor, possibly arising from genetic and epigenetic alterations in normal astroglial cells. Multiple cytogenetic, chromosomal, and genetic alterations have been identified in glioblastoma, with distinct expression of antigens (Ags) and biomarkers that may alter therapeutic potential of this aggressive cancer. Current therapy consists of surgical resection, followed by radiation therapy and chemotherapy. In spite of these treatments, the prognosis for glioblastoma patients is poor. Although recent studies have focused on the development of novel immunotherapeutics against glioblastoma, little is known about glioblastoma-specific immune responses. A better understanding of the molecular interactions among glioblastoma tumors, host immune cells, and the tumor microenvironment may give rise to novel integrated approaches for the simultaneous control of tumor escape pathways and the activation of antitumor immune responses. This review provides a detailed overview concerning genetic alterations in glioblastoma, their effects on Ag and biomarker expression, and the future design of chemoimmunotherapeutics against glioblastoma.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | |
Collapse
|
22
|
Challa S, Chan FKM. Going up in flames: necrotic cell injury and inflammatory diseases. Cell Mol Life Sci 2010; 67:3241-53. [PMID: 20532807 PMCID: PMC3051829 DOI: 10.1007/s00018-010-0413-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 05/09/2010] [Accepted: 05/17/2010] [Indexed: 12/31/2022]
Abstract
Recent evidence indicates that cell death can be induced through multiple mechanisms. Strikingly, the same death signal can often induce apoptotic as well as non-apoptotic cell death. For instance, inhibition of caspases often converts an apoptotic stimulus to one that causes necrosis. Because a dedicated molecular circuitry distinct from that controlling apoptosis is required for necrotic cell injury, terms such as "programmed necrosis" or "necroptosis" have been used to distinguish stimulus-dependent necrosis from those induced by non-specific traumas (e.g., heat shock) or secondary necrosis induced as a consequence of apoptosis. In several experimental models, programmed necrosis/necroptosis has been shown to be a crucial control point for pathogen- or injury-induced inflammation. In this review, we will discuss the molecular mechanisms that regulate programmed necrosis/necroptosis and its biological significance in pathogen infections, drug-induced cell injury, and trauma-induced tissue damage.
Collapse
Affiliation(s)
- Sreerupa Challa
- Department of Pathology, Immunology and Virology Program Diabetes and Endocrinology Center, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Francis Ka-Ming Chan
- Department of Pathology, Immunology and Virology Program Diabetes and Endocrinology Center, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
- Department of Pathology, University of Massachusetts Medical School, Room S2-125, Worcester, MA 01655 USA
| |
Collapse
|
23
|
Wilms' tumor 1 silencing decreases the viability and chemoresistance of glioblastoma cells in vitro: a potential role for IGF-1R de-repression. J Neurooncol 2010; 103:87-102. [PMID: 20820871 DOI: 10.1007/s11060-010-0374-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 08/18/2010] [Indexed: 12/27/2022]
Abstract
Wilms' tumor 1 (WT1) is a transcription factor with a multitude of downstream targets that have wide-ranging effects in non-glioma cell lines. Though its expression in glioblastomas is now well-documented, the role of WT1 in these tumors remains poorly defined. We hypothesized that WT1 functions as an oncogene to enhance glioblastoma viability and chemoresistance. WT1's role was examined by studying the effect of WT1 silencing and overexpression on DNA damage, apoptosis and cell viability. Results indicated that WT1 silencing adversely affected glioblastoma viability, at times, in synergy with 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and cisplatin. To investigate other mechanisms through which WT1 could affect viability, we measured cell cycle distribution, senescence, and autophagy. WT1 silencing had no effect on these processes. Lastly, we examined WT1 regulation of IGF-1R expression. Counterintuitively, upregulation of IGF-1R was evident after WT1 silencing. In conclusion, WT1 functions as a survival factor in glioblastomas, possibly through inhibition of IGF-1R expression.
Collapse
|
24
|
Sun Q, Chen T, Wang X, Wei X. Taxol induces paraptosis independent of both protein synthesis and MAPK pathway. J Cell Physiol 2010; 222:421-32. [PMID: 19918793 DOI: 10.1002/jcp.21982] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our recent studies have shown that high concentration of taxol induced a caspase-independent paraptosis-like cell death and cytoplasmic vacuolization derived predominantly from endoplasmic reticulum (ER) swelling in human lung carcinoma cell lines (ASTC-a-1). In this report, we further explored the relationship between taxol-induced cell death and vacuolization, and the roles of protein synthesis, mitogen-activated protein kinase kinases (MEK), c-jun N-terminal kinase (JNK) and P38 in taxol-induced paraptosis. Enhanced green fluorescent protein (EGFP) was used to probe the cell morphological change, while ER-targeted red fluorescent protein (er-RFP) was used to probe ER spatial distribution. Real-time monitoring of the ER swelling dynamics during the formation of vacuolization inside single living cells co-expressing EGFP and er-RFP further demonstrated that taxol-induced cytoplasmic vacuolization was from the ER restructuring due to fusion and swelling. PI staining showed that taxol-induced vacuolization was not necrosis. These results further demonstrated that the taxol-induced cell death was neither apoptosis nor necrosis, and fitted the criteria of paraptosis characterized by cytoplasmic vacuolization, caspase-independence, lack of apoptotic morphology and insensitivity to broad caspase inhibitor. Our data further indicated that taxol-induced paraptosis required neither protein synthesis nor the participation of MEK, JNK, and P38, which was different from the insulin-like growth factor I receptor (IGFIR)-induced paraptosis. These results suggest that high concentration of taxol activates an alternative paraptotic cell death pathway.
Collapse
Affiliation(s)
- Qingrui Sun
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, South China Normal University, Guangzhou, China
| | | | | | | |
Collapse
|
25
|
Tardito S, Isella C, Medico E, Marchiò L, Bevilacqua E, Hatzoglou M, Bussolati O, Franchi-Gazzola R. The thioxotriazole copper(II) complex A0 induces endoplasmic reticulum stress and paraptotic death in human cancer cells. J Biol Chem 2009; 284:24306-19. [PMID: 19561079 DOI: 10.1074/jbc.m109.026583] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The copper(II) complex A0 induces a type of non-apoptotic cell death also known as paraptosis. Paraptosis involves extensive endoplasmic reticulum vacuolization in the absence of caspase activation. A wide panel of human cancer cell lines was used to demonstrate differences in cytotoxicity by the paraptosis-inducing drug A0 and the metal-based pro-apoptotic drug cisplatin. Gene expression profiling of the human fibrosarcoma HT1080 cells showed that, while cisplatin induced p53 targets, A0 up-regulated genes involved in the unfolded protein response (UPR) and response to heavy metals. The cytotoxic effects of A0 were associated with inhibition of the ubiquitin-proteasome system and accumulation of ubiquitinylated proteins, in a manner dependent on protein synthesis. Cycloheximide inhibited the accumulation of ubiquitinylated proteins and hampered A0-induced cell death process. The occurrence of the UPR during A0-induced death process was shown by the increased abundance of spliced XBP1 mRNA, transient eIF2alpha phosphorylation, and a series of downstream events, including attenuation of global protein synthesis and increased expression of ATF4, CHOP, BIP, and GADD34. Mouse embryonic fibroblasts expressing a mutant eIF2alpha, which could not be phosphorylated, were more resistant to A0 than wild type cells, pointing to a pro-death role of eIF2alpha phosphorylation. A0 may thus represent the prototypical member of a new class of compounds that cause paraptotic cell death via mechanisms involving eIF2alpha phosphorylation and the UPR.
Collapse
Affiliation(s)
- Saverio Tardito
- Unit of General and Clinical Pathology, Department of Experimental Medicine, University of Parma, Parma, Italy
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Hoa N, Myers MP, Douglass TG, Zhang JG, Delgado C, Driggers L, Callahan LL, VanDeusen G, Pham JTH, Bhakta N, Ge L, Jadus MR. Molecular mechanisms of paraptosis induction: implications for a non-genetically modified tumor vaccine. PLoS One 2009; 4:e4631. [PMID: 19247476 PMCID: PMC2645013 DOI: 10.1371/journal.pone.0004631] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2008] [Accepted: 01/02/2009] [Indexed: 01/07/2023] Open
Abstract
Paraptosis is the programmed cell death pathway that leads to cellular necrosis. Previously, rodent and human monocytes/macrophages killed glioma cells bearing the membrane macrophage colony stimulating factor (mM-CSF) through paraptosis, but the molecular mechanism of this killing process was never identified. We have demonstrated that paraptosis of rat T9 glioma cells can be initiated through a large potassium channel (BK)-dependent process initiated by reactive oxygen species. Macrophage mediated cytotoxicity upon the mM-CSF expressing T9-C2 cells was not prevented by the addition of the caspase inhibitor, zVAD-fmk. By a combination of fluorescent confocal and electron microscopy, flow cytometry, electrophysiology, pharmacology, and genetic knock-down approaches, we demonstrated that these ion channels control cellular swelling and vacuolization of rat T9 glioma cells. Cell lysis is preceded by a depletion of intracellular ATP. Six-hour exposure to BK channel activation caused T9 cells to over express heat shock proteins (Hsp 60, 70, 90 and gp96). This same treatment forced HMGB1 translocation from the nuclear region to the periphery. These last molecules are "danger signals" that can stimulate immune responses. Similar inductions of mitochondrial swelling and increased Hsp70 and 90 expressions by BK channel activation were observed with the non-immunogenic F98 glioma cells. Rats injected with T9 cells which were killed by prolonged BK channel activation developed immunity against the T9 cells, while the injection of x-irradiated apoptotic T9 cells failed to produce the vaccinating effect. These results are the first to show that glioma cellular death induced by prolonged BK channel activation improves tumor immunogenicity; this treatment reproduces the vaccinating effects of mM-CSF transduced cells. Elucidation of strategies as described in this study may prove quite valuable in the development of clinical immunotherapy against cancer.
Collapse
Affiliation(s)
- Neil Hoa
- Diagnostic and Molecular Medicine Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
| | - Michael P. Myers
- Chemistry and Biochemistry Department, California State University Long Beach, Long Beach, California, United States of America
| | - Thomas G. Douglass
- Biology Department, California State University Long Beach, Long Beach, California, United States of America
| | - Jian Gang Zhang
- Diagnostic and Molecular Medicine Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, California, United States of America
| | - Christina Delgado
- Diagnostic and Molecular Medicine Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
| | - Lara Driggers
- Diagnostic and Molecular Medicine Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
| | - Linda L. Callahan
- Nursing Department, California State University Long Beach, Long Beach, California, United States of America
| | - Gerald VanDeusen
- Chemistry and Biochemistry Department, California State University Long Beach, Long Beach, California, United States of America
| | - Jimmy T. H. Pham
- Chemistry and Biochemistry Department, California State University Long Beach, Long Beach, California, United States of America
| | - Nirav Bhakta
- Chemistry and Biochemistry Department, California State University Long Beach, Long Beach, California, United States of America
| | - Lisheng Ge
- Diagnostic and Molecular Medicine Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
| | - Martin R. Jadus
- Diagnostic and Molecular Medicine Healthcare Group, Veterans Affairs Medical Center, Long Beach, California, United States of America
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, California, United States of America
- Neuro-Oncology Program, Chao Comprehensive Cancer Center, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
27
|
Wang L, Zheng GG, Ma CH, Lin YM, Zhang HY, Ma YY, Chong JH, Wu KF. A special linker between macrophage and hematopoietic malignant cells: membrane form of macrophage colony-stimulating factor. Cancer Res 2008; 68:5639-47. [PMID: 18632616 DOI: 10.1158/0008-5472.can-07-5804] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The membrane form of macrophage colony-stimulating factor (mM-CSF) is an alternative splicing variant of this cytokine. Although its high expression was detected in hematopoietic malignancies, its physiologic and pathologic roles in hematopoietic system have not been established. In this report, stable transfectant clones expressing mM-CSF (Namalwa-M and Ramos-M) were obtained, which showed reduced proliferation potential in vitro. Moreover, the in vivo study showed that Namalwa-M and Ramos-M exhibited enhanced oncogenicity in tumor size in nude mice model, which could be inhibited by M-CSF monoclonal antibody. A remarkable increase in infiltrating macrophage and the vessel densities was found in tumor tissues formed by lymphoma cell lines that stably expressed mM-CSF, which suggested the involvement of macrophages in this process. The in vitro results using coculture system showed that macrophages could promote Namalwa-M and Ramos-M proliferation and activate extracellular signal-regulated kinase/mitogen-activated protein kinase signal pathway. In addition, the expression of murine origin vascular endothelial growth factor, basic fibroblast growth factor, and hepatocyte growth factor was elevated in Namalwa-M formed tumor tissues. These results suggested that mM-CSF should be a positive regulator in the development of hematopoietic malignancies by abnormally activating infiltrating macrophages, which in turn promote the malignant development. Thus, mM-CSF may be a critical linker between macrophages and malignant cells in the development of hematopoietic malignancies.
Collapse
Affiliation(s)
- Lin Wang
- State Key Laboratory for Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Douglass TG, Driggers L, Zhang JG, Hoa N, Delgado C, Williams CC, Dan Q, Sanchez R, Jeffes EWB, Wepsic HT, Myers MP, Koths K, Jadus MR. Macrophage colony stimulating factor: not just for macrophages anymore! A gateway into complex biologies. Int Immunopharmacol 2008; 8:1354-76. [PMID: 18687298 DOI: 10.1016/j.intimp.2008.04.016] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2008] [Accepted: 04/21/2008] [Indexed: 12/21/2022]
Abstract
Macrophage colony stimulating factor (M-CSF, also called colony stimulating factor-1) has traditionally been viewed as a growth/differentiation factor for monocytes, macrophages, and some female-specific tumors. As a result of alternative mRNA splicing and post-translational processing, several forms of M-CSF protein are produced: a secreted glycoprotein, a longer secreted form containing proteoglycan, and a short membrane-bound isoform. These different forms of M-CSF all initiate cell signaling in cells bearing the M-CSF receptor, called c-fms. Here we review the biology of M-CSF, which has important roles in bone physiology, the intestinal tract, cancer metastases to the bone, macrophage-mediated tumor cell killing and tumor immunity. Although this review concentrates mostly on the membrane form of human M-CSF (mM-CSF), the biology of the soluble forms and the M-CSF receptor will also be discussed for comparative purposes. The mechanisms of the biological effects of the membrane-bound M-CSF reveal that this cytokine is unexpectedly involved in many complex molecular events. Recent experiments suggest that a tumor vaccine based on membrane-bound M-CSF-transduced tumor cells, combined with anti-angiogenic therapy, should be evaluated further for use in clinical trials.
Collapse
Affiliation(s)
- Thomas G Douglass
- Biology Department, California State University Long Beach, 1250 Bellflower Blvd, Long Beach CA 90840, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Schiltz PM, Lee GJ, Zhang JG, Hoa N, Wepsic HT, Dillman RO, Jadus MR. Human Allogeneic and Murine Xenogeneic Dendritic Cells Are Cytotoxic to Human Tumor Cells via Two Distinct Pathways. Cancer Biother Radiopharm 2007; 22:672-83. [DOI: 10.1089/cbr.2007.356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Patric M. Schiltz
- Hoag Hospital Memorial Presbyterian Hospital Comprehensive Cancer Center, Newport Beach, CA
| | - Gregory J. Lee
- Hoag Hospital Memorial Presbyterian Hospital Comprehensive Cancer Center, Newport Beach, CA
| | - Jian Gang Zhang
- Diagnostic and Molecular Medicine Health Care Group, Veterans Affairs Medical Center, Long Beach, CA
| | - Neil Hoa
- Diagnostic and Molecular Medicine Health Care Group, Veterans Affairs Medical Center, Long Beach, CA
| | - H. Terry Wepsic
- Diagnostic and Molecular Medicine Health Care Group, Veterans Affairs Medical Center, Long Beach, CA
| | - Robert O. Dillman
- Hoag Hospital Memorial Presbyterian Hospital Comprehensive Cancer Center, Newport Beach, CA
| | - Martin R. Jadus
- Diagnostic and Molecular Medicine Health Care Group, Veterans Affairs Medical Center, Long Beach, CA
| |
Collapse
|
30
|
Ehtesham M, Black KL, Yu JS. Recent progress in immunotherapy for malignant glioma: treatment strategies and results from clinical trials. Cancer Control 2007; 11:192-207. [PMID: 15153843 DOI: 10.1177/107327480401100307] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Despite advances in surgical and adjuvant radiation therapy and chemotherapy strategies, malignant gliomas continue to be associated with poor prognoses. METHODS We review immune-mediated treatment approaches for malignant glioma and the relevance of recent clinical trials and their outcomes. We specifically address the increasing evidence implicating the role of cytotoxic T cells in ensuring adequate immune-mediated clearance of neoplastic cells and the need for the optimization of therapies that can elicit and support such antitumor T-cell activity. RESULTS The poor outcome of this disease has spurred the search for novel experimental therapies that can address and overcome the root biological phenomena associated with the lethality of this disease. The use of immunotherapy to bolster the otherwise impaired antitumor immune responses in glioma patients has received increasing attention. CONCLUSIONS An effective treatment paradigm for malignant gliomas may eventually require a multifaceted approach combining two or more different immunotherapeutic strategies. Such scenarios may involve the use of local cytokine gene therapy to enhance glioma-cell immunogenicity in conjunction with dendritic cell-based active vaccination to stimulate systemic tumoricidal T-cell immunity. Given the heterogeneity of this disease process and the potential risk of immunoediting out a selected, treatment-refractory tumor cell population, the concurrent use of multiple modalities that target disparate tumor characteristics may be of greatest therapeutic relevance.
Collapse
Affiliation(s)
- Moneeb Ehtesham
- Maxine Dunitz Neurosurgical, Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
31
|
Delgado C, Hoa N, Callahan LL, Schiltz PM, Jahroudi RA, Zhang JG, Wepsic HT, Jadus MR. Generation of human innate immune responses towards membrane macrophage colony stimulating factor (mM-CSF) expressing U251 glioma cells within immunodeficient (NIH-nu/beige/xid) mice. Cytokine 2007; 38:165-76. [PMID: 17689258 DOI: 10.1016/j.cyto.2007.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Revised: 06/05/2007] [Accepted: 06/07/2007] [Indexed: 01/12/2023]
Abstract
The response of human peripheral blood mononuclear cells (PBMC) to cloned human HLA-A2+ U251 glioma cells (U251-2F11/TK) expressing membrane macrophage colony stimulating factor (mM-CSF) was investigated in vitro and in vivo. Enriched human monocytes derived from cancer patients produced a respiratory burst following 20min of interaction with mM-CSF expressing U251 glioma cells. This respiratory burst response was not observed in the enriched human monocytes following similar exposure to the viral vector control U251 (U251-VV) cells. Reactive oxygen species such as H(2)O(2) and HOCl produced death of the U251 cells. The U251-2F11/TK cells failed to grow in severely compromised combined immunodeficient (NIH-bg-nu-xidBR) mice that were depleted of murine monocyte/macrophages then reconstituted with human HLA-A2+ PBMC. Reactive oxygen species (ROS) were produced by PBMC, both in vitro and in vivo in response tomM-CSF expressing U251 cells. U251-2F11/TK cells failed to form subcutaneous tumors in macrophage depleted mice reconstituted with human PBMC; whereas, progressive growth of such tumors was observed with the U251-VV cells. U251-2F11/TK tumors formed if the initial inoculums of PBMC were depleted of monocytes. From this work it can be concluded that mM-CSF transduced U251-2F11/TK glioma cells can safely stimulate human innate immune responses in vivo.
Collapse
Affiliation(s)
- Christina Delgado
- Department of Diagnostic and Molecular Medicine, Box 113, Veterans Affairs Medical Center, 5901 E. 7th Street, Long Beach, CA 90822, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Hoa NT, Zhang JG, Delgado CL, Myers MP, Callahan LL, Vandeusen G, Schiltz PM, Wepsic HT, Jadus MR. Human monocytes kill M-CSF-expressing glioma cells by BK channel activation. J Transl Med 2007; 87:115-29. [PMID: 17318194 DOI: 10.1038/labinvest.3700506] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In this study, human monocytes/macrophages were observed to kill human U251 glioma cells expressing membrane macrophage colony-stimulating factor (mM-CSF) via a swelling and vacuolization process called paraptosis. Human monocytes responded to the mM-CSF-transduced U251 glioma cells, but not to viral vector control U251 glioma cells (U251-VV), by producing a respiratory burst within 20 min. Using patch clamp techniques, functional big potassium (BK) channels were observed on the membrane of the U251 glioma cell. It has been previously reported that oxygen indirectly regulates BK channel function. In this study, it was demonstrated that prolonged BK channel activation in response to the respiratory burst induced by monocytes initiates paraptosis in selected glioma cells. Forced BK channel opening within the glioma cells by BK channel activators (phloretin or pimaric acid) induced U251 glioma cell swelling and vacuolization occurred within 30 min. U251 glioma cell cytotoxicity, induced by using BK channel activators, required between 8 and 12 h. Swelling and vacuolization induced by phloretin and pimaric acid was prevented by iberiotoxin, a specific BK channel inhibitor. Confocal fluorescence microscopy demonstrated BK channels co-localized with the endoplasmic reticulum and mitochondria, the two targeted organelles affected in paraptosis. Iberiotoxin prevented monocytes from producing death in mM-CSF-expressing U251glioma cells in a 24 h assay. This study demonstrates a novel mechanism whereby monocytes can induce paraptosis via the disruption of internal potassium ion homeostasis.
Collapse
Affiliation(s)
- Neil T Hoa
- Department of Diagnostic and Molecular Medicine, Veterans Affairs Medical Center, Long Beach, CA 90822, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Krantic S, Mechawar N, Reix S, Quirion R. Apoptosis-inducing factor: A matter of neuron life and death. Prog Neurobiol 2007; 81:179-96. [PMID: 17267093 DOI: 10.1016/j.pneurobio.2006.12.002] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2006] [Revised: 10/29/2006] [Accepted: 12/07/2006] [Indexed: 01/17/2023]
Abstract
The mitochondrial flavoprotein apoptosis-inducing factor (AIF) is the main mediator of caspase-independent apoptosis-like programmed cell death. Upon pathological permeabilization of the outer mitochondrial membrane, AIF is translocated to the nucleus, where it participates in chromatin condensation and is associated to large-scale DNA fragmentation. Heavy down-regulation of AIF expression in mutant mice or reduced AIF expression achieved with small interfering RNA (siRNA) provides neuroprotection against acute neurodegenerative insults. Paradoxically, in addition to its pro-apoptotic function, AIF likely plays an anti-apoptotic role by regulating the production of reactive oxygen species (ROS) via its putative oxidoreductase and peroxide scavenging activities. In this review, we discuss accumulating evidence linking AIF to both acute and chronic neurodegenerative processes by emphasising mechanisms underlying the dual roles apparently played by AIF in these processes.
Collapse
Affiliation(s)
- Slavica Krantic
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Parc Scientifique Luminy, BP13, 13 273 Marseille, France
| | | | | | | |
Collapse
|
34
|
Zhang JG, Eguchi J, Kruse CA, Gomez GG, Fakhrai H, Schroter S, Ma W, Hoa N, Minev B, Delgado C, Wepsic HT, Okada H, Jadus MR. Antigenic profiling of glioma cells to generate allogeneic vaccines or dendritic cell-based therapeutics. Clin Cancer Res 2007; 13:566-575. [PMID: 17255279 PMCID: PMC4030524 DOI: 10.1158/1078-0432.ccr-06-1576] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE Allogeneic glioma cell lines that are partially matched to the patient at class I human leukocyte antigen (HLA) loci and that display tumor-associated antigens (TAA) or antigenic precursors [tumor antigen precursor proteins (TAPP)] could be used for generating whole tumor cell vaccines or, alternatively, for extraction of TAA peptides to make autologous dendritic cell vaccines. EXPERIMENTAL DESIGN Twenty human glioma cell lines were characterized by molecular phenotyping and by flow cytometry for HLA class I antigen expression. Twelve of the 20 cell lines, as well as analyses of freshly resected glioma tissues, were further characterized for protein and/or mRNA expression of 16 tumor antigen precursor proteins or TAA. RESULTS These 20 human glioma cell lines potentially cover 77%, 85%, and 78% of the U.S. Caucasian population at HLA-A, HLA-B, and HLA-C alleles, respectively. All cells exhibited multiple TAA expressions. Most glioma cells expressed antigen isolated from immunoselected melanoma-2 (Aim-2), B-cyclin, EphA2, GP100, beta1,6-N-acetylglucosaminyltransferase V (GnT-V), IL13Ralpha2, Her2/neu, hTert, Mage, Mart-1, Sart-1, and survivin. Real-time PCR technology showed that glioblastoma specimens expressed most of the TAA as well. Tumor-infiltrating lymphocytes and CD8(+) CTL killed T2 cells when loaded with specific HLA-A2(+) restricted TAA, or gliomas that were both HLA-A2(+) and also positive for specific TAA (Mart-1, GP100, Her2/neu, and tyrosinase) but not those cells negative for HLA-A2 and/or lacking the specific epitope. CONCLUSIONS These data provide proof-in-principle for the use of allogeneic, partially HLA patient-matched glioma cells for vaccine generation or for peptide pulsing with allogeneic glioma cell extracts of autologous patient dendritic cells to induce endogenous CTL in brain tumor patients.
Collapse
Affiliation(s)
- Jian Gang Zhang
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, California
- Pathology Department, Neurooncology Program, Chao Cancer Center, University of California, Irvine, Irvine, California
| | - Junichi Eguchi
- Neurological Surgery, University of Pittsburgh School of Medicine, Brain Tumor Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Carol A. Kruse
- La Jolla Institute for Molecular Medicine, San Diego, California
| | - German G. Gomez
- La Jolla Institute for Molecular Medicine, San Diego, California
| | | | | | - Wenxue Ma
- University of California, San Diego Cancer Center, La Jolla, California
| | - Neil Hoa
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, California
- Pathology Department, Neurooncology Program, Chao Cancer Center, University of California, Irvine, Irvine, California
| | - Boris Minev
- University of California, San Diego Cancer Center, La Jolla, California
| | - Christina Delgado
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, California
- Pathology Department, Neurooncology Program, Chao Cancer Center, University of California, Irvine, Irvine, California
| | - H. Terry Wepsic
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, California
- Pathology Department, Neurooncology Program, Chao Cancer Center, University of California, Irvine, Irvine, California
| | - Hideho Okada
- Neurological Surgery, University of Pittsburgh School of Medicine, Brain Tumor Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Martin R. Jadus
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, California
- Pathology Department, Neurooncology Program, Chao Cancer Center, University of California, Irvine, Irvine, California
| |
Collapse
|
35
|
Chitu V, Stanley ER. Colony-stimulating factor-1 in immunity and inflammation. Curr Opin Immunol 2005; 18:39-48. [PMID: 16337366 DOI: 10.1016/j.coi.2005.11.006] [Citation(s) in RCA: 477] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2005] [Accepted: 11/24/2005] [Indexed: 11/17/2022]
Abstract
Colony-stimulating factor-1 (CSF-1, also known as macrophage-CSF) is the primary regulator of the survival, proliferation, differentiation and function of mononuclear phagocytes. Studies that involve CSF-1-deficient mice demonstrate that there is a variable requirement for CSF-1 in the development of individual mononuclear phagocyte populations. However, these cells uniformly express the CSF-1 receptor, and their morphology, phagocytosis and responsiveness to infectious and non-infectious stimuli is regulated by CSF-1. CSF-1 plays important roles in innate immunity, cancer and inflammatory diseases, including systemic lupus erythematosus, arthritis, atherosclerosis and obesity. In several conditions, activation of macrophages involves a CSF-1 autocrine loop. In addition, secreted and cell-surface isoforms of CSF-1 can have differential effects in inflammation and immunity.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA
| | | |
Collapse
|
36
|
Zhang JG, Dan Q, Fong TC, Williams CC, Avina MD, Tarbiyat-Boldaji M, Khalaghizadeh S, Irwin M, Nguyen A, Zhuang JL, Hoa N, Wepsic HT, Jadus MR. Macrophage colony-stimulating factor expression in retrovirally transduced cells is dependent upon both the adherence status of the target cells and its 5′ flanking untranslated region. Biochem Biophys Res Commun 2005; 330:1275-84. [PMID: 15823581 DOI: 10.1016/j.bbrc.2005.03.113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2005] [Indexed: 11/20/2022]
Abstract
Numerous cell types retrovirally transduced with macrophage colony-stimulating factor (M-CSF) using LXSN-based vectors showed a variable expression of the transgene. Expression of M-CSF correlated with the cells' adherent status. Transduced adherent cells produced the M-CSF, whereas the non-adherent cells synthesized little M-CSF. Studies showed that the 5'-UTR of the M-CSF gene regulated transgenic M-CSF gene expression. Ligation of this 5'-UTR to the enhanced green fluorescent protein gene (EGFP) caused the expression of EGFP to show the same dichotomy as previously seen with the M-CSF. Transgenic M-CSF was expressed within non-adherent cells when the 5'-UTR was removed from the LXSN vector. Quantitative real-time polymerase chain reaction analysis confirmed that lesser production of M-CSF mRNA occurred within the non-adherent cells than in the adherent cells. This difference was eliminated when the 5'-UTR was removed from the retroviral vector. Our work suggests that this 5'-UTR of the M-CSF gene could be an important way to get transgenic expression within adherent cells, but not in non-adherent cells.
Collapse
Affiliation(s)
- Jian-Gang Zhang
- Diagnostic and Molecular Medicine Healthcare Group, Box 113 Veterans Affairs Medical Center, 5901 E. 7th Street, Long Beach, CA 90822, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Despite advances in surgical and adjuvant therapy, the prognosis for malignant gliomas remains dismal. Malignant gliomas, like other malignancies, are able to overcome host immune defences through a variety of mechanisms that have become increasingly well-characterised over the past decade. However, this 'immunologically privileged' status of the brain is not absolute. Systemic immunisation with brain-specific antigens can induce immune responses that are manifested in the CNS, such as experimental allergic encephalomyelitis. The efficacy of peripheral immunisation against brain tumours has also been demonstrated in preclinical models. Based on these observations, clinical trials of peripheral immunisations with brain tumour-derived antigens have been initiated. A limitation of this approach is that the immunological environment within brain tumours is suboptimal for functions of antitumour immune effector cells. As a means to overcome this issue, delivery of cytokine genes to the tumour site may reverse the inhibitory immunological environment of the brain tumours and enhance the efficacy of peripheral vaccine-induced immune effector cells. The brain tumour environment may also be rendered more immunologically favourable by the delivery of additional antigen-presenting cells that can provide infiltrating effector cells with secondary activation signals. Indeed, the authors' recent data indicate that the injection of intracranial tumours with dendritic cells secreting interferon-alpha enhances the efficacy of peripheral vaccinations with tumour-specific antigens by cross-priming tumour antigen-specific T cells in the cervical lymph nodes. This review highlights the recent literature on cytokine gene therapy for brain tumours, and proposes the effective use of cytokine gene delivery both at the site of vaccines (i.e., the site of antigen presentation) and within the target brain tumours (i.e., the site where the effector cells exert their antitumour immunity). Successful immunogene therapy for brain tumours requires detailed understanding of cytokine functions and the use of them at the appropriate stages/sites of the immunological milieu.
Collapse
Affiliation(s)
- Hideho Okada
- University of Pittsburgh Medical Center/Cancer Institute, Department of Neurological Surgery, Brain Tumor Program, G12.a Research Pavilion at the Hillman Cancer Center, 5117 Centre Ave, Pittsburgh, PA 15213-1863, USA.
| | | |
Collapse
|
38
|
Jeffes EWB, Zhang JG, Hoa N, Petkar A, Delgado C, Chong S, Obenaus A, Sanchez R, Khalaghizadeh S, Khomenko T, Knight BA, Alipanah R, Nguyen TV, Shah C, Vohra S, Zhuang JL, Liu J, Wepsic HT, Jadus MR. Antiangiogenic Drugs Synergize with a Membrane Macrophage Colony-Stimulating Factor-Based Tumor Vaccine to Therapeutically Treat Rats with an Established Malignant Intracranial Glioma. THE JOURNAL OF IMMUNOLOGY 2005; 174:2533-43. [PMID: 15728459 DOI: 10.4049/jimmunol.174.5.2533] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Combining a T9/9L glioma vaccine, expressing the membrane form of M-CSF, with a systemic antiangiogenic drug-based therapy theoretically targeted toward growth factor receptors within the tumor's vasculature successfully treated >90% of the rats bearing 7-day-old intracranial T9/9L gliomas. The antiangiogenic drugs included (Z)-3-[4-(dimethylamino)benzylidenyl]indolin-2-one (a platelet-derived growth factor receptor beta and a fibroblast growth factor receptor 1 kinase inhibitor) and oxindole (a vascular endothelial growth factor receptor 2 kinase inhibitor). A total of 20-40% of the animals treated with the antiangiogenic drugs alone survived, while all nontreated controls and tumor vaccine-treated rats died within 40 days. In vitro, these drugs inhibited endothelial cells from proliferating in response to the angiogenic factors produced by T9/9L glioma cells and prevented endothelial cell tubulogenesis. FITC-labeled tomato lectin staining demonstrated fewer and constricted blood vessels within the intracranial tumor after drug therapy. Magnetic resonance imaging demonstrated that the intracranial T9 glioma grew much slower in the presence of these antiangiogenic drugs. These drugs did not affect in vitro glioma cell growth nor T cell mitogenesis. Histological analysis revealed that the tumor destruction occurred at the margins of the tumor, where there was a heavy lymphocytic infiltrate. Real-time PCR showed more IL-2-specific mRNA was present within the gliomas in the vaccinated rats treated with the drugs. Animals that rejected the established T9/9L glioma by the combination therapy proved immune against an intracranial rechallenge by T9/9L glioma, but showed no resistance to an unrelated MADB106 breast cancer.
Collapse
Affiliation(s)
- Edward W B Jeffes
- Diagnostic and Molecular Health Care Group, Veterans Affairs Medical Center, Long Beach, CA 90822, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|