1
|
Meier P, Legrand AJ, Adam D, Silke J. Immunogenic cell death in cancer: targeting necroptosis to induce antitumour immunity. Nat Rev Cancer 2024; 24:299-315. [PMID: 38454135 DOI: 10.1038/s41568-024-00674-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 03/09/2024]
Abstract
Most metastatic cancers remain incurable due to the emergence of apoptosis-resistant clones, fuelled by intratumour heterogeneity and tumour evolution. To improve treatment, therapies should not only kill cancer cells but also activate the immune system against the tumour to eliminate any residual cancer cells that survive treatment. While current cancer therapies rely heavily on apoptosis - a largely immunologically silent form of cell death - there is growing interest in harnessing immunogenic forms of cell death such as necroptosis. Unlike apoptosis, necroptosis generates second messengers that act on immune cells in the tumour microenvironment, alerting them of danger. This lytic form of cell death optimizes the provision of antigens and adjuvanticity for immune cells, potentially boosting anticancer treatment approaches by combining cellular suicide and immune response approaches. In this Review, we discuss the mechanisms of necroptosis and how it activates antigen-presenting cells, drives cross-priming of CD8+ T cells and induces antitumour immune responses. We also examine the opportunities and potential drawbacks of such strategies for exposing cancer cells to immunological attacks.
Collapse
Affiliation(s)
- Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, UK.
| | - Arnaud J Legrand
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, UK
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| | - John Silke
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Zheng C, Guo H, Mo Y, Liu G. Integrating Bioinformatics and Drug Sensitivity Analyses to Identify Molecular Characteristics Associated with Targeting Necroptosis in Breast Cancer and their Clinical Prognostic Significance. Recent Pat Anticancer Drug Discov 2024; 19:681-694. [PMID: 37653627 DOI: 10.2174/1574892819666230831112815] [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: 04/14/2023] [Revised: 06/26/2023] [Accepted: 07/21/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Breast cancer accounts for over 1.8 million new cases worldwide annually, and prompt diagnosis and treatment are imperative to prevent mortality. Necroptosis, a form of programmed cell death, is thought to be a critical pathway for cancer cell apoptosis, yet, its relationship with breast cancer progression and molecular mechanisms remains largely unexplored. OBJECTIVES Our study aims to investigate the molecular characteristics and clinical prognostic value of necroptosis-related genes in breast cancer using a comprehensive approach that involves integrated bioinformatics analysis along with drug sensitivity assessment. METHODS Transcriptional, clinical, and tumor mutation burden (TMB) data related to breast cancer from the TCGA and GEO databases were integrated, and the necroptosis gene set was downloaded from the GSEA website for analysis. The screening conditions were set as adjusted p < 0.05 and |log2FC(fold change)| > 0.585 to screen for differential expression genes related to breast cancer necroptosis. Survival prognosis analysis was conducted on breast cancer necroptosis genes. Further analysis was conducted on prognosis-related necroptosis genes, including immune infiltration analysis and GO/KEGG enrichment analysis, to explore the potential biological functions and signaling pathway mechanisms of breast cancer necroptosis genes. Drug sensitivity screening was conducted on the prognosis-related necroptosis to identify potential drugs that target the promotion of necroptosis gene expression, and ultimately, single-gene analysis was performed on the core target genes obtained. RESULTS Through integrated bioinformatics analysis, 29 differentially expressed mRNAs related to BRCA-Necroptosis were identified, including 18 upregulated mRNAs and 11 downregulated mRNAs. In addition, single-factor analysis of differential genes showed that the expression of HSPA4, PLK1, TNFRSF1B, FLT3, and LEF1 was closely related to BRCA survival prognosis. Based on the expression of these genes, a breast cancer prognosis model was constructed, and it was found that the area under the curve (AUC) of the curve of the risk genes for necroptosis was the largest, indicating that these genes have a certain clinical predictive significance for the occurrence and prognosis of BRCA. Additionally, there were significant differences in clinical characteristics of BRCA patients with different necroptosis gene expressions. Furthermore, GSVA and immune infiltration analysis revealed that Necroptosis-DEGs mainly affect the occurrence and progression of BRCA by participating in immune functions such as APC co-inhibition, APC costimulation, CCR, checkpoint, as well as infiltrating immune cells such as B cells naive, plasma cells, and T cells CD8. Moreover, the necroptosis gene group column chart indicated a 1-year survival rate of 0.979, a 3-year survival rate of 0.883, and a 5-year survival rate of 0.774. The necroptosis gene group and column chart are important indicators for evaluating BRCA prognosis. Finally, drug sensitivity screening of BRCA-Necroptosis genes showed that compounds such as A- 770041, AC220, AP-24534, Bexarotene, and BMS-509744 have certain effects as potential targeted drugs for the treatment of BRCA necroptosis genes. CONCLUSION Necroptosis genes are implicated in the pathogenesis and progression of breast cancer and are thought to impact the prognosis and response to drug treatments in individuals with BRCA. Consequently, understanding the role of these genes in breast cancer may aid in identifying more precise and efficacious therapeutic targets.
Collapse
Affiliation(s)
- Chang Zheng
- Department of Surgery, Shantou University Medical College, Shantou, 515041, China
| | - Hanbin Guo
- Department of Surgery, Shantou University Medical College, Shantou, 515041, China
| | - Yongpan Mo
- Department of Breast and Thyroid Surgery, Second People's Hospital of Shenzhen, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Guowen Liu
- Department of Breast and Thyroid Surgery, Second People's Hospital of Shenzhen, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| |
Collapse
|
3
|
Di Y, Shen Q, Yang Z, Song G, Fang T, Liu Y, Liu Y, Luo Q, Wang F, Yan X, Bai H, Huang Y, Lv F, Wang S. Biosynthesis of Multifunctional Transformable Peptides for Inducing Tumor Cell Apoptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303035. [PMID: 37605329 DOI: 10.1002/smll.202303035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/12/2023] [Indexed: 08/23/2023]
Abstract
Engineered nanomaterials hold great promise to improve the specificity of disease treatment. Herein, a fully protein-based material is obtained from nonpathogenic Escherichia coli (E. coli), which is capable of morphological transformation from globular to fibrous in situ for inducing tumor cell apoptosis. The protein-based material P1 is comprised of a β-sheet-forming peptide KLVFF, pro-apoptotic protein BAK, and GFP along with targeting moieties. The self-assembled nanoparticles of P1 transform into nanofibers in situ in the presence of cathepsin B, and the generated nanofibrils favor the dimerization of functional BH3 domain of BAK on the mitochondrial outer membrane, leading to efficient anticancer activity both in vitro and in vivo via mitochondria-dependent apoptosis through Bcl-2 pathway. To precisely manipulate the morphological transformation of biosynthetic molecules in living cells, a spatiotemporally controllable anticancer system is constructed by coating P1-expressing E. coli with cationic conjugated polyelectrolytes to release the peptides in situ under light irradiation. The biosynthetic peptide-based enzyme-catalytic transformation strategy in vivo would offer a novel perspective for targeted delivery and shows great potential in precision disease therapeutics.
Collapse
Affiliation(s)
- Yufei Di
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qi Shen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Zhiwen Yang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Gang Song
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Tiantian Fang
- Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yazhou Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yamei Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Qun Luo
- College of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Fuyi Wang
- College of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Haotian Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yiming Huang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Fengting Lv
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
4
|
Zhu R, Wang Y, Ouyang Z, Hao W, Zhou F, Lin Y, Cheng Y, Zhou R, Hu W. Targeting regulated chondrocyte death in osteoarthritis therapy. Biochem Pharmacol 2023; 215:115707. [PMID: 37506921 DOI: 10.1016/j.bcp.2023.115707] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
In vivo articular cartilage degeneration is an essential hallmark of osteoarthritis (OA), involving chondrocyte senescence, extracellular matrix degradation, chondrocyte death, cartilage loss, and bone erosion. Among them, chondrocyte death is one of the major factors leading to cartilage degeneration. Many studies have reported that various cell death modes, including apoptosis, ferroptosis, and autophagy, play a key role in OA chondrocyte death. Currently, there is insufficient understanding of OA pathogenesis, and there remains a lack of treatment methods to prevent OA and inhibit its progression. Studies suggest that OA prevention and treatment are mainly directed to arrest premature or excessive chondrocyte death. In this review, we a) discuss the forms of death of chondrocytes and the associations between them, b) summarize the critical factors in chondrocyte death, c) discuss the vital role of chondrocyte death in OA, d) and, explore new approaches for targeting the regulation of chondrocyte death in OA treatment.
Collapse
Affiliation(s)
- Rendi Zhu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yan Wang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ziwei Ouyang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wenjuan Hao
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Fuli Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yi Lin
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yuanzhi Cheng
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Renpeng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| |
Collapse
|
5
|
Anosike NL, Adejuwon JF, Emmanuel GE, Adebayo OS, Etti-Balogun H, Nathaniel JN, Omotosho OI, Aschner M, Ijomone OM. Necroptosis in the developing brain: role in neurodevelopmental disorders. Metab Brain Dis 2023; 38:831-837. [PMID: 36964816 DOI: 10.1007/s11011-023-01203-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 03/17/2023] [Indexed: 03/26/2023]
Abstract
Cell death is vital to various organismal developmental processes including brain development. Apoptosis, the most recognized programmed cell death, has been linked to several developmental processes and implicated in pruning cells to provide the ultimate tissue integrity. However, more recently, other forms of non-apoptotic programmed cell death have been identified, of which necroptosis is of predominant interest. Necroptosis is a regulated form of necrosis, activated under apoptotic-deficient conditions. Tumour necrosis factor (TNF) is a major activator of necroptosis, and the process is mediated by several kinases including receptor-interacting protein kinase (RIPK) and mixed lineage kinase domain-like protein (MLKL). Potential roles for necroptosis during brain development have been muted. Necroptosis has been implicated in mediating neurological disorders, and contributing to the severity of these disorders. Here we will review the literature on the role of necroptosis in neurodevelopment, summarizing its molecular mechanisms and highlighting potential implications for disorders of the developing brain.
Collapse
Affiliation(s)
- Nnenna Loveth Anosike
- The Neuro- Lab, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Joy Funsho Adejuwon
- The Neuro- Lab, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
- Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Godslove Emeka Emmanuel
- The Neuro- Lab, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
- Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Oluwatosin Samuel Adebayo
- The Neuro- Lab, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
- Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Hassanat Etti-Balogun
- The Neuro- Lab, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
- Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Jannie Nathaniel Nathaniel
- The Neuro- Lab, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
- Department of Physiology, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Omolabake Ifeoluwa Omotosho
- The Neuro- Lab, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
- Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Neurosciences, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Paediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Omamuyovwi Meashack Ijomone
- The Neuro- Lab, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria.
- Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Akure, Nigeria.
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
6
|
Vanmeerbeek I, Govaerts J, Laureano RS, Sprooten J, Naulaerts S, Borras DM, Laoui D, Mazzone M, Van Ginderachter JA, Garg AD. The Interface of Tumour-Associated Macrophages with Dying Cancer Cells in Immuno-Oncology. Cells 2022; 11:3890. [PMID: 36497148 PMCID: PMC9741298 DOI: 10.3390/cells11233890] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Tumour-associated macrophages (TAMs) are essential players in the tumour microenvironment (TME) and modulate various pro-tumorigenic functions such as immunosuppression, angiogenesis, cancer cell proliferation, invasion and metastasis, along with resistance to anti-cancer therapies. TAMs also mediate important anti-tumour functions and can clear dying cancer cells via efferocytosis. Thus, not surprisingly, TAMs exhibit heterogeneous activities and functional plasticity depending on the type and context of cancer cell death that they are faced with. This ultimately governs both the pro-tumorigenic and anti-tumorigenic activity of TAMs, making the interface between TAMs and dying cancer cells very important for modulating cancer growth and the efficacy of chemo-radiotherapy or immunotherapy. In this review, we discuss the interface of TAMs with cancer cell death from the perspectives of cell death pathways, TME-driven variations, TAM heterogeneity and cell-death-inducing anti-cancer therapies. We believe that a better understanding of how dying cancer cells influence TAMs can lead to improved combinatorial anti-cancer therapies, especially in combination with TAM-targeting immunotherapies.
Collapse
Affiliation(s)
- Isaure Vanmeerbeek
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Raquel S. Laureano
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Jenny Sprooten
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Daniel M. Borras
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Damya Laoui
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, 1050 Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumour Inflammation and Angiogenesis, VIB Center for Cancer Biology, 3000 Leuven, Belgium
- Laboratory of Tumour Inflammation and Angiogenesis, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Jo A. Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| | - Abhishek D. Garg
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
7
|
Tu M, Tan VP, Yu JD, Tripathi R, Bigham Z, Barlow M, Smith JM, Brown JH, Miyamoto S. RhoA signaling increases mitophagy and protects cardiomyocytes against ischemia by stabilizing PINK1 protein and recruiting Parkin to mitochondria. Cell Death Differ 2022; 29:2472-2486. [PMID: 35760846 PMCID: PMC9751115 DOI: 10.1038/s41418-022-01032-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 01/31/2023] Open
Abstract
Mitophagy, a mitochondria-specific form of autophagy, removes dysfunctional mitochondria and is hence an essential process contributing to mitochondrial quality control. PTEN-induced kinase 1 (PINK1) and the E3 ubiquitin ligase Parkin are critical molecules involved in stress-induced mitophagy, but the intracellular signaling mechanisms by which this pathway is regulated are unclear. We tested the hypothesis that signaling through RhoA, a small GTPase, induces mitophagy via modulation of the PINK1/Parkin pathway as a protective mechanism against ischemic stress. We demonstrate that expression of constitutively active RhoA as well as sphingosine-1-phosphate induced activation of endogenous RhoA in cardiomyocytes result in an accumulation of PINK1 at mitochondria. This is accompanied by translocation of Parkin to mitochondria and ubiquitination of mitochondrial proteins leading to recognition of mitochondria by autophagosomes and their lysosomal degradation. Expression of RhoA in cardiomyocytes confers protection against ischemia, and this cardioprotection is attenuated by siRNA-mediated PINK1 knockdown. In vivo myocardial infarction elicits increases in mitochondrial PINK1, Parkin, and ubiquitinated mitochondrial proteins. AAV9-mediated RhoA expression potentiates these responses and a concurrent decrease in infarct size is observed. Interestingly, induction of mitochondrial PINK1 accumulation in response to RhoA signaling is neither mediated through its transcriptional upregulation nor dependent on depolarization of the mitochondrial membrane, the canonical mechanism for PINK1 accumulation. Instead, our results reveal that RhoA signaling inhibits PINK1 cleavage, thereby stabilizing PINK1 protein at mitochondria. We further show that active RhoA localizes at mitochondria and interacts with PINK1, and that the mitochondrial localization of RhoA is regulated by its downstream effector protein kinase D. These findings demonstrate that RhoA activation engages a unique mechanism to regulate PINK1 accumulation, induce mitophagy and protect against ischemic stress, and implicates regulation of RhoA signaling as a potential strategy to enhance mitophagy and confer protection under stress conditions.
Collapse
Affiliation(s)
- Michelle Tu
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0636, USA
| | - Valerie P Tan
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0636, USA
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Justin D Yu
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0636, USA
| | - Raghav Tripathi
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0636, USA
| | - Zahna Bigham
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0636, USA
| | - Melissa Barlow
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0636, USA
| | - Jeffrey M Smith
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0636, USA
| | - Joan Heller Brown
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0636, USA
| | - Shigeki Miyamoto
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0636, USA.
| |
Collapse
|
8
|
Andreone L, Fuertes F, Sétula C, Barcala Tabarrozzi AE, Orellano MS, Dewey RA, Bottino R, De Bosscher K, Perone MJ. Compound A attenuates proinflammatory cytokine-induced endoplasmic reticulum stress in beta cells and displays beneficial therapeutic effects in a mouse model of autoimmune diabetes. Cell Mol Life Sci 2022; 79:587. [PMID: 36370223 DOI: 10.1007/s00018-022-04615-5] [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: 07/22/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/15/2022]
Abstract
Type 1 diabetes (T1D) is characterized by an immune-mediated progressive destruction of the insulin-producing β-cells. Proinflammatory cytokines trigger endoplasmic reticulum (ER) stress and subsequent insulin secretory deficiency in cultured β-cells, mimicking the islet microenvironment in T1D. β-cells undergo physiologic ER stress due to the high rate of insulin production and secretion under stimulated conditions. Severe and uncompensated ER stress in β-cells is induced by several pathological mechanisms before onset and during T1D. We previously described that the small drug Compound A (CpdA), a selective glucocorticoid receptor (GR/NR3C1, nuclear receptor subfamily 3, group C, member 1) ligand with demonstrated inflammation-suppressive activity in vivo, is an effective modulator of effector T and dendritic cells and of macrophages, yet, in a GR-independent manner. Here, we focus on CpdA's therapeutic potential in T1D cellular and animal models. We demonstrate that CpdA improves the unfolded protein response (UPR) by attenuating ER stress and favoring the survival and function of β-cells exposed to an environment of proinflammatory cytokines. CpdA administration to NODscid mice adoptively transferred with diabetogenic splenocytes (from diabetic NOD mice) led to a delay of disease onset and reduction of diabetes incidence. Histological analysis of the pancreas showed a reduction in islet leukocyte infiltration (insulitis) and preservation of insulin expression in CpdA-treated normoglycemic mice in comparison with control group. These new findings together with our previous reports justify further studies on the administration of this small molecule as a novel therapeutic strategy with dual targets (effector immune and β-cells) during autoimmune diabetes.
Collapse
Affiliation(s)
- Luz Andreone
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Florencia Fuertes
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Carolina Sétula
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Andres E Barcala Tabarrozzi
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Miranda S Orellano
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina
| | - Ricardo A Dewey
- Laboratorio de Terapia Génica Y Células Madre, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM, Buenos Aires, Argentina
| | - Rita Bottino
- Imagine Pharma, Pittsburgh, Pennsylvania, PA and Allegheny Health Network, Pittsburgh, PA, USA
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-Department of Medical Protein Research, VIB, Ghent University, Ghent, Belgium
| | - Marcelo J Perone
- Laboratory of Immuno-Endocrinology, Diabetes and Metabolism, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ), Pilar, Buenos Aires, Argentina.
| |
Collapse
|
9
|
Memari F, Mirzavi F, Jalili-Nik M, Afshari AR, Ghorbani A, Soukhtanloo M. Tumor-Inhibitory Effects of Zerumbone Against HT-29 Human Colorectal Cancer Cells. Int J Toxicol 2022; 41:402-411. [PMID: 35719111 DOI: 10.1177/10915818221104417] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Colorectal cancer (CRC) is the second cause of cancer-associated death globally. Recently, herbal medicinal products and, in particular, zerumbone have been widely studied and used for cancer treatment as they induce significant anti-cancer effects. However, there is limited information about the anti-cancer effects of zerumbone in CRC. Therefore, we aimed to investigate the in vitro anti-cancer effects of the zerumbone in CRC, focusing on cell apoptosis and migration. Anti-proliferative and anti-migratory effects of zerumbone on HT-29 cells were evaluated using MTT and scratch wound healing assay, respectively. Quantitative real-time PCR (qRT-PCR) was performed to determine the mRNA expression levels of migration and apoptosis-related genes. Apoptosis and cell cycle distribution were evaluated by flow cytometry. The intracellular level of reactive oxygen species (ROS) was measured using a ROS assay kit. Additionally, matrix metalloproteinase-2/-9 (MMP-2/-9) activity was determined using gelatin zymography. Zerumbone suppressed the viability of the HT-29 cells dose-dependently while having less cytotoxicity on normal NIH/3T3 cells. Zerumbone induced apoptosis in HT-29 cells and arrested the cell cycle in the G2/M phase. These effects were associated with alteration in the expression of apoptosis-related genes (up-regulation of Bax and down-regulation of Bcl-2 genes). Zerumbone also enhanced the generation of ROS in HT-29 cells. Furthermore, zerumbone significantly inhibited the migration of HT-29 cells and decreased MMP-2/-9 mRNA expression and activity. Our findings provide a potential use for zerumbone to induce apoptosis and suppress metastasis in HT-29 cells; thus, it could be developed as a promising natural agent for future CRC therapy.
Collapse
Affiliation(s)
- Fezzeh Memari
- Department of Clinical Biochemistry, Faculty of Medicine, 113380Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, 196469Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Jalili-Nik
- Department of Clinical Biochemistry, Faculty of Medicine, 113380Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, 196469North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Ahmad Ghorbani
- Department of Pharmacology, Faculty of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmacological Research Center of Medicinal Plants, 113380Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, 113380Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmacological Research Center of Medicinal Plants, 113380Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Mohammed RN, Khosravi M, Rahman HS, Adili A, Kamali N, Soloshenkov PP, Thangavelu L, Saeedi H, Shomali N, Tamjidifar R, Isazadeh A, Aslaminabad R, Akbari M. Anastasis: cell recovery mechanisms and potential role in cancer. Cell Commun Signal 2022; 20:81. [PMID: 35659306 PMCID: PMC9166643 DOI: 10.1186/s12964-022-00880-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 04/07/2022] [Indexed: 12/13/2022] Open
Abstract
Balanced cell death and survival are among the most important cell development and homeostasis pathways that can play a critical role in the onset or progress of malignancy steps. Anastasis is a natural cell recovery pathway that rescues cells after removing the apoptosis-inducing agent or brink of death. The cells recuperate and recover to an active and stable state. So far, minimal knowledge is available about the molecular mechanisms of anastasis. Still, several involved pathways have been explained: recovery through mitochondrial outer membrane permeabilization, caspase cascade arrest, repairing DNA damage, apoptotic bodies formation, and phosphatidylserine. Anastasis can facilitate the survival of damaged or tumor cells, promote malignancy, and increase drug resistance and metastasis. Here, we noted recently known mechanisms of the anastasis process and underlying molecular mechanisms. Additionally, we summarize the consequences of anastatic mechanisms in the initiation and progress of malignancy, cancer cell metastasis, and drug resistance. Video Abstract
Collapse
|
11
|
In vivo Study of a Newly Synthesized Chromen-4-one Derivative as an Antitumor Agent against HCC. J Gastrointest Cancer 2021; 53:980-989. [PMID: 34698995 DOI: 10.1007/s12029-021-00724-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Chromenes are a wide group of natural compounds that can be synthesized chemically. The chromen-4-one nucleus acts as a skeleton for varieties of additional active groups that makes the chromene activity vary between antioxidant and anti-inflammatory agents. In the present study, a newly synthesized chromene compound exhibits different behaviors other than anti-inflammatory and antioxidant activities that it is the first time that a member of chromen-4-one compound can control the cancer progress. Inflammation is the first step in tumor development where the severity grade can potentiate tumor growth and progression. In many tumors, pro-inflammatory genes record high expression level such as tumor necrosis factor (TNF-α) and vascular endothelial growth factors (VEGF). These pro-inflammatory factors act as rate limiting steps in tumor initiation, and controlling its expression acts as an early therapeutic way to control the tumor proliferation. The chromone derivatives have biological activities such as anti-inflammatory and anti-tumor activity. METHODS In the present study, hepatocellular cancer (HCC) induced by diethylnitrosamine (DEN) in rats and then treated with the new chromene derivative and the parameters TNF-α, VEGF, p53, Cyt C, MMP-9, Bcl2, and Bax were measured. RESULTS The treatment strategy Ch compound is to downregulate pro-inflammatory gene expression of early genes as TNF-α as well as VEGF and subsequently control other factors such as p53, Cyt C, and MMP-9. Also, retrieve the balance between Bcl2 and Bax proteins in DEN-induced HCC in rats. CONCLUSION The ability of the new Ch derivative to control the primary initiators of HCC such as TNF-α offers this derivative an anti-tumor activity and encourages further researches to follow and monitor its effect on the molecular level.
Collapse
|
12
|
Yuan W, Wu J, Zhang Q, Liang Y, Zhang M, Qin H, Li CS. Shen-fu injection alleviates acute renal injury by reducing cytokine levels and modulating apoptosis in a porcine hemorrhagic shock model. Acta Cir Bras 2021; 36:e360405. [PMID: 34076082 PMCID: PMC8184256 DOI: 10.1590/acb360405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/16/2021] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Shen-fu injection (SFI) was used to intervene in the resuscitation of porcine hemorrhagic shock (HS) model to study its protective effects on acute kidney injury. METHODS After 60 min of HS, 28 animals were randomly assigned into four groups. The groups were as follows: hemorrhagic shock group (HS); HS resuscitation with shed-blood group (HSR); HS resuscitation with shed-blood and SFI (1 mL·kg-1) group (HSR-SFI); and the sham operation group (Sham). The bloods were analyzed for serum creatinine (sCr), cystatin C (CysC) and neutrophil gelatinase-associated lipocalin (NGAL). BAX, Bcl-2, and caspase-3 protein expressions by Western blot analysis and immunohistochemical staining. The renal tissues were removed and pathologic changes were observed. RESULTS Mean aortic pressure (MAP) in HSR-SFI groups were higher than that in HSR groups after shock. At the 6th hour after shock, the urine volume per hour in the HSR-SFI groups was more than that in the HSR groups. The sCr, NGAL, CysC and cytokine levels of HSR-SFI groups were lower. The Bcl-2 expression was increased in the HSR-SFI groups. The BAX and caspase-3 expressions were reduced. The histopathologic score in the HSR-SFI was lower. CONCLUSIONS SFI may reduce the risk of acute kidney injury (AKI) following hemorrhagic shock by attenuating systemic inflammatory responses, and regulating the expression of apoptosis-related proteins.
Collapse
Affiliation(s)
- Wei Yuan
- Capital Medical University, China
| | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
INTRODUCTION Levels of the apoptosis regulator Fas ligand (FasL) are associated with severity of sepsis, but its association with the mortality of sepsis and necroptosis, a regulated cell death mechanism, is not yet clear. We aimed to assess the association of FasL level with outcomes of sepsis and receptor interacting protein kinase-3 (RIPK3), an essential necroptosis mediator, for determining the relationship between FasL and necroptosis. METHODS Plasma FasL and RIPK3 levels were measured by ELISA from prospectively enrolled critically-ill adult patients. The best cut-off level of FasL for 28-day mortality prediction was determined by Youden's index. The association between plasma levels of FasL and RIPK3 was assessed by a linear regression method. RESULTS Among 188 patients, 58 (30.9%) were diagnosed with sepsis and 84 (44.7%) with septic shock, respectively. Plasma levels of FasL increased in the group order of control, sepsis, and septic shock groups (P for trend < 0.001). For 142 patients with sepsis, organ dysfunction and septic shock were more prevalent in the group with plasma FasL levels that were higher than the best cut-off level. A significant difference in mortality between high and low FasL patients was observed up to 90 days (Log-rank P = 0.013). FasL levels did not significantly change over day 3 and day 7. FasL levels were not correlated with those of RIPK3. CONCLUSIONS The plasma level of FasL was associated with severity of sepsis and was predictive of mortality. However, it was not correlated with RIPK3 level.
Collapse
|
14
|
Paricalcitol Attenuates Contrast-Induced Acute Kidney Injury by Regulating Mitophagy and Senescence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7627934. [PMID: 33299530 PMCID: PMC7704155 DOI: 10.1155/2020/7627934] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/16/2020] [Accepted: 10/28/2020] [Indexed: 12/26/2022]
Abstract
Contrast-induced acute kidney injury (CI-AKI) is the third most common cause of hospital-acquired renal failure, with an incidence of 11%. However, the disease mechanism remains unclear, and no effective treatment is available. Paricalcitol has been reported to be effective in animal models of kidney injury. We hypothesized that paricalcitol could play a renoprotective role against CI-AKI. Rats were divided into control, paricalcitol, contrast, and paricalcitol-plus-contrast groups. We used a previously published protocol to produce CI-AKI. Paricalcitol (0.3 μg/kg) was administered intraperitoneally before 24 h and 30 min before indomethacin. We used HK-2 cells to evaluate the effects of paricalcitol on mitophagy and senescence. Ioversol triggered renal dysfunction, increasing blood urea nitrogen and serum creatinine. Significant tubular damage, increased 8-OHdG expression, and apoptosis were apparent. Ioversol injection induced high expression levels of the mitophagy markers Pink1, Parkin, and LC3 and the senescence markers β-galactosidase and p16INK4A. Paricalcitol pretreatment prevented renal dysfunction and reduced tissue damage by reducing both mitophagy and senescence. Cellular morphological changes were found, and expression of LC3B and HMGB1 was increased by ioversol in HK-2 cells. Paricalcitol countered these effects. This study showed that mitochondria might drive injury phenotypes in CI-AKI, and that paricalcitol protects against CI-AKI by decreasing mitochondrial damage.
Collapse
|
15
|
Norcantharidin: research advances in pharmaceutical activities and derivatives in recent years. Biomed Pharmacother 2020; 131:110755. [PMID: 33152920 DOI: 10.1016/j.biopha.2020.110755] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Cantharidin (CTD) is the main bioactive component of Cantharides, which is called Banmao in Traditional Chinese Medicine (TCM). Norcantharidin (NCTD) is a structural modifier of CTD. To compare with CTD, NCTD has lighter side effects and stronger bioactivity in anti-cancer through inhibiting cell proliferation, causing apoptosis and autophagy, overwhelming migration and metastasis, affecting immunity as well as lymphangiogenesis. Examples of these effects include suppressing Protein Phosphatase 2A and modulating Wnt/beta catenin signal, with Caspase family proteins, AMPK pathway and c-Met/EGFR pathway involving respectively. Moreover, NCTD has the effects of immune enhancement, anti-platelet aggregation and inhibition of renal interstitial fibrosis with distinct signaling pathways. The immunological effects induced by NCTD are related to the regulation of macrophage polarization and LPS-mediated immune response. The antiplatelet activity that NCTD induced is relevant to the inhibition of platelet signaling and the downregulation of α2 integrin. Furthermore, some of novel derivatives designed and synthesized artificially show stronger biological activities (e.g., anticancer effect, enzyme inhibition effect, antioxidant effect) and lower toxicity than NCTD itself. Plenty of literatures have reported various pharmacological effects of NCTD, particularly the anticancer effect, which has been widely concerned in clinical application and laboratory research. In this review, the pharmaceutical activities and derivatives of NCTD are discussed, which can be reference for further study.
Collapse
|
16
|
Michalek S, Brunner T. Nuclear-mitochondrial crosstalk: On the role of the nuclear receptor liver receptor homolog-1 (NR5A2) in the regulation of mitochondrial metabolism, cell survival, and cancer. IUBMB Life 2020; 73:592-610. [PMID: 32931651 DOI: 10.1002/iub.2386] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
Liver receptor homolog-1 (LRH-1, NR5A2) is an orphan nuclear receptor with widespread activities in the regulation of development, stemness, metabolism, steroidogenesis, and proliferation. Many of the LRH-1-regulated processes target the mitochondria and associated activities. While under physiological conditions, a balanced LRH-1 expression and regulation contribute to the maintenance of a physiological equilibrium, deregulation of LRH-1 has been associated with inflammation and cancer. In this review, we discuss the role and mechanism(s) of how LRH-1 regulates metabolic processes, cell survival, and cancer in a nuclear-mitochondrial crosstalk, and evaluate its potential as a pharmacological target.
Collapse
Affiliation(s)
- Svenja Michalek
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
17
|
Hsu SK, Chang WT, Lin IL, Chen YF, Padalwar NB, Cheng KC, Teng YN, Wang CH, Chiu CC. The Role of Necroptosis in ROS-Mediated Cancer Therapies and Its Promising Applications. Cancers (Basel) 2020; 12:E2185. [PMID: 32764483 PMCID: PMC7465132 DOI: 10.3390/cancers12082185] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past decades, promising therapies targeting different signaling pathways have emerged. Among these pathways, apoptosis has been well investigated and targeted to design diverse chemotherapies. However, some patients are chemoresistant to these therapies due to compromised apoptotic cell death. Hence, exploring alternative treatments aimed at different mechanisms of cell death seems to be a potential strategy for bypassing impaired apoptotic cell death. Emerging evidence has shown that necroptosis, a caspase-independent form of cell death with features between apoptosis and necrosis, can overcome the predicament of drug resistance. Furthermore, previous studies have also indicated that there is a close correlation between necroptosis and reactive oxygen species (ROS); both necroptosis and ROS play significant roles both under human physiological conditions such as the regulation of inflammation and in cancer biology. Several small molecules used in experiments and clinical practice eliminate cancer cells via the modulation of ROS and necroptosis. The molecular mechanisms of these promising therapies are discussed in detail in this review.
Collapse
Affiliation(s)
- Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Wen-Tsan Chang
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - I-Ling Lin
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Nitin Balkrushna Padalwar
- Department of Chemistry, National Institute of Technology Tiruchirappalli, Tiruchirappalli 620015, Tamilnadu, India;
| | - Kai-Chun Cheng
- Department of Ophthalmology, Kaohsiung Municipal Hsiaokang Hospital, Kaohsiung 812, Taiwan;
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan;
| | - Chi-Huei Wang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- The Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
18
|
Zerumbone Promotes Cytotoxicity in Human Malignant Glioblastoma Cells through Reactive Oxygen Species (ROS) Generation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3237983. [PMID: 32454937 PMCID: PMC7225859 DOI: 10.1155/2020/3237983] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/13/2020] [Accepted: 04/23/2020] [Indexed: 01/28/2023]
Abstract
Glioblastoma multiforme (GBM) is the most hostile tumor in the central nervous system. Unfortunately, the prognosis of GBM patients is poor following surgical interventions, chemotherapy, and radiotherapy. Consequently, more efficient and effective treatment options for the treatment of GBM need to be explored. Zerumbone, as a sesquiterpene derived from Zingiber zerumbet Smith, has substantial cytotoxic and antiproliferative activities in some types of cancer. Here, we show that exposure of GBM cells (U-87 MG) to Zerumbone demonstrated significant growth inhibition in a concentration-dependent manner. Zerumbone also induced apoptosis and caused cell cycle arrest of human GBM U-87 MG cells in the G2/M phase of the cell cycle. In detail, the apoptotic process triggered by Zerumbone involved the upregulation of proapoptotic Bax and the suppression of antiapoptotic Bcl-2 genes expression as determined by qRT-PCR. Moreover, Zerumbone enhanced the generation of reactive oxygen species (ROS), and N-acetyl cysteine (NAC), as an antioxidant, reversed the ROS-induced cytotoxicity of U-87 MG cells. The Western blot analysis suggested that Zerumbone activated the NF-κB p65, which was partly inhibited by NAC treatment. Collectively, our results confirmed that Zerumbone induces cytotoxicity by ROS generation. Thus, the study raises the possibility of Zerumbone as a potential natural agent for treating GBM due to its ability to induce cytotoxicity.
Collapse
|
19
|
Xia C, He Z, Cai Y, Liang S. Vorinostat upregulates MICA via the PI3K/Akt pathway to enhance the ability of natural killer cells to kill tumor cells. Eur J Pharmacol 2020; 875:173057. [PMID: 32135122 DOI: 10.1016/j.ejphar.2020.173057] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/26/2022]
Abstract
Vorinostat has good therapeutic efficacy against primary cutaneous T-cell lymphoma in the refractory stage. However, the molecular mechanism by which it inhibits solid tumors has not been clarified. To investigate the tumor inhibitory mechanism of vorinostat in cervical cancer, this study used Cell Counting Kit-8, flow cytometry, cell invasion and migration assays and the wound healing assay to evaluate the effects of vorinostat on cervical cancer cell proliferation, apoptosis, cell cycle, migration, and invasion. Real-time quantitative PCR and immunoblotting were used to detect gene and protein expression, respectively, of major histocompatibility class I-related chain A, phosphoinositide 3-kinase, phosphorylated PI3K p55 (Tyr199), and p-Akt (Ser473). The lactate dehydrogenase cytotoxicity assay was used to evaluate the ability of natural killer-92 cells to lyse cervical cancer cells. A xenograft nude mouse model was established to analyze the anti-tumor effect of vorinostat in vivo. Our results showed that vorinostat inhibited the proliferation, migration, and invasion of cervical cancer cells. Vorinostat also induced apoptosis and cell-cycle arrest in the S phase, inhibited PI3K (p110α), p-PI3K p55 (Tyr199), and p-Akt (Ser473) protein expression and upregulated MICA expression in vitro and in vivo, and promoted NK-92 cell-mediated cervical cancer cell lysis. The ability of vorinostat to upregulate MICA expression in cervical cancer cells was related to PI3K/Akt signaling. In brief, vorinostat upregulated MICA through the PI3K/Akt pathway and enhanced the sensitivity of cervical cancer cells to the NK cell-mediated cytolytic reaction. The results of this study demonstrate that vorinostat has anti-solid tumor effects on cervical cancer.
Collapse
Affiliation(s)
- Chenglai Xia
- The College of Pharmacy in Guangzhou Medical University, Guangzhou, 510150, China; Foshan Maternal and Child Health Research Institute, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China.
| | - Zhihong He
- The College of Pharmacy in Guangzhou Medical University, Guangzhou, 510150, China
| | - Yantao Cai
- Department of Dermatology and Rheumatology, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, 528000, China
| | - Shaofen Liang
- The College of Pharmacy in Guangzhou Medical University, Guangzhou, 510150, China; Foshan Maternal and Child Health Research Institute, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, 528000, China
| |
Collapse
|
20
|
Mechanisms underlying FLASH radiotherapy, a novel way to enlarge the differential responses to ionizing radiation between normal and tumor tissues. RADIATION MEDICINE AND PROTECTION 2020. [DOI: 10.1016/j.radmp.2020.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
21
|
Delineation of cell death mechanisms induced by synergistic effects of statins and erlotinib in non-small cell lung cancer cell (NSCLC) lines. Sci Rep 2020; 10:959. [PMID: 31969600 PMCID: PMC6976657 DOI: 10.1038/s41598-020-57707-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022] Open
Abstract
Hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) have been shown to overcome tyrosine kinase inhibitor (TKI) resistance in epithelial growth factor receptor (EGFR) mutated non-small cell lung cancer (NSCLC) cells in vivo and in vitro. However, little is known about the putative induction of non-apoptotic cell death pathways by statins. We investigated the effects of pitavastatin and fluvastatin alone or in combination with erlotinib in three NSCLC cell lines and examined the activation of different cell death pathways. We assessed apoptosis via fluorometric caspase assay and poly (ADP-ribose) polymerase 1 (PARP) cleavage. Furthermore, annexinV/propidium iodide (PI) flow cytometry was performed. Small molecule inhibitors benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (zVAD), necrostatin 1 (Nec1), ferrostatin 1 (Fer1), Ac-Lys-Lys-Norleucinal (Calp1) were used to characterise cell death pathway(s) putatively (co-)activated by pitavastatin/erlotinib co-treatment. Synergism was calculated by additivity and isobolographic analyses. Pitavastatin and fluvastatin induced cell death in EGFR TKI resistant NSCLC cells lines A549, Calu6 and H1993 as shown by caspase 3 activation and PARP cleavage. Co-treatment of cells with pitavastatin and the EGFR TKI erlotinib resulted in synergistically enhanced cytotoxicity compared to pitavastatin monotherapy. Flow cytometry indicated the induction of alternative regulated cell death pathways. However, only co-treatment with mevalonic acid (Mev) or the pan-caspase inhibitor zVAD could restore cell viability. The results show that cytotoxicity mediated by statin/erlotinib co-treatment is synergistic and can overcome erlotinib resistance in K-ras mutated NSCLC and relies only on apoptosis.
Collapse
|
22
|
Wong RA, Luo X, Lu M, An Z, Haas-Kogan DA, Phillips JJ, Shokat KM, Weiss WA, Fan QW. Cooperative Blockade of PKCα and JAK2 Drives Apoptosis in Glioblastoma. Cancer Res 2019; 80:709-718. [PMID: 31806641 DOI: 10.1158/0008-5472.can-18-2808] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/21/2019] [Accepted: 11/27/2019] [Indexed: 01/05/2023]
Abstract
The mTOR signaling is dysregulated prominently in human cancers including glioblastoma, suggesting mTOR as a robust target for therapy. Inhibitors of mTOR have had limited success clinically, however, in part because their mechanism of action is cytostatic rather than cytotoxic. Here, we tested three distinct mTOR kinase inhibitors (TORKi) PP242, KU-0063794, and sapanisertib against glioblastoma cells. All agents similarly decreased proliferation of glioblastoma cells, whereas PP242 uniquely induced apoptosis. Apoptosis induced by PP242 resulted from off-target cooperative inhibition of JAK2 and protein kinase C alpha (PKCα). Induction of apoptosis was also decreased by additional on-target inhibition of mTOR, due to induction of autophagy. As EGFR inhibitors can block PKCα, EGFR inhibitors erlotinib and osimertinib were tested separately in combination with the JAK2 inhibitor AZD1480. Combination therapy induced apoptosis of glioblastoma tumors in both flank and in patient-derived orthotopic xenograft models, providing a preclinical rationale to test analogous combinations in patients. SIGNIFICANCE: These findings identify PKCα and JAK2 as targets that drive apoptosis in glioblastoma, potentially representing a clinically translatable approach for glioblastoma.
Collapse
Affiliation(s)
- Robyn A Wong
- Department of Neurology, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Xujun Luo
- Department of Neurology, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mimi Lu
- Department of Neurology, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Zhenyi An
- Department of Neurology, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, Harvard Medical School and Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Boston, Massachusetts
| | - Joanna J Phillips
- Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco, California
| | - Kevan M Shokat
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California
| | - William A Weiss
- Department of Neurology, University of California, San Francisco, California. .,Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Pediatrics, University of California, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco, California
| | - Qi Wen Fan
- Department of Neurology, University of California, San Francisco, California. .,Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| |
Collapse
|
23
|
Veeramani Kandan P, Dhineshkumar E, Karthikeyan R, Anbuselvan C, Maqbool I, Kanimozhi G, Arul Prakasam B, Rajendra Prasad N. Isolation and characterization of opuntiol from Opuntia Ficus indica (L. Mill) and its antiproliferative effect in KB oral carcinoma cells. Nat Prod Res 2019; 35:3146-3150. [PMID: 31711321 DOI: 10.1080/14786419.2019.1690484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In this study, we isolated and characterized a novel bioactive flavonol from the cactus pad of Opuntia Ficus indica Indica (L. Mill) (OFI) by chromatography techniques. The isolated compound was characterized by FT-IR, 1H and 13C NMR spectroscopy. Single-crystal XRD results illustrate that the obtained flavonol was opuntiol (6-hydroxymethyl-4-methoxy-2H-pyran-2-one) and it was found to be near planar except for the H atoms of the methylene and methyl groups. The crystal packing was stabilized by C-H….O and O-H….O intermolecular hydrogen bonds. The isolated opuntiol significantly inhibited KB cells proliferation and its IC50 value was found to be 30 µM. Further, we noticed that opuntiol significantly induced ROS generation and subsequently altered MMP in KB cells. Western blot analysis and morphological observations by fluorescence microscope indicate the apoptotic inducing potential of opuntiol in KB cells.
Collapse
Affiliation(s)
- Ponniresan Veeramani Kandan
- Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
| | - Ezhumalai Dhineshkumar
- Faculty of Science, Department of Chemistry, Annamalai University, Annamalainagar, India
| | - Ramasamy Karthikeyan
- Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
| | | | - Illiyas Maqbool
- Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
| | - Govindasamy Kanimozhi
- Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India.,Department of Biochemistry, Dharumapurm Gnanambigai Government Arts College for Women, Mayiladuthurai, India
| | | | - Nagarajan Rajendra Prasad
- Faculty of Science, Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
| |
Collapse
|
24
|
Tan VP, Smith JM, Tu M, Yu JD, Ding EY, Miyamoto S. Dissociation of mitochondrial HK-II elicits mitophagy and confers cardioprotection against ischemia. Cell Death Dis 2019; 10:730. [PMID: 31570704 PMCID: PMC6768853 DOI: 10.1038/s41419-019-1965-7] [Citation(s) in RCA: 23] [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: 04/10/2019] [Revised: 07/11/2019] [Accepted: 09/09/2019] [Indexed: 12/23/2022]
Abstract
Preservation of mitochondrial integrity is critical for maintaining cellular homeostasis. Mitophagy is a mitochondria-specific type of autophagy which eliminates damaged mitochondria thereby contributing to mitochondrial quality control. Depolarization of the mitochondrial membrane potential is an established mechanism for inducing mitophagy, mediated through PINK1 stabilization and Parkin recruitment to mitochondria. Hexokinase-II (HK-II) which catalyzes the first step in glucose metabolism, also functions as a signaling molecule to regulate cell survival, and a significant fraction of cellular HK-II is associated with mitochondria (mitoHK-II). We demonstrate here that pharmacological interventions and adenoviral expression of a mitoHK-II dissociating peptide which reduce mitoHK-II levels lead to robust increases in mitochondrial Parkin and ubiquitination of mitochondrial proteins in cardiomyocytes and in a human glioblastoma cell line 1321N1, independent of mitochondrial membrane depolarization or PINK1 accumulation. MitoHK-II dissociation-induced mitophagy was demonstrated using Mito-Keima in cardiomyocytes and in 1321N1 cells. Subjecting cardiomyocytes or the in vivo heart to ischemia leads to modest dissociation of mitoHK-II. This response is potentiated by expression of the mitoHK-II dissociating peptide, which increases Parkin recruitment to mitochondria and, importantly, provides cardioprotection against ischemic stress. These results suggest that mitoHK-II dissociation is a physiologically relevant cellular event that is induced by ischemic stress, the enhancement of which protects against ischemic damage. The mechanism which underlies the effects of mitoHK-II dissociation can be attributed to the ability of Bcl2-associated athanogene 5 (BAG5), an inhibitor of Parkin, to localize to mitochondria and form a molecular complex with HK-II. Overexpression of BAG5 attenuates while knockdown of BAG5 sensitizes the effect of mitoHK-II dissociation on mitophagy. We suggest that HK-II, a glycolytic molecule, can function as a sensor for metabolic derangements at mitochondria to trigger mitophagy, and modulating the intracellular localization of HK-II could be a novel way of regulating mitophagy to prevent cell death induced by ischemic stress.
Collapse
Affiliation(s)
- Valerie P Tan
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Jeffrey M Smith
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Michelle Tu
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Justin D Yu
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Eric Y Ding
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Shigeki Miyamoto
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA.
| |
Collapse
|
25
|
Zhang X, Ye Y, Fu M, Zheng B, Qiu Q, Huang Z. Implication of viral microRNAs in the genesis and diagnosis of Epstein-Barr virus-associated tumors. Oncol Lett 2019; 18:3433-3442. [PMID: 31516561 DOI: 10.3892/ol.2019.10713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 06/26/2019] [Indexed: 12/13/2022] Open
Abstract
The Epstein-Barr virus (EBV) is tightly associated with a variety of human tumors, including Burkitt lymphoma and acquired immune deficiency syndrome-related lymphoma of B-cell origin, as well as nasopharyngeal carcinoma and gastric cancer of epithelial origin. The virus latently infects the host cells and expresses proteins and non-coding RNAs to achieve malignancy. MicroRNAs (miRNAs or miRs) are small RNAs consisting of 19-25 nucleotides, which directly bind to the 3'-untranslated region of mRNAs to promote degradation and inhibit translation of mRNAs. EBV-encoded miRs are generated from two regions of the viral genome, within the apoptosis regulator BHRF1 gene locus and near the BamHI A region in a latency type-dependent manner. In addition, EBV-encoded miRs epigenetically regulate the expression of molecules that are effectors of the cell cycle progression, migration, apoptosis and innate immunity, serving a vital role in supporting viral replication and occurrence of EBV-associated tumors. The feasibility of using such miRs as biomarkers for the diagnosis and prognosis of EBV-associated tumors is currently under investigation.
Collapse
Affiliation(s)
- Xiangning Zhang
- Department of Pathophysiology, Chinese-American Collaborative Cancer Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Yudong Ye
- Department of Otolaryngology, Dongguan City People's Hospital, Dongguan, Guangdong 523059, P.R. China
| | - Ming Fu
- Department of Otolaryngology, Dongguan City People's Hospital, Dongguan, Guangdong 523059, P.R. China
| | - Biying Zheng
- Department of Clinical Microbiology, College of Laboratory Medicine, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Qianhui Qiu
- Department of Otolaryngology, Pearl River Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Zunnan Huang
- Department of Pathophysiology, Chinese-American Collaborative Cancer Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
26
|
Zuo A, Zhao P, Zheng Y, Hua H, Wang X. Tripterine inhibits proliferation, migration and invasion of breast cancer MDA-MB-231 cells by up-regulating microRNA-15a. Biol Chem 2019; 400:1069-1078. [PMID: 30913029 DOI: 10.1515/hsz-2018-0469] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/19/2019] [Indexed: 01/17/2023]
Abstract
Breast cancer is the most commonly diagnosed cancer in women worldwide. Tripterine is an important active component isolated from Triperygium wilfordii Hook F. This study investigated the effects of tripterine on breast cancer cell proliferation, migration, invasion and apoptosis, as well as microRNA-15a (miR-15a) expression. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was performed to measure the expression of miR-15a. Cell transfection was conducted to change the expression of miR-15a. Viability, proliferation, migration, invasion and apoptosis of MDA-MB-231 cells were assessed using the cell counting kit-8 (CCK-8) assay, BrdU incorporation assay, Annexin V-FITC/PI apoptosis detection kit and two-chamber Transwell assay, respectively. Expression of key factors involving in cell proliferation, migration, invasion and apoptosis, as well as the PI3K/AKT and JNK pathways, were evaluated using Western blotting. We found that tripterine inhibited MDA-MB-231 cell viability, proliferation, migration and invasion, but induced cell apoptosis. Moreover, tripterine up-regulated the expression of miR-15a in a concentration-dependent manner and miR-15a participated in the effects of tripterine on MDA-MB-231 cell proliferation, migration, invasion and apoptosis. In addition, tripterine inactivated PI3K/AKT and JNK pathways in MDA-MB-231 cells by up-regulating miR-15a. In conclusion, tripterine inhibited proliferation, migration and invasion of breast cancer MDA-MB-231 cells by up-regulating miR-15a and inactivating PI3K/AKT and JNK pathways.
Collapse
Affiliation(s)
- Anjun Zuo
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Peng Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yu Zheng
- Department of General Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Hui Hua
- Department of Thyroid Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xingang Wang
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao 266000, China
| |
Collapse
|
27
|
Ethiraj P, Sambandam Y, Hathaway-Schrader JD, Haque A, Novince CM, Reddy SV. RANKL triggers resistance to TRAIL-induced cell death in oral squamous cell carcinoma. J Cell Physiol 2019; 235:1663-1673. [PMID: 31309556 DOI: 10.1002/jcp.29086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022]
Abstract
Oral squamous cell carcinoma (OSCC) occurs as a malignancy of the oral cavity. RANK ligand (RANKL) is essential for osteoclast formation/bone resorption. Recently, we showed autoregulation of receptor activator of nuclear factor-κB ligand (RANKL) stimulates OSCC cell proliferation. OSCC cells show resistance to tumor necrosis factor related apoptosis inducing ligand (TRAIL) treatment. Therefore, we hypothesize that RANKL promotes resistance for TRAIL induction of OSCC apoptotic cell death. In this study, SCC14A and SCC74A cells cultured with TRAIL revealed high-level expression of RANKL which increased resistance to TRAIL inhibition of tumor cell proliferation. RANKL stimulation inhibited terminal deoxynucleotidyl transferase dUTP nick end labeling positive staining in TRAIL-treated cells. CRISPR/Cas-9 knockout of RANKL (RANKL-KO) increased caspase-9, caspase-3 activity and cytochrome c release in OSCC cells. RANKL inhibited proapoptotic proteins BAD and BAX expression. TRAIL treatment suppressed the SQSTM1/p62 and RANKL restored the expression. Interestingly, RANKL alone significantly increased proteasome activity. RANKL-KO in OSCC cells inhibited autophagic activity as evidenced by decreased light chain 3B-II and beclin-1 expression. Thus, RANKL stimulation of OSCC tumor cells triggered resistance for TRAIL-induced OSCC cell death. Taken together, blockade of RANKL may inhibit OSCC tumor progression and enhance the potential of TRAIL induced OSCC tumor cell apoptosis.
Collapse
Affiliation(s)
- Purushoth Ethiraj
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Yuvaraj Sambandam
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Jessica D Hathaway-Schrader
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Chad M Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Sakamuri V Reddy
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
28
|
Melittin Inducing the Apoptosis of Renal Tubule Epithelial Cells through Upregulation of Bax/Bcl-2 Expression and Activation of TNF- α Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9450368. [PMID: 31772938 PMCID: PMC6854966 DOI: 10.1155/2019/9450368] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023]
Abstract
Background Acute kidney injury (AKI) caused by bee stings is common, with characteristics of acute onset, severe illness, and high mortality. Melittin, a major component of bee venom, has been considered to play a key role in bee sting related AKI. This study aims to illustrate whether melittin could lead to apoptosis of renal tubular epithelial cells (RTECs) and to investigate its mechanism. Methods In vivo, 45 mice were randomly divided into the melittin group (n=30, injected with melittin into the tail vein according to the total dose of 4.0 ug/g weight) and the control group (n=15, injected with the same volume of saline into the tail vein). In vitro, human RTECs (HK-2) were cultured and treated with melittin (2ug/ml or 4ug/ml) and TNF-α (10ng/ml). Biochemical analysis, HE stains, and electron microscope were performed to evaluate renal function and pathological changes. TUNEL stains and flow cytometry were performed to detect apoptosis. Real-time PCR was performed to detect mRNA levels of Bax, Bcl-2, and TNF-α. Simple western assay and immunohistochemical (IH) and immunofluorescent (IF) stains were performed for protein detection. Results Melittin successfully induced AKI in mice. Compared with the control group, obvious injury and apoptosis of RTECs were observed in the melittin group; the mRNA and protein expressions of Bax were significantly increased, while the expression of Bcl-2 was significantly decreased. The serum TNF-αlevel in melittin group was significantly higher than that in control group. In vitro, the results confirmed that melittin can cause HK-2 cells apoptosis. The trends of expression of Bax and Bcl-2 were consistent with the results in vivo. The levels of TNF-α mRNA and protein by PCR and Western blot were significantly higher in melittin group than those in control group. Conclusion Melittin can lead to the apoptosis of RTECs, which may be mediated by upregulating the expression of Bax/Bcl-2 and activating the TNF-α signaling pathway.
Collapse
|
29
|
Slobodnyuk K, Radic N, Ivanova S, Llado A, Trempolec N, Zorzano A, Nebreda AR. Autophagy-induced senescence is regulated by p38α signaling. Cell Death Dis 2019; 10:376. [PMID: 31092814 PMCID: PMC6520338 DOI: 10.1038/s41419-019-1607-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/14/2019] [Accepted: 04/23/2019] [Indexed: 12/30/2022]
Abstract
Apoptosis and senescence are two mutually exclusive cell fate programs that can be activated by stress. The factors that instruct cells to enter into senescence or apoptosis are not fully understood, but both programs can be regulated by the stress kinase p38α. Using an inducible system that specifically activates this pathway, we show that sustained p38α activation suffices to trigger massive autophagosome formation and to enhance the basal autophagic flux. This requires the concurrent effect of increased mitochondrial reactive oxygen species production and the phosphorylation of the ULK1 kinase on Ser-555 by p38α. Moreover, we demonstrate that macroautophagy induction by p38α signaling determines that cancer cells preferentially enter senescence instead of undergoing apoptosis. In agreement with these results, we present evidence that the induction of autophagy by p38α protects cancer cells from chemotherapy-induced apoptosis by promoting senescence. Our results identify a new mechanism of p38α-regulated basal autophagy that controls the fate of cancer cells in response to stress.
Collapse
Affiliation(s)
- Konstantin Slobodnyuk
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Nevenka Radic
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Saška Ivanova
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain.,Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Faculty of Biology, 08028, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain
| | - Anna Llado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Natalia Trempolec
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain.,Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Faculty of Biology, 08028, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain. .,ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|
30
|
Fang S, Agostinis P, Salven P, Garg AD. Decoding cancer cell death-driven immune cell recruitment: An in vivo method for site-of-vaccination analyses. Methods Enzymol 2019; 636:185-207. [PMID: 32178819 DOI: 10.1016/bs.mie.2019.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anticancer vaccines have recently received renewed attention for immunotherapy of at least a subset of cancer-types. Such vaccines mostly involve either killed cancer or tumor cells alone, or combinations thereof with specific (co-incubated) innate immune cells. In recent years, the immunogenic characteristics of the dead or dying cancer cells have emerged as decisive factors behind the success of anticancer vaccines. This has amplified the importance of accounting for immunology of cell death while preparing anticancer vaccines. This, in turn, has increased the emphasis on the immune reactions at the site-of-vaccination since the therapeutic efficacy of the killed cancer/tumor cell vaccines is contingent upon the nature and characteristics of these reactions at the site-of-injection. In this article, we present a systematic methodology that exploits the murine ear pinna model to study differential immune cell recruitment by dead/dying cancer cells injected in vivo, thereby modeling the site-of-injection relevant for anticancer vaccines.
Collapse
Affiliation(s)
- Shentong Fang
- Wihuri Research Institute and Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Patrizia Agostinis
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium; Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Petri Salven
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Abhishek D Garg
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium.
| |
Collapse
|
31
|
Porro C, Cianciulli A, Trotta T, Lofrumento DD, Calvello R, Panaro MA. Formyl-methionyl-leucyl-phenylalanine Induces Apoptosis in Murine Neurons: Evidence for NO-Dependent Caspase-9 Activation. BIOLOGY 2019; 8:biology8010004. [PMID: 30621183 PMCID: PMC6466069 DOI: 10.3390/biology8010004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022]
Abstract
Formyl-methionyl-leucyl-phenylalanine (fMLP) may be present in the brain in the course of some infectious diseases of the central nervous system (CNS), although little is known about its role. This investigation was performed to study the effect of fMLP on neuron apoptosis. Our results showed that fMLP treatment of primary cultures of neurons was able to induce morphological features of apoptosis in cell cultures, as well as activation of the intrinsic apoptotic pathway, through the upregulation of caspase-9 and caspase-3. This effect contextually occurred to the pro-apoptotic protein Bax activation and cytochrome c release. The in vitro fMLP treatment was also able to induce, in a dose-dependent manner, the increase of inducible nitric oxide synthase (iNOS) expression accompanied by an up-regulation of nitric oxide (NO) release. When neuron cultures were pre-treated with 1400 W, a selective iNOS inhibitor, all of the apoptotic features were significantly reversed. Overall, these results demonstrated that fMLP treatment of neurons leads to intrinsic apoptosis activation, through iNOS expression regulation, suggesting a role for fMLP in CNS neurodegenerative processes.
Collapse
Affiliation(s)
- Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy.
| | - Antonia Cianciulli
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona, 4, 70126 Bari, Italy.
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy.
| | - Dario Domenico Lofrumento
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento, 73100 Lecce, Italy.
| | - Rosa Calvello
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona, 4, 70126 Bari, Italy.
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona, 4, 70126 Bari, Italy.
| |
Collapse
|
32
|
Brosseau C, Durand M, Colas L, Durand E, Foureau A, Cheminant MA, Bouchaud G, Castan L, Klein M, Magnan A, Brouard S. CD9 + Regulatory B Cells Induce T Cell Apoptosis via IL-10 and Are Reduced in Severe Asthmatic Patients. Front Immunol 2018; 9:3034. [PMID: 30622536 PMCID: PMC6308143 DOI: 10.3389/fimmu.2018.03034] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/07/2018] [Indexed: 12/28/2022] Open
Abstract
CD9 was recently identified as a marker of murine IL-10-competent regulatory B cells. Functional impairments or defects in CD9+ IL-10-secreting regulatory B cells are associated with enhanced asthma-like inflammation and airway hyperresponsiveness. In mouse models, all asthma-related features can be abrogated by CD9+ B cell adoptive transfer. We aimed herein to decipher the profiles, features, and molecular mechanisms of the regulatory properties of CD9+ B cells in human and mouse. The profile of CD9+ B cells was analyzed using blood from severe asthmatic patients and normal and asthmatic mice by flow cytometry. The regulatory effects of mouse CD9+ B cells on effector T cell death, cell cycle arrest, apoptosis, and mitochondrial depolarization were determined using yellow dye, propidium iodide, Annexin V, and JC-1 staining. MAPK phosphorylation was analyzed by western blotting. Patients with severe asthma and asthmatic mice both harbored less CD19+CD9+ B cells, although these cells displayed no defect in their capacity to induce T cell apoptosis. Molecular mechanisms of regulation of CD9+ B cells characterized in mouse showed that they induced effector T cell cycle arrest in sub G0/G1, leading to apoptosis in an IL-10-dependent manner. This process occurred through MAPK phosphorylation and activation of both the intrinsic and extrinsic pathways. This study characterizes the molecular mechanisms underlying the regulation of CD9+ B cells to induce effector T cell apoptosis in mice and humans via IL-10 secretion. Defects in CD9+ B cells in blood from patients with severe asthma reveal new insights into the lack of regulation of inflammation in these patients.
Collapse
Affiliation(s)
- Carole Brosseau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France.,Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France.,Institut du Thorax, CHU de Nantes, Nantes, France
| | - Maxim Durand
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France.,Faculté de Médecine, Université de Nantes, Nantes, France
| | - Luc Colas
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France.,Institut du Thorax, CHU de Nantes, Nantes, France.,Faculté de Médecine, Université de Nantes, Nantes, France
| | - Eugénie Durand
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Aurore Foureau
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France.,Institut du Thorax, CHU de Nantes, Nantes, France
| | - Marie-Aude Cheminant
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France
| | - Gregory Bouchaud
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France.,INRA Centre Angers-Nantes, Nantes, France
| | - Laure Castan
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France.,Faculté de Médecine, Université de Nantes, Nantes, France.,INRA Centre Angers-Nantes, Nantes, France
| | - Martin Klein
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France.,Faculté de Médecine, Université de Nantes, Nantes, France
| | - Antoine Magnan
- Institut du thorax, Inserm UMR 1087, CNRS UMR 6291, Université de Nantes, Nantes, France.,Institut du Thorax, CHU de Nantes, Nantes, France
| | - Sophie Brouard
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France.,Centre d'Investigation Clinique (CIC) Biothérapie, CHU Nantes, Nantes, France
| |
Collapse
|
33
|
Gong YN, Crawford JC, Heckmann BL, Green DR. To the edge of cell death and back. FEBS J 2018; 286:430-440. [PMID: 30506628 DOI: 10.1111/febs.14714] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/28/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022]
Abstract
Programmed cell death plays a central role in maintaining homeostasis. Various studies have demonstrated that programmed cell death is not a one-way street; cells can survive even when the core cell death processes are underway. Cell death initiation, prevention, and recovery function in a coordinated fashion to establish and maintain a homeostatic environment. In this review, we discuss how dying cells can be rescued from death's grip and the subsequent physiological consequences. We suggest a fundamental question to be answered-at least at the single cell level is, can we predict if a certain cell is more or less likely to survive or die? And importantly, what physiological and pathological consequences, as well as therapeutic approaches can we delineate from this ability to predict cell death versus survival.
Collapse
Affiliation(s)
- Yi-Nan Gong
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
34
|
He F, Ma N, Midorikawa K, Hiraku Y, Oikawa S, Zhang Z, Huang G, Takeuchi K, Murata M. Taurine exhibits an apoptosis-inducing effect on human nasopharyngeal carcinoma cells through PTEN/Akt pathways in vitro. Amino Acids 2018; 50:1749-1758. [PMID: 30225664 DOI: 10.1007/s00726-018-2651-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is a distinctive type of head and neck malignancy with a high incidence in southern China. Previous studies have confirmed that taurine shows an anti-cancer effect on a variety of human tumors by inhibiting cell proliferation and inducing apoptosis. However, the underlying molecular mechanism of its anti-cancer effect on NPC is not well understood. To clarify these anti-cancer mechanisms, we performed cell viability and colony formation assays. Apoptotic cells were quantified by flow cytometry. The expression levels of apoptosis-related proteins were evaluated by Western blot. The results showed that taurine markedly inhibited cell proliferation in NPC cells, but only slightly in an immortalized normal nasopharyngeal cell line. Taurine suppressed colony formation and induced apoptosis of NPC cell lines in a dose-dependent manner. Furthermore, taurine increased the active form of caspase-9/3 in a dose-dependent manner. Taurine down-regulated the anti-apoptotic protein Bcl-xL and up-regulated the pro-apoptotic protein Bax and GRP78, a major endoplasmic reticulum (ER) chaperone. These results suggest the involvement of mitochondrial and ER stress signaling in apoptosis. In addition, taurine increased the levels of PTEN (phosphatase and tensin homolog deleted on chromosome 10) and p53, and reduced phosphorylated Akt (protein kinase B). In conclusion, taurine may inhibit cell proliferation and induce apoptosis in NPC through PTEN activation with concomitant Akt inactivation.
Collapse
Affiliation(s)
- Feng He
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Otolaryngology Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ning Ma
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, Japan
| | - Kaoru Midorikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yusuke Hiraku
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Shinji Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Zhe Zhang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Guangwu Huang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Kazuhiko Takeuchi
- Department of Otolaryngology Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu, Japan.
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan.
| |
Collapse
|
35
|
Deng W, Han W, Fan T, Wang X, Cheng Z, Wan B, Chen J. Scutellarin inhibits human renal cancer cell proliferation and migration via upregulation of PTEN. Biomed Pharmacother 2018; 107:1505-1513. [PMID: 30257368 DOI: 10.1016/j.biopha.2018.08.127] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/18/2018] [Accepted: 08/24/2018] [Indexed: 10/28/2022] Open
Abstract
BACKGROUND Scutellarin is a naturally flavone glycoside that has been shown to exhibit anti-proliferative and anti-apoptotic activities among various human malignancies. However, the anti-cancer effect of Scutellarin in Renal cell carcinoma (RCC) and the underlying mechanism remains unclear. METHODS AND MATERIALS RCC cell lines ACHN and 786-O were treated with different concentrations (0-210 μM) of Scutellarin in vitro. Cell viability and proliferation were investigated by MTT and colony formation assays. Cell invasion and migration were detected by Transwell assays. Cell apoptosis and cell cycle distribution was measured by flow cytometry. Western blot was used to investigate the expression levels of crucial proteins. Xenograft tumor model was established to evaluate tumor growth in vivo. RESULTS Scutellarin significantly inhibited RCC cell proliferation in a dose- and time- dependent manner. Treatment of RCC cells with Scutellarin (30, 60, and 90 μM) markedly induced apoptosis and cell cycle arrested at G0/G1 phase in a concentration-dependent characteristic. Cell invasion and migration capacities of RCC cells were also dose-dependently suppressed by Scutellarin treatment. Western blot assays revealed that the crucial proteins including cyclin D1, CDK2, Bcl2, MMP-2, and MMP-9 were significantly reduced while Bax, cleaved caspase 3 and p21 were increased by Scutellarin in RCC cells. In vivo assay indicated that Scutellarin possessed anti-cancer effect on xenograft without triggering toxic effect. Mechanically, Scutellarin dramatically increased the protein level of phosphatase and tensin homologue (PTEN) and inhibited the activity of P13K/AKT/mTOR signaling. Ectopic expression of PTEN enhanced the inhibitory effect of Scutellarin on RCC proliferation while knockdown of PTEN abrogated it through regulating its downstream P13K/AKT/mTOR signaling pathway. CONCLUSION Scutellarin inhibited RCC cell proliferation and invasion partially by enhancing the expression of PTEN through inhibition of P13K/AKT/mTOR pathway, suggesting that Scutellarin might serve as a potential therapeutic agent in RCC treatment.
Collapse
Affiliation(s)
- Wenting Deng
- College of Pharmacy, Xi'an Medical University, No. 1 Xinwang Road of Weiyang District, 710021, Xi'an, Shaanxi, China.
| | - Wei Han
- Department of Medical Equipment, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, 710068, Xi'an, Shaanxi, China
| | - Tao Fan
- College of Pharmacy, Xi'an Medical University, No. 1 Xinwang Road of Weiyang District, 710021, Xi'an, Shaanxi, China
| | - Xiaoku Wang
- College of Pharmacy, Xi'an Medical University, No. 1 Xinwang Road of Weiyang District, 710021, Xi'an, Shaanxi, China
| | - Zhao Cheng
- College of Pharmacy, Xi'an Medical University, No. 1 Xinwang Road of Weiyang District, 710021, Xi'an, Shaanxi, China
| | - Bo Wan
- College of Pharmacy, Xi'an Medical University, No. 1 Xinwang Road of Weiyang District, 710021, Xi'an, Shaanxi, China
| | - Jinlian Chen
- College of Pharmacy, Xi'an Medical University, No. 1 Xinwang Road of Weiyang District, 710021, Xi'an, Shaanxi, China
| |
Collapse
|
36
|
Protein phosphatase 2A activation mechanism contributes to JS-K induced caspase-dependent apoptosis in human hepatocellular carcinoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:142. [PMID: 29986744 PMCID: PMC6038275 DOI: 10.1186/s13046-018-0823-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022]
Abstract
Background JS-K is a nitric oxide (NO) donor and could generate intracellularly high levels of NO. The study explores PP2A as a tumor suppressor is a major determinant mediating JS-K-caused apoptosis in human hepatocellular carcinoma (HCC) cells. Methods The human HCC cell lines (PLC5, Huh-7, Bel-7402, SMMC-7721 and HepG2) were used to assess effects of JS-K on cell viability, apoptosis induction and PP2A activation. Effects of JS-K on cell morphology, mitochondrial membrane potential, apoptosis and NO levels were determined in HCC cells expressing PP2A. Simultaneously, the expression of PP2A family including PP2A-A(α/β), PP2A-B55, PP2A-C(α/β) and the substrates of PP2A, such as β-catenin, c-Myc and p-Bcl-2 (Ser70) were detected in sensitive HCC cells. Furthermore, the role of NO in mediating the expression of PP2A was further validated with Z-VAD-FMK (a caspase inhibitor), Carboxy-PTIO (a NO scavenger), okadaic acid (OA, a PP2A inhibitor) and FTY720 (a PP2A agonist) in JS-K treated cells. In addition, the genetic manuplation of PP2A including overexpression and knockdown have been also performed in JS-K treated cells. Moreover, the rat model of primary hepatic carcinoma was established with diethylnitrosamine for 16 weeks to verify the anti-tumor effects of JS-K in vivo. Immunohistochemical and Western blot analysis were used to determine the expression of proteins in rat primary hepatic carcinoma tissues. Results JS-K significantly inhibited cell proliferation, increased apoptosis rate and activated PP2A activity in five HCC cells viability, especially SMMC7721 and HepG2 cells. It was characterized by loss of mitochondrial membrane potential, significant externalization of phosphatidylserine, nuclear morphological changes. Moreover, JS-K enhanced Bax-to-Bcl-2 ratio, released cytochrome c (Cyt c) from mitochondria, activated cleaved-caspase-9/3 and the cleavage of PARP, and decreased the expression of X-linked inhibitor of apoptosis protein (XIAP). Both Z-VAD-FMK and Carboxy-PTIO suppressed the activation of cleaved-caspase-9/3 and of cleaved-PARP in JS-K-treated sensitive HCC cells. Simultaneously, JS-K treatment could lead to the activation of protein phosphatase 2A-C (PP2A-C) but not PP2A-A and PP2A-B55, which subsequently inactivated and dephosphorylated the PP2A substrates including β-catenin, c-Myc, and p-Bcl-2 (Ser70). However, silencing PP2A-C could abolish both the activation of PP2A-C and down-regulation of β-catenin, c-Myc and p-Bcl-2 (Ser70) in sensitive HCC cells. Conversely, PP2A overexpression could enhance the effects of JS-K on activation of PP2A and down-regulation of β-catenin, c-Myc and p-Bcl-2 (Ser70). In addition, adding okadaic acid (OA), a PP2A inhibitor, abolished the effects of JS-K on apoptosis induction, PP2A activation and the substrates of PP2A dephosphorylation; FTY720, a PP2A agonist, enhanced the effects of JS-K including apoptosis induction, PP2A activation and the substrates of PP2A dephosphorylation. The mice exhibited a lower number and smaller tumor nodules in response to JS-K-treated group. A marked increase in the number of hepatocytes with PCNA-positive nuclei (proliferating cells) was evident in DEN group and tended to decrease with JS-K treatment. Furthermore, JS-K treatment could induce PP2A activation and the substrates of PP2A inactivation such as β-catenin, c-Myc and p-Bcl-2(Ser70) in DEN-induced hepatocarcinogenesis. Conclusions High levels of NO released from JS-K induces a caspase-dependent apoptosis through PP2A activation.
Collapse
|
37
|
Gregory CD, Paterson M. An apoptosis-driven 'onco-regenerative niche': roles of tumour-associated macrophages and extracellular vesicles. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2017.0003. [PMID: 29158317 PMCID: PMC5717442 DOI: 10.1098/rstb.2017.0003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2017] [Indexed: 12/31/2022] Open
Abstract
The cell-death programme, apoptosis, is well established as a tumour suppressor mechanism. Paradoxically, high levels of apoptosis in tumours are closely coupled with poor prognosis. Indeed, where it has been studied, cell loss is a striking feature of high-grade cancers, illustrating the importance of considering malignant disease as an imbalance between cell gain and cell loss that favours cell gain rather than as a unidirectional disorder of cell gain alone. In addition to orchestrating cell loss, apoptosis can signal regenerative responses—for example compensatory proliferation—in neighbouring cells. Accumulating evidence suggests that normal tissue repair and regenerative processes are hijacked in the malignant tissue microenvironment such that cancer may be likened to a ‘wound that fails to stop repairing’. We have proposed that a critical requirement for the successful growth, progression and re-growth of malignant tumours is a complex milieu, conceptually termed the ‘onco-regenerative niche’, which is composed, in addition to transformed neoplastic cells, of a network of normal cells and factors activated as if in tissue repair and regeneration. Our work is based around the hypothesis that tumour cell apoptosis, macrophage activation and endothelial activation are key, interlinked elements of the onco-regenerative niche and that apoptotic tumour cell–derived extracellular vesicles provide critical intercellular communication vehicles of the niche. In aggressive B-cell lymphoma, tumour cell apoptosis promotes both angiogenesis and the accumulation of pro-tumour macrophages in the lymphoma microenvironment. Furthermore, apoptotic lymphoma-derived extracellular vesicles have potent pro-tumour potential. These findings have important implications for the roles of apoptosis in regulation of malignant diseases and for the efficacy of apoptosis-inducing anti-cancer therapies. This article is part of the discussion meeting issue ‘Extracellular vesicles and the tumour microenvironment’.
Collapse
Affiliation(s)
- Christopher D Gregory
- MRC Centre for Inflammation Research, University of Edinburgh College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Margaret Paterson
- MRC Centre for Inflammation Research, University of Edinburgh College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
38
|
Liu L, Huang Z, Chen J, Wang J, Wang S. Protein phosphatase 2A mediates JS-K-induced apoptosis by affecting Bcl-2 family proteins in human hepatocellular carcinoma HepG2 cells. J Cell Biochem 2018; 119:6633-6643. [PMID: 29693750 DOI: 10.1002/jcb.26845] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 03/09/2018] [Indexed: 12/18/2022]
Abstract
Protein phosphatase 2A (PP2A) is an important enzyme within various signal transduction pathways. The present study was investigated PP2A mediates JS-K-induced apoptosis by affecting Bcl-2 family protein. JS-K showed diverse inhibitory effects in five HCC cell lines, especially HepG2 cells. JS-K caused a dose- and time-dependent reduction in cell viability and increased in levels of LDH release. Meanwhile, JS-K- induced apoptosis was characterized by mitochondrial membrane potential reduction, Hoechst 33342+ /PI+ dual staining, release of cytochrome c (Cyt c), and activation of cleaved caspase-9/3. Moreover, JS-K-treatment could lead to the activation of protein phosphatase 2A-C (PP2A-C), decrease of anti-apoptotic Bcl-2 family-protein expression including p-Bcl-2 (Ser70), Bcl-2, Bcl-xL, and Mcl-1 as well as the increase of pro-apoptosis Bcl-2 family-protein including Bim, Bad, Bax, and Bak. Furthermore, JS-K caused a marked increase of intracellular NO levels while pre-treatment with Carboxy-PTIO (a NO scavenger) reduced the cytotoxicity effects and the apoptosis rate. Meanwhile, pre-treatment with Carboxy-PTIO attenuated the JS-K-induced up-regulation of PP2A, Cyt c, and cleaved-caspase-9/3 activation. The silencing PP2A-C by siRNA could abolish the activation of PP2A-C, down-regulation of anti-apoptotic Bcl-2 family-protein (p-Bcl-2, Bcl-2, Bcl-xL, and Mcl-1), increase of pro-apoptosis Bcl-2 family-protein (Bim, Bad, Bax, and Bak) and apoptotic-related protein (Cyt c, cleaved caspase-9/3) that were caused by JS-K in HepG2 cells. In addition, pre-treatment with OA (a PP2A inhibitor) also attenuated the above effects induced by JS-K. In summary, NO release from JS-K induces apoptosis through PP2A activation, which contributed to the regulation of Bcl-2 family proteins.
Collapse
Affiliation(s)
- Ling Liu
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Zile Huang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Jingjing Chen
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Jiangang Wang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Shuying Wang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
39
|
Nagaya T, Okuyama S, Ogata F, Maruoka Y, Knapp DW, Karagiannis SN, Fazekas-Singer J, Choyke PL, LeBlanc AK, Jensen-Jarolim E, Kobayashi H. Near infrared photoimmunotherapy targeting bladder cancer with a canine anti-epidermal growth factor receptor (EGFR) antibody. Oncotarget 2018; 9:19026-19038. [PMID: 29721181 PMCID: PMC5922375 DOI: 10.18632/oncotarget.24876] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/06/2018] [Indexed: 12/23/2022] Open
Abstract
Anti-epidermal growth factor receptor (EGFR) antibody therapy is used in EGFR expressing cancers including lung, colon, head and neck, and bladder cancers, however results have been modest. Near infrared photoimmunotherapy (NIR-PIT) is a highly selective tumor treatment that employs an antibody-photo-absorber conjugate which is activated by NIR light. NIR-PIT is in clinical trials in patients with recurrent head and neck cancers using cetuximab-IR700 as the conjugate. However, its use has otherwise been restricted to mouse models. This is an effort to explore larger animal models with NIR-PIT. We describe the use of a recombinant canine anti-EGFR monoclonal antibody (mAb), can225IgG, conjugated to the photo-absorber, IR700DX, in three EGFR expressing canine transitional cell carcinoma (TCC) cell lines as a prelude to possible canine clinical studies. Can225-IR700 conjugate showed specific binding and cell-specific killing after NIR-PIT on EGFR expressing cells in vitro. In the in vivo study, can225-IR700 conjugate demonstrated accumulation of the fluorescent conjugate with high tumor-to-background ratio. Tumor-bearing mice were separated into 4 groups: (1) no treatment; (2) 100 µg of can225-IR700 i.v. only; (3) NIR light exposure only; (4) 100 µg of can225-IR700 i.v., NIR light exposure. Tumor growth was significantly inhibited by NIR-PIT treatment compared with the other groups (p < 0.001), and significantly prolonged survival was achieved (p < 0.001 vs. other groups) in the treatment groups. In conclusion, NIR-PIT with can225-IR700 is a promising treatment for canine EGFR-expressing cancers, including invasive transitional cell carcinoma in pet dogs, that could provide a pathway to translation to humans.
Collapse
Affiliation(s)
- Tadanobu Nagaya
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Shuhei Okuyama
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Fusa Ogata
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yasuhiro Maruoka
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Deborah W. Knapp
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Sophia N. Karagiannis
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences, King’s College London, London, UK
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, UK
| | - Judit Fazekas-Singer
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
| | - Peter L. Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Amy K. LeBlanc
- Comparative Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Erika Jensen-Jarolim
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
40
|
Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Annicchiarico-Petruzzelli M, Antonov AV, Arama E, Baehrecke EH, Barlev NA, Bazan NG, Bernassola F, Bertrand MJM, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Boya P, Brenner C, Campanella M, Candi E, Carmona-Gutierrez D, Cecconi F, Chan FKM, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Cohen GM, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D'Angiolella V, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, DeBerardinis RJ, Deshmukh M, Di Daniele N, Di Virgilio F, Dixit VM, Dixon SJ, Duckett CS, Dynlacht BD, El-Deiry WS, Elrod JW, Fimia GM, Fulda S, García-Sáez AJ, Garg AD, Garrido C, Gavathiotis E, Golstein P, Gottlieb E, Green DR, Greene LA, Gronemeyer H, Gross A, Hajnoczky G, Hardwick JM, Harris IS, Hengartner MO, Hetz C, Ichijo H, Jäättelä M, Joseph B, Jost PJ, Juin PP, Kaiser WJ, Karin M, Kaufmann T, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Knight RA, Kumar S, Lee SW, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lowe SW, Luedde T, Lugli E, MacFarlane M, Madeo F, Malewicz M, Malorni W, Manic G, Marine JC, Martin SJ, Martinou JC, Medema JP, Mehlen P, Meier P, Melino S, Miao EA, Molkentin JD, Moll UM, Muñoz-Pinedo C, Nagata S, Nuñez G, Oberst A, Oren M, Overholtzer M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pereira DM, Pervaiz S, Peter ME, Piacentini M, Pinton P, Prehn JHM, Puthalakath H, Rabinovich GA, Rehm M, Rizzuto R, Rodrigues CMP, Rubinsztein DC, Rudel T, Ryan KM, Sayan E, Scorrano L, Shao F, Shi Y, Silke J, Simon HU, Sistigu A, Stockwell BR, Strasser A, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Thorburn A, Tsujimoto Y, Turk B, Vanden Berghe T, Vandenabeele P, Vander Heiden MG, Villunger A, Virgin HW, Vousden KH, Vucic D, Wagner EF, Walczak H, Wallach D, Wang Y, Wells JA, Wood W, Yuan J, Zakeri Z, Zhivotovsky B, Zitvogel L, Melino G, Kroemer G. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 2018; 25:486-541. [PMID: 29362479 PMCID: PMC5864239 DOI: 10.1038/s41418-017-0012-4] [Citation(s) in RCA: 3988] [Impact Index Per Article: 664.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the Nomenclature Committee on Cell Death (NCCD) has formulated guidelines for the definition and interpretation of cell death from morphological, biochemical, and functional perspectives. Since the field continues to expand and novel mechanisms that orchestrate multiple cell death pathways are unveiled, we propose an updated classification of cell death subroutines focusing on mechanistic and essential (as opposed to correlative and dispensable) aspects of the process. As we provide molecularly oriented definitions of terms including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence, and mitotic catastrophe, we discuss the utility of neologisms that refer to highly specialized instances of these processes. The mission of the NCCD is to provide a widely accepted nomenclature on cell death in support of the continued development of the field.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Paris Descartes/Paris V University, Paris, France.
| | - Ilio Vitale
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dieter Adam
- Institute of Immunology, Kiel University, Kiel, Germany
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Emad S Alnemri
- Department of Biochemistry and Molecular Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lucia Altucci
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Ivano Amelio
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - David W Andrews
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | - Alexey V Antonov
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Nickolai A Barlev
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Francesca Bernassola
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Mathieu J M Bertrand
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Katiuscia Bianchi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Albert Ludwigs University, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Center for Biological Investigation (CIB), Spanish National Research Council (CSIC), Madrid, Spain
| | - Catherine Brenner
- INSERM U1180, Châtenay Malabry, France
- University of Paris Sud/Paris Saclay, Orsay, France
| | - Michelangelo Campanella
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
- University College London Consortium for Mitochondrial Research, London, UK
| | - Eleonora Candi
- Biochemistry Laboratory, Dermopatic Institute of Immaculate (IDI) IRCCS, Rome, Italy
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | | | - Francesco Cecconi
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Francis K-M Chan
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Navdeep S Chandel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John A Cidlowski
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Aaron Ciechanover
- Technion Integrated Cancer Center (TICC), The Ruth and Bruce Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Gerald M Cohen
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Juan R Cubillos-Ruiz
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Vincenzo D'Angiolella
- Cancer Research UK and Medical Research Council Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vincenzo De Laurenzi
- Department of Medical, Oral and Biotechnological Sciences, CeSI-MetUniversity of Chieti-Pescara "G. d'Annunzio", Chieti, Italy
| | - Ruggero De Maria
- Institute of General Pathology, Catholic University "Sacro Cuore", Rome, Italy
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nicola Di Daniele
- Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Vishva M Dixit
- Department of Physiological Chemistry, Genentech, South San Francisco, CA, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Colin S Duckett
- Baylor Scott & White Research Institute, Baylor College of Medicine, Dallas, TX, USA
| | - Brian D Dynlacht
- Department of Pathology, New York University School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - John W Elrod
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine at Temple University School of Medicine, Philadelphia, PA, USA
| | - Gian Maria Fimia
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry, Tübingen University, Tübingen, Germany
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Carmen Garrido
- INSERM U1231 "Lipides Nutrition Cancer", Dijon, France
- Faculty of Medicine, University of Burgundy France Comté, Dijon, France
- Cancer Centre Georges François Leclerc, Dijon, France
| | - Evripidis Gavathiotis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Pierre Golstein
- Immunology Center of Marseille-Luminy, Aix Marseille University, Marseille, France
| | - Eyal Gottlieb
- Technion Integrated Cancer Center (TICC), The Ruth and Bruce Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lloyd A Greene
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Hinrich Gronemeyer
- Team labeled "Ligue Contre le Cancer", Department of Functional Genomics and Cancer, Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France
- CNRS UMR 7104, Illkirch, France
- INSERM U964, Illkirch, France
- University of Strasbourg, Illkirch, France
| | - Atan Gross
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Gyorgy Hajnoczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J Marie Hardwick
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Isaac S Harris
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Cellular and Molecular Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Marja Jäättelä
- Cell Death and Metabolism Unit, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Philipp J Jost
- III Medical Department for Hematology and Oncology, Technical University Munich, Munich, Germany
| | - Philippe P Juin
- Team 8 "Stress adaptation and tumor escape", CRCINA-INSERM U1232, Nantes, France
- University of Nantes, Nantes, France
- University of Angers, Angers, France
- Institute of Cancer Research in Western France, Saint-Herblain, France
| | - William J Kaiser
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, University of California San Diego, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Oliver Kepp
- Paris Descartes/Paris V University, Paris, France
- Faculty of Medicine, Paris Sud/Paris XI University, Kremlin-Bicêtre, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Campus, Villejuif, France
- Team 11 labeled "Ligue Nationale contre le Cancer", Cordeliers Research Center, Paris, France
- INSERM U1138, Paris, France
- Pierre et Marie Curie/Paris VI University, Paris, France
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Richard N Kitsis
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Daniel J Klionsky
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Richard A Knight
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Sam W Lee
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - John J Lemasters
- Center for Cell Death, Injury and Regeneration, Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
- Center for Cell Death, Injury and Regeneration, Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Beth Levine
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andreas Linkermann
- Division of Nephrology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Stuart A Lipton
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Richard A Lockshin
- Department of Biology, St. John's University, Queens, NY, USA
- Queens College of the City University of New York, Queens, NY, USA
| | - Carlos López-Otín
- Departament of Biochemistry and Molecular Biology, Faculty of Medicine, University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Scott W Lowe
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tom Luedde
- Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Humanitas Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Marion MacFarlane
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Frank Madeo
- Department Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Michal Malewicz
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Walter Malorni
- National Centre for Gender Medicine, Italian National Institute of Health (ISS), Rome, Italy
| | - Gwenola Manic
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Seamus J Martin
- Departments of Genetics, Trinity College, University of Dublin, Dublin 2, Ireland
| | - Jean-Claude Martinou
- Department of Cell Biology, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- Cancer Genomics Center, Amsterdam, The Netherlands
| | - Patrick Mehlen
- Apoptosis, Cancer and Development laboratory, CRCL, Lyon, France
- Team labeled "La Ligue contre le Cancer", Lyon, France
- LabEx DEVweCAN, Lyon, France
- INSERM U1052, Lyon, France
- CNRS UMR5286, Lyon, France
- Department of Translational Research and Innovation, Léon Bérard Cancer Center, Lyon, France
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Mary-Jean Mitchell Green Building, Chester Beatty Laboratories, London, UK
| | - Sonia Melino
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Rome, Italy
| | - Edward A Miao
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffery D Molkentin
- Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Cristina Muñoz-Pinedo
- Cell Death Regulation Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Shigekazu Nagata
- Laboratory of Biochemistry and Immunology, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Gabriel Nuñez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
- Center for Innate Immunity and Immune Disease, Seattle, WA, USA
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute, Rehovot, Israel
| | - Michael Overholtzer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michele Pagano
- Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University School of Medicine, New York, NY, USA
| | - Theocharis Panaretakis
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Manolis Pasparakis
- Institute for Genetics, Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Campus Vienna BioCentre, Vienna, Austria
| | - David M Pereira
- REQUIMTE/LAQV, Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
- National University Cancer Institute, National University Health System (NUHS), Singapore, Singapore
| | - Marcus E Peter
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
- LTTA center, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Health Science Foundation, Cotignola, Italy
| | - Jochen H M Prehn
- Department of Physiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hamsa Puthalakath
- Department of Biochemistry, La Trobe University, Victoria, Australia
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine (IBYME), National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
- Department of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, Stuttgart, Germany
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cecilia M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, UK
| | - Thomas Rudel
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Emre Sayan
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Luca Scorrano
- Department of Biology, University of Padua, Padua, Italy
- Venetian Institute of Molecular Medicine, Padua, Italy
| | - Feng Shao
- National Institute of Biological Sciences, Beijing, China
| | - Yufang Shi
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences, Shanghai, China
- Jiangsu Key Laboratory of Stem Cells and Medicinal Biomaterials, Institutes for Translational Medicine, Soochow University, Suzhou, China
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - John Silke
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Division of Inflammation, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Antonella Sistigu
- Institute of General Pathology, Catholic University "Sacro Cuore", Rome, Italy
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Chemistry, Columbia University, New York, NY, USA
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Gyorgy Szabadkai
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Department of Cell and Developmental Biology, University College London Consortium for Mitochondrial Research, London, UK
- Francis Crick Institute, London, UK
| | | | - Daolin Tang
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- Center for DAMP Biology, Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory for Protein Modification and Degradation of Guangdong Province, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas Medical School, University of Crete, Heraklion, Greece
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado, Aurora, CO, USA
| | | | - Boris Turk
- Department Biochemistry and Molecular Biology, "Jozef Stefan" Institute, Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Tom Vanden Berghe
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Andreas Villunger
- Division of Developmental Immunology, Innsbruck Medical University, Innsbruck, Austria
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA, USA
| | - Erwin F Wagner
- Genes, Development and Disease Group, Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, London, UK
| | - David Wallach
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Ying Wang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Will Wood
- School of Cellular and Molecular Medicine, Faculty of Biomedical Sciences, University of Bristol, Bristol, UK
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zahra Zakeri
- Department of Biology, Queens College of the City University of New York, Queens, NY, USA
| | - Boris Zhivotovsky
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Laurence Zitvogel
- Faculty of Medicine, Paris Sud/Paris XI University, Kremlin-Bicêtre, France
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Gerry Melino
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Guido Kroemer
- Paris Descartes/Paris V University, Paris, France.
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Campus, Villejuif, France.
- Team 11 labeled "Ligue Nationale contre le Cancer", Cordeliers Research Center, Paris, France.
- INSERM U1138, Paris, France.
- Pierre et Marie Curie/Paris VI University, Paris, France.
- Biology Pole, European Hospital George Pompidou, AP-HP, Paris, France.
| |
Collapse
|
41
|
Ros U, Peña-Blanco A, Hänggi K, Kunzendorf U, Krautwald S, Wong WWL, García-Sáez AJ. Necroptosis Execution Is Mediated by Plasma Membrane Nanopores Independent of Calcium. Cell Rep 2017; 19:175-187. [PMID: 28380356 PMCID: PMC5465952 DOI: 10.1016/j.celrep.2017.03.024] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/22/2016] [Accepted: 03/06/2017] [Indexed: 01/14/2023] Open
Abstract
Necroptosis is a form of regulated necrosis that
results in cell death and content release after plasma membrane permeabilization.
However, little is known about the molecular events responsible for the disruption of
the plasma membrane. Here, we find that early increase in cytosolic calcium in
TNF-induced necroptosis is mediated by treatment with a Smac mimetic via the
TNF/RIP1/TAK1 survival pathway. This does not require the activation of the necrosome
and is dispensable for necroptosis. Necroptosis induced by the activation of TLR3/4
pathways does not trigger early calcium flux. We also demonstrate that necroptotic
plasma membrane rupture is mediated by osmotic forces and membrane pores around 4 nm
in diameter. This late permeabilization step represents a hallmark in necroptosis
execution that is cell and treatment independent and requires the RIP1/RIP3/MLKL
core. In support of this, treatment with osmoprotectants reduces cell damage in an
in vivo necroptosis model of ischemia-reperfusion injury. Early calcium signaling in TSZ necroptosis correlates with
cellular levels of cIAP1/2 Calcium flux is induced by a Smac mimetic and is
dispensable for necroptosis execution Pores ∼4 nm in diameter mediate final plasma membrane
disruption in necroptosis
Collapse
Affiliation(s)
- Uris Ros
- Interfaculty Institute of Biochemistry, Tübingen University, 72076 Tübingen, Germany.
| | - Aida Peña-Blanco
- Interfaculty Institute of Biochemistry, Tübingen University, 72076 Tübingen, Germany
| | - Kay Hänggi
- Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Ulrich Kunzendorf
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - W Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry, Tübingen University, 72076 Tübingen, Germany; Max-Planck Institute for Intelligent Systems, 70569 Stuttgart, Germany.
| |
Collapse
|
42
|
Die Another Day: Inhibition of Cell Death Pathways by Cytomegalovirus. Viruses 2017; 9:v9090249. [PMID: 28869497 PMCID: PMC5618015 DOI: 10.3390/v9090249] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/27/2017] [Accepted: 08/28/2017] [Indexed: 12/26/2022] Open
Abstract
Multicellular organisms have evolved multiple genetically programmed cell death pathways that are essential for homeostasis. The finding that many viruses encode cell death inhibitors suggested that cellular suicide also functions as a first line of defence against invading pathogens. This theory was confirmed by studying viral mutants that lack certain cell death inhibitors. Cytomegaloviruses, a family of species-specific viruses, have proved particularly useful in this respect. Cytomegaloviruses are known to encode multiple death inhibitors that are required for efficient viral replication. Here, we outline the mechanisms used by the host cell to detect cytomegalovirus infection and discuss the methods employed by the cytomegalovirus family to prevent death of the host cell. In addition to enhancing our understanding of cytomegalovirus pathogenesis we detail how this research has provided significant insights into the cross-talk that exists between the various cell death pathways.
Collapse
|
43
|
Cancer's Achilles' Heel: Apoptosis and Necroptosis to the Rescue. Int J Mol Sci 2016; 18:ijms18010023. [PMID: 28025559 PMCID: PMC5297658 DOI: 10.3390/ijms18010023] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/05/2016] [Accepted: 12/19/2016] [Indexed: 12/24/2022] Open
Abstract
Apoptosis, and the more recently discovered necroptosis, are two avenues of programmed cell death. Cancer cells survive by evading these two programs, driven by oncogenes and tumor suppressor genes. While traditional therapy using small molecular inhibitors and chemotherapy are continuously being utilized, a new and exciting approach is actively underway by identifying and using synergistic relationship between driver and rescue genes in a cancer cell. Through these synthetic lethal relationships, we are gaining tremendous insights into tumor vulnerabilities and specific molecular avenues for induction of programmed cell death. In this review, we briefly discuss the two cell death processes and cite examples of such synergistic manipulations for therapeutic purposes.
Collapse
|
44
|
Bernier J. Immuno-oncology: Allying forces of radio- and immuno-therapy to enhance cancer cell killing. Crit Rev Oncol Hematol 2016; 108:97-108. [DOI: 10.1016/j.critrevonc.2016.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 11/01/2016] [Accepted: 11/01/2016] [Indexed: 12/13/2022] Open
|