1
|
Alotaibi F, Alshammari K, Alotaibi BA, Alsaab H. Destabilizing the genome as a therapeutic strategy to enhance response to immune checkpoint blockade: a systematic review of clinical trials evidence from solid and hematological tumors. Front Pharmacol 2024; 14:1280591. [PMID: 38264532 PMCID: PMC10803447 DOI: 10.3389/fphar.2023.1280591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/11/2023] [Indexed: 01/25/2024] Open
Abstract
Background: Genomic instability is increased alterations in the genome during cell division and is common among most cancer cells. Genome instability enhances the risk of initial carcinogenic transformation, generating new clones of tumor cells, and increases tumor heterogeneity. Although genome instability contributes to malignancy, it is also an "Achilles' heel" that constitutes a therapeutically-exploitable weakness-when sufficiently advanced, it can intrinsically reduce tumor cell survival by creating DNA damage and mutation events that overwhelm the capacity of cancer cells to repair those lesions. Furthermore, it can contribute to extrinsic survival-reducing events by generating mutations that encode new immunogenic antigens capable of being recognized by the immune system, particularly when anti-tumor immunity is boosted by immunotherapy drugs. Here, we describe how genome-destabilization can induce immune activation in cancer patients and systematically review the induction of genome instability exploited clinically, in combination with immune checkpoint blockade. Methods: We performed a systematic review of clinical trials that exploited the combination approach to successfully treat cancers patients. We systematically searched PubMed, Cochrane Central Register of Controlled Trials, Clinicaltrials.gov, and publication from the reference list of related articles. The most relevant inclusion criteria were peer-reviewed clinical trials published in English. Results: We identified 1,490 studies, among those 164 were clinical trials. A total of 37 clinical trials satisfied the inclusion criteria and were included in the study. The main outcome measurements were overall survival and progression-free survival. The majority of the clinical trials (30 out of 37) showed a significant improvement in patient outcome. Conclusion: The majority of the included clinical trials reported the efficacy of the concept of targeting DNA repair pathway, in combination with immune checkpoint inhibitors, to create a "ring of synergy" to treat cancer with rational combinations.
Collapse
Affiliation(s)
- Faizah Alotaibi
- College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Alahsa, Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Kanaan Alshammari
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- Oncology Department, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Badi A. Alotaibi
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Hashem Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, Saudi Arabia
| |
Collapse
|
2
|
Laureano RS, Vanmeerbeek I, Sprooten J, Govaerts J, Naulaerts S, Garg AD. The cell stress and immunity cycle in cancer: Toward next generation of cancer immunotherapy. Immunol Rev 2024; 321:71-93. [PMID: 37937803 DOI: 10.1111/imr.13287] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/05/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
The cellular stress and immunity cycle is a cornerstone of organismal homeostasis. Stress activates intracellular and intercellular communications within a tissue or organ to initiate adaptive responses aiming to resolve the origin of this stress. If such local measures are unable to ameliorate this stress, then intercellular communications expand toward immune activation with the aim of recruiting immune cells to effectively resolve the situation while executing tissue repair to ameliorate any damage and facilitate homeostasis. This cellular stress-immunity cycle is severely dysregulated in diseased contexts like cancer. On one hand, cancer cells dysregulate the normal cellular stress responses to reorient them toward upholding growth at all costs, even at the expense of organismal integrity and homeostasis. On the other hand, the tumors severely dysregulate or inhibit various components of organismal immunity, for example, by facilitating immunosuppressive tumor landscape, lowering antigenicity, and increasing T-cell dysfunction. In this review we aim to comprehensively discuss the basis behind tumoral dysregulation of cellular stress-immunity cycle. We also offer insights into current understanding of the regulators and deregulators of this cycle and how they can be targeted for conceptualizing successful cancer immunotherapy regimen.
Collapse
Affiliation(s)
- Raquel S Laureano
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Anoushirvani AA, Jafarian Yazdi A, Amirabadi S, Asouri SA, Shafabakhsh R, Sheida A, Hosseini Khabr MS, Jafari A, Tamehri Zadeh SS, Hamblin MR, Kalantari L, Talaei Zavareh SA, Mirzaei H. Role of non-coding RNAs in neuroblastoma. Cancer Gene Ther 2023; 30:1190-1208. [PMID: 37217790 DOI: 10.1038/s41417-023-00623-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/25/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023]
Abstract
Neuroblastoma is known as the most prevalent extracranial malignancy in childhood with a neural crest origin. It has been widely accepted that non-coding RNAs (ncRNAs) play important roles in many types of cancer, including glioma and gastrointestinal cancers. They may regulate the cancer gene network. According to recent sequencing and profiling studies, ncRNAs genes are deregulated in human cancers via deletion, amplification, abnormal epigenetic, or transcriptional regulation. Disturbances in the expression of ncRNAs may act either as oncogenes or as anti-tumor suppressor genes, and can lead to the induction of cancer hallmarks. ncRNAs can be secreted from tumor cells inside exosomes, where they can be transferred to other cells to affect their function. However, these topics still need more study to clarify their exact roles, so the present review addresses different roles and functions of ncRNAs in neuroblastoma.
Collapse
Affiliation(s)
- Ali Arash Anoushirvani
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Sanaz Amirabadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran
| | - Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ameneh Jafari
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX: 15179/64311, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University, Kashan, Iran.
| |
Collapse
|
4
|
Werry F, Mazur E, Theyse LFH, Edlich F. Apoptosis Regulation in Osteoarthritis and the Influence of Lipid Interactions. Int J Mol Sci 2023; 24:13028. [PMID: 37685835 PMCID: PMC10488181 DOI: 10.3390/ijms241713028] [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: 07/28/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common chronic diseases in human and animal joints. The joints undergo several morphological and histological changes during the development of radiographically visible osteoarthritis. The most discussed changes include synovial inflammation, the massive destruction of articular cartilage and ongoing joint destruction accompanied by massive joint pain in the later stadium. Either the increased apoptosis of chondrocytes or the insufficient apoptosis of inflammatory macrophages and synovial fibroblasts are likely to underly this process. In this review, we discuss the current state of research on the pathogenesis of OA with special regard to the involvement of apoptosis.
Collapse
Affiliation(s)
- Frederike Werry
- Institute of Biochemistry, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany;
| | - Emilia Mazur
- Soft Tissue & Orthopaedic Surgery Service, Department for Small Animals, College of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany;
| | - Lars F. H. Theyse
- Soft Tissue & Orthopaedic Surgery Service, Department for Small Animals, College of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany;
| | - Frank Edlich
- Institute of Biochemistry, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany;
| |
Collapse
|
5
|
Gupta P, Makkar TK, Goel L, Pahuja M. Role of inflammation and oxidative stress in chemotherapy-induced neurotoxicity. Immunol Res 2022; 70:725-741. [PMID: 35859244 DOI: 10.1007/s12026-022-09307-7] [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] [Received: 03/29/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022]
Abstract
Chemotherapeutic agents may adversely affect the nervous system, including the neural precursor cells as well as the white matter. Although the mechanisms are not completely understood, several hypotheses connecting inflammation and oxidative stress with neurotoxicity are now emerging. The proposed mechanisms differ depending on the class of drug. For example, toxicity due to cisplatin occurs due to activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which alters hippocampal long-term potentiation. Free radical injury is also involved in the cisplatin-mediated neurotoxicity as dysregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) has been seen which protects against the free radical injury by regulating glutathione S-transferases and hemeoxygenase-1 (HO-1). Thus, correcting the imbalance between NF-κB and Nrf2/HO-1 pathways may alleviate cisplatin-induced neurotoxicity. With newer agents like bortezomib, peripheral neuropathy occurs due to up-regulation of TNF-α and IL-6 in the sensory neurons. Superoxide dismutase dysregulation is also involved in bortezomib-induced neuropathy. This article reviews the available literature on inflammation and oxidative stress in neurotoxicity caused by various classes of chemotherapeutic agents. It covers the conventional medicines like platinum compounds, vinca alkaloids, and methotrexate, as well as the newer therapeutic agents like immunomodulators and immune checkpoint inhibitors. A better understanding of the pathophysiology will lead to further advancement in strategies for management of chemotherapy-induced neurotoxicity.
Collapse
Affiliation(s)
- Pooja Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India. .,Coordinator, AIIMS Adverse Drug Reaction Monitoring Centre, Pharmacovigilance Program of India, New Delhi, India.
| | - Tavneet Kaur Makkar
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Lavisha Goel
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Monika Pahuja
- Division of Basic Medical Sciences, Indian Council of Medical Research, New Delhi, India
| |
Collapse
|
6
|
Cheng W, Cui C, Liu G, Ye C, Shao F, Bagchi AK, Mehta JL, Wang X. NF-κB, A Potential Therapeutic Target in Cardiovascular Diseases. Cardiovasc Drugs Ther 2022; 37:571-584. [PMID: 35796905 DOI: 10.1007/s10557-022-07362-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2022] [Indexed: 11/03/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally. Atherosclerosis is the basis of major CVDs - myocardial ischemia, heart failure, and stroke. Among numerous functional molecules, the transcription factor nuclear factor κB (NF-κB) has been linked to downstream target genes involved in atherosclerosis. The activation of the NF-κB family and its downstream target genes in response to environmental and cellular stress, hypoxia, and ischemia initiate different pathological events such as innate and adaptive immunity, and cell survival, differentiation, and proliferation. Thus, NF-κB is a potential therapeutic target in the treatment of atherosclerosis and related CVDs. Several biologics and small molecules as well as peptide/proteins have been shown to regulate NF-κB dependent signaling pathways. In this review, we will focus on the function of NF-κB in CVDs and the role of NF-κB inhibitors in the treatment of CVDs.
Collapse
Affiliation(s)
- Weijia Cheng
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China.,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Can Cui
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China.,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Gang Liu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chenji Ye
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Fang Shao
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450046, China
| | - Ashim K Bagchi
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA
| | - Jawahar L Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA.
| | - Xianwei Wang
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China. .,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
7
|
Nandi SK, Basu S, Bhattacharjya A, Ghosh RD, Bose CK, Mukhopadhyay S, Bhattacharya R. Interplay of gut microbiome, fatty acids and the endocannabinoid system in regulating development, progression, immunomodulation and chemoresistance of cancer. Nutrition 2022; 103-104:111787. [DOI: 10.1016/j.nut.2022.111787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 05/17/2022] [Accepted: 06/25/2022] [Indexed: 11/25/2022]
|
8
|
Ceramella J, Mariconda A, Sirignano M, Iacopetta D, Rosano C, Catalano A, Saturnino C, Sinicropi MS, Longo P. Novel Au Carbene Complexes as Promising Multi-Target Agents in Breast Cancer Treatment. Pharmaceuticals (Basel) 2022; 15:507. [PMID: 35631334 PMCID: PMC9146163 DOI: 10.3390/ph15050507] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/09/2022] [Accepted: 04/19/2022] [Indexed: 01/27/2023] Open
Abstract
Over the past decade, metal complexes based on N-heterocyclic carbenes (NHCs) have attracted great attention due to their wide and exciting applications in material sciences and medicinal chemistry. In particular, the gold-based complexes are the focus of research efforts for the development of new anticancer compounds. Literature data and recent results, obtained by our research group, reported the design, the synthesis and the good anticancer activity of some silver and gold complexes with NHC ligands. In particular, some of these complexes were active towards some breast cancer cell lines. Considering this evidence, here we report some new Au-NHC complexes prepared in order to improve solubility and biological activity. Among them, the compounds 1 and 6 showed an interesting anticancer activity towards the breast cancer MDA-MB-231 and MCF-7 cell lines, respectively. In addition, in vitro and in silico studies demonstrated that they were able to inhibit the activity of the human topoisomerases I and II and the actin polymerization reaction. Moreover, a downregulation of vimentin expression and a reduced translocation of NF-kB into the nucleus was observed. The interference with these vital cell structures induced breast cancer cells' death by triggering the extrinsic apoptotic pathway.
Collapse
Affiliation(s)
- Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy; (J.C.); (M.S.S.)
| | - Annaluisa Mariconda
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy; (A.M.); (C.S.)
| | - Marco Sirignano
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.S.); (P.L.)
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy; (J.C.); (M.S.S.)
| | - Camillo Rosano
- U.O. Proteomica e Spettrometria di Massa, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 1632 Genova, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy;
| | - Carmela Saturnino
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy; (A.M.); (C.S.)
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy; (J.C.); (M.S.S.)
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (M.S.); (P.L.)
| |
Collapse
|
9
|
Jiang H, Wang S, Hou L, Huang JA, Su B. Resveratrol inhibits cell apoptosis by suppressing long noncoding RNA (lncRNA) XLOC_014869 during lipopolysaccharide-induced acute lung injury in rats. J Thorac Dis 2022; 13:6409-6426. [PMID: 34992821 PMCID: PMC8662516 DOI: 10.21037/jtd-21-1113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/28/2021] [Indexed: 12/29/2022]
Abstract
Background Acute lung injury (ALI) is a common clinical complication with a high mortality rate. Resveratrol (Res) has been shown to protect against ALI, but the role of long noncoding RNAs (lncRNAs) in this process is still unclear. Methods Male rats (n=20) aged 7–8 weeks were randomly divided into four groups: control, lipopolysaccharide (LPS), LPS + Res, and LPS + dexamethasone (Dexa). Intragastric administration of Res (0.5 mg/kg) or Dexa (1.5 mg/kg) was performed 1 h before intraperitoneal injection of LPS (5 mg/kg). Lung tissue, serum, and bronchoalveolar lavage fluid were sampled 6 h after LPS treatment for inflammatory factor detection, pathological detection, lncRNA sequencing and bioinformatical analysis, and TdT-mediated dUTP Nick-End Labeling. Quantitative real time polymerase chain reaction and western blotting were used to verify the sequencing results. LPS, Res, and RNA interference were used in rat alveolar epithelial cells experiments to confirm the protective of Res/lncRNA against ALI. Results Res pretreatment inhibited lung injury and the increase of inflammatory cytokines induced by LPS. The differentially expressed lncRNAs and mRNAs (P<0.05 and |fold change| >2) were mainly involved in the signaling pathway of immunity, infection, signaling molecules and interactions. Among the lncRNAs and mRNAs, 26 mRNAs and 23 lncRNAs had high levels in lungs treated with LPS but decreased with Res, and 17 mRNAs and 27 lncRNAs were at lower levels in lungs treated with LPS but increased with Res. lncRNA and adjacent mRNA analysis showed that lncRNAs XLOC_014869 and the adjacent gene Fos, and the possible downstream genes Jun and Faslg were increased by LPS, but these changes were attenuated by Res. Pretreatment with Res reduced LPS-induced lung tissue apoptosis. Similarly, Res treatment and knockdown of lncRNA XLOC_014869 reduced LPS-induced apoptosis and the levels of Fos, c-Jun, and Fas-L. Conclusions Res can inhibit the increase of lncRNAs XLOC_014869 caused by LPS stimulation and inhibit lung cell apoptosis. These effects may be due to lncRNA XLOC_014869 mediation of the pro-apoptotic factors (Fos, c-Jun, and Fas-L).
Collapse
Affiliation(s)
- Hongbin Jiang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shanmei Wang
- Department of Emergency, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jian-An Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Bo Su
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Bruscoli S, Riccardi C, Ronchetti S. GILZ as a Regulator of Cell Fate and Inflammation. Cells 2021; 11:cells11010122. [PMID: 35011684 PMCID: PMC8750894 DOI: 10.3390/cells11010122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/21/2021] [Accepted: 12/29/2021] [Indexed: 12/30/2022] Open
Abstract
One of the human body’s initial responses to stress is the adrenal response, involving the release of mediators that include adrenaline and glucocorticoids (GC). GC are involved in controlling the inflammatory and immune response mechanisms. Of these, the molecular mechanisms that contribute to anti-inflammatory effects warrant more investigation. Previously, we found that GC induced GILZ (glucocorticoid-induced leucine zipper) quickly and widely in thymocytes, T lymphocytes, and other leukocytes. GILZ regulates the activation of cells and is an essential mediator of endogenous GC and the majority of GC anti-inflammatory effects. Further research in this regard could lead to the development of an anti-inflammatory treatment that yields the therapeutic outcomes of GC but without their characteristic adverse effects. Here, we examine the mechanisms of GILZ in the context of GC. Specifically, we review its role in the proliferation and differentiation of cells and in apoptosis. We also examine its involvement in immune cells (macrophages, neutrophils, dendritic cells, T and B lymphocytes), and in non-immune cells, including cancer cells. In conclusion, GILZ is an anti-inflammatory molecule that could mediate the immunomodulatory activities of GC, with less adverse effects, and could be a target molecule for designing new therapies to treat inflammatory diseases.
Collapse
|
11
|
Chung KS, Yoo CB, Lee JH, Lee HH, Park SE, Han HS, Lee SY, Kwon BM, Choi JH, Lee KT. Regulation of ROS-Dependent JNK Pathway by 2'-Hydroxycinnamaldehyde Inducing Apoptosis in Human Promyelocytic HL-60 Leukemia Cells. Pharmaceutics 2021; 13:pharmaceutics13111794. [PMID: 34834209 PMCID: PMC8618870 DOI: 10.3390/pharmaceutics13111794] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/28/2022] Open
Abstract
The present study demonstrated that 2'-hydroxycinnamaldehyde (2'-HCA) induced apoptosis in human promyelocytic leukemia HL-60 cells through the activation of mitochondrial pathways including (1) translocation of Bim and Bax from the cytosol to mitochondria, (2) downregulation of Bcl-2 protein expression, (3) cytochrome c release into the cytosol, (4) loss of mitochondrial membrane potential (ΔΨm), and (5) caspase activation. 2'-HCA also induced the activation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase1/2 (ERK1/2) in HL-60 cells. The pharmacological and genetic inhibition of JNK effectively prevented 2'-HCA-induced apoptosis and activator protein-1 (AP-1)-DNA binding. In addition, 2'-HCA resulted in the accumulation of reactive oxygen species (ROS) and depletion of intracellular glutathione (GSH) and protein thiols (PSH) in HL-60 cells. NAC treatment abrogated 2'-HCA-induced JNK phosphorylation, AP-1-DNA binding, and Bim mitochondrial translocation, suggesting that oxidative stress may be required for 2'-HCA-induced intrinsic apoptosis. Xenograft mice inoculated with HL-60 leukemia cells demonstrated that the intraperitoneal administration of 2'-HCA inhibited tumor growth by increasing of TUNEL staining, the expression levels of nitrotyrosine and pro-apoptotic proteins, but reducing of PCNA protein expression. Taken together, our findings suggest that 2'-HCA induces apoptosis via the ROS-dependent JNK pathway and could be considered as a potential therapeutic agent for leukemia.
Collapse
Affiliation(s)
- Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
| | - Chae-Bin Yoo
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
| | - Jeong-Hun Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea;
| | - Hwi-Ho Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
| | - Sang-Eun Park
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmarcy, Kyung Hee University, Seoul 02447, Korea
| | - Hee-Soo Han
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea;
| | - Su-Yeon Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmarcy, Kyung Hee University, Seoul 02447, Korea
| | - Byoung-Mok Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea;
| | - Jung-Hye Choi
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea;
- Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea; (K.-S.C.); (C.-B.Y.); (J.-H.L.); (H.-H.L.); (S.-E.P.); (H.-S.H.); (S.-Y.L.)
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul 02447, Korea;
- Correspondence: ; Tel.: +82-2-961-0860
| |
Collapse
|
12
|
Manikandan DB, Arumugam M, Veeran S, Sridhar A, Krishnasamy Sekar R, Perumalsamy B, Ramasamy T. Biofabrication of ecofriendly copper oxide nanoparticles using Ocimum americanum aqueous leaf extract: analysis of in vitro antibacterial, anticancer, and photocatalytic activities. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:33927-33941. [PMID: 33410001 DOI: 10.1007/s11356-020-12108-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/14/2020] [Indexed: 06/12/2023]
Abstract
Nanotechnology tends to be a swiftly growing field of research that actively influences and inhibits the growth of bacteria/cancer. Noble metal nanoparticles (NPs) such as silver, copper, and gold have been used to damage bacterial and cancer growth over recent years; however, the toxicity of higher NPs concentrations remains a major issue. The copper oxide nanoparticles (CuONPs) were therefore fabricated using a simple green chemistry approach. Biofabricated CuONPs were characterized using UV-visible, FE-SEM with EDS, HR-TEM, FT-IR, XRD, Raman spectroscopy, and XPS analysis. Formations of CuONPs have been observed by UV-visible absorbance peak at 360.74 nm. The surface morphology of the CuONPs showed the spherical structure and size (~ 68 nm). The EDS spectrum of CuONPs has proved to be the key signals of copper (Cu) and oxygen (O) components. FT-IR analysis, to validate the important functional biomolecules (O-H, C=C, C-H, C-O) are responsible for reduction and stabilization of CuONPs. The monoclinic end-centered crystalline structures of CuONPs were confirmed with XRD planes. The electrochemical oxygen states of the CuONPs have been studied using spectroscopy of the Raman and X-ray photoelectron. After successful preparation, CuONPs examined their antibacterial, anticancer, and photocatalytic activities. Green-fabricated CuONPs were promising antibacterial candidate against human pathogenic gram-negative bacteria Escherichia coli, Vibrio cholerae, Salmonella typhimurium, Klebsiella pneumoniae, Aeromonas hydrophila, and Pseudomonas aeruginosa. CuONPs were demonstrated the excellent anticancer activity against A549 human lung adenocarcinoma cell line. Furthermore, CuONPs exhibited photocatalytic degradation of azo dyes such as eosin yellow (EY), rhodamine 123 (Rh 123), and methylene blue (MB). Biofabricated CuONPs may therefore be an important biomedical research for the aid of bacterial/cancer diseases and photocatalytic degradation of azo dyes.
Collapse
Affiliation(s)
- Dinesh Babu Manikandan
- Laboratory of Aquabiotics/Nanoscience, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Manikandan Arumugam
- Laboratory of Aquabiotics/Nanoscience, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Srinivasan Veeran
- Laboratory of Aquabiotics/Nanoscience, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Arun Sridhar
- Laboratory of Aquabiotics/Nanoscience, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Rajkumar Krishnasamy Sekar
- Laboratory of Aquabiotics/Nanoscience, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Balaji Perumalsamy
- National Centre for Alternatives to Animal Experiments, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Thirumurugan Ramasamy
- Laboratory of Aquabiotics/Nanoscience, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India.
- National Centre for Alternatives to Animal Experiments, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India.
| |
Collapse
|
13
|
Abstract
Phototherapeutic modalities induce apoptosis of keratinocytes and immune cells, impact cytokine production, downregulate the IL-23/Th17 axis, and induce regulatory T cells. As in anti-IL-17 or anti-IL-23 antibody treatment, the dual action of phototherapy on skin and the immune system is likely responsible for sustained resolution of lesions in diseases such as psoriasis. In cutaneous T cell lymphoma, phototherapy may function by causing tumor cell apoptosis and eliminating the neoplastic and inflammatory infiltrate. Further research on phototherapeutic mechanisms will help advance, optimize, and refine dermatologic treatments and may open up novel avenues for treatment strategies in dermatology and beyond.
Collapse
Affiliation(s)
- Zizi Yu
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Peter Wolf
- Department of Dermatology, Research Unit for Photodermatology, Medical University of Graz, Auenbruggerplatz 8, Graz A-8036, Austria.
| |
Collapse
|
14
|
Zhou X, Lu H, Li F, Hao X, Han L, Dong Q, Chen X. MicroRNA-429 inhibits neuroblastoma cell proliferation, migration and invasion via the NF-κB pathway. Cell Mol Biol Lett 2020; 25:5. [PMID: 32082390 PMCID: PMC7020518 DOI: 10.1186/s11658-020-0202-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 01/31/2020] [Indexed: 12/12/2022] Open
Abstract
Background MicroRNAs (miRNAs or miRs) can participate in the development and progression of neuroblastoma. Many studies have indicated that miR-429 can participate in tumor development. However, the mechanism underlying miR-429-mediated progression of neuroblastoma remains largely unclear. Methods Colony formation and apoptosis assays were used to determine the effect of miR-429 on cell proliferation. Its impact on cell migration was determined using the wound-healing and Transwell assays. The target gene of miR-429 was confirmed via western blotting and luciferase reporter assays. A nude mouse xenograft model with miR-429 overexpression was used to assess the effect on tumor growth. Results Our findings indicate that miR-429 is downregulated in neuroblastoma cell lines. We also found that it can induce apoptosis and inhibit proliferation in cells of those lines. MiR-429 can bind to the 3′-UTR of IKKβ mRNA and overexpression of IKKβ can reverse cell proliferation, blocking the effect of miR-429. Furthermore, miR-429 overexpression inhibited neuroblastoma growth in our nude mouse xenograft model. Conclusion We provide important insight into miR-429 as a tumor suppressor through interaction with IKKβ, which is a catalytic subunit of the IKK complex that activates NF-κB nuclear transport. Our results demonstrate that miR-429 may be a new target for the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Xianjun Zhou
- 1Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| | - Hongting Lu
- 1Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| | - Fujiang Li
- 1Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| | - Xiwei Hao
- 1Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| | - Lulu Han
- 2Department of Operation Room, the Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, 266000 Shandong China
| | - Qian Dong
- 1Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| | - Xin Chen
- 1Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| |
Collapse
|
15
|
IL-24 Inhibits Lung Cancer Growth by Suppressing GLI1 and Inducing DNA Damage. Cancers (Basel) 2019; 11:cancers11121879. [PMID: 31783569 PMCID: PMC6966580 DOI: 10.3390/cancers11121879] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/14/2019] [Accepted: 11/19/2019] [Indexed: 01/18/2023] Open
Abstract
Aberrant expression of GLI1 is responsible for aggressive tumor behavior and survival due to its effects on the DNA damage response (DDR). We investigated whether interleukin (IL)-24, a tumor suppressor, inhibits GLI1 and the associated DDR pathway in human NSCLCs. IL-24 treatment reduces mRNA and protein expression of GLI1 in lung tumor cells, but not in normal cells. GLI1 reporter assay and mRNA studies demonstrated that IL-24 regulates GLI1 at the post-transcriptional level by favoring mRNA degradation. Associated with GLI1 inhibition was marked suppression of the ATM-mediated DDR pathway resulting in increased DNA damage, as evidenced by γ-H2AX foci and Comet assay. Furthermore, attenuation of GLI1-associated DDR by IL-24 increased caspase-3 and PARP activity, resulting in cancer cell apoptosis. GLI1 inhibition and overexpression confirmed that IL-24-mediated anti-tumor effects involved the GLI-dependent pathway. Finally, we observed that IL-24-mediated alteration in GLI1 is independent of the canonical hedgehog-signaling pathway. Our study provides evidence that IL-24 treatment induces DNA damage, and reduces GLI1 expression and offers an opportunity for testing IL-24-based therapy for inhibiting GLI1 in lung cancer.
Collapse
|
16
|
Zhu H, Li Y, Liu Y, Han B. Bivalent SMAC Mimetics for Treating Cancer by Antagonizing Inhibitor of Apoptosis Proteins. ChemMedChem 2019; 14:1951-1962. [DOI: 10.1002/cmdc.201900410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/10/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Hongping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of PharmacyChengdu University of Traditional Chinese Medicine 1166 Liutai Avenue Chengdu 611137 China
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University 168 Huaguan Road Chengdu 610052 China
| | - Yi Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University 168 Huaguan Road Chengdu 610052 China
| | - Yue Liu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University 168 Huaguan Road Chengdu 610052 China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of PharmacyChengdu University of Traditional Chinese Medicine 1166 Liutai Avenue Chengdu 611137 China
| |
Collapse
|
17
|
Kay J, Thadhani E, Samson L, Engelward B. Inflammation-induced DNA damage, mutations and cancer. DNA Repair (Amst) 2019; 83:102673. [PMID: 31387777 DOI: 10.1016/j.dnarep.2019.102673] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/15/2019] [Accepted: 07/18/2019] [Indexed: 12/22/2022]
Abstract
The relationships between inflammation and cancer are varied and complex. An important connection linking inflammation to cancer development is DNA damage. During inflammation reactive oxygen and nitrogen species (RONS) are created to combat pathogens and to stimulate tissue repair and regeneration, but these chemicals can also damage DNA, which in turn can promote mutations that initiate and promote cancer. DNA repair pathways are essential for preventing DNA damage from causing mutations and cytotoxicity, but RONS can interfere with repair mechanisms, reducing their efficacy. Further, cellular responses to DNA damage, such as damage signaling and cytotoxicity, can promote inflammation, creating a positive feedback loop. Despite coordination of DNA repair and oxidative stress responses, there are nevertheless examples whereby inflammation has been shown to promote mutagenesis, tissue damage, and ultimately carcinogenesis. Here, we discuss the DNA damage-mediated associations between inflammation, mutagenesis and cancer.
Collapse
Affiliation(s)
- Jennifer Kay
- Department of Biological Engineering, United States.
| | | | - Leona Samson
- Department of Biological Engineering, United States; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States
| | | |
Collapse
|
18
|
Hennessy RJ, Pham K, Delconte R, Rautela J, Hodgkin PD, Huntington ND. Quantifying NK cell growth and survival changes in response to cytokines and regulatory checkpoint blockade helps identify optimal culture and expansion conditions. J Leukoc Biol 2019; 105:1341-1354. [PMID: 31079418 DOI: 10.1002/jlb.ma0718-296r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/01/2019] [Accepted: 04/28/2019] [Indexed: 01/02/2023] Open
Abstract
NK cells are innate lymphocytes critical for immune surveillance, particularly in eradication of metastatic cancer cells and acute antiviral responses. In contrast to T cells, NK cell-mediated immunity is rapid, with spontaneous cytotoxicity and cytokine/chemokine production upon pathogen detection. The renaissance in cancer immunology has cast NK cell biology back into the spotlight with an urgent need for deeper understanding of the regulatory networks that govern NK cell antitumor activity. To this end, we have adapted and refined a series of quantitative cellular calculus methods, previously applied to T and B lymphocytes, to dissect the biologic outcomes of NK cells following stimulation with cytokines (IL-15, IL-12, IL-18) or deletion of genes that regulate NK cell proliferation (Cish), survival (Bcl2l11), and activation-induced-cell-death (AICD; Fas). Our methodology is well suited to delineate effects on division rate, intrinsic apoptosis, and AICD, permitting variables such as population half-life, rate of cell division, and their combined influence on population numbers in response to stimuli to be accurately measured and modelled. Changes in these variables that result from gene deletion, concentration of stimuli, time, and cell density give insight into the dynamics of NK cell responses and serve as a platform to dissect the mechanism of action of putative checkpoints in NK cell activation and novel NK cell immunotherapy agents.
Collapse
Affiliation(s)
- Robert J Hennessy
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Kim Pham
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca Delconte
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Jai Rautela
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Philip D Hodgkin
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas D Huntington
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| |
Collapse
|
19
|
Zhu J, Petit PF, Van den Eynde BJ. Apoptosis of tumor-infiltrating T lymphocytes: a new immune checkpoint mechanism. Cancer Immunol Immunother 2019; 68:835-847. [PMID: 30406374 PMCID: PMC11028327 DOI: 10.1007/s00262-018-2269-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022]
Abstract
Immunotherapy based on checkpoint inhibitors is providing substantial clinical benefit, but only to a minority of cancer patients. The current priority is to understand why the majority of patients fail to respond. Besides T-cell dysfunction, T-cell apoptosis was reported in several recent studies as a relevant mechanism of tumoral immune resistance. Several death receptors (Fas, DR3, DR4, DR5, TNFR1) can trigger apoptosis when activated by their respective ligands. In this review, we discuss the immunomodulatory role of the main death receptors and how these are shaping the tumor microenvironment, with a focus on Fas and its ligand. Fas-mediated apoptosis of T cells has long been known as a mechanism allowing the contraction of T-cell responses to prevent immunopathology, a phenomenon known as activation-induced cell death, which is triggered by induction of Fas ligand (FasL) expression on T cells themselves and qualifies as an immune checkpoint mechanism. Recent evidence indicates that other cells in the tumor microenvironment can express FasL and trigger apoptosis of tumor-infiltrating lymphocytes (TIL), including endothelial cells and myeloid-derived suppressor cells. The resulting disappearance of TIL prevents anti-tumor immunity and may in fact contribute to the absence of TIL that is typical of "cold" tumors that fail to respond to immunotherapy. Interfering with the Fas-FasL pathway in the tumor microenvironment has the potential to increase the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Jingjing Zhu
- Ludwig Institute for Cancer Research, 1200, Brussels, Belgium
- de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75 B1.74.03, 1200, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology, 1200, Brussels, Belgium
| | - Pierre-Florent Petit
- Ludwig Institute for Cancer Research, 1200, Brussels, Belgium
- de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75 B1.74.03, 1200, Brussels, Belgium
| | - Benoit J Van den Eynde
- Ludwig Institute for Cancer Research, 1200, Brussels, Belgium.
- de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75 B1.74.03, 1200, Brussels, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology, 1200, Brussels, Belgium.
| |
Collapse
|
20
|
Kaina B, Christmann M. DNA repair in personalized brain cancer therapy with temozolomide and nitrosoureas. DNA Repair (Amst) 2019; 78:128-141. [PMID: 31039537 DOI: 10.1016/j.dnarep.2019.04.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 12/16/2022]
Abstract
Alkylating agents have been used since the 60ties in brain cancer chemotherapy. Their target is the DNA and, although the DNA of normal and cancer cells is damaged unselectively, they exert tumor-specific killing effects because of downregulation of some DNA repair activities in cancer cells. Agents exhibiting methylating properties (temozolomide, procarbazine, dacarbazine, streptozotocine) induce at least 12 different DNA lesions. These are repaired by damage reversal mechanisms involving the alkyltransferase MGMT and the alkB homologous protein ALKBH2, and through base excision repair (BER). There is a strong correlation between the MGMT expression level and therapeutic response in high-grade malignant glioma, supporting the notion that O6-methylguanine and, for nitrosoureas, O6-chloroethylguanine are the most relevant toxic damages at therapeutically relevant doses. Since MGMT has a significant impact on the outcome of anti-cancer therapy, it is a predictive marker of the effectiveness of methylating anticancer drugs, and clinical trials are underway aimed at assessing the influence of MGMT inhibition on the therapeutic success. Other DNA repair factors involved in methylating drug resistance are mismatch repair, DNA double-strand break (DSB) repair by homologous recombination (HR) and DSB signaling. Base excision repair and ALKBH2 might also contribute to alkylating drug resistance and their downregulation may have an impact on drug sensitivity notably in cells expressing a high amount of MGMT and at high doses of temozolomide, but the importance in a therapeutic setting remains to be shown. MGMT is frequently downregulated in cancer cells (up to 40% in glioblastomas), which is due to CpG promoter methylation. Astrocytoma (grade III) are frequently mutated in isocitrate dehydrogenase (IDH1). These tumors show a surprisingly good therapeutic response. IDH1 mutation has an impact on ALKBH2 activity thus influencing DNA repair. A master switch between survival and death is p53, which often retains transactivation activity (wildtype) in malignant glioma. The role of p53 in regulating survival via DNA repair and the routes of death are discussed and conclusions as to cancer therapeutic options were drawn.
Collapse
Affiliation(s)
- Bernd Kaina
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany.
| | - Markus Christmann
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| |
Collapse
|
21
|
A network-centric approach to drugging TNF-induced NF-κB signaling. Nat Commun 2019; 10:860. [PMID: 30808860 PMCID: PMC6391473 DOI: 10.1038/s41467-019-08802-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 01/30/2019] [Indexed: 01/01/2023] Open
Abstract
Target-centric drug development strategies prioritize single-target potency in vitro and do not account for connectivity and multi-target effects within a signal transduction network. Here, we present a systems biology approach that combines transcriptomic and structural analyses with live-cell imaging to predict small molecule inhibitors of TNF-induced NF-κB signaling and elucidate the network response. We identify two first-in-class small molecules that inhibit the NF-κB signaling pathway by preventing the maturation of a rate-limiting multiprotein complex necessary for IKK activation. Our findings suggest that a network-centric drug discovery approach is a promising strategy to evaluate the impact of pharmacologic intervention in signaling. Chemical perturbation of specific protein–protein interactions is notoriously difficult, yet necessary when complete inhibition of a signalling pathway is detrimental to the cell. Here, the authors use a systems approach and identify two first-in-class small molecules that specifically inhibit TNF-induced NF-κB activation.
Collapse
|
22
|
Focus on Cdc42 in Breast Cancer: New Insights, Target Therapy Development and Non-Coding RNAs. Cells 2019; 8:cells8020146. [PMID: 30754684 PMCID: PMC6406589 DOI: 10.3390/cells8020146] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 12/25/2022] Open
Abstract
Breast cancer is the most common malignant tumors in females. Although the conventional treatment has demonstrated a certain effect, some limitations still exist. The Rho guanosine triphosphatase (GTPase) Cdc42 (Cell division control protein 42 homolog) is often upregulated by some cell surface receptors and oncogenes in breast cancer. Cdc42 switches from inactive guanosine diphosphate (GDP)-bound to active GTP-bound though guanine-nucleotide-exchange factors (GEFs), results in activation of signaling cascades that regulate various cellular processes such as cytoskeletal changes, proliferation and polarity establishment. Targeting Cdc42 also provides a strategy for precise breast cancer therapy. In addition, Cdc42 is a potential target for several types of non-coding RNAs including microRNAs and lncRNAs. These non-coding RNAs is extensively involved in Cdc42-induced tumor processes, while many of them are aberrantly expressed. Here, we focus on the role of Cdc42 in cell morphogenesis, proliferation, motility, angiogenesis and survival, introduce the Cdc42-targeted non-coding RNAs, as well as present current development of effective Cdc42-targeted inhibitors in breast cancer.
Collapse
|
23
|
Peng Q, Liu Y, Dong M, Xu F, Huang J, Chen J, Li X, Zhang J, Zhang W. Interaction between NF-κB and AP-1 and their intracellular localization at labor in human late pregnant myometrial cells in vivo and in vitro. Medicine (Baltimore) 2018; 97:e12494. [PMID: 30235753 PMCID: PMC6160212 DOI: 10.1097/md.0000000000012494] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Preterm birth (PTB) is the most important cause of neonatal morbidity and mortality next to congenital anomalies in the developed world. NF-κB and AP-1 were reported to play an important role in parturition initiation. However, the interaction relationship between the 2 molecules in labor initiation has not yet been reported.This study aimed to investigate the interaction between NF-κB and AP-1 and their intracellular translocation during labor in human late pregnant myometrial cells (HLPMCs).Co-immunoprecipitation (Co-IP), Western blot analysis, immunohistochemistry (IHC), and immunocytofluorescence (ICF) techniques were applied to explore the interaction between NF-κB and AP-1 and the alteration in their intracellular localization before and after labor onset.The protein expression levels of NF-κBp65 and AP-1(c-jun) in the natural labor group were observed significantly higher than that in the non-labor group. Pearson's correlation analysis showed a positive correlation between the protein expression of NF-κBp65 and AP-1(c-jun). Interactions were found between the 2 molecules in HLPMCs both in natural labor and non-labor group and were also found in primary culture HLPMCs before and after neuromedin B (NMB) stimulation. NF-κBp65 and AP-1(c-jun) were localized mainly in the cytoplasm before labor onset or NMB stimulation and were translocated into the nucleus upon labor initiation and NMB stimulation.These results demonstrated that upregulated protein expression of NF-κBp65 and AP-1(c-jun), the enhanced interaction between the 2 molecules, and their translocation to nucleus might be correlated to labor initiation.
Collapse
|
24
|
Manzamine A Exerts Anticancer Activity against Human Colorectal Cancer Cells. Mar Drugs 2018; 16:md16080252. [PMID: 30060617 PMCID: PMC6117705 DOI: 10.3390/md16080252] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/24/2018] [Accepted: 07/27/2018] [Indexed: 01/06/2023] Open
Abstract
Marine sponges are known to produce numerous bioactive secondary metabolites as defense strategies to avoid predation. Manzamine A is a sponge-derived β-carboline-fused pentacyclic alkaloid with various bioactivities, including recently reported anticancer activity on pancreatic cancer. However, its cytotoxicity and mode of action against other tumors remain unclear. In this study, we exhibit that manzamine A reduced cell proliferation in several colorectal cancer (CRC) cell lines. To further investigate the manzamine A triggered molecular regulation, we analyzed the gene expression with microarray and revealed that pathways including cell cycle, DNA repair, mRNA metabolism, and apoptosis were dysregulated. We verified that manzamine A induced cell cycle arrest at G0/G1 phase via inhibition of cyclin-dependent kinases by p53/p21/p27 and triggered a caspase-dependent apoptotic cell death through mitochondrial membrane potential depletion. Additionally, we performed bioinformatics analysis and demonstrated that manzamine A abolished epithelial–mesenchymal transition process. Several mesenchymal transcriptional factors, such as Snail, Slug, and Twist were suppressed and epithelial marker E-cadherin was induced simultaneously in HCT116 cells by manzamine A, leading to the epithelial-like phenotype and suppression of migration. These findings suggest that manzamine A may serve as a starting point for the development of an anticancer drug for the treatment of metastatic CRC.
Collapse
|
25
|
Verma AK, Ghosh S, Basu A. Chandipura Virus Induced Neuronal Apoptosis via Calcium Signaling Mediated Oxidative Stress. Front Microbiol 2018; 9:1489. [PMID: 30034380 PMCID: PMC6043780 DOI: 10.3389/fmicb.2018.01489] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/15/2018] [Indexed: 01/06/2023] Open
Abstract
Chandipura Virus (CHPV) a negative-stranded RNA virus belonging to the Rhabdoviridae family, has been previously reported to bring about neuronal apoptosis by stimulating oxidative stress. Our in silico data suggested the involvement of Angiotensin II in intracellular Ca2+ secretion within CHPV infected cells that further lead to enhancement of ROS level and mitochondrial dysfunction. ROS is also known to phosphorylate p38 that leads to neuronal apoptosis through FasL-FADD pathway during CHPV infection. Minocycline a broad-spectrum antibiotic well-known for its anti-oxidative and anti-inflammatory role was used in the present study to investigate its efficacy against CHPV. The results obtained from the present study showed minocycline to be effective in mitigating the levels of cytoplasmic Ca2+, ROS, phosphorylation of p38 molecules and hence cellular apoptosis. Thus minocycline apart from being an anti-inflammatory and anti-oxidative agent, our study showed that minocycline has an additional Ca2+ chelation activity.
Collapse
|
26
|
Li A, Wang Q, He G, Jin J, Huang G. DEP domain containing 1 suppresses apoptosis via inhibition of A20 expression, which activates the nuclear factor κB signaling pathway in HepG2 cells. Oncol Lett 2018; 16:949-955. [PMID: 29963168 PMCID: PMC6019891 DOI: 10.3892/ol.2018.8770] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 03/26/2018] [Indexed: 12/21/2022] Open
Abstract
A previous study revealed that DEP domain containing 1 (DEPDC1) is involved in the carcinogenesis of bladder cancer via forming a complex with zinc finger protein 224 (ZNF224) to suppress A20 expression, resulting in the activation of the nuclear factor (NF)-κB signaling pathway; however, the role of DEPDC1 in liver cancer remains unclear. Hep G2 cells were treated with 11R-DEP: 611–628, a peptide capable of disrupting the DEPDC1-ZNF224 complex. Cell proliferation was examined using an MTT assay and apoptosis was analyzed via detection of the apoptotic marker caspase-3 using western blot analysis. A20 expression was examined via reverse transcription-quantitative polymerase chain reaction and NF-κB subcellular localization was determined via immunofluorescence staining. microRNA (miR)-130a was overexpressed in HepG2 cells and its effects on proliferation and apoptosis were examined. The results demonstrated that 11R-DEP: 611–628 (3 µM) and miR-130a inhibited cell proliferation and promoted apoptosis in HepG2 cells by activating A20 expression, which blocks the nuclear transportation of NF-κB. In addition, the results demonstrated that the 11R-DEP: 611–628 (3 µM) treatment resulted in downregulation of DEPDC1 expression, indicating that DEPDC1 expression is regulated by the DEPDC1-ZNF224 complex. In conclusion, the data indicated that DEPDC1 suppresses apoptosis to promote cell proliferation through the NF-κB signaling pathway in HepG2 cells and that DEPDC1 is a potential target for the treatment of liver cancer.
Collapse
Affiliation(s)
- Aili Li
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Qingqing Wang
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Gaofeng He
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, Guangxi 541001, P.R. China.,Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Junfei Jin
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, Guangxi 541001, P.R. China.,Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Guojin Huang
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China.,China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| |
Collapse
|
27
|
Su L, Jiang Y, Xu Y, Li X, Gao W, Xu C, Zeng C, Song J, Weng W, Liang W. Xihuang pill promotes apoptosis of Treg cells in the tumor microenvironment in 4T1 mouse breast cancer by upregulating MEKK1/SEK1/JNK1/AP-1 pathway. Biomed Pharmacother 2018; 102:1111-1119. [PMID: 29710529 DOI: 10.1016/j.biopha.2018.03.063] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/09/2018] [Accepted: 03/11/2018] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE To determine the role of the MEKK1/SEK1/JNK1/AP-1 pathway in the action of Xihuang pill (XHP) in reducing regulatory T (Treg) cell numbers in the tumor microenvironment in a 4T1 mouse breast cancer model, and to clarify the anti-tumor mechanism of XHP in breast cancer. METHODS We established a mouse 4T1 breast cancer model. Model mice were administered XHP for 2 weeks, and tumor tissues were then removed, weighed, sliced, and homogenized. Treg cells in the tumor microenvironment were isolated by magnetic cell sorting and analyzed by immunohistochemistry and flow cytometry. Treg cell apoptosis was detected by TdT-mediated dUTP nick end labeling. mRNA expression levels of MEKK1, SEK1, JNK1, and AP-1 in Treg cells in the tumor microenvironment were detected by quantitative real-time PCR and their protein expression levels were detected by immunofluorescence staining and western blot. RESULTS Tumor weights were significantly lower in the XHP groups compared with the untreated control group. The overall number of Treg cells in the tumor microenvironment decreased while the number of apoptotic Treg cells increased with increasing doses of XHP. mRNA and protein expression levels of MEKK1, SEK1, JNK1, and AP-1 in Treg cells in the tumor microenvironment increased with increasing doses of XHP. CONCLUSION XHP might promote Treg cell apoptosis in the tumor microenvironment and further inhibit the tumor growth of 4T1 mouse breast cancer. The mechanism of XHP may be related to upregulation of gene and protein expression of MEKK1, SEK1, JNK1, and AP-1 in Treg cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Liang Su
- Xin Hua Affiliated Hospital of Dalian University, Dalian 116000, China
| | - Yiming Jiang
- Xin Hua Affiliated Hospital of Dalian University, Dalian 116000, China
| | - Yu Xu
- Medical College of Dalian University, Dalian 116622, China
| | - Xinye Li
- Medical College of Dalian University, Dalian 116622, China
| | - Wenbin Gao
- Department of Medical Oncology, The 3rd Affiliated Hospital of Shenzhen University, Shenzhen 518001, China
| | - Chunwei Xu
- Department of Pathology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, China
| | - Changqian Zeng
- Medical College of Dalian University, Dalian 116622, China
| | - Jie Song
- Medical College of Dalian University, Dalian 116622, China
| | - Wencai Weng
- Xin Hua Affiliated Hospital of Dalian University, Dalian 116000, China
| | - Wenbo Liang
- Medical College of Dalian University, Dalian 116622, China.
| |
Collapse
|
28
|
Morgan R, El-Tanani M, Hunter KD, Harrington KJ, Pandha HS. Targeting HOX/PBX dimers in cancer. Oncotarget 2018; 8:32322-32331. [PMID: 28423659 PMCID: PMC5458287 DOI: 10.18632/oncotarget.15971] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/23/2017] [Indexed: 12/30/2022] Open
Abstract
The HOX and PBX gene families encode transcription factors that have key roles in establishing the identity of cells and tissues in early development. Over the last 20 years it has become apparent that they are also dysregulated in a wide range of solid and haematological malignancies and have a predominantly pro-oncogenic function. A key mode of transcriptional regulation by HOX and PBX proteins is through their interaction as a heterodimer or larger complex that enhances their binding affinity and specificity for DNA, and there is growing evidence that this interaction is a potential therapeutic target in malignancies that include prostate, breast, renal, ovarian and lung cancer, melanoma, myeloma, and acute myeloid leukaemia. This review summarizes the roles of HOX and PBX genes in cancer and assesses the therapeutic potential of HOX/PBX dimer inhibition, including the availability of biomarkers for its application in precision medicine.
Collapse
Affiliation(s)
- Richard Morgan
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, UK
| | - Mohamed El-Tanani
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, UK
| | - Keith D Hunter
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Kevin J Harrington
- Targeted Therapy Team, Chester Beatty Laboratories, The Institute of Cancer Research, London, UK
| | - Hardev S Pandha
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
29
|
Al-Saeedi M, Steinebrunner N, Kudsi H, Halama N, Mogler C, Büchler MW, Krammer PH, Schemmer P, Müller M. Neutralization of CD95 ligand protects the liver against ischemia-reperfusion injury and prevents acute liver failure. Cell Death Dis 2018; 9:132. [PMID: 29374146 PMCID: PMC5833836 DOI: 10.1038/s41419-017-0150-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/11/2022]
Abstract
Ischemia-reperfusion injury is a common pathological process in liver surgery and transplantation, and has considerable impact on the patient outcome and survival. Death receptors are important mediators of ischemia-reperfusion injury, notably the signaling pathways of the death receptor CD95 (Apo-1/Fas) and its corresponding ligand CD95L. This study investigates, for the first time, whether the inhibition of CD95L protects the liver against ischemia-reperfusion injury. Warm ischemia was induced in the median and left liver lobes of C57BL/6 mice for 45 min. CD95Fc, a specific inhibitor of CD95L, was applied prior to ischemia. Hepatic injury was assessed via consecutive measurements of liver serum enzymes, histopathological assessment of apoptosis and necrosis and caspase assays at 3, 6, 12, 18 and 24 h after reperfusion. Serum levels of liver enzymes, as well as characteristic histopathological changes and caspase assays indicated pronounced features of apoptotic and necrotic liver damage 12 and 24 h after ischemia-reperfusion injury. Animals treated with the CD95L-blocker CD95Fc, exhibited a significant reduction in the level of serum liver enzymes and showed both decreased histopathological signs of parenchymal damage and decreased caspase activation. This study demonstrates that inhibition of CD95L with the CD95L-blocker CD95Fc, is effective in protecting mice from liver failure due to ischemia-reperfusion injury of the liver. CD95Fc could therefore emerge as a new pharmacological therapy for liver resection, transplantation surgery and acute liver failure.
Collapse
Affiliation(s)
- Mohammed Al-Saeedi
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Niels Steinebrunner
- Department of Gastroenterology, Intoxications, and Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Hassan Kudsi
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Niels Halama
- Medical Oncology, National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Carolin Mogler
- Department of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus W Büchler
- Department of General, Visceral, and Transplant Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter H Krammer
- Division of Immunogenetics, German Cancer Research Center, Heidelberg, Germany
| | - Peter Schemmer
- Department of Surgery, Division of Transplant Surgery, Medical University of Graz, Graz, Austria.
| | - Martina Müller
- Department of Internal Medicine I, Gastroenterology, Endocrinology, Rheumatology, and Infectious Diseases, Regensburg University Hospital, Regensburg, Germany
| |
Collapse
|
30
|
Li J, Liu Y, Duan P, Yu R, Gu Z, Li L, Liu Z, Su L. NF‑κB regulates HSF1 and c‑Jun activation in heat stress‑induced intestinal epithelial cell apoptosis. Mol Med Rep 2017; 17:3388-3396. [PMID: 29257252 DOI: 10.3892/mmr.2017.8199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 07/13/2017] [Indexed: 11/05/2022] Open
Abstract
Heat stress may induce intestinal epithelial cell apoptosis; however, the molecular mechanisms have not yet been identified. The present study used IEC‑6 rat small intestinal epithelial cells to investigate heat stress‑induced production of reactive oxygen species (ROS), which may be involved in nuclear factor (NF)‑κB activation during heat stress. IEC‑6 cells were transfected with NF‑κB p65‑specific small interfering RNA (siRNA), and observed a significant increase in cell apoptosis and caspase‑3 cleavage; however, in cells transfected with adenovirus that constitutively overexpressed p65, the opposite results were obtained. Furthermore, p65 knockdown increased the heat stress‑induced expression and activity of heat shock transcription factor 1 (HSF1); conversely, p65 overexpression slightly decreased HSF1 activity. The levels of heat stress‑induced c‑Jun phosphorylation were also examined: Knockdown of p65 resulted in a reduction of c‑Jun phosphorylation, whereas p65 overexpression resulted in increased phosphorylation. Furthermore, siRNA‑mediated knockdown of HSF1 in IEC‑6 cells significantly increased heat stress‑induced apoptosis. Cells pretreated with c‑Jun peptide, an inhibitor of c‑Jun activation, exhibited a significant reduction in apoptosis. These findings indicated that heat stress stimulation in IEC‑6 cells induced the pro‑apoptotic role of NF‑κB by regulating HSF1 and c‑Jun activation.
Collapse
Affiliation(s)
- Jun Li
- Graduate School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yanan Liu
- Department of Intensive Care Unit, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Pengkai Duan
- Department of Intensive Care Unit, Key Laboratory of Tropical Zone Trauma Care and Tissue Repair of PLA, General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Rongguo Yu
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, Fujian 350000, P.R. China
| | - Zhengtao Gu
- Department of Intensive Care Unit, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Li Li
- Department of Intensive Care Unit, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhifeng Liu
- Department of Intensive Care Unit, Key Laboratory of Tropical Zone Trauma Care and Tissue Repair of PLA, General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Lei Su
- Graduate School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
31
|
Baicalin hydrate inhibits cancer progression in nasopharyngeal carcinoma by affecting genome instability and splicing. Oncotarget 2017; 9:901-914. [PMID: 29416665 PMCID: PMC5787522 DOI: 10.18632/oncotarget.22868] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/20/2017] [Indexed: 01/08/2023] Open
Abstract
Baicalin hydrate (BH), a natural compound, has been investigated for many years because of its traditional medicinal properties. However, the anti-tumor activities of BH and its epigenetic role in NPC have not been elucidated. In this study, we identified that BH inhibits NPC cell growth in vivo and in vitro by inducing apoptosis and cell cycle arrest. BH epigenetically regulated genome instability by up-regulating the expression of satellite 2 (Sat2), alpha satellite (α-Sat), and major satellite (Major-Sat). BH also increased the level of IKKα, Suv39H1, and H3K9me3 and decreased LSH expression. Interestingly, BH promoted the splicing of Suv39H1 via the enhancement of m6A RNA methylation, rather than DNA methylation. Taken together, our results demonstrated that BH has an anti-tumor role in NPC and revealed a unique role of BH in genome instability and splicing in response to DNA damage.
Collapse
|
32
|
Zhang Q, Gupta S, Schipper DL, Kowalczyk GJ, Mancini AE, Faeder JR, Lee REC. NF-κB Dynamics Discriminate between TNF Doses in Single Cells. Cell Syst 2017; 5:638-645.e5. [PMID: 29128333 DOI: 10.1016/j.cels.2017.10.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/30/2017] [Accepted: 10/13/2017] [Indexed: 01/28/2023]
Abstract
Although cytokine-dependent dynamics of nuclear factor κB (NF-κB) are known to encode information that regulates cell fate decisions, it is unclear whether single-cell responses are switch-like or encode more information about cytokine dose. Here, we measure the dynamic subcellular localization of NF-κB in response to a range of tumor necrosis factor (TNF) stimulation conditions to determine the prevailing mechanism of single-cell dose discrimination. Using an information theory formalism that accounts for signaling dynamics and non-responsive cell subpopulations, we find that the information transmission capacity of single cells exceeds that predicted from a switch-like response. Instead, we observe that NF-κB dynamics within single cells contain sufficient information to encode multiple, TNF-dependent cellular states, and have an activation threshold that varies across the population. By comparing single-cell responses to an internal, experimentally observed reference, we demonstrate that cells can grade responses to TNF across several orders of magnitude in concentration. This suggests that cells contain additional control points to fine-tune their cytokine responses beyond the decision to activate.
Collapse
Affiliation(s)
- Qiuhong Zhang
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sanjana Gupta
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David L Schipper
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Gabriel J Kowalczyk
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Allison E Mancini
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - James R Faeder
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Robin E C Lee
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
33
|
Marine Sponge Natural Products with Anticancer Potential: An Updated Review. Mar Drugs 2017; 15:md15100310. [PMID: 29027954 PMCID: PMC5666418 DOI: 10.3390/md15100310] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/28/2017] [Accepted: 10/09/2017] [Indexed: 12/11/2022] Open
Abstract
Despite the huge investment into research and the significant effort and advances made in the search for new anticancer drugs in recent decades, cancer cure and treatment continue to be a formidable challenge. Many sources, including plants, animals, and minerals, have been explored in the oncological field because of the possibility of identifying novel molecular therapeutics. Marine sponges are a prolific source of secondary metabolites, a number of which showed intriguing tumor chemopreventive and chemotherapeutic properties. Recently, Food and Drug Administration-approved drugs derived from marine sponges have been shown to reduce metastatic breast cancer, malignant lymphoma, and Hodgkin's disease. The chemopreventive and potential anticancer activity of marine sponge-derived compounds could be explained by multiple cellular and molecular mechanisms, including DNA protection, cell-cycle modulation, apoptosis, and anti-inflammatory activities as well as their ability to chemosensitize cancer cells to traditional antiblastic chemotherapy. The present article aims to depict the multiple mechanisms involved in the chemopreventive and therapeutic effects of marine sponges and critically explore the limitations and challenges associated with the development of marine sponge-based anticancer strategy.
Collapse
|
34
|
Zou H, Su R, Ruan J, Shao H, Qian K, Ye J, Yao Y, Nair V, Qin A. Double-stranded RNA induces chicken T-cell lymphoma apoptosis by TRIF and NF-κB. Sci Rep 2017; 7:7547. [PMID: 28790362 PMCID: PMC5548913 DOI: 10.1038/s41598-017-07919-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 05/10/2017] [Indexed: 12/20/2022] Open
Abstract
Toll-like receptor-3 (TLR3), a member of the pathogen recognition receptor family, has been reported to activate immune response and to exhibit pro-apoptotic activity against some tumor cells. However it is unclear whether TLR3 has same function against chicken lymphoma. In this paper we investigated the effect of TLR3 activation on a Marek’s disease lymphoma-derived chicken cell line, MDCC-MSB1. The TLR3 agonist poly (I:C) activated TLR3 pathway and inhibited tumor cells proliferation through caspase-dependent apoptosis. Using pharmacological approaches, we found that an interferon-independent mechanism involving Toll-IL-1-receptor domain-containing adapter-inducing IFN-α (TRIF) and nuclear factor κB (NF-κB) causes the apoptosis of MDCC-MSB1 cells. This is the first report about the function of TLR3 in chicken T-cell lymphoma, especially in signal pathway. The mechanisms underlying TLR3-mediated apoptosis may contribute to the development of new drug to treat lymphomas and oncovirus infections.
Collapse
Affiliation(s)
- Haitao Zou
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China
| | - Ruixue Su
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China
| | - Jing Ruan
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China
| | - Hongxia Shao
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,Jiangsu Key Lab of Zoonosis, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China
| | - Kun Qian
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,Jiangsu Key Lab of Zoonosis, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,UK-China Centre of Excellence for Research on Avian Diseases, 169 Huanghe 2nd Road, Binzhou, Shandong, P. R. China
| | - Jianqiang Ye
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China.,Jiangsu Key Lab of Zoonosis, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China
| | - Yongxiu Yao
- The Pirbright Institute, Ash road, Pirbright, Working, Surrey, GU24 0NF, United Kingdom.,UK-China Centre of Excellence for Research on Avian Diseases, 169 Huanghe 2nd Road, Binzhou, Shandong, P. R. China
| | - Venugopal Nair
- The Pirbright Institute, Ash road, Pirbright, Working, Surrey, GU24 0NF, United Kingdom.,UK-China Centre of Excellence for Research on Avian Diseases, 169 Huanghe 2nd Road, Binzhou, Shandong, P. R. China
| | - Aijian Qin
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China. .,Jiangsu Key Lab of Zoonosis, No. 12 East Wenhui Road, Yangzhou, Jiangsu, 225009, P. R. China. .,UK-China Centre of Excellence for Research on Avian Diseases, 169 Huanghe 2nd Road, Binzhou, Shandong, P. R. China.
| |
Collapse
|
35
|
Rasool RU, Nayak D, Chakraborty S, Faheem MM, Rah B, Mahajan P, Gopinath V, Katoch A, Iqra Z, Yousuf SK, Mukherjee D, Kumar LD, Nargotra A, Goswami A. AKT is indispensable for coordinating Par-4/JNK cross talk in p21 downmodulation during ER stress. Oncogenesis 2017; 6:e341. [PMID: 28530706 PMCID: PMC5523074 DOI: 10.1038/oncsis.2017.41] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/22/2017] [Accepted: 04/17/2017] [Indexed: 12/25/2022] Open
Abstract
The double-edged role of p21 to command survival and apoptosis is emerging. The current investigation highlights ER stress-mediated JNK activation that plausibly triggers cell death by attenuating endogenous p21 level. Here, we demonstrated that ER stress activator 3-AWA diminishes the p21 levels in cancer cells by averting the senescent phenotype to commence G2/M arrest. In essence, the deceleration in p21 level occurs through ER stress/JNK/Caspase-3 axis via activation/induction of proapoptotic Par-4 and inhibition of AKT. The molecular dynamics studies identified important interactions, which may be responsible for the AKT inhibition and efficacy of 3-AWA towards AKT binding pocket. Interestingly, the p21 deceleration was rescued by incubating the cells with 3-AWA in the presence of an ER stress inhibitor, Salubrinal. Furthermore, we demonstrated that p21 expression decreases solitarily in Par-4+/+ MEFs; albeit, ER stress-induced JNK activation was observed in both Par-4+/+ and Par-4−/− MEFs. Par-4 knockdown or overexpression studies established that ectopic Par-4 along with ER stress are not sufficient to downregulate p21 in PC-3 cells but are adequate for DU-145 cells and that the ER stress inflicted activation of JNK, inhibition of AKT and Par-4 induction are all crucial to p21 downmodulation by 3-AWA. By using isogenic cell lines, such as HCT-116 p53+/+ and HCT-116 p53−/−, we found that deceleration in p21 expression due to ER stress is p53 independent. Moreover, in orthotopic carcinogen-induced rat colorectal carcinoma model, we found that 3-AWA inhibits colorectal tumor growth and formation of colorectal polyps at a tolerable dose, similar to the first-line drug for colorectal cancer-5-fluorouracil.
Collapse
Affiliation(s)
- R U Rasool
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - D Nayak
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - S Chakraborty
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - M M Faheem
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - B Rah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - P Mahajan
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - V Gopinath
- Cancer Biology Division, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - A Katoch
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Z Iqra
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - S K Yousuf
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - D Mukherjee
- Natural Product Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - L D Kumar
- Cancer Biology Division, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - A Nargotra
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - A Goswami
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| |
Collapse
|
36
|
Krishnaraj J, Kowshik J, Sebastian R, Raghavan SC, Nagini S. Exposure to welding fumes activates DNA damage response and redox-sensitive transcription factor signalling in Sprague-Dawley rats. Toxicol Lett 2017; 274:8-19. [DOI: 10.1016/j.toxlet.2017.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/30/2017] [Accepted: 04/03/2017] [Indexed: 10/19/2022]
|
37
|
Liver receptor homolog-1 (NR5a2) regulates CD95/Fas ligand transcription and associated T-cell effector functions. Cell Death Dis 2017; 8:e2745. [PMID: 28406481 PMCID: PMC5477591 DOI: 10.1038/cddis.2017.173] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 01/25/2023]
Abstract
CD95/Fas ligand (FasL) is a cell death-promoting member of the tumor necrosis factor family with important functions in the regulation of T-cell homeostasis and cytotoxicity. In T cells, FasL expression is tightly regulated on a transcriptional level involving a complex set of different transcription factors. The orphan nuclear receptor liver receptor homolog-1 (LRH-1/NR5a2) is involved in the regulation of development, lipid metabolism and proliferation and is predominantly expressed in epithelial tissues. However, its expression in T lymphocytes has never been reported so far. Based on in silico analysis, we identified potential LRH-1 binding sites within the FASLG promoter. Here, we report that LRH-1 is expressed in primary and secondary lymphatic tissues, as well as in CD4+ and CD8+ T cells. LRH-1 directly binds to its binding sites in the FASLG promoter, and thereby drives FASLG promoter activity. Mutations in the LRH-1 binding sites reduce FASLG promoter activity. Pharmacological inhibition of LRH-1 decreases activation-induced FasL mRNA expression, as well as FasL-mediated activation-induced T-cell apoptosis and T-cell cytotoxicity. In a mouse model of Concanavalin A-induced and FasL-mediated hepatitis pharmacological inhibition of LRH-1 resulted in decreased hepatic FasL expression and a significant reduction of liver damage. In summary, these data show for the first time LRH-1 expression in T cells, its role in FASLG transcription and the potential of pharmacological inhibition of LRH-1 in the treatment of FasL-mediated immunopathologies.
Collapse
|
38
|
Ge QL, Liu SH, Ai ZH, Tao MF, Ma L, Wen SY, Dai M, Liu F, Liu HS, Jiang RZ, Xue ZW, Jiang YH, Sun XH, Hu YM, Zhao YX, Chen X, Tao Y, Zhu XL, Ding WJ, Yang BQ, Liu DD, Zhang XR, Teng YC. RelB/NF-κB links cell cycle transition and apoptosis to endometrioid adenocarcinoma tumorigenesis. Cell Death Dis 2016; 7:e2402. [PMID: 27711077 PMCID: PMC5133976 DOI: 10.1038/cddis.2016.309] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 08/18/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
Abstract
Dysfunction of nuclear factor-κB (NF-κB) signaling has been causally associated with numerous human malignancies. Although the NF-κB family of genes has been implicated in endometrial carcinogenesis, information regarding the involvement of central regulators of NF-κB signaling in human endometrial cancer (EC) is limited. Here, we investigated the specific roles of canonical and noncanonical NF-κB signaling in endometrial tumorigenesis. We found that NF-κB RelB protein, but not RelA, displayed high expression in EC samples and cell lines, with predominant elevation in endometrioid adenocarcinoma (EEC). Moreover, tumor cell-intrinsic RelB was responsible for the abundant levels of c-Myc, cyclin D1, Bcl-2 and Bcl-xL, which are key regulators of cell cycle transition, apoptosis and proliferation in EEC. In contrast, p27 expression was enhanced by RelB depletion. Thus, increased RelB in human EC is associated with enhanced EEC cell growth, leading to endometrial cell tumorigenicity. Our results reveal that regulatory RelB in noncanonical NF-κB signaling may serve as a therapeutic target to block EC initiation.
Collapse
Affiliation(s)
- Qiu-Lin Ge
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - San-Hong Liu
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Hong Ai
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - Min-Fang Tao
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - Li Ma
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - Shan-Yun Wen
- Department of Obstetrics and Gynecology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Miao Dai
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - Fei Liu
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - Han-Shao Liu
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong-Zhen Jiang
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - Zhuo-Wei Xue
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Hang Jiang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Hua Sun
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Ming Hu
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Xu Zhao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Chen
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Tao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Lu Zhu
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - Wen-Jing Ding
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Bing-Qing Yang
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| | - Dan-Dan Liu
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Ren Zhang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yin-Cheng Teng
- Department of Obstetrics and Gynecology, The Sixth People's Hospital affiliated with Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
39
|
Abstract
Background: Apoptosis, or programmed cell death, is an essential physiological process that controls cell numbers during physiological processes, and eliminates abnormal cells that can potentially harm an organism. Objective: This review summarizes our current state of knowledge of apoptosis induction in skin by UV radiation. Methods: A review of the literature was undertaken focusing on cell death in the skin secondary to UV radiation. Results: It is evident that a number of apoptotic pathways, both intrinsic and extrinsic, are induced following exposure to damaging UV radiation. Conclusion: Although our understanding of the apoptotic processes is gradually increasing, many important aspects remain obscure. These include interconnections between pathways, wavelength-specific differences and cell type differences.
Collapse
Affiliation(s)
- Jeffrey Chow
- Department of Laboratory Medicine and Pathology, 4B1.19 Walter C Mackenzie Health Science Centre, University of Alberta, 8440-112th Street, Edmonton, AB, CanadaT6G 2B7
| | - Victor A. Tron
- Department of Laboratory Medicine and Pathology, 4B1.19 Walter C Mackenzie Health Science Centre, University of Alberta, 8440-112th Street, Edmonton, AB, CanadaT6G 2B7
| |
Collapse
|
40
|
Apoptosis induced by temozolomide and nimustine in glioblastoma cells is supported by JNK/c-Jun-mediated induction of the BH3-only protein BIM. Oncotarget 2016; 6:33755-68. [PMID: 26418950 PMCID: PMC4741800 DOI: 10.18632/oncotarget.5274] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/03/2015] [Indexed: 12/16/2022] Open
Abstract
The outcome of cancer therapy strongly depends on the complex network of cell signaling pathways, including transcription factor activation following drug exposure. Here we assessed whether and how the MAP kinase (MAPK) cascade and its downstream target, the transcription factor AP-1, influence the sensitivity of malignant glioma cells to the anticancer drugs temozolomide (TMZ) and nimustine (ACNU). Both drugs induce apoptosis in glioma cells at late times following treatment. Activation of the MAPK cascade precedes apoptosis, as shown by phosphorylation of Jun kinase (JNK) and c-Jun, a main component of AP-1. Pharmacological inhibition and siRNA mediated knockdown of JNK and c-Jun reduced the level of apoptosis in LN-229 glioma cells treated with TMZ or ACNU. Analyzing the underlying molecular mechanism, we identified the pro-apoptotic gene BIM as a critical target of AP-1, which is upregulated following TMZ and ACNU. Importantly, shRNA mediated downregulation of BIM in the malignant glioma cell lines LN-229 and U87MG led to an attenuated cleavage of caspase-9 and, consequently, reduced the level of apoptosis following TMZ and ACNU treatment. Overall, we identified JNK/c-Jun activation and BIM induction as a late pro-apoptotic response of glioma cells treated with alkylating anticancer drugs.
Collapse
|
41
|
Guanidine Alkaloids from the Marine Sponge Monanchora pulchra Show Cytotoxic Properties and Prevent EGF-Induced Neoplastic Transformation in Vitro. Mar Drugs 2016; 14:md14070133. [PMID: 27428983 PMCID: PMC4962023 DOI: 10.3390/md14070133] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/05/2016] [Accepted: 07/08/2016] [Indexed: 12/13/2022] Open
Abstract
Guanidine alkaloids from sponges Monanchora spp. represent diverse bioactive compounds, however, the mechanisms underlying bioactivity are very poorly understood. Here, we report results of studies on cytotoxic action, the ability to inhibit EGF-induced neoplastic transformation, and the effects on MAPK/AP-1 signaling of eight rare guanidine alkaloids, recently isolated from the marine sponge Monanchora pulchra, namely: monanchocidin A (1), monanchocidin B (2), monanchomycalin C (3), ptilomycalin A (4), monanchomycalin B (5), normonanchocidin D (6), urupocidin A (7), and pulchranin A (8). All of the compounds induced cell cycle arrest (apart from 8) and programmed death of cancer cells. Ptilomycalin A-like compounds 1–6 activated JNK1/2 and ERK1/2, following AP-1 activation and caused p53-independent programmed cell death. Compound 7 induced p53-independent cell death without activation of AP-1 or caspase-3/7, and the observed JNK1/2 activation did not contribute to the cytotoxic effect of the compound. Alkaloid 8 induced JNK1/2 (but not ERK1/2) activation leading to p53-independent cell death and strong suppression of AP-1 activity. Alkaloids 1–4, 7, and 8 were able to inhibit the EGF-induced neoplastic transformation of JB6 P+ Cl41 cells. Our results suggest that investigated guanidine marine alkaloids hold potential to eliminate human cancer cells and prevent cancer cell formation and spreading.
Collapse
|
42
|
Herdegen T, Mielke K, Kallunki T. Review : c-Jun and the c-Jun Amino-Terminal Kinases: Bipotential Components of the Neuronal Stress Response. Neuroscientist 2016. [DOI: 10.1177/107385849900500311] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Expression of the inducible transcription factor c-Jun in neurons is a common finding after neuronal injury or 'stress,' such as ischemia, excitotoxicity, axon transection, UV irradiation, stimulation by cytokines, or production of such lipid messengers as ceramide. The neuronal 'stress response' displays striking similarities to the stress response of other cell types such as lymphocytes or tumor cells and is characterized by the activation of programs that lead to apoptosis or survival. It is accepted knowledge that c-Jun can act as neuronal 'killer' under in vitro conditions (with the death inducing ligand fas-ligand as novel AP-1 controlled target gene), but there is also growing evidence that c-Jun is linked to neuronal repair or survival. The control of this dichotomous function of c-Jun is not fully understood. Similar to the expression of c-Jun, the transcriptional activation of c-Jun by amino-terminal phosphorylation and the activation of the catalyzing c- Jun amino-terminal kinases (JNK), also called stress activated protein kinases, can also be linked to both neuronal survival and apoptosis. We suggest a model for the control of gene transcription after neuronal stress with activation of JNK and phosphorylation of c-Jun as transcriptional prerequisites, and with asso ciated partners as transcriptional effectors, e.g., by the expression and/or suppression of other transcription factors as activating transcription factor 2 (ATF-2), c-Fos, or JunD. This scenario is complicated by the observation that activity of JNK does not lead automatically to c-Jun phosphorylation. This review summa rizes the role of c-Jun and JNK as down-stream mediators of neuronal stressors and places the function of these molecules in the context of other stressful stimuli and intraneuronal responses. NEUROSCIENTIST 5:147-154, 1999
Collapse
Affiliation(s)
| | - Kirsten Mielke
- Department of Pharmacology University of Kiel Kiel,
Germany
| | - Tuula Kallunki
- Department of Pharmacology University of San Diego La
Jolla, California
| |
Collapse
|
43
|
Muxel SM, Laranjeira-Silva MF, Carvalho-Sousa CE, Floeter-Winter LM, Markus RP. The RelA/cRel nuclear factor-κB (NF-κB) dimer, crucial for inflammation resolution, mediates the transcription of the key enzyme in melatonin synthesis in RAW 264.7 macrophages. J Pineal Res 2016; 60:394-404. [PMID: 26887983 DOI: 10.1111/jpi.12321] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/12/2016] [Indexed: 12/14/2022]
Abstract
Lipopolysaccharide (LPS) modulates the transcription of the gene that codifies the enzyme arylalkylamine-N-acetyltransferase (AA-NAT) through nuclear translocation of the transcription factor nuclear factor-κ-light-chain-enhancer of activated B cells (NF-κB). AA-NAT converts serotonin to N-acetylserotonin, the ultimate precursor of melatonin. Activation of kappa B elements (aa-nat-κB), localized in the promoter (nat-κB1 and nat-κB2), leads to Aa-nat transcription in RAW 264.7 macrophages. Competitive electrophoretic mobility shift assay (EMSA) with oligonucleotide probes corresponding to each of the two elements, as well as a NF-κB consensus corresponding probe, revealed different specificities for each κB element. In addition, activator protein-1 (AP-1) as well as signal transducers and activator of transcription-1 and 3 (STAT-1; STAT-3) competed with NF-κB for binding to nat-κB1, while only STAT-3 competed with NF-κB for binding to nat-κB2. According to co-immunoprecipitation (ChiP) assays, these two sites are able to distinguish NF-κB subunits. The sequence nat-κB1 bound dimers containing p52, RelA, and cRel, while nat-κB2 bound preferentially p50, p52, and RelA, and did not bind cRel. The expression of RelA and cRel is essential for the induction of Aa-nat expression and melatonin synthesis. Considering that the expression of cRel is induced by the earlier expressed p50/RelA, the differential effects of NF-κB dimers may be intimately associated with the temporal regulation of inflammatory responses, with the resolution phase being associated with paracrine and autocrine melatonin effects. Such data suggest that the proven effects of exogenous melatonin in the resolution phase of inflammation are paralleled by the effects of locally synthesized melatonin in immune cells.
Collapse
Affiliation(s)
- Sandra Marcia Muxel
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | - Regina P Markus
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
44
|
Nuclear Transcription Factor Kappa B Downregulation Reduces Chemoresistance in Bone Marrow-derived Cells Through P-glycoprotein Modulation. Arch Med Res 2016; 47:78-88. [DOI: 10.1016/j.arcmed.2016.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 05/20/2016] [Indexed: 01/03/2023]
|
45
|
Ishikura S, Ogawa M, Doi K, Matsuzaki H, Iwaihara Y, Tanaka Y, Tsunoda T, Hideshima H, Okamura T, Shirasawa S. Zfat-deficient CD4⁺ CD8⁺ double-positive thymocytes are susceptible to apoptosis with deregulated activation of p38 and JNK. J Cell Biochem 2016; 116:149-57. [PMID: 25169027 DOI: 10.1002/jcb.24954] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/22/2014] [Indexed: 11/08/2022]
Abstract
Zfat, which is a nuclear protein harboring an AT-hook domain and 18-repeats of C2H2 zinc-finger motif, is highly expressed in immune-related tissues, including the thymus and spleen. T cell specific deletion of the Zfat gene by crossing Zfat(f/f) mice with LckCre mice yields a significant reduction in the number of CD4(+) CD8(+) double-positive (DP) thymocytes. However, physiological role for Zfat in T cell development in the thymus remains unknown. Here, we found that Zfat-deficient DP thymocytes in Zfat(f/f)-LckCre mice were susceptible to apoptosis both at an unstimulated state and in response to T cell receptor (TCR)-stimulation. The phosphorylation levels of p38 and JNK were elevated in Zfat-deficient thymocytes at an unstimulated state with an enhanced phosphorylation of ATF2 and with an over-expression of Gadd45α⋅ On the other hand, the activation of JNK in the Zfat-deficient thymocytes, but not p38, was strengthened and prolonged in response to TCR-stimulation. All these results demonstrate that Zfat critically participates in the development of DP thymocytes through regulating the activities of p38 and JNK.
Collapse
Affiliation(s)
- Shuhei Ishikura
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Central Research Institute for Advanced Molecular Medicine, Fukuoka University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yu X, Deng Q, Li W, Xiao L, Luo X, Liu X, Yang L, Peng S, Ding Z, Feng T, Zhou J, Fan J, Bode AM, Dong Z, Liu J, Cao Y. Neoalbaconol induces cell death through necroptosis by regulating RIPK-dependent autocrine TNFα and ROS production. Oncotarget 2015; 6:1995-2008. [PMID: 25575821 PMCID: PMC4385831 DOI: 10.18632/oncotarget.3038] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/02/2015] [Indexed: 02/04/2023] Open
Abstract
Necroptosis/regulated necrosis is a caspase-independent, but receptor interacting protein kinase (RIPK)-dependent form of cell death. In previous studies, neoalbaconol (NA), a constituent extracted from Albatrellus confluens, was demonstrated to induce necroptosis in some cancer cell lines. The molecular mechanism of NA-induced necroptosis is described in this research study. We determined that NA-induced cell death is partly dependent on tumor necrosis factor α (TNFα) feed-forward signaling. More importantly, NA abolished the ubiquitination of RIPK1 by down-regulating E3 ubiquitin ligases, cellular inhibitors of apoptosis protein 1/2 (cIAP1/2) and TNFα receptor-associated factors (TRAFs). The suppression of RIPK1 ubiquitination induced the activation of the non-canonical nuclear factor-κB (NF-κB) pathway and stimulated the transcription of TNFα. Moreover, we also found that NA caused RIPK3-mediated reactive oxygen species (ROS) production and contribution to cell death. Taken together, these results suggested that two distinct mechanisms are involved in NA-induced necroptosis and include RIPK1/NF-κB-dependent expression of TNFα and RIPK3-dependent generation of ROS.
Collapse
Affiliation(s)
- Xinfang Yu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, China.,Key Laboratory of Chinese Ministry of Education, Central South University, Hunan, China.,Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Central South University, Hunan, China
| | - Qipan Deng
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, China.,Key Laboratory of Chinese Ministry of Education, Central South University, Hunan, China.,Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Central South University, Hunan, China
| | - Wei Li
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, China.,Key Laboratory of Chinese Ministry of Education, Central South University, Hunan, China.,Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Central South University, Hunan, China
| | - Lanbo Xiao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, China.,Key Laboratory of Chinese Ministry of Education, Central South University, Hunan, China.,Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Central South University, Hunan, China
| | - Xiangjian Luo
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, China.,Key Laboratory of Chinese Ministry of Education, Central South University, Hunan, China.,Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Central South University, Hunan, China
| | - Xiaolan Liu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, China.,Key Laboratory of Chinese Ministry of Education, Central South University, Hunan, China.,Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Central South University, Hunan, China
| | - Lifang Yang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, China.,Key Laboratory of Chinese Ministry of Education, Central South University, Hunan, China.,Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Central South University, Hunan, China
| | - Songling Peng
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, China.,Key Laboratory of Chinese Ministry of Education, Central South University, Hunan, China.,Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Central South University, Hunan, China
| | - Zhihui Ding
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan, China
| | - Tao Feng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan, China
| | - Jian Zhou
- Liver Cancer Institute, Liver Surgery Department, Zhongshan Hospital
| | - Jia Fan
- Liver Cancer Institute, Liver Surgery Department, Zhongshan Hospital
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Jikai Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan, China
| | - Ya Cao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Hunan, China.,Key Laboratory of Chinese Ministry of Education, Central South University, Hunan, China.,Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Central South University, Hunan, China
| |
Collapse
|
47
|
Broekgaarden M, Weijer R, van Gulik TM, Hamblin MR, Heger M. Tumor cell survival pathways activated by photodynamic therapy: a molecular basis for pharmacological inhibition strategies. Cancer Metastasis Rev 2015; 34:643-90. [PMID: 26516076 PMCID: PMC4661210 DOI: 10.1007/s10555-015-9588-7] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Photodynamic therapy (PDT) has emerged as a promising alternative to conventional cancer therapies such as surgery, chemotherapy, and radiotherapy. PDT comprises the administration of a photosensitizer, its accumulation in tumor tissue, and subsequent irradiation of the photosensitizer-loaded tumor, leading to the localized photoproduction of reactive oxygen species (ROS). The resulting oxidative damage ultimately culminates in tumor cell death, vascular shutdown, induction of an antitumor immune response, and the consequent destruction of the tumor. However, the ROS produced by PDT also triggers a stress response that, as part of a cell survival mechanism, helps cancer cells to cope with the PDT-induced oxidative stress and cell damage. These survival pathways are mediated by the transcription factors activator protein 1 (AP-1), nuclear factor E2-related factor 2 (NRF2), hypoxia-inducible factor 1 (HIF-1), nuclear factor κB (NF-κB), and those that mediate the proteotoxic stress response. The survival pathways are believed to render some types of cancer recalcitrant to PDT and alter the tumor microenvironment in favor of tumor survival. In this review, the molecular mechanisms are elucidated that occur post-PDT to mediate cancer cell survival, on the basis of which pharmacological interventions are proposed. Specifically, pharmaceutical inhibitors of the molecular regulators of each survival pathway are addressed. The ultimate aim is to facilitate the development of adjuvant intervention strategies to improve PDT efficacy in recalcitrant solid tumors.
Collapse
Affiliation(s)
- Mans Broekgaarden
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ruud Weijer
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Thomas M van Gulik
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA, USA
| | - Michal Heger
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
48
|
Evison BJ, Sleebs BE, Watson KG, Phillips DR, Cutts SM. Mitoxantrone, More than Just Another Topoisomerase II Poison. Med Res Rev 2015; 36:248-99. [PMID: 26286294 DOI: 10.1002/med.21364] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 02/06/2023]
Abstract
Mitoxantrone is a synthetic anthracenedione originally developed to improve the therapeutic profile of the anthracyclines and is commonly applied in the treatment of breast and prostate cancers, lymphomas, and leukemias. A comprehensive overview of the drug's molecular, biochemical, and cellular pharmacology is presented here, beginning with the cardiotoxic nature of its predecessor doxorubicin and how these properties shaped the pharmacology of mitoxantrone itself. Although mitoxantrone is firmly established as a DNA topoisomerase II poison within mammalian cells, it is now clear that the drug interacts with a much broader range of biological macromolecules both covalently and noncovalently. Here, we consider each of these interactions in the context of their wider biological relevance to cancer therapy and highlight how they may be exploited to further enhance the therapeutic value of mitoxantrone. In doing so, it is now clear that mitoxantrone is more than just another topoisomerase II poison.
Collapse
Affiliation(s)
- Benny J Evison
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Keith G Watson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Don R Phillips
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| | - Suzanne M Cutts
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| |
Collapse
|
49
|
Konstantakou EG, Voutsinas GE, Velentzas AD, Basogianni AS, Paronis E, Balafas E, Kostomitsopoulos N, Syrigos KN, Anastasiadou E, Stravopodis DJ. 3-BrPA eliminates human bladder cancer cells with highly oncogenic signatures via engagement of specific death programs and perturbation of multiple signaling and metabolic determinants. Mol Cancer 2015. [PMID: 26198749 PMCID: PMC4511243 DOI: 10.1186/s12943-015-0399-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Urinary bladder cancer is one of the most fatal and expensive diseases of industrialized world. Despite the strenuous efforts, no seminal advances have been achieved for its clinical management. Given the importance of metabolic reprogramming in cancer cell survival and growth, we have herein employed 3-BrPA, a halogenated derivative of pyruvate and historically considered inhibitor of glycolysis, to eliminate bladder cancer cells with highly oncogenic molecular signatures. METHODS Bladder cancer cells were exposed to 3-BrPA in the absence or presence of several specific inhibitors. Cell viability was determined by MTT and flow-cytometry assays; cell death, signaling activity and metabolic integrity by Western blotting and immunofluorescence; mutant-gene profiling by DNA sequencing; and gene expression by RT-sqPCR. RESULTS 3-BrPA could activate dose-dependent apoptosis (type 1 PCD) and regulated necrosis (type 3 PCD) of T24 (grade III; H-Ras(G12V); p53(ΔY126)), but not RT4 (grade I), cells, with PARP, MLKL, Drp1 and Nec-7-targeted components critically orchestrating necrotic death. However, similarly to RIPK1 and CypD, p53 presented with non-essential contribution to 3-BrPA-induced cellular collapse, while reactivation of mutant p53 with PRIMA-1 resulted in strong synergism of the two agents. Given the reduced expression of MPC components (likely imposing mitochondrial dysfunction) in T24 cells, the suppression of constitutive autophagy (required by cells carrying oncogenic Ras; also, type 2 PCD) and derangement of glucose-homeostasis determinants by 3-BrPA critically contribute to drug-directed depletion of ATP cellular stores. This bioenergetic crisis is translated to severe dysregulation of Akt/FoxO/GSK-3, mTOR/S6, AMPK and MAPK (p44/42, p38 and SAPK/JNK) signaling pathways in 3-BrPA-treated T24 cells. Sensitivity to 3-BrPA (and tolerance to glucose deprivation) does not rely on B-Raf(V600E) or K-Ras(G13D) mutant oncogenic proteins, but partly depends on aberrant signaling activities of Akt, MAPK and AMPK kinases. Interestingly, MCT1- and macropinocytosis-mediated influx of 3-BrPA in T24 represents the principal mechanism that regulates cellular responsiveness to the drug. Besides its capacity to affect transcription in gene-dependent manner, 3-BrPA can also induce GLUT4-specific splicing silencing in both sensitive and resistant cells, thus dictating alternative routes of drug trafficking. CONCLUSIONS Altogether, it seems that 3-BrPA represents a promising agent for bladder cancer targeted therapy.
Collapse
Affiliation(s)
- Eumorphia G Konstantakou
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Zografou 15784, Athens, Greece.
| | - Gerassimos E Voutsinas
- Laboratory of Environmental Mutagenesis and Carcinogenesis, Institute of Biosciences and Applications, NCSR Demokritos, Athens, Greece.
| | - Athanassios D Velentzas
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Zografou 15784, Athens, Greece.
| | - Aggeliki-Stefania Basogianni
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Zografou 15784, Athens, Greece.
| | - Efthimios Paronis
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Evangelos Balafas
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Nikolaos Kostomitsopoulos
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Konstantinos N Syrigos
- Oncology Unit GPP, Sotiria General Hospital, Athens School of Medicine, Athens, Greece. .,Yale School of Medicine, New Haven, Connecticut, USA.
| | - Ema Anastasiadou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Dimitrios J Stravopodis
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Zografou 15784, Athens, Greece.
| |
Collapse
|
50
|
Saeed Y, Rehman A, Xie B, Xu J, Hong M, Hong Q, Deng Y. Astroglial U87 Cells Protect Neuronal SH-SY5Y Cells from Indirect Effect of Radiation by Reducing DNA Damage and Inhibiting Fas Mediated Apoptotic Pathway in Coculture System. Neurochem Res 2015; 40:1644-54. [DOI: 10.1007/s11064-015-1642-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 06/11/2015] [Accepted: 06/16/2015] [Indexed: 12/17/2022]
|