1
|
Al Saihati HA, Badr OA, Dessouky AA, Mostafa O, Samir Farid A, Aborayah NH, Abdullah Aljasir M, Baioumy B, Mahmoud Taha N, El-Sherbiny M, Hamed Al-Serwi R, Ramadan MM, Salim RF, Shaheen D, E M Ali F, Ebrahim N. Exploring the cytoprotective role of mesenchymal stem Cell-Derived exosomes in chronic liver Fibrosis: Insights into the Nrf2/Keap1/p62 signaling pathway. Int Immunopharmacol 2024; 141:112934. [PMID: 39178516 DOI: 10.1016/j.intimp.2024.112934] [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: 06/01/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024]
Abstract
Hepatic fibrosis is a common pathology present in most chronic liver diseases. Autophagy is a lysosome-mediated intracellular catabolic and recycling process that plays an essential role in maintaining normal hepatic functions. Nuclear factor erythroid 2-like 2 (Nrf2) is a transcription factor responsible for the regulation of cellular anti-oxidative stress response. This study was designed to assess the cytoprotective effect of mesenchymal stem cell-derived exosomes (MSC-exos) on endothelial-mesenchymal transition (EMT) in Carbon Tetrachloride (CCL4) induced liver fibrosis. Rats were treated with 0.1 ml of CCL4 twice weekly for 8 weeks, followed by administration of a single dose of MSC-exos. Rats were then sacrificed after 4 weeks, and liver samples were collected for gene expression analyses, Western blot, histological studies, immunohistochemistry, and transmission electron microscopy. Our results showed that MSC-exos administration decreased collagen deposition, apoptosis, and inflammation. Exosomes modulate the Nrf2/Keap1/p62 pathway, restoring autophagy and Nrf2 levels through modulation of the non-canonical pathway of Nrf2/Keap1/p62. Additionally, MSC-exos regulated miR-153-3p, miR-27a, miR-144 and miRNA-34a expression. In conclusion, the present study shed light on MSC-exos as a cytoprotective agent against EMT and tumorigenesis in chronic liver inflammation.
Collapse
Affiliation(s)
- Hajir A Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Saudi Arabia.
| | - Omnia A Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Egypt.
| | - Arigue A Dessouky
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, 44519 Zagazig, Egypt.
| | - Ola Mostafa
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt.
| | - Ayman Samir Farid
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Qalyubia, Egypt.
| | - Nashwa H Aborayah
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Egypt, Department of Pharmacology, Mutah University, Mutah 61710, Jordan.
| | - Mohammad Abdullah Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia.
| | - Bodour Baioumy
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Egypt.
| | | | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia; Department of Anatomy, Faculty of Medicine, Mansoura University, Egypt.
| | - Rasha Hamed Al-Serwi
- Department of Basic Dental Sciences, College of Dentistry, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Mahmoud M Ramadan
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah City, United Arab Emirates; Department of Cardiology, Faculty of Medicine, Mansoura University, Mansoura City, Egypt.
| | - Rabab F Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha Universit, Egypt.
| | - Dalia Shaheen
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Nesrine Ebrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Stem Cell Unit, Egypt.
| |
Collapse
|
2
|
Cagnin S, Pontisso P, Martini A. SerpinB3: A Multifaceted Player in Health and Disease-Review and Future Perspectives. Cancers (Basel) 2024; 16:2579. [PMID: 39061218 PMCID: PMC11274807 DOI: 10.3390/cancers16142579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
SerpinB3, a member of the serine-protease inhibitor family, has emerged as a crucial player in various physiological and pathological processes. Initially identified as an oncogenic factor in squamous cell carcinomas, SerpinB3's intricate involvement extends from fibrosis progression and cancer to cell protection in acute oxidative stress conditions. This review explores the multifaceted roles of SerpinB3, focusing on its implications in fibrosis, metabolic syndrome, carcinogenesis and immune system impairment. Furthermore, its involvement in tissue protection from oxidative stress and wound healing underscores its potential as diagnostic and therapeutic tool. Recent studies have described the therapeutic potential of targeting SerpinB3 through its upstream regulators, offering novel strategies for cancer treatment development. Overall, this review underscores the importance of further research to fully elucidate the mechanisms of action of SerpinB3 and to exploit its therapeutic potential across various medical conditions.
Collapse
Affiliation(s)
| | - Patrizia Pontisso
- Department of Medicine, University of Padova, 35123 Padova, Italy; (S.C.); (A.M.)
| | | |
Collapse
|
3
|
Bajinka O, Ouedraogo SY, Golubnitschaja O, Li N, Zhan X. Energy metabolism as the hub of advanced non-small cell lung cancer management: a comprehensive view in the framework of predictive, preventive, and personalized medicine. EPMA J 2024; 15:289-319. [PMID: 38841622 PMCID: PMC11147999 DOI: 10.1007/s13167-024-00357-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/20/2024] [Indexed: 06/07/2024]
Abstract
Energy metabolism is a hub of governing all processes at cellular and organismal levels such as, on one hand, reparable vs. irreparable cell damage, cell fate (proliferation, survival, apoptosis, malignant transformation etc.), and, on the other hand, carcinogenesis, tumor development, progression and metastazing versus anti-cancer protection and cure. The orchestrator is the mitochondria who produce, store and invest energy, conduct intracellular and systemically relevant signals decisive for internal and environmental stress adaptation, and coordinate corresponding processes at cellular and organismal levels. Consequently, the quality of mitochondrial health and homeostasis is a reliable target for health risk assessment at the stage of reversible damage to the health followed by cost-effective personalized protection against health-to-disease transition as well as for targeted protection against the disease progression (secondary care of cancer patients against growing primary tumors and metastatic disease). The energy reprogramming of non-small cell lung cancer (NSCLC) attracts particular attention as clinically relevant and instrumental for the paradigm change from reactive medical services to predictive, preventive and personalized medicine (3PM). This article provides a detailed overview towards mechanisms and biological pathways involving metabolic reprogramming (MR) with respect to inhibiting the synthesis of biomolecules and blocking common NSCLC metabolic pathways as anti-NSCLC therapeutic strategies. For instance, mitophagy recycles macromolecules to yield mitochondrial substrates for energy homeostasis and nucleotide synthesis. Histone modification and DNA methylation can predict the onset of diseases, and plasma C7 analysis is an efficient medical service potentially resulting in an optimized healthcare economy in corresponding areas. The MEMP scoring provides the guidance for immunotherapy, prognostic assessment, and anti-cancer drug development. Metabolite sensing mechanisms of nutrients and their derivatives are potential MR-related therapy in NSCLC. Moreover, miR-495-3p reprogramming of sphingolipid rheostat by targeting Sphk1, 22/FOXM1 axis regulation, and A2 receptor antagonist are highly promising therapy strategies. TFEB as a biomarker in predicting immune checkpoint blockade and redox-related lncRNA prognostic signature (redox-LPS) are considered reliable predictive approaches. Finally, exemplified in this article metabolic phenotyping is instrumental for innovative population screening, health risk assessment, predictive multi-level diagnostics, targeted prevention, and treatment algorithms tailored to personalized patient profiles-all are essential pillars in the paradigm change from reactive medical services to 3PM approach in overall management of lung cancers. This article highlights the 3PM relevant innovation focused on energy metabolism as the hub to advance NSCLC management benefiting vulnerable subpopulations, affected patients, and healthcare at large. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00357-5.
Collapse
Affiliation(s)
- Ousman Bajinka
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Serge Yannick Ouedraogo
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, University Hospital Bonn, Venusberg Campus 1, Rheinische Friedrich-Wilhelms-University of Bonn, 53127 Bonn, Germany
| | - Na Li
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
4
|
Neves S, Pacheco S, Vaz F, James P, Simões T, Penque D. Occupational second-hand smoke exposure: A comparative shotgun proteomics study on nasal epithelia from healthy restaurant workers. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 108:104459. [PMID: 38685369 DOI: 10.1016/j.etap.2024.104459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/05/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
Non-smokers exposed to second-hand smoke (SHS) present risk of developing tobacco smoke-associated pathologies. To investigate the airway molecular response to SHS exposure that could be used in health risk assessment, comparative shotgun proteomics was performed on nasal epithelium from a group of healthy restaurant workers, non-smokers (never and former) exposed and not exposed to SHS in the workplace. HIF1α-glycolytic targets (GAPDH, TPI) and proteins related to xenobiotic metabolism, cell proliferation and differentiation leading to cancer (ADH1C, TUBB4B, EEF2) showed significant modulation in non-smokers exposed. In never smokers exposed, enrichment of glutathione metabolism pathway and EEF2-regulating protein synthesis in genotoxic response were increased, while in former smokers exposed, proteins (LYZ, ATP1A1, SERPINB3) associated with tissue damage/regeneration, apoptosis inhibition and inflammation that may lead to asthma, COPD or cancer, were upregulated. The identified proteins are potential response and susceptibility/risk biomarkers for SHS exposure.
Collapse
Affiliation(s)
- Sofia Neves
- Laboratory of Proteomics, Human Genetics Department, National Institute of Health Dr. Ricardo Jorge, INSA I.P, Lisbon, Portugal; Center for Toxicogenomics and Human Health, ToxOmics, NOVA Medical School-FCM, UNL, Lisbon, Portugal.
| | - Solange Pacheco
- Laboratory of Proteomics, Human Genetics Department, National Institute of Health Dr. Ricardo Jorge, INSA I.P, Lisbon, Portugal
| | - Fátima Vaz
- Laboratory of Proteomics, Human Genetics Department, National Institute of Health Dr. Ricardo Jorge, INSA I.P, Lisbon, Portugal; Center for Toxicogenomics and Human Health, ToxOmics, NOVA Medical School-FCM, UNL, Lisbon, Portugal
| | - Peter James
- Protein Technology Laboratory, Department of Immunotechnology, Lund University, Sweden
| | - Tânia Simões
- CECAD Cologne-Excellence in Aging Research University of Cologne, Germany
| | - Deborah Penque
- Laboratory of Proteomics, Human Genetics Department, National Institute of Health Dr. Ricardo Jorge, INSA I.P, Lisbon, Portugal; Center for Toxicogenomics and Human Health, ToxOmics, NOVA Medical School-FCM, UNL, Lisbon, Portugal
| |
Collapse
|
5
|
Biasiolo A, Sandre M, Ferro S, Quarta S, Ruvoletto M, Villano G, Turato C, Guido M, Marin O, Pontisso P. Epitope-Specific Anti-SerpinB3 Antibodies for SerpinB3 Recognition and Biological Activity Inhibition. Biomolecules 2023; 13:biom13050739. [PMID: 37238609 DOI: 10.3390/biom13050739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
SerpinB3 is a serine protease inhibitor that plays a relevant role in disease progression and cancer by increasing fibrosis, cell proliferation, and invasion, besides conferring resistance to apoptosis. The mechanisms underlying these biological activities are not yet fully understood. The aim of this study was to generate antibodies directed against different SerpinB3 epitopes to better investigate their biological role. Five exposed epitopes were identified using the software DNASTAR Lasergene and the corresponding synthetic peptides were used for NZW rabbit immunization. Anti-P#2 and anti-P#4 antibodies were able to recognize both SerpinB3 and SerpinB4 by ELISA. Anti-P#5 antibody, produced against the reactive site loop of SerpinB3, showed the greatest specific reactivity for human SerpinB3. This antibody was able to recognize SerpinB3 at nuclear level, while anti-P#3 antibody recognized SerpinB3 only at cytoplasmic level, both by immunofluorescence and by immunohistochemistry. The biological activity of each antibody preparation was assessed in HepG2 cells overexpressing SerpinB3 and anti-P#5 antibody reduced proliferation by 12% cell and cell invasion by 75%, while trivial results were obtained with the other antibody preparations. These findings indicate that the reactive site loop of SerpinB3 is essential for the invasiveness features induced by this serpin and it could become a novel druggable target.
Collapse
Affiliation(s)
- Alessandra Biasiolo
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Michele Sandre
- Department of Biomedical Sciences, University of Padova, Via Colombo 3, 35131 Padova, Italy
| | - Stefania Ferro
- Department of Biomedical Sciences, University of Padova, Via Colombo 3, 35131 Padova, Italy
| | - Santina Quarta
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Mariagrazia Ruvoletto
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Gianmarco Villano
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Cristian Turato
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maria Guido
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Oriano Marin
- Department of Biomedical Sciences, University of Padova, Via Colombo 3, 35131 Padova, Italy
| | - Patrizia Pontisso
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
6
|
Huang Z, Chen Y, Chen R, Zhou B, Wang Y, Hong L, Wang Y, Wang J, Xu X, Huang Z, Chen W. HPV Enhances HNSCC Chemosensitization by Inhibiting SERPINB3 Expression to Disrupt the Fanconi Anemia Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2202437. [PMID: 36382555 PMCID: PMC9811475 DOI: 10.1002/advs.202202437] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/06/2022] [Indexed: 06/16/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the most common malignant tumor of the head and neck, and the prognosis of patients is poor due to chemotherapeutic resistance. Interestingly, patients with HNSCC induced by human papillomavirus (HPV) infection are more sensitive to chemotherapy and display a better prognosis than HPV-negative patients. The biological relevance of HPV infection and the mechanism underlying chemosensitivity to cisplatin remain unknown. Herein, SERPINB3 is identified as an important target for regulation of cisplatin sensitivity by HPV-E6/E7 in HNSCC. Downregulation of SERPINB3 inhibits cisplatin-induced DNA damage repair and enhances the cytotoxicity of cisplatin. In detail, decreasing SERPINB3 expression reduces the USP1-mediated deubiquitination of FANCD2-FANCI in the Fanconi anemia pathway, thereby interfering with cisplatin-induced DNA interstrand crosslinks repair and further contributing to HNSCC cell apoptosis. To translate this finding, pH-responsive nanoparticles are used to deliver SERPINB3 small interfering RNA in combination with cisplatin, and this treatment successfully reverses cisplatin chemotherapeutic resistance in a patient-derived xenograft model from HPV-negative HNSCC. Taken together, these findings suggest that targeting SERPINB3 based on HPV-positive HNSCC is a potential strategy to overcome cisplatin resistance in HPV-negative HNSCC and improves the prognosis of this disease.
Collapse
Affiliation(s)
- Zixian Huang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Yongju Chen
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Rui Chen
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Bin Zhou
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Yongqiang Wang
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Lei Hong
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Yuepeng Wang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Jianguang Wang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Xiaoding Xu
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Zhiquan Huang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Weiliang Chen
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| |
Collapse
|
7
|
Luke CJ, Markovina S, Good M, Wight IE, Thomas BJ, Linneman JM, Lanik WE, Koroleva O, Coffman MR, Miedel MT, Gong Q, Andress A, Campos Guerrero M, Wang S, Chen L, Beatty WL, Hausmann KN, White FV, Fitzpatrick JAJ, Orvedahl A, Pak SC, Silverman GA. Lysoptosis is an evolutionarily conserved cell death pathway moderated by intracellular serpins. Commun Biol 2022; 5:47. [PMID: 35022507 PMCID: PMC8755814 DOI: 10.1038/s42003-021-02953-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/07/2021] [Indexed: 01/02/2023] Open
Abstract
Lysosomal membrane permeabilization (LMP) and cathepsin release typifies lysosome-dependent cell death (LDCD). However, LMP occurs in most regulated cell death programs suggesting LDCD is not an independent cell death pathway, but is conscripted to facilitate the final cellular demise by other cell death routines. Previously, we demonstrated that Caenorhabditis elegans (C. elegans) null for a cysteine protease inhibitor, srp-6, undergo a specific LDCD pathway characterized by LMP and cathepsin-dependent cytoplasmic proteolysis. We designated this cell death routine, lysoptosis, to distinguish it from other pathways employing LMP. In this study, mouse and human epithelial cells lacking srp-6 homologues, mSerpinb3a and SERPINB3, respectively, demonstrated a lysoptosis phenotype distinct from other cell death pathways. Like in C. elegans, this pathway depended on LMP and released cathepsins, predominantly cathepsin L. These studies suggested that lysoptosis is an evolutionarily-conserved eukaryotic LDCD that predominates in the absence of neutralizing endogenous inhibitors.
Collapse
Affiliation(s)
- Cliff J Luke
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA.
- Siteman Cancer Center, and Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA.
| | - Stephanie Markovina
- Siteman Cancer Center, and Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
- Radiation Oncology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Misty Good
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Ira E Wight
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Brian J Thomas
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - John M Linneman
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Wyatt E Lanik
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Olga Koroleva
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Maggie R Coffman
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Mark T Miedel
- Department of Computational and Systems biology, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qingqing Gong
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Arlise Andress
- Radiation Oncology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Marlene Campos Guerrero
- Radiation Oncology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Songyan Wang
- Radiation Oncology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - LiYun Chen
- Radiation Oncology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Wandy L Beatty
- Molecular Microbiology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Kelsey N Hausmann
- Molecular Microbiology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Frances V White
- Department of Pathology and Immunology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - James A J Fitzpatrick
- Cell Biology and Physiology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
- Neuroscience, and Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Anthony Orvedahl
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Stephen C Pak
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA
| | - Gary A Silverman
- Departments of Pediatrics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA.
- Siteman Cancer Center, and Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA.
- Cell Biology and Physiology, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA.
- Genetics, Washington University School of Medicine and the Children's Discovery Institute of St. Louis Children's Hospital, St. Louis, MO, USA.
| |
Collapse
|
8
|
Lee SH, Cho WJ, Najy AJ, Saliganan AD, Pham T, Rakowski J, Loughery B, Ji CH, Sakr W, Kim S, Kato I, Chung WK, Kim HE, Kwon YT, Kim HRC. p62/SQSTM1-induced caspase-8 aggresomes are essential for ionizing radiation-mediated apoptosis. Cell Death Dis 2021; 12:997. [PMID: 34697296 PMCID: PMC8546074 DOI: 10.1038/s41419-021-04301-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 09/03/2021] [Accepted: 10/07/2021] [Indexed: 12/28/2022]
Abstract
The autophagy–lysosome pathway and apoptosis constitute vital determinants of cell fate and engage in a complex interplay in both physiological and pathological conditions. Central to this interplay is the archetypal autophagic cargo adaptor p62/SQSTM1/Sequestosome-1 which mediates both cell survival and endoplasmic reticulum stress-induced apoptosis via aggregation of ubiquitinated caspase-8. Here, we investigated the role of p62-mediated apoptosis in head and neck squamous cell carcinoma (HNSCC), which can be divided into two groups based on human papillomavirus (HPV) infection status. We show that increased autophagic flux and defective apoptosis are associated with radioresistance in HPV(-) HNSCC, whereas HPV(+) HNSCC fail to induce autophagic flux and readily undergo apoptotic cell death upon radiation treatments. The degree of radioresistance and tumor progression of HPV(-) HNSCC respectively correlated with autophagic activity and cytosolic levels of p62. Pharmacological activation of the p62-ZZ domain using small molecule ligands sensitized radioresistant HPV(-) HNSCC cells to ionizing radiation by facilitating p62 self-polymerization and sequestration of cargoes leading to apoptosis. The self-polymerizing activity of p62 was identified as the essential mechanism by which ubiquitinated caspase-8 is sequestered into aggresome-like structures, without which irradiation fails to induce apoptosis in HNSCC. Our results suggest that harnessing p62-dependent sequestration of ubiquitinated caspase-8 provides a novel therapeutic avenue in patients with radioresistant tumors.
Collapse
Affiliation(s)
- Su Hyun Lee
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Cellular Degradation Biology Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Won Jin Cho
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Abdo J Najy
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Allen-Dexter Saliganan
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Tri Pham
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Joseph Rakowski
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Division of Radiation Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Brian Loughery
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Chang Hoon Ji
- Cellular Degradation Biology Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.,AUTOTAC Bio Inc., Changkkyunggung-ro 254, Jongno-gu, Seoul, 03080, Korea
| | - Wael Sakr
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Seongho Kim
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Ikuko Kato
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Weon Kuu Chung
- Department of Radiation Oncology, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Harold E Kim
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA.,Division of Radiation Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA
| | - Yong Tae Kwon
- Cellular Degradation Biology Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea. .,AUTOTAC Bio Inc., Changkkyunggung-ro 254, Jongno-gu, Seoul, 03080, Korea. .,SNU Dementia Research Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea. .,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
| | - Hyeong-Reh C Kim
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA. .,Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, 48201, USA.
| |
Collapse
|
9
|
Low P66shc with High SerpinB3 Levels Favors Necroptosis and Better Survival in Hepatocellular Carcinoma. BIOLOGY 2021; 10:biology10050363. [PMID: 33922660 PMCID: PMC8145214 DOI: 10.3390/biology10050363] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022]
Abstract
Simple Summary Cell proliferation and escape from apoptosis are important pathological features of hepatocellular carcinoma, one of the tumors with the highest mortality rate worldwide. The aim of the study was to evaluate the expression of the pro-apoptotic p66shc and the anti-apoptotic SerpinB3 molecules in relation to clinical outcome in patients with hepatocellular carcinoma and to evaluate their effect on cell fate and tumor growth. In patients with hepatocellular carcinoma the best survival was observed in the subgroup with p66shc levels below median values and SerpinB3 levels above median values. Mice p66shc knockout showed high levels of SerpinB3, while in hepatoma cells overexpressing SerpinB3, p66shc expression was trivial. Hepatoma cells overexpressing SerpinB3 were more prone to die after oxidizing treatments. These cells injected in nude mice developed tumors five times smaller than those from controls. Tumors originating from hepatoma cells overexpressing SerpinB3 showed typical features of necroptosis. In conclusion, in patients affected by hepatocellular carcinoma, the pattern characterized by p66shc downregulation and elevated SerpinB3 levels was associated with markedly better survival. This pattern favored necroptosis in experimental high-stress conditions. Abstract Cell proliferation and escape from apoptosis are important pathological features of hepatocellular carcinoma (HCC), one of the tumors with the highest mortality rate worldwide. The aim of the study was to evaluate the expression of the pro-apoptotic p66shc and the anti-apoptotic SerpinB3 in HCCs in relation to clinical outcome, cell fate and tumor growth. p66shc and SerpinB3 were evaluated in 67 HCC specimens and the results were correlated with overall survival. Proliferation and cell death markers were analyzed in hepatoma cells overexpressing SerpinB3, under different stress conditions. p66shc−/− mice and xenograft models were also used to assess the effects of p66shc and SerpinB3 on tumor growth. In patients with HCC, the best survival was observed in the subgroup with p66shc levels below median values and SerpinB3 levels above median values. Mice p66shc−/− showed high levels of SerpinB3, while in HepG2 cells overexpressing SerpinB3, p66shc expression was trivial. HepG2 overexpressing SerpinB3 cells were more prone to die after oxidizing treatments, such as diamide or high concentration H2O2. These cells injected in nude mice developed tumors five times smaller than those from control HepG2 cells. Tumors originating from HepG2 overexpressing SerpinB3 cells showed decreased activated Caspase-8, with concomitant increase of RIP3K and decreased levels of cleaved RIP3K, typical features of necroptosis. In conclusion, in patients affected by HCC, the pattern characterized by p66shc downregulation and elevated SerpinB3 levels was associated with markedly better survival. This pattern favored necroptosis in experimental high-stress conditions.
Collapse
|
10
|
Cho WJ, Kessel D, Rakowski J, Loughery B, Najy AJ, Pham T, Kim S, Kwon YT, Kato I, Kim HE, Kim HRC. Photodynamic Therapy as a Potent Radiosensitizer in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13061193. [PMID: 33801879 PMCID: PMC7998908 DOI: 10.3390/cancers13061193] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 01/17/2023] Open
Abstract
Simple Summary Despite the advances in multimodality treatment strategies, more than 30% of patients with advanced head and neck squamous cell carcinoma (HNSCC) experience recurrence of the disease that is usually derived from the residual tumor. The goal of our study is to understand the molecular basis underlying radiotherapy resistance in advanced HNSCC and to identify a mechanism-based radiosensitizer. We found that the autophagic cell survival pathway is upregulated in therapy-resistant HNSCC. Photodynamic therapy (PDT) directed at the endoplasmic reticulum (ER)/mitochondria induces programmed cell death such as paraptosis and apoptosis in an autophagic adaptor p62-dependent manner, promoting radiotoxicity. Abstract Despite recent advances in therapeutic modalities such as radiochemotherapy, the long-term prognosis for patients with advanced head and neck squamous cell carcinoma (HNSCC), especially nonviral HNSCC, remains very poor, while survival of patients with human papillomavirus (HPV)-associated HNSCC is greatly improved after radiotherapy. The goal of this study is to develop a mechanism-based treatment protocol for high-risk patients with HPV-negative HNSCC. To achieve our goal, we have investigated molecular mechanisms underlying differential radiation sensitivity between HPV-positive and -negative HNSCC cells. Here, we found that autophagy is associated with radioresistance in HPV-negative HNSCC, whereas apoptosis is associated with radiation sensitive HPV-positive HNSCC. Interestingly, we found that photodynamic therapy (PDT) directed at the endoplasmic reticulum (ER)/mitochondria initially induces paraptosis followed by apoptosis. This led to a substantial increase in radiation responsiveness in HPV-negative HNSCC, while the same PDT treatment had a minimal effect on HPV-positive cells. Here, we provide evidence that the autophagic adaptor p62 mediates signal relay for the induction of apoptosis, promoting ionizing radiation (XRT)-induced cell death in HPV-negative HNSCC. This work proposes that ER/mitochondria-targeted PDT can serve as a radiosensitizer in intrinsically radioresistant HNSCC that exhibits an increased autophagic flux.
Collapse
Affiliation(s)
- Won Jin Cho
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (W.J.C.); (A.J.N.); (T.P.)
| | - David Kessel
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Correspondence: (D.K.); (H.-R.C.K.); Tel.: +1-313-577-1766 (D.K.); +1-313-577-2407 (H.-R.C.K.)
| | - Joseph Rakowski
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (J.R.); (B.L.); (S.K.); (I.K.); (H.E.K.)
- Division of Radiation Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Brian Loughery
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (J.R.); (B.L.); (S.K.); (I.K.); (H.E.K.)
- Division of Radiation Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Abdo J. Najy
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (W.J.C.); (A.J.N.); (T.P.)
| | - Tri Pham
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (W.J.C.); (A.J.N.); (T.P.)
| | - Seongho Kim
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (J.R.); (B.L.); (S.K.); (I.K.); (H.E.K.)
| | - Yong Tae Kwon
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea;
| | - Ikuko Kato
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (J.R.); (B.L.); (S.K.); (I.K.); (H.E.K.)
| | - Harold E. Kim
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (J.R.); (B.L.); (S.K.); (I.K.); (H.E.K.)
- Division of Radiation Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Hyeong-Reh C. Kim
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (W.J.C.); (A.J.N.); (T.P.)
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (J.R.); (B.L.); (S.K.); (I.K.); (H.E.K.)
- Correspondence: (D.K.); (H.-R.C.K.); Tel.: +1-313-577-1766 (D.K.); +1-313-577-2407 (H.-R.C.K.)
| |
Collapse
|
11
|
Gong J, Song Y, Xu L, Che X, Hou K, Guo T, Cheng Y, Liu Y, Qu X. Upregulation of Serine Proteinase Inhibitor Clade B Member 3 (SERPINB3) Expression by Stromal Cell-Derived Factor (SDF-1)/CXCR4/Nuclear Factor kappa B (NF-κB) Promotes Migration and Invasion of Gastric Cancer Cells. Med Sci Monit 2020; 26:e927411. [PMID: 33110054 PMCID: PMC7604976 DOI: 10.12659/msm.927411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/25/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Serine proteinase inhibitor clade B member 3 (SERPINB3) is a neutral glycoprotein. Its overexpression is related to the promotion of cell proliferation and activation via the nuclear factor kappa B (NF-kappaB) pathway in several tumors. Whether it can participate in stromal cell-derived factor (SDF-1)/NF-kappaB-induced metastasis of gastric cancer has not been reported. MATERIAL AND METHODS We analyzed the ability of SDF-1 to induce migration and invasion in vitro by knocking down the expression of SERPINB3 with siRNAs in gastric cancer cells. We also explored the effects of a CXCR4 antagonist and NF-kappaB inhibitor on SERPINB3 expression. We verified the effect of SERPINB3 on prognosis in gastric cancer specimens by immunohistochemistry. RESULTS In vitro experiments confirmed that SDF-1 upregulated the expression of SERPINB3 and promoted metastasis in gastric cancer cells. This phenomenon was reversed by knockdown of SERPINB3, a chemokine receptor 4 (CXCR4) antagonist, and an NF-kappaB inhibitor, which downregulated the expression of SERPINB3. In patients with gastric cancer, a significant positive correlation was observed between CXCR4 and SERPINB3 expression (r=0.222, P=0.029). Moreover, double positivity for SERPINB3 and CXCR4 was certified to be an independent prognostic factor (HR=3.332, P<0.001). CXCR4-positive patients who also expressed SERPINB3 were inclined to suffer from lymph node metastasis, confirming that SERPINB3 is a downstream molecule of CXCR4. CONCLUSIONS In vitro and pathological results showed that SDF-1/CXCR4 activated the NF-kappaB pathway and upregulated SERPINB3 to facilitate the migration and invasion of gastric cancer cells.
Collapse
Affiliation(s)
- Jing Gong
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Ling Xu
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Xiaofang Che
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Kezuo Hou
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Tianshu Guo
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yu Cheng
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yunpeng Liu
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Xiujuan Qu
- Department of Medical Oncology, Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
12
|
Thermozier S, Zhang X, Hou W, Fisher R, Epperly MW, Liu B, Bahar I, Wang H, Greenberger JS. Radioresistance of Serpinb3a-/- Mice and Derived Hematopoietic and Marrow Stromal Cell Lines. Radiat Res 2019; 192:267-281. [PMID: 31295086 PMCID: PMC6759811 DOI: 10.1667/rr15379.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Serpins are a group of serine-proteases involved in multiple signal transduction pathways in mammalian cells. In particular, Serpinb3a is involved in the lysosomal necrosis cell death pathway with components that overlap with radiation-induced apoptosis. We investigated the radiation response of Serpinb3a-/- mice compared to Serpinb3a+/+ mice on the Balb/c background. Serpinb3a-/- mice showed significant radioresistance to a dose of 8.0 Gy total-body irradiation, compared to Serpinb3a+/+ Balb/c mice. Long-term bone marrow cultures from Serpinb3a-/- mice showed increased longevity. In clonogenic survival assays, fresh bone marrow hematopoietic progenitors, as well as clonal interleukin-3 (IL-3)-dependent hematopoietic progenitor and bone marrow stromal cell lines from Serpinb3a-/- mice were radioresistant. Serpinb3a-/- mouse bone marrow-derived stromal cell lines had increased baseline and postirradiation antioxidant capacity. Serpinb3a-/- bone marrow stromal cells showed increased radiation-induced RNA transcripts for MnSOD and p21, and decreased levels of p53 and TGF-b. Both irradiated Serpinb3a-/- mouse bone marrow stromal cell lines and plasma removed from total-body irradiated mice had decreased levels of expression of stress response and inflammation-associated proteins. Abrogation of Serpinb3a may be a potential new target for mitigation of radiation effects.
Collapse
Affiliation(s)
- Stephanie Thermozier
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Xichen Zhang
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Bing Liu
- Department of Computational Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Ivet Bahar
- Department of Computational Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
13
|
Gatto M, Luisetto R, Ghirardello A, Cavicchioli L, Codolo G, Biasiolo A, Maggioni G, Saccon F, Beggio M, Cappon A, Venturini R, Pontisso P, Doria A. SERPINB3 Delays Glomerulonephritis and Attenuates the Lupus-Like Disease in Lupus Murine Models by Inducing a More Tolerogenic Immune Phenotype. Front Immunol 2018; 9:2081. [PMID: 30254646 PMCID: PMC6141748 DOI: 10.3389/fimmu.2018.02081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 08/22/2018] [Indexed: 12/24/2022] Open
Abstract
Objective: To explore the effects of SERPINB3 administration in murine lupus models with a focus on lupus-like nephritis. Methods: 40 NZB/W F1 mice were subdivided into 4 groups and intraperitoneally injected with recombinant SERPINB3 (7.5 μg/0.1 mL or 15 μg/0.1 mL) or PBS (0.1 mL) before (group 1 and 2) or after (group 3 and 4) the development of proteinuria (≥100 mg/dl). Two additional mice groups were provided by including 20 MRL/lpr mice which were prophylactically injected with SERPINB3 (10 mice, group 5) or PBS (10 mice, group 6). Time of occurrence and levels of anti-dsDNA and anti-C1q antibodies, proteinuria and serum creatinine, overall- and proteinuria-free survival were assessed in mice followed up to natural death. Histological analysis was performed in kidneys of both lupus models. The Th17:Treg cell ratio was assessed by flow-cytometry in splenocytes of treated and untreated MRL/lpr mice. Statistical analysis was performed using non parametric tests and Kaplan-Meier curves, when indicated. Results: Autoantibody levels and proteinuria were significantly decreased and time of occurrence significantly delayed in SERPINB3-treated mice vs. controls. In agreement with these findings, proteinuria-free and overall survival were significantly improved in SERPINB3-treated groups vs. controls. Histological analysis demonstrated a lower prevalence of severe tubular lesions in kidneys of group 5 vs. group 6. SERPINB3-treated mice showed an overall trend toward a reduced prevalence of severe lesions in both strains. Th17:Treg ratio was significantly decreased in splenocytes of MRL/lpr mice treated with SERPINB3, compared to untreated control mice. Conclusions: SERPINB3 significantly improves disease course and delays the onset of severe glomerulonephritis in lupus-prone mice, possibly inducing a more tolerogenic immune phenotype.
Collapse
Affiliation(s)
- Mariele Gatto
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Anna Ghirardello
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Laura Cavicchioli
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Gaia Codolo
- Department of Biology, University of Padova, Padova, Italy
| | - Alessandra Biasiolo
- Internal Medicine and Hepatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Giuseppe Maggioni
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Francesca Saccon
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Marianna Beggio
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Andrea Cappon
- Internal Medicine and Hepatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Roberta Venturini
- Laboratory Medicine Unit, University-Hospital of Padova, Padova, Italy
| | - Patrizia Pontisso
- Internal Medicine and Hepatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| |
Collapse
|
14
|
Izuhara K, Yamaguchi Y, Ohta S, Nunomura S, Nanri Y, Azuma Y, Nomura N, Noguchi Y, Aihara M. Squamous Cell Carcinoma Antigen 2 (SCCA2, SERPINB4): An Emerging Biomarker for Skin Inflammatory Diseases. Int J Mol Sci 2018; 19:E1102. [PMID: 29642409 PMCID: PMC5979376 DOI: 10.3390/ijms19041102] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 12/19/2022] Open
Abstract
Squamous cell carcinoma antigens 1 and 2 (SCCA1 and 2, SERPIN B3 and B4), members of the ovalbumin serpin (ov-serpin)/clade B serpin family, were originally discovered as tumor-specific antigens and are used as tumor markers for various kinds of squamous cell carcinomas. Recently, our understanding of the underlying mechanisms of how SCCA1/2 enhance tumor growth has greatly increased. Moreover, it has been shown that SCCA1/2 are involved in the pathogenesis of several inflammatory diseases: asthma, psoriasis, and atopic dermatitis (AD). IL-22 and IL-17, signature cytokines of type 17 inflammation, as well as IL-4 and IL-13, signature cytokines of type 2 inflammation, both of which are positively correlated with the pathogenesis of psoriasis and allergic diseases, respectively, can induce expression of SCCA1/2 in airway epithelial cells and/or keratinocytes, leading to high expression of SCCA1/2 in these diseases. Based on these findings, several trials have been performed to examine the potential of applying SCCA1/2 to biomarkers for these diseases. The findings show that SCCA2 is useful to aid diagnosis, estimate clinical severity and disease type, and assess responses to treatment in psoriasis and AD. These results suggest that SCCA2 has emerged as a novel biomarker for skin inflammatory diseases.
Collapse
Affiliation(s)
- Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga 849-8501, Japan.
| | - Yukie Yamaguchi
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan.
| | - Shoichiro Ohta
- Department of Medical Technology and Sciences, School of Health Sciences at Fukuoka, International University of Health and Welfare, Okawa 831-8501, Japan.
| | - Satoshi Nunomura
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga 849-8501, Japan.
| | - Yasuhiro Nanri
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga 849-8501, Japan.
| | | | | | | | - Michiko Aihara
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan.
| |
Collapse
|
15
|
Fassan M, Realdon S, Vianello L, Quarta S, Ruol A, Castoro C, Scarpa M, Zaninotto G, Guzzardo V, Chiarion Sileni V, Pontisso P, Rugge M. Squamous cell carcinoma antigen (SCCA) is up-regulated during Barrett's carcinogenesis and predicts esophageal adenocarcinoma resistance to neoadjuvant chemotherapy. Oncotarget 2018; 8:24372-24379. [PMID: 28042960 PMCID: PMC5421854 DOI: 10.18632/oncotarget.14108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/22/2016] [Indexed: 12/15/2022] Open
Abstract
Squamous Cell Carcinoma Antigen (SCCA) is consistently overexpressed in many different solid tumors, and has been associated with both tumor aggressiveness and chemoresistance. No data, however, is currently available on SCCA expression during esophageal Barrett's carcinogenesis, nor on SCCA expression's role on esophageal adenocarcinoma chemoresistance. The SCCA immunohistochemical expression was assessed in a series of 100 biopsy samples covering the whole histological spectrum of Barrett's oncogenesis. Squamous native mucosa was characterized by a moderate to strong cytoplasmic and nuclear SCCA expression in suprabasal, medium, and superficial layers. On the other hand, almost half of the considered lesions did not express SCCA; the other half featured weak to moderate SCCA expression. The relationship between SCCA protein expression and tumor response to neoadjuvant chemotherapy was assessed in 90 esophageal adenocarcinoma specimens (40 biopsy and 50 surgery specimens), stratified according to Mandard tumor regression grade. As observed in other settings, the presence of SCCA expression clustered in the group of tumors characterized by a lower responsiveness to neoadjuvant treatments. The present results suggest an involvement of SCCA in a subset of Barrett-related tumors, and prompt to consider the SCCA-protein expression as response-predictive marker of neoadjuvant therapy in esophageal adenocarcinomas.
Collapse
Affiliation(s)
- Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Stefano Realdon
- Gastroenterology Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padua, Italy
| | - Luca Vianello
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Santina Quarta
- Department of Medicine (DIMED), 5th Medical Clinic, University of Padua, Padua, Italy
| | - Alberto Ruol
- Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), 3rd Surgical Clinic, University of Padua, Padua, Italy
| | - Carlo Castoro
- Esophageal and Digestive Tract Surgical Unit, Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| | - Marco Scarpa
- Esophageal and Digestive Tract Surgical Unit, Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| | - Giovanni Zaninotto
- Imperial College London, Department of Surgery and Cancer, Division of Surgery, London, UK
| | - Vincenza Guzzardo
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Vanna Chiarion Sileni
- Melanoma & Esophageal Oncology Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padua, Italy
| | - Patrizia Pontisso
- Department of Medicine (DIMED), 5th Medical Clinic, University of Padua, Padua, Italy
| | - Massimo Rugge
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy.,Veneto Tumour Registry, Veneto Region, Padua, Italy
| |
Collapse
|
16
|
|
17
|
Gómez-Sintes R, Ledesma MD, Boya P. Lysosomal cell death mechanisms in aging. Ageing Res Rev 2016; 32:150-168. [PMID: 26947122 DOI: 10.1016/j.arr.2016.02.009] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 02/22/2016] [Accepted: 02/29/2016] [Indexed: 12/14/2022]
Abstract
Lysosomes are degradative organelles essential for cell homeostasis that regulate a variety of processes, from calcium signaling and nutrient responses to autophagic degradation of intracellular components. Lysosomal cell death is mediated by the lethal effects of cathepsins, which are released into the cytoplasm following lysosomal damage. This process of lysosomal membrane permeabilization and cathepsin release is observed in several physiopathological conditions and plays a role in tissue remodeling, the immune response to intracellular pathogens and neurodegenerative diseases. Many evidences indicate that aging strongly influences lysosomal activity by altering the physical and chemical properties of these organelles, rendering them more sensitive to stress. In this review we focus on how aging alters lysosomal function and increases cell sensitivity to lysosomal membrane permeabilization and lysosomal cell death, both in physiological conditions and age-related pathologies.
Collapse
Affiliation(s)
- Raquel Gómez-Sintes
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biologicas, CIB-CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - María Dolores Ledesma
- Department of Molecular Neurobiology, Centro Biologia Molecular Severo Ochoa, CSIC-UAM, C/Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biologicas, CIB-CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
18
|
Sun Y, Sheshadri N, Zong WX. SERPINB3 and B4: From biochemistry to biology. Semin Cell Dev Biol 2016; 62:170-177. [PMID: 27637160 DOI: 10.1016/j.semcdb.2016.09.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 12/15/2022]
Abstract
Human SERPINB3 and SERPINB4 are evolutionary duplicated serine/cysteine protease inhibitors. Genomic analysis indicates that these paralogous genes were encoded from independent loci arising from tandem gene duplication. Although the two molecules share 92% identity of their amino acid sequences, they are distinct in the Reactive Center Loop (RCL) including a hinge region and catalytic sequences which accounts for altered substrate specificity. Elevated expression of the two molecules has been reported to contribute to numerous pathological conditions such as inflammatory diseases and cancer. In this review, we focus on summarizing the biochemical features of SERPINB3/B4 and discussing the mechanistic basis for their biological functions and implications in human diseases.
Collapse
Affiliation(s)
- Yu Sun
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Namratha Sheshadri
- National Cancer Institute, Center for Cancer Research, Frederick, MD 21702, United States
| | - Wei-Xing Zong
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, United States.
| |
Collapse
|
19
|
Saxena M, Busca A, Holcik M, Kumar A. Bacterial DNA Protects Monocytic Cells against HIV-Vpr-Induced Mitochondrial Membrane Depolarization. THE JOURNAL OF IMMUNOLOGY 2016; 196:3754-67. [PMID: 26969755 DOI: 10.4049/jimmunol.1402379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 02/19/2016] [Indexed: 12/26/2022]
Abstract
Monocytes and macrophages are important HIV reservoirs, as they exhibit marked resistance to apoptosis upon infection. However, the mechanism underlying resistance to apoptosis in these cells is poorly understood. Using HIV-viral protein R-52-96 aa peptide (Vpr), we show that primary monocytes and THP-1 cells treated with Vpr are highly susceptible to mitochondrial depolarization, but develop resistance following stimulation with bacterial DNA or CpG oligodeoxynucleotide. We have shown that Vpr-induced mitochondrial depolarization is mediated by TNFR-associated factor-1 (TRAF-1) and TRAF-2 degradation and subsequent activation of caspase-8, Bid, and Bax. To provide the mechanism governing such resistance to mitochondrial depolarization, our results show that prior stimulation with CpG oligodeoxynucleotide or Escherichia coli DNA prevented: 1) TRAF-1/2 downregulation; 2) activation of caspase-8, Bid, and Bax; and 3) subsequent mitochondrial depolarization and release of apoptosis-inducing factor and cytochrome c Furthermore, this protection was mediated by upregulation of antiapoptotic protein (c-IAP-2) through calmodulin-dependent kinase-II activation. Thus, c-IAP-2 may prevent Vpr-mediated mitochondrial depolarization through stabilizing TRAF-1/2 expression and sequential inhibition of caspase-8, Bid, and Bax.
Collapse
Affiliation(s)
- Mansi Saxena
- Department of Biochemistry, Microbiology and Immunology, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada
| | - Aurelia Busca
- Department of Biochemistry, Microbiology and Immunology, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada
| | - Martin Holcik
- Research Institute, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada; and
| | - Ashok Kumar
- Department of Biochemistry, Microbiology and Immunology, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada; Research Institute, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada; and Department of Pathology and Laboratory Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada
| |
Collapse
|
20
|
Wei Y, Zhou F, Zhang D, Chen Q, Xing D. A graphene oxide based smart drug delivery system for tumor mitochondria-targeting photodynamic therapy. NANOSCALE 2016; 8:3530-8. [PMID: 26799192 DOI: 10.1039/c5nr07785k] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Subcellular organelles play critical roles in cell survival. In this work, a novel photodynamic therapy (PDT) drug delivery and phototoxicity on/off nano-system based on graphene oxide (NGO) as the carrier is developed to implement subcellular targeting and attacking. To construct the nanodrug (PPa-NGO-mAb), NGO is modified with the integrin αvβ3 monoclonal antibody (mAb) for tumor targeting. Pyropheophorbide-a (PPa) conjugated with polyethylene-glycol is used to cover the surface of the NGO to induce phototoxicity. Polyethylene-glycol phospholipid is loaded to enhance water solubility. The results show that the phototoxicity of PPa on NGO can be switched on and off in organic and aqueous environments, respectively. The PPa-NGO-mAb assembly is able to effectively target the αvβ3-positive tumor cells with surface ligand and receptor recognition; once endocytosized by the cells, they are observed escaping from lysosomes and subsequently transferring to the mitochondria. In the mitochondria, the 'on' state PPa-NGO-mAb performs its effective phototoxicity to kill cells. The biological and physical dual selections and on/off control of PPa-NGO-mAb significantly enhance mitochondria-mediated apoptosis of PDT. This smart system offers a potential alternative to drug delivery systems for cancer therapy.
Collapse
Affiliation(s)
- Yanchun Wei
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou 510631, P. R. China.
| | | | | | | | | |
Collapse
|
21
|
Dai L, Liu J, Luo Z, Li M, Cai K. Tumor therapy: targeted drug delivery systems. J Mater Chem B 2016; 4:6758-6772. [DOI: 10.1039/c6tb01743f] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The review highlights the main targeted drug delivery systems for tumor therapy, including the targeting sites, strategies, mechanisms and preclinical/clinical trials.
Collapse
Affiliation(s)
- Liangliang Dai
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Junjie Liu
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Zhong Luo
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Menghuan Li
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education College of Bioengineering
- Chongqing University
- Chongqing 400044
- P. R. China
| |
Collapse
|
22
|
Moschovi M, Critselis E, Cen O, Adamaki M, Lambrou GI, Chrousos GP, Vlahopoulos S. Drugs acting on homeostasis: challenging cancer cell adaptation. Expert Rev Anticancer Ther 2015; 15:1405-1417. [PMID: 26523494 DOI: 10.1586/14737140.2015.1095095] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Cancer treatment aims to exploit properties that define malignant cells. In recent years, it has become apparent that malignant cells often survive cancer treatment and ensuing cell stress by switching on auxiliary turnover pathways, changing cellular metabolism and, concomitantly, the gene expression profile. The changed profile impacts the material exchange of cancer cells with affected tissues. Herein, we show that pathways of proteostasis and energy generation regulate common transcription factors. Namely, when one pathway of intracellular turnover is blocked, it triggers alternative turnover mechanisms, which induce transcription factor proteins that control expression of cytokines and regulators of apoptosis, cell division, differentiation, metabolism, and response to hormones. We focus on several alternative turnover mechanisms that can be blocked by drugs already used in clinical practice for the treatment of other non-cancer related diseases. We also discuss paradigms on the challenges posed by cancer cell adaptation mechanisms.
Collapse
Affiliation(s)
- Maria Moschovi
- a 1 University of Athens, Horemio Research Institute, First Department of Pediatrics, Thivon & Levadeias, Goudi, Athens, 11527, Greece
- b 2 University of Athens, Pediatric Hematology/Oncology Unit, First Department of Pediatrics, University of Athens, "Aghia Sofia" Children's Hospital, Thivon & Levadeias, 11527 Goudi, Athens, Greece
| | - Elena Critselis
- a 1 University of Athens, Horemio Research Institute, First Department of Pediatrics, Thivon & Levadeias, Goudi, Athens, 11527, Greece
| | - Osman Cen
- c 3 Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago Ave, Chicago, IL 60611, USA
| | - Maria Adamaki
- a 1 University of Athens, Horemio Research Institute, First Department of Pediatrics, Thivon & Levadeias, Goudi, Athens, 11527, Greece
- b 2 University of Athens, Pediatric Hematology/Oncology Unit, First Department of Pediatrics, University of Athens, "Aghia Sofia" Children's Hospital, Thivon & Levadeias, 11527 Goudi, Athens, Greece
| | - George I Lambrou
- a 1 University of Athens, Horemio Research Institute, First Department of Pediatrics, Thivon & Levadeias, Goudi, Athens, 11527, Greece
| | - George P Chrousos
- a 1 University of Athens, Horemio Research Institute, First Department of Pediatrics, Thivon & Levadeias, Goudi, Athens, 11527, Greece
| | - Spiros Vlahopoulos
- a 1 University of Athens, Horemio Research Institute, First Department of Pediatrics, Thivon & Levadeias, Goudi, Athens, 11527, Greece
| |
Collapse
|
23
|
Turato C, Pontisso P. SERPINB3 (serpin peptidase inhibitor, clade B (ovalbumin), member 3). ACTA ACUST UNITED AC 2015; 19:202-209. [PMID: 25984243 DOI: 10.4267/2042/56413] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Review on SERPINB3, with data on DNA/RNA, on the protein encoded and where the gene is implicated.
Collapse
|
24
|
Ciscato F, Sciacovelli M, Villano G, Turato C, Bernardi P, Rasola A, Pontisso P. SERPINB3 protects from oxidative damage by chemotherapeutics through inhibition of mitochondrial respiratory complex I. Oncotarget 2015; 5:2418-27. [PMID: 24810714 PMCID: PMC4058015 DOI: 10.18632/oncotarget.1411] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
SERPINB3 (SB3) is a serine protease inhibitor overexpressed in several malignancies of epithelial origin, including primary liver cancer, where it inhibits apoptosis through poorly defined mechanisms. In the present study we analyze the effect of SB3 on hepatoma cell death elicited by a panel of chemotherapeutic agents. We report that SB3 shields cells from the toxicity of drugs with a pro-oxidant action such as doxorubicin, cisplatin and EM20-25. The rapid rise in ROS levels prompted by these compounds causes opening of the mitochondrial permeability transition pore (PTP), irreversibly committing cells to death. We find that a fraction of SB3 locates in mitochondrial inner compartments, and that this mitochondrial fraction increases under conditions of oxidative stress. Mitochondrial SB3 inhibits ROS generation and the ensuing PTP induction and cell death through an inhibitory interaction with respiratory Complex I. These findings identify a novel mechanism of action of SB3 that contributes to tumor cell resistance to anti-neoplastic drugs
Collapse
Affiliation(s)
- Francesco Ciscato
- CNR Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Sheshadri N, Catanzaro JM, Bott AJ, Sun Y, Ullman E, Chen EI, Pan JA, Wu S, Crawford HC, Zhang J, Zong WX. SCCA1/SERPINB3 promotes oncogenesis and epithelial-mesenchymal transition via the unfolded protein response and IL6 signaling. Cancer Res 2014; 74:6318-29. [PMID: 25213322 DOI: 10.1158/0008-5472.can-14-0798] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The serine/cysteine protease inhibitor SCCA1 (SERPINB3) is upregulated in many advanced cancers with poor prognosis, but there is limited information about whether it makes functional contributions to malignancy. Here, we show that SCCA1 expression promoted oncogenic transformation and epithelial-mesenchymal transition (EMT) in mammary epithelial cells, and that SCCA1 silencing in breast cancer cells halted their proliferation. SCCA1 overexpression in neu(+) mammary tumors increased the unfolded protein response (UPR), IL6 expression, and inflammatory phenotypes. Mechanistically, SCCA1 induced a prolonged nonlethal increase in the UPR that was sufficient to activate NF-κB and expression of the protumorigenic cytokine IL6. Overall, our findings established that SCCA1 contributes to tumorigenesis by promoting EMT and a UPR-dependent induction of NF-κB and IL6 autocrine signaling that promotes a protumorigenic inflammation.
Collapse
Affiliation(s)
- Namratha Sheshadri
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York
| | - Joseph M Catanzaro
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York
| | - Alex J Bott
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York
| | - Yu Sun
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York
| | - Erica Ullman
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York
| | - Emily I Chen
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | - Ji-An Pan
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York
| | - Song Wu
- Department of Applied Math and Statistics, Stony Brook University, Stony Brook, New York
| | | | - Jianhua Zhang
- Department of Pathology, University of Alabama, Birmingham VA Medical Center, Birmingham, Alabama
| | - Wei-Xing Zong
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York.
| |
Collapse
|
26
|
Ni HM, Woolbright BL, Williams J, Copple B, Cui W, Luyendyk JP, Jaeschke H, Ding WX. Nrf2 promotes the development of fibrosis and tumorigenesis in mice with defective hepatic autophagy. J Hepatol 2014; 61:617-25. [PMID: 24815875 PMCID: PMC4143992 DOI: 10.1016/j.jhep.2014.04.043] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 04/27/2014] [Accepted: 04/29/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Autophagy is an intracellular lysosomal degradation process that plays an important role in regulating normal physiological functions of the liver. The purpose of the present study was to investigate the mechanism(s) by which the loss of hepatic autophagy leads to liver inflammation, fibrosis and tumorigenesis. METHODS Hepatocyte-specific Atg5 knockout mice were generated by crossing Atg5 Flox/Flox mice with albumin Cre mice. These mice were also crossed with Nrf2 knockout mice to generate Atg5 Flox/Flox, Albumin Cre(+)/Nrf2(-/-) double knockout mice. These mice were housed for various time points up to 15 months, and blood and liver tissues were harvested for biochemical and histological analysis. RESULTS Hepatocyte-specific deletion of Atg5 resulted in increased apoptosis, inflammation and fibrosis in the liver. Increased apoptosis in hepatocyte-specific Atg5 knockout mice was likely due to accumulation of aberrant polyubiquitinated proteins (proteotoxicity) and disruption of the homeostasis of pro-and anti-apoptotic proteins. All of these pathological changes started as early as one month and persisted for 12-15 months. At 9-15 months of age, these mice also developed hepatocellular adenomas. Interestingly, deletion of Nrf2 in Atg5 liver-specific knockout mice markedly abolished these pathological changes, indicating a key role for this transcription factor in the mechanism of hepatic pathology. CONCLUSIONS Our results provide genetic evidence that loss of autophagy in hepatocytes causes cell death resulting in liver inflammation, fibrosis and tumorigenesis. We also demonstrate that persistent activation of Nrf2 is critical for liver inflammation, fibrosis and eventual tumorigenesis that occur in mice with defects in hepatocyte autophagy.
Collapse
Affiliation(s)
- Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Benjamin L Woolbright
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Jessica Williams
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Bryan Copple
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Wei Cui
- Department of Pathology & Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - James P Luyendyk
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States; Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, United States
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States.
| |
Collapse
|
27
|
Gomes S, Marques PI, Matthiesen R, Seixas S. Adaptive evolution and divergence of SERPINB3: a young duplicate in great Apes. PLoS One 2014; 9:e104935. [PMID: 25133778 PMCID: PMC4136820 DOI: 10.1371/journal.pone.0104935] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/14/2014] [Indexed: 11/23/2022] Open
Abstract
A series of duplication events led to an expansion of clade B Serine Protease Inhibitors (SERPIN), currently displaying a large repertoire of functions in vertebrates. Accordingly, the recent duplicates SERPINB3 and B4 located in human 18q21.3 SERPIN cluster control the activity of different cysteine and serine proteases, respectively. Here, we aim to assess SERPINB3 and B4 coevolution with their target proteases in order to understand the evolutionary forces shaping the accelerated divergence of these duplicates. Phylogenetic analysis of primate sequences placed the duplication event in a Hominoidae ancestor (∼30 Mya) and the emergence of SERPINB3 in Homininae (∼9 Mya). We detected evidence of strong positive selection throughout SERPINB4/B3 primate tree and target proteases, cathepsin L2 (CTSL2) and G (CTSG) and chymase (CMA1). Specifically, in the Homininae clade a perfect match was observed between the adaptive evolution of SERPINB3 and cathepsin S (CTSS) and most of sites under positive selection were located at the inhibitor/protease interface. Altogether our results seem to favour a coevolution hypothesis for SERPINB3, CTSS and CTSL2 and for SERPINB4 and CTSG and CMA1. A scenario of an accelerated evolution driven by host-pathogen interactions is also possible since SERPINB3/B4 are potent inhibitors of exogenous proteases, released by infectious agents. Finally, similar patterns of expression and the sharing of many regulatory motifs suggest neofunctionalization as the best fitted model of the functional divergence of SERPINB3 and B4 duplicates.
Collapse
Affiliation(s)
- Sílvia Gomes
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- * E-mail: (SG); (SS)
| | - Patrícia I. Marques
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Rune Matthiesen
- National Health Institute Doutor Ricardo Jorge (INSA), Lisboa, Portugal
| | - Susana Seixas
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- * E-mail: (SG); (SS)
| |
Collapse
|
28
|
Sivaprasad U, Kinker KG, Ericksen MB, Lindsey M, Gibson AM, Bass SA, Hershey NS, Deng J, Medvedovic M, Khurana Hershey GK. SERPINB3/B4 contributes to early inflammation and barrier dysfunction in an experimental murine model of atopic dermatitis. J Invest Dermatol 2014; 135:160-169. [PMID: 25111616 PMCID: PMC4268075 DOI: 10.1038/jid.2014.353] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 07/08/2014] [Accepted: 07/31/2014] [Indexed: 01/21/2023]
Abstract
Serine proteases are critical for epidermal barrier homeostasis, and their aberrant expression and/or activity is associated with chronic skin diseases. Elevated levels of the serine protease inhibitors SERPINB3 and SERPINB4 are seen in patients with atopic dermatitis and psoriasis. However, their mechanistic role in the skin is unknown. To evaluate the contribution of Serpinb3a (mouse homolog of SERPINB3 and SERPINB4) in atopic dermatitis, we examined the effect of topical Aspergillus fumigatus extract exposure in wild-type and Serpinb3a-null mice on transepidermal water loss (TEWL), sensitization, and inflammation. Allergen exposure induced Serpinb3a expression in the skin, along with increased TEWL, epidermal thickness, and skin inflammation, all of which were attenuated in the absence of Serpinb3a. Attenuated TEWL correlated with decreased expression of the pro-inflammatory marker S100A8. Silencing of SERPINB3/B4 in human keratinocytes decreased S100A8 expression, supporting a role for SERPINB3/B4 in the initiation of the acute inflammatory response. RNA-seq analysis following allergen exposure identified a network of pro-inflammatory genes induced in wild-type mice that was absent in Serpinb3a-null mice. In conclusion, Serpinb3a deficiency attenuates barrier dysfunction and the early inflammatory response following cutaneous allergen exposure, supporting a role for Serpinb3a (mice) and SERPINB3/B4 (humans) early in atopic dermatitis.
Collapse
Affiliation(s)
- Umasundari Sivaprasad
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kayla G Kinker
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mark B Ericksen
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mark Lindsey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Aaron M Gibson
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stacey A Bass
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Nicolas S Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jingyuan Deng
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mario Medvedovic
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA.
| |
Collapse
|
29
|
Catanzaro JM, Sheshadri N, Pan JA, Sun Y, Shi C, Li J, Powers RS, Crawford HC, Zong WX. Oncogenic Ras induces inflammatory cytokine production by upregulating the squamous cell carcinoma antigens SerpinB3/B4. Nat Commun 2014; 5:3729. [PMID: 24759783 PMCID: PMC4025922 DOI: 10.1038/ncomms4729] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 03/26/2014] [Indexed: 12/14/2022] Open
Abstract
Mounting evidence indicates that oncogenic Ras can modulate cell autonomous inflammatory cytokine production, although the underlying mechanism remains unclear. Here we show that squamous cell carcinoma antigens 1 and 2 (SCCA1/2), members of the Serpin family of serine/cysteine protease inhibitors, are transcriptionally up-regulated by oncogenic Ras via MAPK and the ETS family transcription factor PEA3. Increased SCCA expression leads to inhibition of protein turnover, unfolded protein response, activation of NF-κB, and is essential for Ras-mediated cytokine production and tumor growth. Analysis of human colorectal and pancreatic tumor samples reveals a positive correlation between Ras mutation, enhanced SCCA expression, and IL-6 expression. These results indicate that SCCA is a Ras-responsive factor that has a role in Ras-associated cytokine production and tumorigenesis.
Collapse
Affiliation(s)
- Joseph M Catanzaro
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Namratha Sheshadri
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Ji-An Pan
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Yu Sun
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Chanjuan Shi
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Jinyu Li
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, New York 11797, USA
| | - R Scott Powers
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, New York 11797, USA
| | - Howard C Crawford
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224, USA
| | - Wei-Xing Zong
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| |
Collapse
|
30
|
Gatto M, Iaccarino L, Ghirardello A, Bassi N, Pontisso P, Punzi L, Shoenfeld Y, Doria A. Serpins, immunity and autoimmunity: old molecules, new functions. Clin Rev Allergy Immunol 2014; 45:267-80. [PMID: 23325331 DOI: 10.1007/s12016-013-8353-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Serine protease inhibitors (serpins) are evolutionary old, structurally conserved molecules which encompass nearly all branches of life. More than 1,000 serpins were characterized to date which are subdivided into 16 subgroups (A-P) according to their common ancestry; among them, 37 are found in humans. Serpins were termed after their capability to inhibit serine proteases, but mounting evidence suggests that they may achieve a greater deal of functions, ranging from embryological growth to synaptic plasticity, development of both myeloid and lymphoid immune cells, and modulation of apoptosis. Serpins are mainly extracellular molecules, although some of them (namely, ov-serpins or clade B serpins) mostly act inside the cells, being either ubiquitously or tissue-specifically expressed. Among newly characterized serpin functions, regulation of cellular proliferation through apoptosis modulation and proteasome disturbance seems to play a major role. Accordingly, several serpins were found to be hyperexpressed in tumor cells. Indeed, apoptosis dysregulation is likely to be a cornerstone in both tumorigenesis and autoimmunity, since uncontrolled cellular viability results in tumor proliferation, while inefficient disposal of apoptotic debris may favor the rescue of autoreactive immune cells. Such a process was widely documented in systemic lupus erythematosus (SLE). Interestingly, alterations in the expression of some serpins, e.g., the ov-serpin SERPINB3, are being unraveled in patients affected with SLE and other autoimmune disorders, suggesting that a failure in serpin function might affect immune homeostasis and self-tolerance, thereby contributing to autoimmunity. Here, we provide an overview of serpin origin, function, and dysfunction, focusing on human serpins and ov-serpins, with a hub on SERPINB3.
Collapse
Affiliation(s)
- Mariele Gatto
- Division of Rheumatology, Department of Medicine, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhang YB, Zhao W, Zeng RX. Autophagic degradation of caspase-8 protects U87MG cells against H2O2-induced oxidative stress. Asian Pac J Cancer Prev 2014; 14:4095-9. [PMID: 23991959 DOI: 10.7314/apjcp.2013.14.7.4095] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Oxidative stress induces apoptosis in many cellular systems including glioblastoma cells, with caspase-8 activation was regarded as a major contribution to H2O2-induced cell death. This study focused on the role of the autophagic protein p62 in H2O2-induced apoptosis in U87MG cells. Oxidative stress was applied with H2O2, and cell apoptosis and viability were measured with use of caspase inhibitors or autophagic mediators or siRNA p62, GFP-p62 and GFP-p62-UBA (del) transfection. We found that H2O2 -induced U87MG cell death was correlated with caspase-8. To understand the role of p62 in MG132-induced cell death, the levels of p62/SQSTM1 or autophagy in U87MG cells were modulated with biochemical or genetic methods. The results showed that the over-expression of wild type p62/SQSTM1 significantly reduced H2O2 induced cell death, but knockdown of p62 aggravated the process. In addition, inhibition of autophagy promoted p62 and active caspase-8 increasing H2O2 -induced apoptosis while induction of autophagy manifested the opposite effect. We further demonstrated that the function of p62/SQSTM1 required its C-terminus UBA domain to attenuate H2O2 cytotoxity by inhibition of caspase-8 activity. Our results indicated that p62/SQSTM1 was a potential contributor to mediate caspase-8 activation by autophagy in oxidative stress process.
Collapse
Affiliation(s)
- Yi-Bo Zhang
- Department of Pathogen Biology, School of Basic Medical Sciences, Liaoning Medical University, Jinzhou, China.
| | | | | |
Collapse
|
32
|
Biswas S, Torchilin VP. Nanopreparations for organelle-specific delivery in cancer. Adv Drug Deliv Rev 2014; 66:26-41. [PMID: 24270008 DOI: 10.1016/j.addr.2013.11.004] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 10/30/2013] [Accepted: 11/13/2013] [Indexed: 01/07/2023]
Abstract
To efficiently deliver therapeutics into cancer cells, a number of strategies have been recently investigated. The toxicity associated with the administration of chemotherapeutic drugs due to their random interactions throughout the body necessitates the development of drug-encapsulating nanopreparations that significantly mask, or reduce, the toxic side effects of the drugs. In addition to reduced side effects associated with drug encapsulation, nanocarriers preferentially accumulate in tumors as a result of its abnormally leaky vasculature via the Enhanced Permeability and Retention (EPR) effect. However, simple passive nanocarrier delivery to the tumor site is unlikely to be enough to elicit a maximum therapeutic response as the drug-loaded carriers must reach the intracellular target sites. Therefore, efficient translocation of the nanocarrier through the cell membrane is necessary for cytosolic delivery of the cargo. However, crossing the cell membrane barrier and reaching cytosol might still not be enough for achieving maximum therapeutic benefit, which necessitates the delivery of drugs directly to intracellular targets, such as bringing pro-apoptotic drugs to mitochondria, nucleic acid therapeutics to nuclei, and lysosomal enzymes to defective lysosomes. In this review, we discuss the strategies developed for tumor targeting, cytosolic delivery via cell membrane translocation, and finally organelle-specific targeting, which may be applied for developing highly efficacious, truly multifunctional, cancer-targeted nanopreparations.
Collapse
Affiliation(s)
- Swati Biswas
- Center for Pharmaceutical Biotechnology and Nanomedicine, 360 Huntington Avenue, 140 The Fenway, Northeastern University, Boston, 02115, USA; Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad, Andhra Pradesh 500078, India
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, 360 Huntington Avenue, 140 The Fenway, Northeastern University, Boston, 02115, USA.
| |
Collapse
|
33
|
Sridevi P, Nhiayi MK, Setten RL, Wang JYJ. Persistent inhibition of ABL tyrosine kinase causes enhanced apoptotic response to TRAIL and disrupts the pro-apoptotic effect of chloroquine. PLoS One 2013; 8:e77495. [PMID: 24147007 PMCID: PMC3795698 DOI: 10.1371/journal.pone.0077495] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 09/04/2013] [Indexed: 12/14/2022] Open
Abstract
TNF-Related Apoptosis Inducing Ligand (TRAIL) binds to and activates death receptors to stimulate caspase-8 and apoptosis with higher efficiency in cancer than normal cells but the development of apoptosis resistance has limited its clinical efficacy. We found that stable, but not transient knockdown of the ABL tyrosine kinase enhanced the apoptotic response to TRAIL. Re-expression of Abl, but not its nuclear import- or kinase-defective mutant, in the ABL-knockdown cells re-established apoptosis suppression. TRAIL is known to stimulate caspase-8 ubiquitination (Ub-C8), which can facilitate caspase-8 activation or degradation by the lysosomes. In the ABL-knockdown cells, we found a higher basal level of Ub-C8 that was not further increased by lysosomal inhibition. Re-expression of Abl in the ABL-knockdown cells reduced the basal Ub-C8, correlating with apoptosis suppression. We found that lysosomal inhibition by chloroquine (CQ) could also enhance TRAIL-induced apoptosis. However, this pro-apoptotic effect of CQ was lost in the ABL-knockdown cells but restored by Abl re-expression. Interestingly, kinase inhibition at the time of TRAIL stimulation was not sufficient to enhance apoptosis. Instead, persistent treatment for several days with imatinib, an ABL kinase inhibitor, was required to cause the enhanced and the CQ-insensitive apoptotic response to TRAIL. Together, these results show that persistent loss of nuclear ABL tyrosine kinase function can sensitize cells to TRAIL and suggest that long-term exposure to the FDA-approved ABL kinase inhibitors may potentiate apoptotic response to TRAIL-based cancer therapy.
Collapse
Affiliation(s)
- Priya Sridevi
- Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - May K. Nhiayi
- Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Ryan L. Setten
- Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jean Y. J. Wang
- Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Manley S, Williams JA, Ding WX. Role of p62/SQSTM1 in liver physiology and pathogenesis. Exp Biol Med (Maywood) 2013; 238:525-38. [PMID: 23856904 DOI: 10.1177/1535370213489446] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
p62/sequestosome-1/A170/ZIP (hereafter referred to as p62) is a scaffold protein that has multiple functions, such as signal transduction, cell proliferation, cell survival, cell death, inflammation, tumourigenesis and oxidative stress response. While p62 is an autophagy substrate and is degraded by autophagy, p62 serves as an autophagy receptor for selective autophagic clearance of protein aggregates and organelles. Moreover, p62 functions as a signalling hub for various signalling pathways, including NF-κB, Nrf2 and mTOR. In this review, we discuss the pathophysiological role of p62 in the liver, including formation of hepatic inclusion bodies, cholestasis, obesity, insulin resistance, liver cell death and tumourigenesis.
Collapse
Affiliation(s)
- Sharon Manley
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, USA
| | | | | |
Collapse
|
35
|
Weber H, Müller L, Jonas L, Schult C, Sparmann G, Schuff-Werner P. Calpain mediates caspase-dependent apoptosis initiated by hydrogen peroxide in pancreatic acinar AR42J cells. Free Radic Res 2013; 47:432-46. [PMID: 23495712 DOI: 10.3109/10715762.2013.785633] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Several studies have shown that oxidative stress induces apoptosis in many cellular systems including pancreatic acinar cells. However, the exact molecular mechanisms leading to apoptosis remain partially understood. This study aimed to investigate the role of the cytosolic cysteine protease calpain in H2O2-induced apoptosis in pancreatic AR42J cells. Apoptosis was evaluated using flow cytometric analysis of sub-G1 DNA populations, electron-microscopic analysis, caspase-3-specific αII-spectrin breakdown, and measuring the proteolytic activities of the initiator caspase-12 and caspase-8, and the executioner caspase-3. H2O2 induced an increase in the calpain proteolytic activity immediately after starting the experiments that tended to return to a nearly normal level after 8 h and could be attributed to m-calpain. Whereas no caspase-12, caspase-8 and caspase-3 activations could be detected within the first 0.5 h, significantly increased proteolytic activities were observed after 8 h compared with the control. At the same time, the cells showed first ultrastructural hallmarks of apoptosis and a decreased viability. In addition, αII-spectrin fragmentation was identified using immunoblotting that could be attributed to both calpain and caspase-3. Calpain inhibition reduced the activities of caspase-12, caspase-8, and caspase-3 leading to a decrease in the number of apoptotic cells. Immunoblotting analyses of caspase-12 and caspase-8 indicate that calpain may be involved in the activation process of both proteases. The results suggest that H2O2-induced apoptosis of AR42J cells requires activation of m-calpain initiating the endoplasmic reticulum stress-induced caspase-12 pathway and a caspase-8-dependent pathway. The findings also suggest that calpain may be involved in the execution phase of apoptosis.
Collapse
Affiliation(s)
- H Weber
- Institute of Clinical Chemistry and Laboratory Medicine, University of Rostock, Rostock, Germany.
| | | | | | | | | | | |
Collapse
|
36
|
Pan JA, Fan Y, Gandhirajan RK, Madesh M, Zong WX. Hyperactivation of the mammalian degenerin MDEG promotes caspase-8 activation and apoptosis. J Biol Chem 2012; 288:2952-63. [PMID: 23239879 DOI: 10.1074/jbc.m112.441063] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Intracellular calcium overload plays a critical role in numerous pathological syndromes such as heart failure, brain ischemia, and stroke. Hyperactivation of the acid-sensing ion channels including degenerin/epithelial amiloride-sensitive sodium (DEG/ENaC) channels has been shown to elevate intracellular calcium and cause subsequent neuronal cell death that is independent of the canonical Egl-1/Ced-9/Ced-4/Ced-3 apoptotic pathway in Caenorhabditis elegans. In mammalian cells, hyperactivation of the DEG/ENaC channels can also lead to cell death, although the underlying mechanism remains largely unknown. Here, we use a tetracycline-inducible system to express the hyperactivation mutant of a mammalian DEG/ENaC channel protein, MDEG G430F, in murine kidney epithelial cells deficient in the key mitochondrial apoptotic proteins Bax and Bak. Remarkably, expression of MDEG G430F induces increased intracellular calcium, reactive oxygen species (ROS) production, and cell death. The MDEG G430F-induced cell death is blocked by the intracellular calcium chelator 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (acetoxymethyl ester), ROS scavengers, and the caspase inhibitor z-VAD-fmk (where z and fmk are benzyloxycarbonyl and fluoromethyl ketone). Mechanistically, the intracellular calcium overload and ROS increase lead to the inhibition of proteasomal and autophagic protein degradation, which promotes the accumulation of protein aggregates containing caspase-8 and subsequent caspase-8 activation. As protein aggregation upon the inhibition of proteasomal and autophagic degradation pathways is mediated by the ubiquitin-binding protein SQSTM1/p62 and the autophagy-related protein LC3, silencing of p62 and LC3 protects cells from MDEG G430F-induced cell death. Our results uncover a new mechanism of caspase-8-mediated apoptosis induced by intracellular calcium overload that is dependent on the autophagy-related proteins LC3 and p62 upon hyperactivation of DEG/ENaC channels.
Collapse
Affiliation(s)
- Ji-An Pan
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11794, USA
| | | | | | | | | |
Collapse
|
37
|
Inhibition of protein degradation induces apoptosis through a microtubule-associated protein 1 light chain 3-mediated activation of caspase-8 at intracellular membranes. Mol Cell Biol 2011; 31:3158-70. [PMID: 21628531 DOI: 10.1128/mcb.05460-11] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The accumulation of damaged or misfolded proteins, if unresolved, can lead to a detrimental consequence within cells termed proteotoxicity. Since cancerous cells often display elevated protein synthesis and by-product disposal, inhibition of the protein degradation pathways is an emerging approach for cancer therapy. However, the molecular mechanism underlying proteotoxicity remains largely unclear. We show here that inhibition of proteasomal degradation results in an increased oligomerization and activation of caspase-8 on the cytosolic side of intracellular membranes. This enhanced caspase-8 oligomerization and activation are promoted through its interaction with the ubiquitin-binding protein SQSTM1/p62 and the microtubule-associated protein light chain 3 (LC3), which are enriched at intracellular membranes in response to proteotoxic stress. Silencing LC3 by shRNA, or the LC3 mutants defective in membrane localization or p62 interaction fail to induce caspase-8 activation and apoptosis. Our results unveiled a previously unknown mechanism through which disruption of protein homeostasis induces caspase-8 oligomerization, activation, and apoptosis.
Collapse
|