1
|
Huang J, Luo S, Shen J, Lee M, Chen R, Ma S, Sun LQ, Li JJ. Cellular polarity pilots breast cancer progression and immunosuppression. Oncogene 2025; 44:783-793. [PMID: 40057606 PMCID: PMC11913746 DOI: 10.1038/s41388-025-03324-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
Disrupted cellular polarity (DCP) is a hallmark of solid cancer, the malignant disease of epithelial tissues, which occupies ~90% of all human cancers. DCP has been identified to affect not only the cancer cell's aggressive behavior but also the migration and infiltration of immune cells, although the precise mechanism of DCP-affected tumor-immune cell interaction remains unclear. This review discusses immunosuppressive tumor microenvironments (TME) caused by DCP-driven tumor cell proliferation with DCP-impaired immune cell functions. We will revisit the fundamental roles of cell polarity (CP) proteins in sustaining mammary luminal homeostasis, epithelial transformation, and breast cancer progression. Then, the current data on CP involvement in immune cell activation, maturation, migration, and tumor infiltration are evaluated. The CP status on the immune effector cells and their targeted tumor cells are highlighted in tumor immune regulation, including the antigen presentation and the formation of immune synapses (IS). CP-regulated antigen presentation and delivery and the formation of IS between the immune cells, especially between the immune effectors and tumor cells, will be addressed. Alterations of CP on the tumor cells, infiltrated immune effector cells, or both are discussed with these aspects. We conclude that CP-mediated tumor aggressiveness coupled with DCP-impaired immune cell disability may decide the degree of immunosuppressive status and responsiveness to immune checkpoint blockade (ICB). Further elucidating the dynamics of CP- or DCP-mediated immune regulation in TME will provide more critical insights into tumor-immune cell dynamics, which is required to invent more effective approaches for cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Huang
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Shufeng Luo
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Cancer Center, Central South University, China, Hunan, Changsha
| | - Juan Shen
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Maya Lee
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Rachel Chen
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lun-Quan Sun
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Cancer Center, Central South University, China, Hunan, Changsha.
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA.
- NCI-designated Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California, USA.
| |
Collapse
|
2
|
Wen L, Liu Z, Zhou L, Liu Z, Li Q, Geng B, Xia Y. Bone and Extracellular Signal-Related Kinase 5 (ERK5). Biomolecules 2024; 14:556. [PMID: 38785963 PMCID: PMC11117709 DOI: 10.3390/biom14050556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/17/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Bones are vital for anchoring muscles, tendons, and ligaments, serving as a fundamental element of the human skeletal structure. However, our understanding of bone development mechanisms and the maintenance of bone homeostasis is still limited. Extracellular signal-related kinase 5 (ERK5), a recently identified member of the mitogen-activated protein kinase (MAPK) family, plays a critical role in the pathogenesis and progression of various diseases, especially neoplasms. Recent studies have highlighted ERK5's significant role in both bone development and bone-associated pathologies. This review offers a detailed examination of the latest research on ERK5 in different tissues and diseases, with a particular focus on its implications for bone health. It also examines therapeutic strategies and future research avenues targeting ERK5.
Collapse
Affiliation(s)
- Lei Wen
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming 650032, China
| | - Zirui Liu
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Libo Zhou
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Zhongcheng Liu
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Qingda Li
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Bin Geng
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Yayi Xia
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| |
Collapse
|
3
|
Clinical Significance and Regulation of ERK5 Expression and Function in Cancer. Cancers (Basel) 2022; 14:cancers14020348. [PMID: 35053510 PMCID: PMC8773716 DOI: 10.3390/cancers14020348] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/08/2022] [Accepted: 01/08/2022] [Indexed: 02/06/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) is a unique kinase among MAPKs family members, given its large structure characterized by the presence of a unique C-terminal domain. Despite increasing data demonstrating the relevance of the ERK5 pathway in the growth, survival, and differentiation of normal cells, ERK5 has recently attracted the attention of several research groups given its relevance in inflammatory disorders and cancer. Accumulating evidence reported its role in tumor initiation and progression. In this review, we explore the gene expression profile of ERK5 among cancers correlated with its clinical impact, as well as the prognostic value of ERK5 and pERK5 expression levels in tumors. We also summarize the importance of ERK5 in the maintenance of a cancer stem-like phenotype and explore the major known contributions of ERK5 in the tumor-associated microenvironment. Moreover, although several questions are still open concerning ERK5 molecular regulation, different ERK5 isoforms derived from the alternative splicing process are also described, highlighting the potential clinical relevance of targeting ERK5 pathways.
Collapse
|
4
|
Paudel R, Fusi L, Schmidt M. The MEK5/ERK5 Pathway in Health and Disease. Int J Mol Sci 2021; 22:ijms22147594. [PMID: 34299213 PMCID: PMC8303459 DOI: 10.3390/ijms22147594] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
The MEK5/ERK5 mitogen-activated protein kinases (MAPK) cascade is a unique signaling module activated by both mitogens and stress stimuli, including cytokines, fluid shear stress, high osmolarity, and oxidative stress. Physiologically, it is mainly known as a mechanoreceptive pathway in the endothelium, where it transduces the various vasoprotective effects of laminar blood flow. However, it also maintains integrity in other tissues exposed to mechanical stress, including bone, cartilage, and muscle, where it exerts a key function as a survival and differentiation pathway. Beyond its diverse physiological roles, the MEK5/ERK5 pathway has also been implicated in various diseases, including cancer, where it has recently emerged as a major escape route, sustaining tumor cell survival and proliferation under drug stress. In addition, MEK5/ERK5 dysfunction may foster cardiovascular diseases such as atherosclerosis. Here, we highlight the importance of the MEK5/ERK5 pathway in health and disease, focusing on its role as a protective cascade in mechanical stress-exposed healthy tissues and its function as a therapy resistance pathway in cancers. We discuss the perspective of targeting this cascade for cancer treatment and weigh its chances and potential risks when considering its emerging role as a protective stress response pathway.
Collapse
|
5
|
Xu Q, Zhang J, Telfer BA, Zhang H, Ali N, Chen F, Risa B, Pearson AJ, Zhang W, Finegan KG, Ucar A, Giurisato E, Tournier C. The extracellular-regulated protein kinase 5 (ERK5) enhances metastatic burden in triple-negative breast cancer through focal adhesion protein kinase (FAK)-mediated regulation of cell adhesion. Oncogene 2021; 40:3929-3941. [PMID: 33981002 PMCID: PMC8195737 DOI: 10.1038/s41388-021-01798-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 03/23/2021] [Accepted: 04/14/2021] [Indexed: 12/22/2022]
Abstract
There is overwhelming clinical evidence that the extracellular-regulated protein kinase 5 (ERK5) is significantly dysregulated in human breast cancer. However, there is no definite understanding of the requirement of ERK5 in tumor growth and metastasis due to very limited characterization of the pathway in disease models. In this study, we report that a high level of ERK5 is a predictive marker of metastatic breast cancer. Mechanistically, our in vitro data revealed that ERK5 was critical for maintaining the invasive capability of triple-negative breast cancer (TNBC) cells through focal adhesion protein kinase (FAK) activation. Specifically, we found that phosphorylation of FAK at Tyr397 was controlled by a kinase-independent function of ERK5. Accordingly, silencing ERK5 in mammary tumor grafts impaired FAK phosphorylation at Tyr397 and suppressed TNBC cell metastasis to the lung without preventing tumor growth. Collectively, these results establish a functional relationship between ERK5 and FAK signaling in promoting malignancy. Thus, targeting the oncogenic ERK5-FAK axis represents a promising therapeutic strategy for breast cancer exhibiting aggressive clinical behavior.
Collapse
Affiliation(s)
- Qiuping Xu
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jingwei Zhang
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Brian A Telfer
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Hao Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Nisha Ali
- Manchester University NHS FT, Wythenshawe hospital, Manchester, UK
| | - Fuhui Chen
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Blanca Risa
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Adam J Pearson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Wei Zhang
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Katherine G Finegan
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ahmet Ucar
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emanuele Giurisato
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
6
|
Hoang VT, Matossian MD, Ucar DA, Elliott S, La J, Wright MK, Burks HE, Perles A, Hossain F, King CT, Browning VE, Bursavich J, Fang F, Del Valle L, Bhatt AB, Cavanaugh JE, Flaherty PT, Anbalagan M, Rowan BG, Bratton MR, Nephew KP, Miele L, Collins-Burow BM, Martin EC, Burow ME. ERK5 Is Required for Tumor Growth and Maintenance Through Regulation of the Extracellular Matrix in Triple Negative Breast Cancer. Front Oncol 2020; 10:1164. [PMID: 32850332 PMCID: PMC7416559 DOI: 10.3389/fonc.2020.01164] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 06/09/2020] [Indexed: 12/16/2022] Open
Abstract
Conventional mitogen-activated protein kinase (MAPK) family members regulate diverse cellular processes involved in tumor initiation and progression, yet the role of ERK5 in cancer biology is not fully understood. Triple-negative breast cancer (TNBC) presents a clinical challenge due to the aggressive nature of the disease and a lack of targeted therapies. ERK5 signaling contributes to drug resistance and metastatic progression through distinct mechanisms, including activation of epithelial-to-mesenchymal transition (EMT). More recently a role for ERK5 in regulation of the extracellular matrix (ECM) has been proposed, and here we investigated the necessity of ERK5 in TNBC tumor formation. Depletion of ERK5 expression using the CRISPR/Cas9 system in MDA-MB-231 and Hs-578T cells resulted in loss of mesenchymal features, as observed through gene expression profile and cell morphology, and suppressed TNBC cell migration. In vivo xenograft experiments revealed ERK5 knockout disrupted tumor growth kinetics, which was restored using high concentration Matrigel™ and ERK5-ko reduced expression of the angiogenesis marker CD31. These findings implicated a role for ERK5 in the extracellular matrix (ECM) and matrix integrity. RNA-sequencing analyses demonstrated downregulation of matrix-associated genes, integrins, and pro-angiogenic factors in ERK5-ko cells. Tissue decellularization combined with cryo-SEM and interrogation of biomechanical properties revealed that ERK5-ko resulted in loss of key ECM fiber alignment and mechanosensing capabilities in breast cancer xenografts compared to parental wild-type cells. In this study, we identified a novel role for ERK5 in tumor growth kinetics through modulation of the ECM and angiogenesis axis in breast cancer.
Collapse
Affiliation(s)
- Van T. Hoang
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Margarite D. Matossian
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Deniz A. Ucar
- Department of Genetics, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Steven Elliott
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jacqueline La
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Maryl K. Wright
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Hope E. Burks
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Aaron Perles
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Fokhrul Hossain
- Department of Genetics, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Connor T. King
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Valentino E. Browning
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Jacob Bursavich
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Fang Fang
- Medical Sciences, School of Medicine, Indiana University Bloomington, Bloomington, IN, United States
| | - Luis Del Valle
- Department of Pathology, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Akshita B. Bhatt
- Department of Pharmacology, School of Pharmacy, Duquesne University, Pittsburgh, PA, United States
| | - Jane E. Cavanaugh
- Department of Pharmacology, School of Pharmacy, Duquesne University, Pittsburgh, PA, United States
| | - Patrick T. Flaherty
- Department of Medicinal Chemistry, School of Pharmacy, Duquesne University, Pittsburgh, PA, United States
| | - Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Melyssa R. Bratton
- Cellular and Molecular Biology Core, Xavier University, New Orleans, LA, United States
| | - Kenneth P. Nephew
- Medical Sciences, School of Medicine, Indiana University Bloomington, Bloomington, IN, United States
| | - Lucio Miele
- Department of Genetics, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Bridgette M. Collins-Burow
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Cancer Center, New Orleans, LA, United States
| | - Elizabeth C. Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Matthew E. Burow
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
7
|
Impact of ERK5 on the Hallmarks of Cancer. Int J Mol Sci 2019; 20:ijms20061426. [PMID: 30901834 PMCID: PMC6471124 DOI: 10.3390/ijms20061426] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 12/15/2022] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) belongs to the mitogen-activated protein kinase (MAPK) family that consists of highly conserved enzymes expressed in all eukaryotic cells and elicits several biological responses, including cell survival, proliferation, migration, and differentiation. In recent years, accumulating lines of evidence point to a relevant role of ERK5 in the onset and progression of several types of cancer. In particular, it has been reported that ERK5 is a key signaling molecule involved in almost all the biological features of cancer cells so that its targeting is emerging as a promising strategy to suppress tumor growth and spreading. Based on that, in this review, we pinpoint the hallmark-specific role of ERK5 in cancer in order to identify biological features that will potentially benefit from ERK5 targeting.
Collapse
|
8
|
Focus on Cdc42 in Breast Cancer: New Insights, Target Therapy Development and Non-Coding RNAs. Cells 2019; 8:cells8020146. [PMID: 30754684 PMCID: PMC6406589 DOI: 10.3390/cells8020146] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 12/25/2022] Open
Abstract
Breast cancer is the most common malignant tumors in females. Although the conventional treatment has demonstrated a certain effect, some limitations still exist. The Rho guanosine triphosphatase (GTPase) Cdc42 (Cell division control protein 42 homolog) is often upregulated by some cell surface receptors and oncogenes in breast cancer. Cdc42 switches from inactive guanosine diphosphate (GDP)-bound to active GTP-bound though guanine-nucleotide-exchange factors (GEFs), results in activation of signaling cascades that regulate various cellular processes such as cytoskeletal changes, proliferation and polarity establishment. Targeting Cdc42 also provides a strategy for precise breast cancer therapy. In addition, Cdc42 is a potential target for several types of non-coding RNAs including microRNAs and lncRNAs. These non-coding RNAs is extensively involved in Cdc42-induced tumor processes, while many of them are aberrantly expressed. Here, we focus on the role of Cdc42 in cell morphogenesis, proliferation, motility, angiogenesis and survival, introduce the Cdc42-targeted non-coding RNAs, as well as present current development of effective Cdc42-targeted inhibitors in breast cancer.
Collapse
|
9
|
Adam C, Glück L, Ebert R, Goebeler M, Jakob F, Schmidt M. The MEK5/ERK5 mitogen-activated protein kinase cascade is an effector pathway of bone-sustaining bisphosphonates that regulates osteogenic differentiation and mineralization. Bone 2018; 111:49-58. [PMID: 29567200 DOI: 10.1016/j.bone.2018.03.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 01/24/2023]
Abstract
Bisphosphonates play an important role in the treatment of metabolic bone diseases such as osteoporosis. In addition to their anti-resorptive activity by triggering osteoclast apoptosis, nitrogen-containing bisphosphonates (N-BP) may also influence osteogenic differentiation, which might rely on their capacity to inhibit the mevalonate pathway. In vascular endothelial cells inhibition of this pathway by cholesterol-lowering statins activates the MEK5/ERK5 mitogen-activated protein kinase cascade, which plays an important role in cellular differentiation, apoptosis or inflammatory processes. Here we evaluated whether N-BP may also target the MEK5/ERK5 pathway and analysed the consequences of ERK5 activation on osteogenic differentiation. We show that N-BP dose-dependently activate ERK5 in primary human endothelial cells and osteoblasts. The mechanism likely involves farnesyl pyrophosphate synthase inhibition and subsequent functional inhibition of the small GTPase Cdc42 since siRNA-mediated knockdown of both genes could reproduce N-BP-induced ERK5 activation. ERK5 activation resulted in regulation of several bone-relevant genes and was required for calcification and osteogenic differentiation of bone marrow-derived mesenchymal stems cells as evident by the lack of alkaline phosphatase induction and alizarin-red S staining observed upon ERK5 knockdown or upon differentiation initiation in presence of a pharmacological ERK5 inhibitor. Our data provide evidence that N-BP activate the MEK5/ERK5 cascade and reveal an essential role of ERK5 in osteogenic differentiation and mineralization of skeletal precursors.
Collapse
Affiliation(s)
- Christian Adam
- Department of Dermatology, University Hospital Würzburg, Germany
| | - Lucia Glück
- Department of Dermatology, University Hospital Würzburg, Germany
| | - Regina Ebert
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Germany
| | | | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Germany
| | - Marc Schmidt
- Department of Dermatology, University Hospital Würzburg, Germany.
| |
Collapse
|
10
|
Min J, Geng H, Liu Z, Liang Z, Zhang Z, Xie D, Wang Y, Zhang T, Yu D, Zhong C. ERK5 regulates tobacco smoke‑induced urocystic epithelial‑mesenchymal transition in BALB/c mice. Mol Med Rep 2017; 15:3893-3897. [PMID: 28440402 DOI: 10.3892/mmr.2017.6457] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/07/2017] [Indexed: 11/05/2022] Open
Abstract
Tobacco smoke (TS) is an important risk factor of bladder cancer. Epithelial‑mesenchymal transition (EMT) is involved in the initiation and development of cancer. The role of extracellular signal‑regulated kinase (ERK) 5 in regulating TS‑induced EMT remains to be elucidated. The aim of the present study was to investigate the regulatory role of ERK5 in TS‑triggered EMT in the bladder of mice. BALB/c mice were used for an in vivo TS exposure model. Mice were treated for 6 h a day for 12 weeks. The results demonstrated that mice exposed to TS had decreased mRNA and protein expression levels of the epithelial markers E‑cadherin and zonula occludens‑1, whereas expression levels of the mesenchymal markers Vimentin and N‑cadherin were increased. Treatment with XMD8‑92, a highly specific ERK5 inhibitor, effectively abrogated TS‑triggered activation of ERK5, activator protein‑1 and EMT alterations in the bladder of BALB/c mice. The data suggested that ERK5 regulates TS‑mediated urocystic EMT. These findings provide insight into the molecular mechanisms of TS‑associated bladder tumorigenesis.
Collapse
Affiliation(s)
- Jie Min
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Hao Geng
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhiqi Liu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhaofeng Liang
- Department of Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhiqiang Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Dongdong Xie
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yi Wang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Tao Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Dexin Yu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
11
|
Extracellular Signal-Regulated Kinase 5 is Required for Low-Concentration H 2O 2-Induced Angiogenesis of Human Umbilical Vein Endothelial Cells. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6895730. [PMID: 28540300 PMCID: PMC5429924 DOI: 10.1155/2017/6895730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/22/2017] [Accepted: 03/09/2017] [Indexed: 01/28/2023]
Abstract
Background. The aim of this study was to assess the effects of low concentrations of H2O2 on angiogenesis of human umbilical vein endothelial cells (HUVECs) in vitro and explore the underlying mechanisms. Methods. HUVECs were cultured and stimulated with different concentrations of H2O2. Flow cytometric analysis was used to select an optimal concentration of H2O2 for the following experiments. Cell proliferation, migration, and tubule formation were evaluated by Cell Counting Kit-8 (CCK-8) assays, scratch wound assays, and Matrigel tubule formation assays, respectively. For gain and loss of function studies, constitutively active MEK5 (CA-MEK5) and ERK5 shRNA lentiviruses were used to activate or knock down extracellular signal-regulated kinase 5 (ERK5). Results. We found that low concentrations of H2O2 promoted HUVECs proliferation, migration, and tubule formation. ERK5 in HUVECs was significantly activated by H2O2. Enhanced ERK5 activity significantly amplified the proangiogenic effects of H2O2; in contrast, ERK5 knock-down abrogated the effects of H2O2. Conclusions. Our results confirmed that low concentrations of H2O2 promoted HUVECs angiogenesis in vitro, and ERK5 is an essential mediator of this process. Therefore, ERK5 may be a potential therapeutic target for promoting angiogenesis and improving graft survival.
Collapse
|
12
|
Hoang VT, Yan TJ, Cavanaugh JE, Flaherty PT, Beckman BS, Burow ME. Oncogenic signaling of MEK5-ERK5. Cancer Lett 2017; 392:51-59. [PMID: 28153789 PMCID: PMC5901897 DOI: 10.1016/j.canlet.2017.01.034] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/17/2017] [Accepted: 01/23/2017] [Indexed: 12/17/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) regulate diverse cellular processes including proliferation, cell survival, differentiation, and apoptosis. While conventional MAPK constituents have well-defined roles in oncogenesis, the MEK5 pathway has only recently emerged in cancer research. In this review, we consider the MEK5 signaling cascade, focusing specifically on its involvement in drug resistance and regulation of aggressive cancer phenotypes. Moreover, we explore the role of MEK5/ERK5 in tumorigenesis and metastatic progression, discussing the discrepancies in preclinical studies and assessing its viability as a therapeutic target for anti-cancer agents.
Collapse
Affiliation(s)
- Van T Hoang
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Thomas J Yan
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Jane E Cavanaugh
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Patrick T Flaherty
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | | | - Matthew E Burow
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA; Department of Pharmacology, Tulane University, New Orleans, LA, USA; Tulane Cancer Center, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
13
|
Yang D, Zhang Y, Cheng Y, Hong L, Wang C, Wei Z, Cai Q, Yan R. High Expression of Cell Division Cycle 42 Promotes Pancreatic Cancer Growth and Predicts Poor Outcome of Pancreatic Cancer Patients. Dig Dis Sci 2017; 62:958-967. [PMID: 28181096 DOI: 10.1007/s10620-017-4451-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 01/10/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND Cell division cycle 42 (CDC42), an important member of the Rho family, is overexpressed in various human cancers. However, its expression and role in pancreatic cancer (PC) are not well understood. AIM The present study was designed to investigate the expression patterns and underlying cellular mechanisms of CDC42 in PC. METHODS First, immunohistochemical analysis, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were performed to detect CDC42 expression in clinical pancreatic carcinoma and adjacent tissues. Second, differential expression of CDC42 between PC cells and normal cells was evaluated by qRT-PCR and Western blotting. Third, the correlation between CDC42 expression as well as clinicopathological characteristics and patient survival was analyzed. Finally, CDC42 was knocked down to examine its role both in vivo and in vitro. RESULTS The results showed significantly increased CDC42 expression in pancreatic tumor tissues compared with adjacent normal tissues, as revealed by qRT-PCR, Western blotting and immunostaining. Compared to PanC-1 cells, CDC42 expression was downregulated in HPDE6-C7 cells as shown by qRT-PCR and Western blotting. High CDC42 expression was observed in 69.2% (83/120) of pancreatic adenocarcinoma patients and was significantly associated with tumor differentiation (p = 0.013), median tumor size (p = 0.005), tumor infiltration (pT stage, p = 0.04), lymph nodal status (pN stage, p = 0.044) and TNM staging (p = 0.003). Multivariate Cox regression analysis revealed CDC42 expression to be an independent predictor of survival of PC patients (HR 3.0, 95% CI 1.60-5.61, p = 0.001). Finally, we found that CDC42 promoted the proliferation of PanC-1 cells both in vivo and in vitro. CONCLUSIONS Our findings reveal that CDC42 might play an important role in promoting PC development, and the findings suggest that CDC42 might serve as a potential prognostic indicator of PC.
Collapse
Affiliation(s)
- Dejun Yang
- Department of Gastrointestinal Surgery, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, People's Republic of China
| | - Yu Zhang
- Department of Gastrointestinal Surgery, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, People's Republic of China
| | - Yajun Cheng
- Department of Gastrointestinal Surgery, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, People's Republic of China
| | - Liang Hong
- Outpatient Department, Yichuan Community Health Service Center, 43 Lishan Road, Shanghai, 200065, People's Republic of China
| | - Changming Wang
- Department of Gastrointestinal Surgery, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, People's Republic of China
| | - Ziran Wei
- Department of Gastrointestinal Surgery, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, People's Republic of China
| | - Qingping Cai
- Department of Gastrointestinal Surgery, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, People's Republic of China.
| | - Ronglin Yan
- Department of Gastrointestinal Surgery, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, People's Republic of China.
| |
Collapse
|
14
|
Lu L, Chen J, Tang H, Bai L, Lu C, Wang K, Li M, Yan Y, Tang L, Wu R, Ye Y, Jin L, Liang Z. EGCG Suppresses ERK5 Activation to Reverse Tobacco Smoke-Triggered Gastric Epithelial-Mesenchymal Transition in BALB/c Mice. Nutrients 2016; 8:380. [PMID: 27447666 PMCID: PMC4963860 DOI: 10.3390/nu8070380] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/28/2016] [Accepted: 06/14/2016] [Indexed: 12/14/2022] Open
Abstract
Tobacco smoke is an important risk factor of gastric cancer. Epithelial-mesenchymal transition is a crucial pathophysiological process in cancer development. ERK5 regulation of epithelial-mesenchymal transition may be sensitive to cell types and/or the cellular microenvironment and its role in the epithelial-mesenchymal transition process remain elusive. Epigallocatechin-3-gallate (EGCG) is a promising chemopreventive agent for several types of cancers. In the present study we investigated the regulatory role of ERK5 in tobacco smoke-induced epithelial-mesenchymal transition in the stomach of mice and the preventive effect of EGCG. Exposure of mice to tobacco smoke for 12 weeks reduced expression of epithelial markers E-cadherin, ZO-1, and CK5, while the expression of mesenchymal markers Snail-1, Vimentin, and N-cadherin were increased. Importantly, we demonstrated that ERK5 modulated tobacco smoke-mediated epithelial-mesenchymal transition in mice stomach, as evidenced by the findings that tobacco smoke elevated ERK5 activation, and that tobacco smoke-triggered epithelial-mesenchymal transition was reversed by ERK5 inhibition. Treatment of EGCG (100 mg/kg BW) effectively attenuated tobacco smoke-triggered activation of ERK5 and epithelial-mesenchymal transition alterations in mice stomach. Collectively, these data suggested that ERK5 was required for tobacco smoke-triggered gastric epithelial-mesenchymal transition and that EGCG suppressed ERK5 activation to reverse tobacco smoke-triggered gastric epithelial-mesenchymal transition in BALB/c mice. These findings provide new insights into the mechanism of tobacco smoke-associated gastric tumorigenesis and the chemoprevention of tobacco smoke-associated gastric cancer.
Collapse
Affiliation(s)
- Ling Lu
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Jia Chen
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Hua Tang
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Ling Bai
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Chun Lu
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Kehuan Wang
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Manli Li
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Yinmei Yan
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Ling Tang
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Rui Wu
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang 212013, China.
| | - Longtao Jin
- Zhenjiang Matemity and Child Health Care Hospital, Zhenjiang 212001, China.
| | - Zhaofeng Liang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang 212013, China.
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
15
|
Simões AES, Rodrigues CMP, Borralho PM. The MEK5/ERK5 signalling pathway in cancer: a promising novel therapeutic target. Drug Discov Today 2016; 21:1654-1663. [PMID: 27320690 DOI: 10.1016/j.drudis.2016.06.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/18/2016] [Accepted: 06/08/2016] [Indexed: 12/18/2022]
Abstract
Conventional mitogen-activated protein kinase (MAPK) family members are among the most sought-after oncogenic effectors for the development of novel human cancer treatment strategies. MEK5/ERK5 has been the less-studied MAPK subfamily, despite its increasingly demonstrated relevance in the growth, survival, and differentiation of normal cells. MEK5/ERK5 signalling has already been proposed to have pivotal roles in several cancer hallmarks, and to mediate the effects of a range of oncogenes. Accumulating evidence indicates the contribution of MEK5/ERK5 signalling to therapy resistance and the benefits of using MEK5/ERK5 inhibitory strategies in the treatment of human cancer. Here, we explore the major known contributions of MEK5/ERK5 signalling to the onset and progression of several types of cancer, and highlight the potential clinical relevance of targeting MEK5/ERK5 pathways.
Collapse
Affiliation(s)
- André E S Simões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal.
| | - Pedro M Borralho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal.
| |
Collapse
|
16
|
Vu T, Jin L, Datta PK. Effect of Cigarette Smoking on Epithelial to Mesenchymal Transition (EMT) in Lung Cancer. J Clin Med 2016; 5:jcm5040044. [PMID: 27077888 PMCID: PMC4850467 DOI: 10.3390/jcm5040044] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/04/2016] [Accepted: 04/06/2016] [Indexed: 12/15/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a process that allows an epithelial cell to acquire a mesenchymal phenotype through multiple biochemical changes resulting in an increased migratory capacity. During cancer progression, EMT is found to be associated with an invasive or metastatic phenotype. In this review, we focus on the discussion of recent studies about the regulation of EMT by cigarette smoking. Various groups of active compounds found in cigarette smoke such as polycyclic aromatic hydrocarbons (PAH), nicotine-derived nitrosamine ketone (NNK), and reactive oxygen specicies (ROS) can induce EMT through different signaling pathways. The links between EMT and biological responses to cigarette smoke, such as hypoxia, inflammation, and oxidative damages, are also discussed. The effect of cigarette smoke on EMT is not only limited to cancer types directly related to smoking, such as lung cancer, but has also been found in other types of cancer. Altogether, this review emphasizes the importance of understanding molecular mechanisms of the induction of EMT by cigarette smoking and will help in identifying novel small molecules for targeting EMT induced by smoking.
Collapse
Affiliation(s)
- Trung Vu
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Lin Jin
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Pran K Datta
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
17
|
Liang Z, Xie W, Wu R, Geng H, Zhao L, Xie C, Li X, Huang C, Zhu J, Zhu M, Zhu W, Wu J, Geng S, Zhong C. ERK5 negatively regulates tobacco smoke-induced pulmonary epithelial-mesenchymal transition. Oncotarget 2015; 6:19605-19618. [PMID: 25965818 PMCID: PMC4637308 DOI: 10.18632/oncotarget.3747] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/06/2015] [Indexed: 12/21/2022] Open
Abstract
As the primary cause of lung cancer, tobacco smoke (TS) promotes the initiation and progression of lung tumorigenesis. Epithelial-mesenchymal transition (EMT) is a crucial process involved in cell malignant transformation. The role of ERK5, the lesser studied member of MAPKs family, in regulating TS-triggered pulmonary EMT has not been investigated. Normal human bronchial epithelial cells and BALB/c mice were used as in vitro and in vivo TS exposure models. Exposure of normal human bronchial epithelial cells to TS for 7 days induced morphological change, enhanced migratory and invasive capacities, reduced epithelial marker expression and increased mesenchymal marker expression. Importantly, we demonstrated for the first time that ERK5 negatively regulated TS-mediated lung epithelial EMT, as evidenced by the findings that TS suppressed ERK5 activation, and that TS-triggered EMT was mimicked with ERK5 inhibition and reversed by ERK5 overexpression. The negative regulation of ERK5 on pulmonary EMT was further confirmed in mice exposed to TS for 12 weeks. Taken together, our data suggest that ERK5 negatively regulates TS-mediated pulmonary EMT. These findings provide new insight into the molecular mechanisms of TS-associated lung tumorigenesis and may open up new avenues in the search for potential target of lung cancer intervention.
Collapse
Affiliation(s)
- Zhaofeng Liang
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wei Xie
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rui Wu
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hao Geng
- Department of Surgery, The Second Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Li Zhao
- Department of Surgery, The Second Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Chunfeng Xie
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoting Li
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Cong Huang
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jianyun Zhu
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Mingming Zhu
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Weiwei Zhu
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jieshu Wu
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shanshan Geng
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Caiyun Zhong
- Department of Toxicology and Nutritional Science, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|